26.12.2012 Views

West Coast Worm Meeting Abstracts - Caenorhabditis elegans ...

West Coast Worm Meeting Abstracts - Caenorhabditis elegans ...

West Coast Worm Meeting Abstracts - Caenorhabditis elegans ...

SHOW MORE
SHOW LESS

Create successful ePaper yourself

Turn your PDF publications into a flip-book with our unique Google optimized e-Paper software.

<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> <strong>Abstracts</strong><br />

1. Meiotic Pairing and Synapsis [p 11]<br />

Amy MacQueen, Anne M. Villeneuve<br />

2. Conditional mitotic spindle mutants in C. <strong>elegans</strong> [p 12]<br />

Danielle R. Hamill, Bruce Bowerman<br />

3. Mitotic Chromosome Segregation by a Conserved Protein Complex [p 13]<br />

K. Hagstrom, R. Chan, D. Pasqualone, B.J. Meyer<br />

4. HIM-10 a Probable Kinetochore Protein Involved in Mitotic and Meiotic Chromosome Segregation [p<br />

14] M. Howe, D G. Albertson, B. J. Meyer<br />

5. A C. <strong>elegans</strong> chromokinesin required for chromosome segregation [p 15]<br />

Jim Powers, Bill Saxton, Susan Strome<br />

6. Nuclear Envelope Dynamics in <strong>Caenorhabditis</strong> <strong>elegans</strong> [p 16]<br />

Kenneth Lee, Yosef Gruenbaum, Katherine L. Wilson<br />

7. The formin protein CYK-1 acts in parallel to an aurora-like kinase/MKLP-1 pathway to execute<br />

cytokinesis in early <strong>Caenorhabditis</strong> <strong>elegans</strong> embryos [p 17]<br />

Aaron F. Severson, Danielle R. Hamill, Bruce Bowerman<br />

8. The L type Cyclin SAG-4 is required for heat-shock induced protein expression [p 18]<br />

Wen J. Chen, Yvonne M. Hajdu-Cronin, Paul W. Sternberg<br />

9. cdl-1 encodes a stem- loop binding protein (SLBP) homolog and may be essential for core histone<br />

expression. [p 19]<br />

Yuki Kodama, Asako Sugimoto, Joel Rothman, Masayuki Yamamoto<br />

10. UNC-23 is a member of the BAG family of chaperone regulators [p 20]<br />

Poupak Rahmani, Donald Moerman<br />

11. Maternal UNC-45 protein co-localizes with NMY-2, a non-muscle myosin at the cleavage furrow of<br />

early embryos [p 21]<br />

Wanyuan Ao, Dave Pilgrim<br />

12. Polyunsaturated fatty acids requirements for proper functioning of the nervous system [p 22]<br />

Jenny Watts, John Browse<br />

13. Testing functions of phagocytosis receptor homologs in cell corpse elimination and gonadal outgrowth<br />

[p 23]<br />

Sambath Chung, Monica Driscoll<br />

14. Regulation of Cell Fusion in C. <strong>elegans</strong> [p 24]<br />

Scott Alper, Cynthia Kenyon<br />

15. Ethanol sensitivity genes in <strong>Caenorhabditis</strong> <strong>elegans</strong> [p 25]<br />

MinGi Hong, JaeYoung Kwon, InYoung Lee, MinSung Choi, Junho Lee<br />

16. State-dependent learning in C. <strong>elegans</strong>. [p 26]<br />

Jill C. Bettinger, Steven L. McIntire<br />

17. The ut236 mutant in C. <strong>elegans</strong> has defects in the interaction of two sensory signals and an<br />

associative learning. [p 27]<br />

Takeshi Ishihara, Yuichi Iino, Isao Katsura<br />

18. Mutation in the LIM homeobox gene lim-6 disrupts asymmetric function of the ASE chemosensory<br />

neurons [p 28]<br />

J.T. Pierce-Shimomura, M.R. Gaston, B.J. Pearson, S.R. Lockery<br />

19. Information Coding in the C. <strong>elegans</strong> Olfactory System [p 29]<br />

PD Wes, A Sagasti, G Jansen, RHA Plasterk, CI Bargmann<br />

20. Roles of osm-9/capsaicin receptor family members in sensory behaviors [p 30]<br />

D. Tobin, D. Madsen, G. Moulder, R. Barstead, A.V. Maricq, M. deBono, C. Bargmann<br />

21. Execution and regulation of male C. <strong>elegans</strong> spicule muscle contractions during mating [p 31]<br />

L. René García, Paul W. Sternberg<br />

22. Genetic Analysis of Nicotine Adaptation in C. <strong>elegans</strong>. [p 32]<br />

Jinah Kim, Laura E. Waggoner, Kari A. Dickinson, Daniel S. Poole, William R. Schafer<br />

23. Neural control of locomotion in C. <strong>elegans</strong> [p 33]<br />

Saleem Mukhtar, Jane Mendel, Jehoshua (Shuki) Bruck, Paul W. Sternberg<br />

24. Analysis of glutamatergic neurotransmission by knockout of glutamate transporter genes. [p 34]<br />

Itzhak Mano, Monica Driscoll<br />

25. Electrophysiological analysis of C. <strong>elegans</strong> ionotropic glutamate receptors [p 35]<br />

Jerry E. Mellem, Penelope J. Brockie, David M. Madsen, Andres V. Maricq<br />

26. Electrophysiological analysis of unc-18 mutants. [p 36]<br />

J. E. Richmond, R. Weimer, W. S. Davis, E. M. Jorgensen<br />

1


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

27. Electrophysiological analysis of serotonin modulation of body wall neuromuscular physiology. [p 37]<br />

Jon Madison, Joshua Kaplan<br />

28. Serotonin signaling in the pharynx [p 38]<br />

Timothy Niacaris, Leon Avery<br />

29. A Muscarinic Contribution to the Regulation of Feeding [p 39]<br />

Kate Steger, Leon Avery<br />

30. <strong>Worm</strong>Base: From ACeDB to a more complete and usable database [p 40]<br />

Paul W. Sternberg, Erich Schwarz, Norma Foltz, <strong>Worm</strong>Base Consortium<br />

31. The C. <strong>elegans</strong> ORFeome project [p 41]<br />

Jerome Reboul, Philippe Vaglio, Cindy Jackson, Troy Moore, Jean Thierry-Mieg, Danielle<br />

Thierry-Mieg, Jim Hartley, Gary Temple, Mike Brasch, Nia Tzellas, Marc Vidal<br />

32. Analysis of splicing and regulatory elements using the Intronerator [p 42]<br />

W. James Kent, Alan M. Zahler<br />

33. A global profile of germ line gene expression using microarrays reveals germ line-specific regulation<br />

of the X chromosome in males and hermaphrodites [p 43]<br />

Valerie Reinke, Harold E. Smith, Jeremy Nance, Abby F. Dernburg, Anne M. Villeneuve, Samuel<br />

Ward, Stuart K. Kim<br />

34. The promise and peril of genomics: sperm development as model system [p 44]<br />

Harold Smith, Marci Millhouse, Sam Ward<br />

35. Functional Analysis of Chromosome I [p 45]<br />

Andrew Fraser, Ravi Kamath, Peder Zipperlen, Maruxa Martinez-Campos, Julie Ahringer<br />

36. Optimizing the mutagenic properties of the mos1 transposon in C. <strong>elegans</strong> [p 46]<br />

Daniel C. Williams, Jean-Louis Bessereau, Erik M. Jorgensen<br />

37. SNAP-25, a protein implicated genetically in C. <strong>elegans</strong> anesthetic mechanisms, binds the general<br />

anesthetic isoflurane [p 47]<br />

Jason Berilgen, Mike Crowder<br />

38. Happy worms: further characterization of fluoxetine (prozac) resistant mutants [p 48]<br />

Robert K.M. Choy, James H. Thomas<br />

39. unc-43 Ca2+/Calmodulin-dependent kinase II (CaMKII) mutant worms have convulsions in response<br />

to the seizure-inducing drug PTZ [p 49]<br />

Elizabeth M. Newton, James H. Thomas<br />

40. Neurotoxin sensitivity of dopaminergic neurons in C. <strong>elegans</strong>: role of the dopamine transporter and<br />

cell death pathways [p 50]<br />

R. Nass, J. Duerr, , J. Rand, D. M. Miller, R. D. Blakely<br />

41. Gene expression in transgenic C. <strong>elegans</strong> animals expressing the human beta amyloid peptide. [p 51]<br />

Chris Link, Carolyn Johnson, Amy Fluet, Kyle K. Duke, Stuart K. Kim<br />

42. A nematode model for mitochondrial diseases [p 52]<br />

William Y. Tsang, Bernard D. Lemire<br />

43. Bacillus toxin (Bt) susceptibility and resistance in C. <strong>elegans</strong> [p 53]<br />

Lisa Marroquin, Dino Elyassnia, Joel Griffitts, Johanna O’Dell, Jerald Feitelson, Raffi Aroian<br />

44. New dauer genes and pathways [p 54]<br />

Michael Ailion, James H. Thomas<br />

45. Temporal regulation of aging in the nematode C. <strong>elegans</strong> [p 55]<br />

Andrew Dillin, Cynthia Kenyon<br />

46. A longitudinal analysis of adult neurons in C. <strong>elegans</strong> [p 56]<br />

Mark I. Snow, Pamela L. Larsen<br />

47. Germ-line cells that regulate aging in C. <strong>elegans</strong> [p 57]<br />

Nuno Arantes-Oliveira, Javier Apfeld, Cynthia Kenyon<br />

48. A screen for genes that control programmed cell death in the germ line [p 58]<br />

S Milstein, A Gartner, M Hengartner<br />

49. C. <strong>elegans</strong> p53: requirement for radiation-induced programmed cell death, stress resistance, and<br />

normal adult lifespan following diapause. [p 59]<br />

W. Brent Derry, Aaron Putzke, Joel H. Rothman<br />

50. Identification of cell-specific regulators of programmed cell death in C. <strong>elegans</strong>. [p 60]<br />

Shai Shaham, Cori Bargmann<br />

51. Biochemical, structural, and genetic analyses of the activation of programmed cell death [p 61]<br />

Jay Parrish, Betsy Metters, Lin Chen, Ding Xue<br />

52. Analysis of RNA associated with P granules in germ cells of C. <strong>elegans</strong> adults [p 62]<br />

Jennifer A. Schisa, Jason N. Pitt, James R. Priess<br />

53. The splicing Sm proteins colocalize with P granules in germ cells and participate in P granule<br />

localization in the early embryo [p 63]<br />

Scott A. Barbee, Alex L. Lublin, Thomas C. Evans<br />

2


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

54. pod-2 defines a new class of mutants required for antero-posterior asymmetry in the early<br />

<strong>Caenorhabditis</strong> <strong>elegans</strong> embryo [p 64]<br />

Akiko Tagawa, Raffi V. Aroian<br />

55. ooc-5 encodes a putative ATPase required for the reestablishment of asymmetric PAR protein<br />

localization in two-cell embryos [p 65]<br />

Stephen E. Basham, Lesilee S. Rose<br />

56. RIC-8 (Synembryn): A novel regulator of G Protein signaling [p 66]<br />

Kenneth G. Miller, Melanie D. Emerson, John R. McManus, James B. Rand<br />

57. MED-1 AND -2 act at the convergence point of SKN-1 and POS-1 to specify MS and E identity [p 67]<br />

Morris F. Maduro, Regina Broitman-Maduro, Joel H. Rothman<br />

58. Mass spectrometric identification of PLP-1 and its role in mesendoderm specification [p 68]<br />

E. Witze, E. Field, D. Hunt, J.H. Rothman<br />

59. The C. <strong>elegans</strong> NeuroD homolog cnd-1 functions in multiple aspects of motor neuron fate<br />

specification [p 69]<br />

Steven Hallam, Emily Singer, David Waring, Yishi Jin<br />

60. Left-right asymmetry in C. <strong>elegans</strong> intestinal organogenesis involves a LIN-12/Notch signaling<br />

pathway [p 70]<br />

Greg J. Hermann, Ben Leung, James R. Priess<br />

61. The pho-1 Gene and Three Kinds of Gut Polarity [p 71]<br />

Tetsunari Fukushige, James D. McGhee<br />

62. A role for dishevelled in asymmetric cell division. [p 72]<br />

Nancy Hawkins, Gregory Ellis, Bruce Bowerman, Gian Garriga<br />

63. rho-1, a target of the exchange factor unc-73, is required for cell migrations during C. <strong>elegans</strong><br />

development [p 73]<br />

Andrew G. Spencer, Christian J. Malone, Satoshi Orita, Min Han<br />

64. PTP-1, a LAR-like receptor protein tyrosine phosphatase, may act in parallel with C. <strong>elegans</strong> Eph<br />

signaling to direct morphogenesis [p 74]<br />

Robert J. Harrington, Michael Gutch, Michael Hengartner, Nicholas Tonks, Andrew Chisholm<br />

65. GEX-2 and GEX-3 define a conserved protein complex required for tissue morphogenesis and cell<br />

migrations in C. <strong>elegans</strong> [p 75]<br />

Martha Soto, Katsuhisa Kasuya, Hiroshi Qadota, Kozo Kaibuchi, Craig C. Mello<br />

66. Pharyngeal extension: the short and the long of it [p 76]<br />

MF Portereiko, SE Mango<br />

67. A VAB-8/UNC-51/UNC-14 complex mediates axon outgrowth [p 77]<br />

Tina Lai, Gian Garriga<br />

68. Cytoskeletal Signalling in Response to the UNC-6 Axonal Attractant [p 78]<br />

Zemer Gitai, Erik Lundquist, Marc Tessier-Lavigne, Cori Bargmann<br />

69. Identifying genes involved in axonal branching in C. <strong>elegans</strong> [p 79]<br />

Joe C. Hao, Marc Tessier-Lavigne, Cornelia I. Bargmann<br />

70. UNC-119 suppresses supernumerary branching in C. <strong>elegans</strong> [p 80]<br />

Karla Knobel, Warren Davis, Michael Bastiani, Erik Jorgensen<br />

71. UNC-119 and axon outgrowth: Toward a mechanism [p 81]<br />

Wayne Materi, Dave Pilgrim<br />

72. Three distinct functions of beta-spectrin (UNC-70) [p 82]<br />

Marc Hammarlund, Warren S. Davis, Erik M. Jorgensen<br />

73. RPM-1, a conserved novel protein, regulates presynaptic terminal formation [p 83]<br />

Xun Huang, Mei Zhen, Bruce Bamber, Yishi Jin<br />

74. A C. <strong>elegans</strong> Inositol 5- Phosphatase Homologue Involved In Inositol 1,4,5-triphosphate Signaling and<br />

Ovulation. [p 84]<br />

Yen Kim Bui, Paul W. Sternberg<br />

75. Mechanisms regulating the timing and specificity of anchor cell attachment to the vulval epithelium [p<br />

85] David R. Sherwood, Paul W. Sternberg<br />

76. Mutations in cyclin E reveal coordination between cell-cycle control and vulval development. [p 86]<br />

David S. Fay, Min Han<br />

77. Novel cell-cell interactions during vulva development in Pristionchus pacificus [p 87]<br />

Benno Jungblut, Ralf J Sommer<br />

78. Cellular and genetic analysis of Gq mediated signaling pathways in C. <strong>elegans</strong> [p 88]<br />

C. A. Bastiani, S. Gharib, P.W. Sternberg, M.I. Simon<br />

79. Calcium/calmodulin-dependent protein Kinase II regulates C. <strong>elegans</strong> locomotion in concert with a<br />

G-protein signaling network [p 89]<br />

Merrilee Robatzek, James H. Thomas<br />

3


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

80. A novel lateral signaling pathway determines asymmetric olfactory neuron fates [p 90]<br />

Alvaro Sagasti, Cori Bargmann<br />

81. The search for dosage compensation complex binding sites on X chromosomes [p 91]<br />

Raymond C. Chan, Tammy F. Wu, Barbara J. Meyer<br />

82. Recognition and Assembly of SDC Protein Complexes onto Specific DNA Target Sites [p 92]<br />

Diana Chu, Heather Dawes, Jason Lieb, Annie Kuo, Barbara J. Meyer<br />

83. The TBP-like Factor CeTLF is Required to Activate RNA Polymerase II Transcription in C. <strong>elegans</strong><br />

Embryos [p 93]<br />

Linda S. Kaltenbach, Susan E. Mango<br />

84. The intracellular domain of the feminising receptor TRA-2A interacts directly with the transcription<br />

factor TRA-1A [p 94]<br />

David H. Lum, P. Kuwabara, D. Zarkower, A.M. Spence<br />

85. chw-1 encodes a novel protein that interacts with pha-4 [p 95]<br />

Michael Horner, Linda Kaltenbach, Susan Mango<br />

86. The UNC-4 homeoprotein and its transcriptional co-repressor UNC-37/Groucho regulate<br />

neurotransmitter vesicles in cholinergic motor neurons [p 96]<br />

Kim Lickteig, Janet Duerr, Dennis Frisby, David Hall, Jim Rand, David Miller<br />

87. The components of sensory cilia in C. <strong>elegans</strong> [p 97]<br />

Peter Swoboda, Kerry Bubb, James H. Thomas<br />

88. In vivo imaging of HSN outgrowth [p 98]<br />

Carolyn E. Adler, Cornelia I. Bargmann<br />

89. Temporal and spatial requirement of sensory cilia in the regulation of worm lifespan [p 99]<br />

Joy Alcedo, Javier Apfeld, Bella Albinder, Jennifer Dorman, Honor Hsin, Bernadine Tsung, Cynthia<br />

Kenyon<br />

90. THE TTX-3 LIM HOMEOBOX GENE IS A CENTRAL REGULATOR OF INTERNEURON CELL FATE<br />

[p 100]<br />

Z. Altun-Gultekin, O. Hobert<br />

91. The heterochronic gene pathway: Regulatory interactions and regulatory outputs. [p 101]<br />

Victor Ambros, Marta Hristova, Rosalind Lee, Eric Moss<br />

92. Genetic and phenotypic characterization of evl-14 and evl-20, genes involved in C. <strong>elegans</strong> vulva<br />

development [p 102]<br />

Igor Antoshechkin, Min Han<br />

93. <strong>Caenorhabditis</strong> <strong>elegans</strong> T05H10.5, a homologue of yeast ubiquitin fusion degradation protein<br />

(UDF-2), is expressed throughout the nervous system and in the gut [p 103]<br />

Wanyuan Ao, Dave Pilgrim<br />

94. Genetic analysis of neuroendocrine controls of fat metabolism in C. <strong>elegans</strong> [p 104]<br />

Kaveh Ashrafi, Gary Ruvkun<br />

95. zig genes and the PVT guidepost neuron [p 105]<br />

Oscar Aurelio, Oliver Hobert<br />

96. Analysis of GABA receptor plasticity in C. <strong>elegans</strong> [p 106]<br />

Bruce A. Bamber, Janet E. Richmond, Pierrette K. Danieu<br />

97. Isolation of suppressors of a dominant synapse defective mutant, syd-5(ju89) [p 107]<br />

Renee Baran, Yishi Jin<br />

98. Cargo recognition by synaptic vesicle kinesin [p 108]<br />

Ewa Bednarek, Erik M. Jorgensen<br />

99. The exp-1 locus may encode a subunit of an excitatory GABA receptor [p 109]<br />

Asim A. Beg, Erik M. Jorgensen<br />

100. The life span gene clk-2 is essential for embryonic development [p 110]<br />

Claire Bènard, Brent McCright, Yue Zhang, Stephanie Felkai, Siegfried Hekimi<br />

101. Characterization of <strong>Caenorhabditis</strong> <strong>elegans</strong> gamma-tubulin in dividing cells and differentiated tissues<br />

[p 111]<br />

Yves Bobinnec, Makoto Fukuda, Eisuke Nishida<br />

102. Does CEH-20, an Exd/Pbx homolog in C. <strong>elegans</strong>, play a role in worm embryogenesis? [p 111]<br />

Q.F. Boese, W.B. Wood<br />

103. A-domain-containing protein family in C. <strong>elegans</strong>. [p 112]<br />

Michael Brannan, Joaquin Muriel, Kathryn Taylor, Gordon Lithgow, Danny Tuckwell<br />

104. Distribution and Regulation of Glutamate Receptors in the Locomotory Control Circuit of C. <strong>elegans</strong>.<br />

[p 113]<br />

Penelope J. Brockie, David M. Madsen, Yi Zheng, Jerry E. Mellem, Andres V. Maricq<br />

105. Mutations That Affect Synaptic Localization Of Glr-1 [p 114]<br />

Michelle Burbea, Joshua M. Kaplan<br />

106. Regulation of C. <strong>elegans</strong> dauer formation by an RNA quality control pathway component [p 115]<br />

J Burgess, JC Labbe, S Hekimi<br />

4


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

107. Synaptic vesicle localization is misregulated in unc-16 mutants [p 116]<br />

DT Byrd, Y Jin<br />

108. The egl-21 gene encodes a carboxypeptidase E, which is required for pro-neuropeptide processing<br />

[p 117]<br />

Tija Carey, Joshua M. Kaplan<br />

109. How are anterior cell migrations guided by mig-13? [p 118]<br />

QueeLim Ch’ng, Cynthia Kenyon<br />

110. New Screens for Negative Regulators of let-23 [p 119]<br />

Monica Chan, Marie Tiongsen, Romel C. Castro, Vanessa Lee, Gregg Jongeward<br />

111. C. <strong>elegans</strong> MRE-11 is required for meiotic recombination and DNA repair but not for the meiotic G2<br />

DNA damage checkpoint [p 120]<br />

Gregory Chin, Anne Villeneuve<br />

112. Suppressor Analysis of Eph/Ephrin Defective Signaling in C. <strong>elegans</strong> [p 121]<br />

Ian Chin-Sang, Julie McCleery, Andrew Chisholm<br />

113. RNAi Screen for Components of the C. <strong>elegans</strong> Meiotic Machinery [p 122]<br />

Mónica Colaiácovo, Gillian Stanfield, Kirthi Reddy, Anne Villeneuve<br />

114. Exploring the role of PINCH/UNC-97 in muscle development and focal adhesion assembly in<br />

<strong>Caenorhabditis</strong> <strong>elegans</strong> and mammalian tissue culture cell lines [p 123]<br />

Shaun Cordes, May Dang-Lawson, Poupak Rahmani, Linda Matsuuchi, Donald G. Moerman<br />

115. The SAD-1 kinase regulates presynaptic vesicle clustering in C. <strong>elegans</strong> [p 124]<br />

Justin Gage Crump, Mei Zhen, Kang Shen, Yishi Jin, Cornelia I. Bargmann<br />

116. Mutants with altered sensitivity to the effects of ethanol on locomotion [p 125]<br />

Andrew G. Davies, Tod R. Thiele, Catharine Eastman, Steven L. McIntire<br />

117. A screen for DD/DV axonal morphology defects [p 126]<br />

M. Wayne Davis, Erik M. Jorgensen<br />

118. spn-2 AND spn-3 FUNCTION TO ORIENT THE SPINDLE DURING EARLY CLEAVAGES [p 127]<br />

Leah R. DeBella, Lesilee S. Rose<br />

119. Molecules acting in parallel with UNC-34 to control cell migration [p 128]<br />

Megan Dell, N Chugh, N Hawkins, E Kong, J Hardin, G Garriga<br />

120. Insights into the role of C. <strong>elegans</strong> protein UNC-119 in axonogenesis [p 129]<br />

Chantal Denholm, Wayne Materi, Daniel Gietz, David Pilgrim<br />

121. The defecation gene aex-1 may regulate a retrograde signaling pathway at neuromusclular<br />

junctions. [p 130]<br />

Motomichi Doi, Kouichi Iwasaki<br />

122. Cosuppression in the Germline: Silencing is Golden [p 131]<br />

Abby F. Dernburg, Mónica P. Colaiácovo, Jonathan Zalevsky, Anne M. Villeneuve<br />

123. sur-9 a Suppressor of Activated let-60(n1046) in the C.<strong>elegans</strong> Vulva. [p 132]<br />

Dennis Eastburn, Min Han<br />

124. Knockouts In C. <strong>elegans</strong>: Madness and Methodology [p 133]<br />

Mark Edgley, Erin Gilchrist, Greg Mullen, Bin Shen, Margaret Kotarska, Don Moerman, Steven<br />

Jones, Anil Dsouza, Gary Moulder, Malini Viswanathan, Martin Lansdale, Robert Barstead<br />

125. Using DNA microarrays to identify targets of homeobox genes in C. <strong>elegans</strong> [p 134]<br />

Andreas Eizinger, Tibor Vellai, Fritz Müller, Stuart K. Kim<br />

126. ded Genes Disrupt Cell Division Timing and Patterning in C. <strong>elegans</strong> Embryos [p 135]<br />

Sandra Encalada, Paula Martin, Jennifer Phillips, Rebecca Lyzcak, Danielle Hamill, Kathryn Swan,<br />

Bruce Bowerman<br />

127. Voltage-dependent currents in homologous chemosensory neurons with different functions in C.<br />

<strong>elegans</strong> [p 136]<br />

S Faumont, S.R. Lockery<br />

128. VAV is required for pharyngeal muscle contraction in C. <strong>elegans</strong> [p 137]<br />

R.T. Fazzio, J.E. Mellem, M.C. Beckerle, A.V. Maricq<br />

129. Regulation of C. <strong>elegans</strong> Body Size by Sensory Cues [p 138]<br />

Manabi Fujiwara, Hoan Phan, Steven L. McIntire<br />

130. Regulation of intracellular dynamics of MAPKAPK2 in living C.<strong>elegans</strong> [p 139]<br />

Makoto Fukuda, Yves Bobinnec, Eisuke Nishida<br />

131. Role of cki-1 in terminal embryonic differentiation and cell-cycle arrest [p 140]<br />

Masamitsu Fukuyama, W. Brent Derry, Joel H. Rothman<br />

132. sax-1 and sax-2 act in parallel with unc-34 to Maintain Neuron Polarity. [p 141]<br />

Maria E. Gallegos, Jennifer A. Zallen, Cori Bargmann<br />

133. Identifying pharyngeal targets of PHA-4 using DNA microarrays [p 142]<br />

Jeb Gaudet, Michael Horner, Stuart Kim, Susan E. Mango<br />

134. An overview of predicted cytochrome P450 genes in C. <strong>elegans</strong> [p 143]<br />

Erin Gilchrist<br />

5


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

135. Clues toward understanding EGF/ Wnt signal integration in the specification of P12 fate: analysis of<br />

the egl-5 promoter [p 144]<br />

Lisa Girard, Henrique B. Ferreira, Scott Emmons, Paul Sternberg<br />

136. spn-4: a gene required for mitotic spindle orientation in the 2-cell stage C. <strong>elegans</strong> embryo [p 145]<br />

José E. Gomes, Kathryn A. Swan, Christopher A. Shelton, Bruce Bowerman<br />

137. Ca2+-signalling via the neuron-specific Ca2+ sensor NCS-1 is essential for thermotaxis, a form of<br />

associative learning and memory in C. <strong>elegans</strong> [p 146]<br />

Marie Gomez, Edouard De Castro, Ernesto Guarin, Patrick Nef<br />

138. Characterizing the Neural Circuitry of Chemotaxis to Volatile Odorants [p 147]<br />

Jesse Gray, Maria Gallegos, Tim Yu, Cori Bargmann<br />

139. Studies on the Nematicidal Bacillus thuringiensis Toxins [p 148]<br />

Joel S. Griffitts, Raffi V. Aroian<br />

140. Synaptic localization of the glutamate-gated chloride channel GBR-2 [p 149]<br />

Maria E. Grunwald, Joshua M. Kaplan<br />

141. Regulation and function of lin-11 in C. <strong>elegans</strong> vulval development [p 150]<br />

Bhagwati P Gupta, Paul W. Sternberg<br />

142. sur-7, a gene that suppresses activated ras [p 151]<br />

Eric Hague, Min Han<br />

143. Characterization and Suppression of eat-16; sag-1/dgk-1 lethality [p 152]<br />

Yvonne M. Hajdu-Cronin, Wen J. Chen, Paul W. Sternberg<br />

144. Improved Tissue Preservation Using Metal Mirror Freezing or High Pressure Freezing for TEM [p<br />

153]<br />

David H. Hall, Frank Macaluso, Gloria Stepheney, Marie-Christine Paupard<br />

145. Role of the <strong>Caenorhabditis</strong> <strong>elegans</strong> homologs of cdk5 and p35 in migration and axon outgrowth [p<br />

154]<br />

Thomas Harbaugh, Gian Garriga<br />

146. Regulation of egg-laying by sensory cues [p 155]<br />

Laura Anne Hardaker, William R. Schafer<br />

147. Characterization of the C. <strong>elegans</strong> Serotonin-Synthetic Aromatic Amino Acid Decarboxylase Gene<br />

bas-1 [p 156]<br />

Emily Hare, Curtis M. Loer<br />

148. XOL-1 Files [p 157]<br />

Christian A. Hassig, Barbara J. Meyer<br />

149. Y41G9a.1, the C. <strong>elegans</strong> Homologue of Tg737, is Expressed in Ciliated Neurons [p 158]<br />

Courtney J. Haycraft, Patrick D. Taulman, Stephen M. Krum, Bradley K. Yoder<br />

150. let-381 is a forkhead gene [p 159]<br />

Marika Hellqvist-Greberg, Ann M Rose, David L Baillie<br />

151. Genetic analysis of dynamic search behavior in C. <strong>elegans</strong> [p 160]<br />

T.T. Hills, F. Adler, A. V. Maricq<br />

152. Multiple roles for the Ras-MAPK signal transduction pathway in chemotaxis to odorants? [p 161]<br />

Takaaki Hirotsu, Satoshi Saeki, Yuichi Iino<br />

153. mab-26 encodes the C. <strong>elegans</strong> ephrin EFN-4 [p 162]<br />

Thomas Holcomb, Sean E. George, Ian Chin-Sang, Mei Ding, Andrew Chisholm<br />

154. syd-8, a new player in axon guidance. [p 163]<br />

Xun Huang, Mei Zhen, Yishi Jin<br />

155. Analysis of gcy-31, a putative soluble guanylyl cyclase gene in <strong>Caenorhabditis</strong> <strong>elegans</strong> [p 164]<br />

Martin L Hudson, David S. Karow, Michael A. Marletta, David B. Morton<br />

156. Using C. <strong>elegans</strong> to Determine the Mechanism of Action of Pharmaceuticals and Pesticides [p 165]<br />

Tak Hung, Ben Burley, Emery Dora, Dan Elkes, Steve Gendreau, Denise Jacobus, Rachel Kindt,<br />

Mark Lackner, Lisa Moore, Scott Ogg, Dianne Parry, Roxanna Peng, Ellyn Pham, Jenny Kopczynski<br />

157. In vivo characterization of the effects of the unc-64(md130) mutation on anesthetic sensitivity. [p<br />

166]<br />

Hunt S.J., Mike Crowder<br />

158. Regulation of the C. <strong>elegans</strong> epidermal growth factor homolog LIN-3 [p 167]<br />

Byung Joon Hwang, Paul W. Sternberg<br />

159. Characterization of the regulatory elements required for neuron-specific expression of SNAP-25 in<br />

the nematode [p 168]<br />

Soon Baek Hwang, Junho Lee<br />

160. Analysis of 2° vulval lineage execution [p 169]<br />

Takao Inoue, Paul W. Sternberg<br />

161. Developing a C. briggsae genetic map [p 170]<br />

B. Johnsen, S. Gharib, A. Mah, K. Brown, D. Baillie, P. Sternberg<br />

6


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

162. Coenzyme Q and aging in the nematode <strong>Caenorhabditis</strong> <strong>elegans</strong>. [p 171]<br />

Tanya Jonassen, Pamela L. Larsen, Catherine F. Clarke<br />

163. osm-9 signaling: who’s involved? [p 172]<br />

Amanda H. Kahn, David Tobin, Cornelia I. Bargmann<br />

164. Looking for synergy with PHA-4 on the myo-2 promoter [p 173]<br />

John Kalb, Pete Okkema, Jim McGhee<br />

165. Initial Characterization of Soluble Guanylate Cyclases in C. <strong>elegans</strong> [p 174]<br />

David Karow, Jennifer Chang, Scott Nicholls, Ronald Ellis, Martin Hudson, David Morton, Michael<br />

Marletta<br />

166. Multiple regulatory elements activate end-1 expression in the E lineage [p 175]<br />

Jodie J. Kasmir, Morris Maduro, Joel H. Rothman<br />

167. Mutations that perturb the effect of octopamine/serotonin on pharyngeal activity. [p 176]<br />

John Keane, Leon Avery<br />

168. Pheromone Regulation of Neuroendocrine Outputs in C. <strong>elegans</strong> [p 177]<br />

Scott Kennedy, Gabriel Hayes, Gary Ruvkun<br />

169. Calcium Imaging in Excitable Cells of C. <strong>elegans</strong>. [p 178]<br />

Rex Kerr, Varda Lev-Ram, Roger Y. Tsien, William R. Schafer<br />

170. A genetic analysis of the effects of ethanol on egg laying [p 179]<br />

Hongkyun Kim, M. Christina Yu, James Kim, Steven L. McIntire<br />

171. Genes affecting the activity of nicotinic receptors involved in egg-laying behavior [p 180]<br />

Jinah Kim, Daniel S. Poole, Laura E. Waggoner, Alexandra Treschow, William R. Schafer<br />

172. Sensory axon guidance defects in C. <strong>elegans</strong> [p 181]<br />

Susan Kirch, Gage Crump, Cori Bargmann<br />

173. Isolation of a third lin-4 allele from a lin-3A overexpression line [p 182]<br />

Martha Kirouac, Paul Sternberg<br />

174. elt-5 and elt-6 are essential for development of seam cells, the vulva, and the male tail. [p 183]<br />

Kyunghee Koh, Joel H. Rothman<br />

175. A genetic screen for genes involved in gut development and differentiation in <strong>Caenorhabditis</strong><br />

<strong>elegans</strong> [p 184]<br />

Jay D. Kormish, James D. McGhee<br />

176. An E1-like activating enzyme is involved in cell division processes in the early C. <strong>elegans</strong> embryo. [p<br />

185]<br />

Thimo K. Kurz, Danielle R. Hamill, Bruce Bowerman<br />

177. Olfactory Adaptation [p 186]<br />

Noelle L’Etoile, Cori Bargmann<br />

178. You can’t get there from here: a gene required for pharyngeal extension. [p 187]<br />

SK Lange, JR Saam, SE Mango<br />

179. Signaling by the VAB-1 Eph receptor intracellular domain [p 188]<br />

Kristoffer Larsen,, Sean George, Andrew Chisholm<br />

180. mdf-1 suppressors that may play a role in the metaphase to anaphase checkpoint [p 189]<br />

Elaine Law, Risa Kitagawa, Ann M. Rose<br />

181. Characterization of a C. <strong>elegans</strong> Defecation Mutant [p 190]<br />

Anne Lehtela, Garry Wong<br />

182. Organogenesis of the C. <strong>elegans</strong> Intestine [p 191]<br />

Benjamin Leung, Greg J. Hermann, James R. Priess<br />

183. Expression and regulation of daf-16::gfp constructs [p 192]<br />

Kui Lin, Cynthia Kenyon<br />

184. Identification of novel unc-64 (syntaxin) alleles [p 193]<br />

Christine Liu, C. Michael Crowder<br />

185. Mutations that cause neurite sprouting of the DVB motor neuron [p 194]<br />

Loria, P., Boulin, T., Conte, S., Hobert, O.<br />

186. A b -tubulin gene, tbb-2, functions as an activator of mei-1 and mei-2 in female meiotic spindle<br />

formation in <strong>Caenorhabditis</strong> <strong>elegans</strong>. [p 195]<br />

Chenggang Lu, Martin Srayko, Paul E. Mains<br />

187. Global profile of gene expression during aging [p 196]<br />

James Lund, Pamela Larsen, Pat Tedesco, Thomas Johnson, Stuart Kim<br />

188. Conditional mutations affecting mitotic spindle positioning and polarity in the C. <strong>elegans</strong> embryo [p<br />

197]<br />

Rebecca Lyczak, Bruce Bowerman<br />

189. Role of PDZ domain proteins in establishing gut epithelial polarity [p 198]<br />

Kathleen E. Mach, Stuart K. Kim<br />

190. Genetic analysis of NMDA receptor expression in C. <strong>elegans</strong> [p 199]<br />

David M. Madsen, Chingju Lin, Penelope J. Brockie, Andres V. Maricq<br />

7


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

191. Large Scale Reverse Genetic Approach Using RNAi [p 200]<br />

Sarah Mahoney, Alex Phan, Mark Maxwell, Candace Swimmer, Jonathan Heller, Brett Milash, Kate<br />

McKusick, Monique Nicoll<br />

192. Sequence Confirmation of 182 snps between C. <strong>elegans</strong> N2 and CB4856 Strains and Plans for<br />

Generation of 1000 New snps. [p 201]<br />

Penny Mapa, Kathryn Swan, Mike Ellis<br />

193. Building a dictionary for C. <strong>elegans</strong> promoter sequences [p 202]<br />

Steven McCarroll, Hao Li, Cori Bargmann<br />

194. High Pressure Freezing Methods for C. <strong>elegans</strong> Embryo Ultrastructure and EM Immunolabeling [p<br />

203] Kent L. McDonald, Thomas Mueller-Reichert, Akiko Tagawa, Chad A. Rappleye, Raffi Aroian<br />

195. Molecular Identification of Transcriptional Targets of the DAF-16 Winged Helix Transcription Factor<br />

[p 204]<br />

Joshua J. McElwee, James H. Thomas<br />

196. Functional conservation of C. <strong>elegans</strong> UNC-30 and mouse Pitx2 in GABAergic neuron specification<br />

[p 205]<br />

Jason McEwen, Yishi Jin<br />

197. Genes involved in nicotinic neurotransmission in the pharynx [p 206]<br />

Jim McKay, David Raizen, Leon Avery<br />

198. Genetic analysis of the functions of a GSK-3ß homolog called sgg-1 and a ß-TRCP/slimb homolog<br />

during C. <strong>elegans</strong> embryogenesis [p 207]<br />

Marc Meneghini, Greg Ellis, Ann Schlesinger, Bruce Bowerman<br />

199. The Effect of Nonimmobilizers on C. <strong>elegans</strong> [p 208]<br />

Laura B. Metz, Mike Crowder<br />

200. Isolation and characterization of mutations that enhance let-23(sa62gf) during vulval development [p<br />

209] Nadeem Moghal, Paul W. Sternberg<br />

201. The trampoline assay: A new method for measuring the step response of the chemotaxis<br />

mechanism in C. <strong>elegans</strong>. [p 210]<br />

Moravec, M.L, Cervantes, J., Lockery, S.R.<br />

202. Identification of genes regulating body length in the DBL-1 pathway by differential hybridization of<br />

arrayed cDNAs [p 211]<br />

Kiyokazu Morita, Makoto Mochii, Yukiko Sugihara, Satoru Yoshida, Yo Suzuki, William B. Wood, Yuji<br />

Kohara, Naoto Ueno<br />

203. Mutations in the ephrin mab-26/efn-4 cause defects in closure of the gastrulation cleft and in<br />

epidermal enclosure [p 212]<br />

Sarah L. Moseley, Andrew Chisholm<br />

204. Cellular and developmental events required to generate functional muscle in C. <strong>elegans</strong>. [p 213]<br />

K. Norman, S. Cordes, G. Mullen, P. Rahmani, T. Rogalski, D. Moerman<br />

205. Is the DAG kinase DGK-1 an effector of Go alpha (GOA-1)? [p 214]<br />

Stephen Nurrish, Michael Dybbs, Joshua Kaplan<br />

206. Transforming Nematodes into Insects: Understanding Bt-resistance [p 215]<br />

Johanna O’Dell, Raffi Aroian<br />

207. The cytoskeletal protein zk370.3 may contribute to oocyte development and fertilization [p 216]<br />

Alex Parker, Ann M. Rose<br />

208. Oxidant Stress Responses in C. <strong>elegans</strong> [p 217]<br />

Farhang Payvar, Andrew DeMatteo, Tom Hazinski<br />

209. Pharyngeal pumping defects in unc-103 mutants [p 218]<br />

Christina I. Petersen, David J. Reiner, Elizabeth M. Newton., James H. Thomas, Jeffrey R. Balser<br />

210. A requirement for C. <strong>elegans</strong> Rho-binding kinase in early cleavage [p 219]<br />

Alisa J. Piekny, Paul E. Mains<br />

211. Function of the receptor tyrosine kinase CAM-1/KIN-8 in coordinated movement [p 220]<br />

S. Poulson, D. Madsen, A.V. Maricq<br />

212. The Autosomal Sex Signal in C. <strong>elegans</strong>? [p 221]<br />

Jennifer R. Powell, Barbara J. Meyer<br />

213. Got the blues? Try another genetic screen! [p 222]<br />

Chad Rappleye, Rebecca Lyczak, Bruce Bowerman, Raffi Aroian<br />

214. Identification of Components of the Meiotic Machinery in C. <strong>elegans</strong> [p 223]<br />

Kirthi Reddy, Monica Colaiacovo, Gillian Stanfield, Anne Villeneuve<br />

215. Novel and Atypical Receptor Tyrosine Kinases in Morphogenesis. [p 224]<br />

David J. Reiner, Lewis Leng, Barbara J. Meyer<br />

216. Differential effects of heat shock and cold shock following massed and distributed long-term<br />

habituation training in C. <strong>elegans</strong> [p 225]<br />

Jacqueline Rose, Kenneth Eng, Catharine Rankin<br />

8


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

217. A new en masse training procedure to study long-term habituation in C. <strong>elegans</strong> [p 226]<br />

Jacqueline Rose, Catharine Rankin<br />

218. Global patterns of expression patterns in muscle using mRNA-Tagging [p 227]<br />

Peter J. Roy, Stuart Kim<br />

219. Cooperation between unc-26/synaptojanin and the dynamin-related protein DRP-1 during<br />

mitochondrial division [p 228]<br />

Dan Rube, Todd Harris, Erik Jorgensen, Alexander van der Bliek<br />

220. Calcium dynamics of fertilization in C. <strong>elegans</strong> [p 229]<br />

Aravinthan D.T. Samuel, Venkatesh N. Murthy, Michael O. Hengartner<br />

221. Mutants in Thermosensory Neuron Specification and Function [p 230]<br />

John S. Satterlee, Piali Sengupta<br />

222. Vesicular GABA transport in C. <strong>elegans</strong> requires two proteins UNC-47 and UNC-46 [p 231]<br />

Kim Schuske, Erik M. Jorgensen<br />

223. Utilizing two approaches, genetic and genomic, to identify the vesicular glutamate transporter [p 232]<br />

Kim Schuske, Dan Williams, Erik M. Jorgensen<br />

224. Actin-dependent processes in the early C. <strong>elegans</strong> embryo require the profilin gene pfn-1, the FH<br />

gene cyk-1, and bel-1 [p 233]<br />

Aaron F. Severson, Rebecca Lyczak, David L. Baillie, Bruce Bowerman<br />

225. LIN-12 post-transcriptional downregulation during VPC specification [p 234]<br />

DD Shaye, I Greenwald<br />

226. Distint and redundant functions of mu1 medium chains of AP-1 clathrin-associated protein complex<br />

in the nematode <strong>Caenorhabditis</strong> <strong>elegans</strong> [p 235]<br />

Jaegal Shim, Junho Lee<br />

227. Molecular Mechanisms of Daf-12 Action: Identificationof Response Elements and Functional<br />

Analysis of the Protein [p 236]<br />

Yuriy Shostak, Adam Antebi, Marc R. van Gilst, Kieth R. Yamamoto<br />

228. Serotonin-resistant egg-laying mutants and a receptor knockout in progress [p 237]<br />

Stanley Shyn, William Schafer<br />

229. A novel genetic screen for synaptic transmission genes acting in the diacyglycerol pathway [p 238]<br />

Derek S. Sieburth, Wendy Cham, Josh M. Kaplan<br />

230. Evidence of a Mate-finding Cue in the Free-Living Soil Nematode C. <strong>elegans</strong> [p 239]<br />

Jasper M. Simon, Paul W. Sternberg<br />

231. Understanding C27H5.1: From sequence to sense [p 240]<br />

Jessica Smith, David Pilgrim<br />

232. Genetic screens for novel components involved in blastomere asymmetry in the early C. <strong>elegans</strong><br />

embryo [p 241]<br />

Martha Soto, Craig C. Mello<br />

233. Pax be with you - patterning the pharynx [p 242]<br />

Jeff Stevenson, Andrew Chisholm, Susan E. Mango<br />

234. The evolution and expression of FEM-2 [p 243]<br />

Paul Stothard, Dave Hansen, Tamara Checkland, Dave Pilgrim<br />

235. Forming a gut: the view from an elt and two odds [p 244]<br />

Keith Strohmaier, Morris Maduro, Joel Rothman<br />

236. C. <strong>elegans</strong> homologue of protein phosphatase 4 is required in spermatogenesis [p 245]<br />

Eisuke Sumiyoshi, Asako Sugimoto, Masayuki Yamamoto<br />

237. Transcriptional regulation of the tryptophan hydroxylase gene tph-1 [p 246]<br />

Ji Ying Sze<br />

238. Searching for new genes involved in dosage compensation [p 247]<br />

Chun Tsai, Barbara J. Meyer<br />

239. Characterizing the role of let-99 in spindle orientation [p 248]<br />

Meng-Fu Tsou, Adam Hayashi, Lesilee S. Rose<br />

240. Characterization and cloning of the muscle activation gene unc-58 [p 249]<br />

Monika Tzoneva, James H. Thomas<br />

241. The structure/function relationship of clk-1 in the nematode <strong>Caenorhabditis</strong> <strong>elegans</strong> [p 250]<br />

Antonio Ubach, Siegfried Hekimi<br />

242. Characterization of transcriptional regulation by a class of monomeric nuclear receptors found in C.<br />

<strong>elegans</strong> [p 251]<br />

Marc R. Van Gilst, Keith R. Yamamoto<br />

243. UNC-4 targets ACR-5 and DEL-1: Are they determinants of synaptic choice? [p 252]<br />

Stephen E. Von Stetina, David M. Miller, III<br />

244. Nicotine adaptation: a process involving PKC-dependant regulation of nAChR protein levels. [p 253]<br />

Laura Waggoner, Kari Dickonson, Daniel Poole, Bill Schafer<br />

9


245. Analysis of GLR-7, GLR-5, Ionotropic Glutamate Receptor Subunits [p 254]<br />

Craig S. Walker, David M. Madsen, Penelope J. Brockie, Andres V. Maricq<br />

246. Microarray analysis of gene expression patterns in dauer larvae [p 255]<br />

John Wang, Stuart K. Kim<br />

247. Characterization of CAN cell and excretory canal defects in mig-10(ct41) mutants [p 256]<br />

Nicole Washington, Jim Manser<br />

248. ric-7 encodes a novel presynaptic protein required for neurotransmission [p 257]<br />

Robby M. Weimer, Erik M. Jorgensen<br />

249. The requirement of synaptic vesicle loading for synaptic vesicle exocytosis [p 258]<br />

Robby M. Weimer, Janet E. Richmond, Erik M. Jorgensen<br />

250. Unraveling the biological role of DMWD, a gene close to the unstable CTG-repeat in the myotonic<br />

dystrophy locus. [p 259]<br />

J. <strong>West</strong>erlaken, B. Wieringa, P.E. Mains<br />

251. Calcium imaging of the defecation rhythm in C. <strong>elegans</strong> [p 260]<br />

Jeanna M. Wheeler, James H. Thomas<br />

252. Establishing the left/right asymmetry of Q neuroblast polarisation and migration in C. <strong>elegans</strong> [p 261]<br />

Lisa Williams, Lee Honigberg, Cynthia Kenyon<br />

253. A screen for cell migration and axon outgrowth mutants [p 262]<br />

Jim Withee, Gian Garriga<br />

254. Mapping and Characterization of had-1, an HSN Axon Guidance Gene [p 263]<br />

Lianna Wong, Jim Rader, Gian Garriga<br />

255. Recognition of X-chromosome-enriched DNA elements by dosage compensation proteins [p 264]<br />

Tammy F. Wu, Jason D. Lieb, Barbara J. Meyer<br />

256. Rac-like GTPases and cell migration [p 265]<br />

Yi-Chun Wu, Li-Chun Cheng, Nei-Yin Weng, Ting-Wen Cheng<br />

257. Two new genes regulating neuroblast migration in C. <strong>elegans</strong> [p 266]<br />

Lucie Yang, Mary Sym, Queelim Ch’ng, Cynthia Kenyon<br />

258. Identification and characterization of telomere binding proteins in the nematode C. <strong>elegans</strong> [p 267]<br />

Su Young Yi, Seunghyun Kim, Junho Lee<br />

259. Molecular Analysis of the Dosage Compensation Gene dpy-21 [p 268]<br />

Stephanie Yonker, Edith Cookson, Barbara J. Meyer<br />

260. A search for lethal synaptic function mutants using a sensitized background [p 269]<br />

Karen Yook, Erik Jorgensen<br />

261. Identification of downstream target genes in daf-2 pathway [p 270]<br />

Hui Yu, Pamela L. Larsen<br />

262. Fate specification in male P(9-11).p equivalence group [p 271]<br />

Hui Yu, Paul W. Sternberg<br />

263. Loss of a dynamin related protein MGM-1 causes excessive mitochondrial fragmentation [p 272]<br />

Mauro Zappaterra, Alexander van der Bliek<br />

264. Isolation and phenotypic analysis of syd-7 [p 273]<br />

Mei Zhen, Nikki Alvarez, Yishi Jin<br />

265. A screen to identify genes that regulate the activity of the C. <strong>elegans</strong> glutamate receptor GLR-1. [p<br />

274] Yi Zheng, Heng Xie, Pene J. Brockie, Andres V. Maricq<br />

266. A resource for C. <strong>elegans</strong> microarrays [p 275]<br />

Stuart K. Kim, Min Jiang, Kyle Duke<br />

Leon Avery (Leon@eatworms.swmed.edu)<br />

Last modified: Mon Jul 24 15:24:42 2000<br />

<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

10


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

MEIOTIC PAIRING AND SYNAPSIS<br />

Amy MacQueen, Anne M. Villeneuve<br />

Department of Developmental Biology, Stanford University, Stanford CA 94305<br />

Successful meiotic chromosome segregation relies upon a prior association between homologous<br />

chromosomes. We want to understand how homologs establish and maintain functional associations<br />

throughout meiotic prophase. Using cytological tools, we screened through a collection of meiotic<br />

chromosome segregation mutants to identify mutations that specifically disrupt homologous pairing and<br />

synapsis.<br />

We have identified mutations in three complementation groups that cause defects in homologous<br />

synapsis. hal-1 (homolog alignment) mutants contain chromosomes that are asynapsed in the pachytene<br />

region of the germline, where partner chromosomes are normally aligned in parallel tracks with one<br />

another. Fluorescence in situ hybridization (FISH) studies indicate that homologous pairing is abolished in<br />

hal-1 germlines. Further, chromatin in hal-1 early meiotic nuclei does not undergo the distinct<br />

morphological transition that normally accompanies the onset of pairing, suggesting that hal-1 disrupts an<br />

early step required for the initiation of pairing.<br />

Chromatin in nuclei entering meiosis in sys-1 or sys-2 (synapsis) mutants do exhibit typical transition<br />

morphology, and homologs initially pair. However, as nuclei progress to later stages of meiotic prophase,<br />

paired associations are lost. Failure to stabilize homolog pairing suggests a role for the sys-1 and sys-2<br />

genes in chromosome synapsis. Further, the sys-1 gene encodes a protein consisting mainly of a<br />

predicted coiled-coil domain, suggesting that it is likely a structural component of the synaptonemal<br />

complex. Interestingly, a timecourse analysis of sys-1 meiotic nuclei using FISH probes near to either end<br />

of chromosome I revealed that the "pairing center" end consistently achieves a higher degree of<br />

association during meiotic prophase compared with the opposite end of the chromosome. This differential<br />

behavior of opposite ends of a chromosome strengthens prior notions that synapsis along a chromosome<br />

initiates at the "pairing center"end, and suggests a role for the sys-1 gene in synapsis.<br />

Our analysis demonstrates that homolog pairing and synapsis can be subdivided into discrete steps: hal-1<br />

gene function is required for the early establishment of homologous associations, while sys-1 and other<br />

genes of its class are required for the maintenance of these pairing interactions.<br />

11


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

CONDITIONAL MITOTIC SPINDLE MUTANTS IN C. ELEGANS<br />

Danielle R. Hamill, Bruce Bowerman<br />

Institute of Molecular Biology, University of Oregon, Eugene, OR 97403<br />

The large size of the transparent early embryos and the powerful genetics of C. <strong>elegans</strong>, make it<br />

attractive for studies of cell division. Therefore, we have isolated and observed early embryonic cell<br />

divisions in approximately 600 temperature-sensitive, embryonic-lethal mutants in an ongoing screen in<br />

the lab. About 35 mutants appear defective in microtubule- and/or microfilament-dependent processes,<br />

including pronuclear migration, centrosome function, mitotic spindle assembly or orientation, and<br />

cytokinesis. We isolated ts-alleles of several genes known to affect these processes, including an<br />

aurora-like kinase (air-2) and an MKLP1-like kinesin (zen-4) [see abstract by A.F. Severson], as well as<br />

several previously unidentified genes [see abstracts by J.E. Gomes and R. Lyczak].<br />

Here we describe two temperature-sensitive mutants, spd-4 and spd-5, that are required for mitotic<br />

spindle assembly and function (spd=spindle-defective). spd-4 mutant embryos assemble bipolar mitotic<br />

spindles, but they are shorter than wild type and do not elongate. spd-4 mutant embryos also have<br />

defects in DNA segregation and cytokinesis. Pronuclear migration is defective in spd-5 mutant embryos, a<br />

mitotic spindle does not form, and the first cell division fails. Intriguingly, spd-4 and spd-5 show genetic<br />

interactions that suggest they function together in a complex to regulate mitotic spindle assembly in the<br />

early C. <strong>elegans</strong> embryo.<br />

From the map position and phenotype of spd-4, as well as genetic complementation analysis (in<br />

collaboration with D. Schmidt, S. Strome, and W. Saxton, Indiana University) we believe that spd-4 might<br />

encode a dynein heavy chain gene, although this still needs to be confirmed. Injection of dsRNA<br />

corresponding to the Genefinder locus F56A3.4 phenocopies the spd-5 mutant phenotype. We are<br />

sequencing this locus to determine the molecular nature of the lesion. F56A3.4 encodes a coiled-coil<br />

protein with no significant similarity to other proteins apart from this motif. Therefore, if spd-4 is dynein<br />

heavy chain, spd-5 likely represents a novel dynein regulator.<br />

12


MITOTIC CHROMOSOME SEGREGATION BY A CONSERVED<br />

PROTEIN COMPLEX<br />

K. Hagstrom, R. Chan, D. Pasqualone, B.J. Meyer<br />

HHMI & MCB, UC Berkeley, Berkeley, CA 94720<br />

<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

From bacteria to man, the highly conserved SMC (structural maintenance of chromosomes) protein family<br />

is required for chromosome segregation and cell division. In C. <strong>elegans</strong> SMC proteins also direct X<br />

chromosome dosage compensation. We are studying the composition and function of SMC protein<br />

complexes and how individual SMC proteins participate in more than one chromosomal process. For<br />

example, how does the SMC protein MIX-1, essential for both mitosis and dosage compensation, achieve<br />

its dual function within a single cell? MIX-1 requires the SMC protein DPY-27 for its role in dosage<br />

compensation and X localization, but DPY-27 plays no role in mitosis. Thus, it seemed likely that MIX-1<br />

would have a different SMC partner for mitosis. Searching the C. <strong>elegans</strong> genome revealed another SMC<br />

homolog, SLP-2 (SMC-like protein-2.)<br />

The RNAi phenotype and protein localization of SLP-2 suggested its involvement in mitosis. Like MIX-1,<br />

SLP-2 RNAi produces dead embryos with defects such as chromatin bridges and abnormally large nuclei.<br />

Time-lapse microscopy shows a failure in chromosome segregation, and fluorescent in situ hybridization<br />

reveals nuclei with abnormally high DNA content. Thus, loss of SLP-2 prevents chromosome segregation,<br />

but not DNA replication and cell cycle progression. SLP-2 co-localizes with MIX-1 on mitotic<br />

chromosomes in embryos and in the germline. SLP-2 and MIX-1 surround chromosomes as they<br />

condense, then appear on the poleward face of chromosomes aligned at metaphase, where they remain<br />

until they disappear at telophase.<br />

The idea that MIX-1 partners with SLP-2 for mitosis, but with DPY-27 for dosage compensation, is further<br />

supported by immunoprecipitation (IP) results. Co-IP from embryonic extracts is observed between SLP-2<br />

and MIX-1, but not between SLP-2 and DPY-27. Moreover, IPs show that SLP-2 and MIX-1 are part of a<br />

large protein complex. The identities of these subunits are being explored by mass spectrometry. One<br />

subunit (see R. Chan, et al.) shares homology with both a conserved component of the mitotic complex in<br />

other organisms, and a component of the C. <strong>elegans</strong> dosage compensation complex. It will be interesting<br />

to learn if the mitotic and dosage compensation complexes share additional components, and to what<br />

extent the biochemical activities of these complexes are related.<br />

13


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

HIM-10 A PROBABLE KINETOCHORE PROTEIN INVOLVED IN<br />

MITOTIC AND MEIOTIC CHROMOSOME SEGREGATION<br />

M. Howe 1 , D G. Albertson 2 , B. J. Meyer 1<br />

1HHMI & Dept. of Mol. Cell Bio. UCB, Berkeley, CA 94720<br />

2CRI, UCSF, San Francisco, CA 94143<br />

The mitotic and meiotic chromosomes of C. <strong>elegans</strong> are atypical. These mitotic chromosomes are<br />

holocentric, that is, the kinetochore (the structure mediating chromosome attachment to the spindle)<br />

extends along the length of the chromosome. The ultrastructure of these long kinetochores is similar to<br />

those of monocentic chromosomes common to other animals. C. <strong>elegans</strong> meiotic chromosomes have no<br />

discernable kinetochores at the ultrastructural level. Investigation of C. <strong>elegans</strong> chromosomes may<br />

identify conserved features of kinetochores essential to chromosome segregation in mitosis and meiosis.<br />

To understand C. <strong>elegans</strong> kinetochores we have characterized him-10, a gene implicated in mitotic<br />

kinetochore function by an allele that increases free duplication loss. Cloning him-10 revealed that the<br />

gene encodes a novel protein. Protein localization and loss-of-function phenotypes are consistent with<br />

HIM-10 playing a direct role in kinetochore function in mitosis and meiosis.<br />

HIM-10 appears to associate with the kinetochore face of mitotic chromosomes from prophase through<br />

anaphase. HIM-10 staining partially overlaps with a conserved centromeric histone variant HPC-3, with<br />

HIM-10 localizing to the kinetochore region, distal to HPC-3.<br />

him-10 RNAi treatment caused the progeny from injected animals to die as embryos. Fluorescent in situ<br />

hybridization (FISH) to these dead embryos revealed severe aneuploidy suggesting that lethality is due to<br />

aberrant embryonic mitosis. Tubulin staining of dsRNAi embryos showed displaced metaphase<br />

chromosomes, unipolar chromosome attachments, and irregular nuclei. FISH and tubulin staining suggest<br />

that loss of him-10 function causes segregation defects consistent with kinetochore failure.<br />

him-10 is also required during meiosis. The hypomorphic mutation, him-10 (e1511ts), causes a sterility<br />

that can be rescued by mating with wild-type males, suggesting a role for the protein in sperm meiosis.<br />

FISH to dead embryos from the ts sterile adults revealed monosomic and trisomic embryos, suggesting<br />

that the mutation causes chromosome loss in meiosis, not embryonic mitosis. HIM-10 encases<br />

spermatocyte and oocyte chromosomes and duplications, suggesting that a kinetochore-like structure<br />

functions in C. <strong>elegans</strong> meiosis.<br />

14


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

A C. ELEGANS CHROMOKINESIN REQUIRED FOR<br />

CHROMOSOME SEGREGATION<br />

Jim Powers, Bill Saxton, Susan Strome<br />

Biology Department, Indiana University, Bloomington, IN 47401<br />

A search of the C. <strong>elegans</strong> genome, using the motor domain of kinesin heavy chain (unc-116), which is a<br />

microtubule motor, identified 18 distinct kinesins. Two of these, CeChromoK-A and B, are most closely<br />

related to the vertebrate chromokinesins. Vertebrate chromokinesins bind chromosomes and can bind<br />

microtubules in an ATP-sensitive manner. Therefore, they may function as mitotic motors. However,<br />

strong evidence that chromokinesins actually move along microtubules has not been reported. The<br />

CeChromoKs lack the putative DNA-binding domain that is found in vertebrate chromokinesins.<br />

We have tested the functions of CeChromoKs in oogenesis and early embryogenesis by RNA<br />

interference (RNAi). We detected no phenotypes after RNAi of CeChromoK-A. However, RNAi of<br />

CeChromoK-B causes marked defects in mitosis. Anti-tubulin and anti-histone staining and observation of<br />

GFP-tagged histone in embryos suggest that spindle formation is normal, but that chromosomes<br />

congress poorly to form a loose metaphase plate. During anaphase, chromosomes do not disjoin<br />

accurately, stretching along the pole-to-pole axis to form numerous anaphase bridges. Some<br />

chromosomes fragment, giving rise to multiple micronuclei during telophase.<br />

Antibodies to CeChromoK-B show bright staining of mitotic nuclei in the distal germline and of oocyte<br />

nuclei in the oviduct. After fertilization, CeChromoK-B becomes concentrated between paired meiotic<br />

chromosomes, and is left near the spindle equator as anaphase proceeds. A similar pattern is seen<br />

during mitosis. CeChromoK-B becomes associated with chromosomes during late prophase, remains on<br />

the chromosomes during metaphase and early anaphase, and later becomes concentrated at the spindle<br />

equator, appearing to associate with the overlapping microtubules of the developing telophase bridge.<br />

This concentration persists in the midbody after cytokinesis.<br />

Our results suggest that despite the lack of a recognizable DNA-binding domain, CeChromoK-B<br />

associates with mitotic chromatin and is important for chromosome-microtubule interactions that ensure<br />

an ordered metaphase plate and accurate anaphase separation of chromosomes. We suspect that<br />

CeChromoK-B modulates the interactions of microtubules with chromosome arms to resolve bipolar<br />

chromatid attachment or chromosome catenation.<br />

15


NUCLEAR ENVELOPE DYNAMICS IN CAENORHABDITIS<br />

ELEGANS<br />

Kenneth Lee, Yosef Gruenbaum, Katherine L. Wilson<br />

*No Address*<br />

<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

Emerin, MAN1 and LAP2 are integral membrane proteins of the vertebrate nuclear envelope. They share<br />

a 43-residue N-terminal motif, termed the LEM-domain. We found three putative LEM-domain genes in<br />

<strong>Caenorhabditis</strong> <strong>elegans</strong>, designated emr-1, lem-2, and lem-3. We analyzed emr-l, which encodes<br />

Ce-emerin, and lem-2, which encodes Ce-MAN1. Ce-Emerin and Ce-MAN1 migrate on SDS-PAGE as 17<br />

and 52 kDa proteins, respectively. Based on their biochemical extraction properties and<br />

immunolocalization, both Ce-emerin and Ce-MAN1 are integral membrane proteins localized at the<br />

nuclear envelope. We used antibodies against Ce-MAN1, Ce-emerin, nucleoporins, and Ce-lamin to<br />

determine the timing of nuclear envelope breakdown during mitosis in C. <strong>elegans</strong>. The C. <strong>elegans</strong> nuclear<br />

envelope disassembles very late, compared to vertebrates and Drosophila. The nuclear membranes<br />

remained intact everywhere except near spindle poles during metaphase and early anaphase, fully<br />

disassembling only during mid-late anaphase. Disassembly of pore complexes, and to a lesser extent the<br />

lamina, depended on embryo age: pore complexes were absent during metaphase in >30-cell embryos,<br />

but exist until anaphase in 2-24 cell embryos. Intranuclear mRNA splicing factors disassembled after<br />

prophase. The timing of nuclear disassembly in C. <strong>elegans</strong> is novel, and may reflect its evolutionary<br />

position between unicellular and more complex eukaryotes.<br />

16


THE FORMIN PROTEIN CYK-1 ACTS IN PARALLEL TO AN<br />

AURORA-LIKE KINASE/MKLP-1 PATHWAY TO EXECUTE<br />

CYTOKINESIS IN EARLY CAENORHABDITIS ELEGANS<br />

EMBRYOS<br />

Aaron F. Severson, Danielle R. Hamill, Bruce Bowerman<br />

University of Oregon, Eugene, Oregon 97403 USA<br />

<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

The C. <strong>elegans</strong> Formin Homology protein CYK-1 localizes to cleavage furrows in dividing embryonic cells<br />

and, based on an analysis of partial loss-of-function mutations, is required late in cytokinesis (Swan et al.<br />

1998). Analysis of a more severe allele indicates that CYK-1 also is required early in cytokinesis for<br />

contractile ring assembly or function. In addition to CYK-1, embryonic cytokinesis in C. <strong>elegans</strong> requires<br />

the Aurora-like kinase AIR-2 and the mitotic kinesin-like protein ZEN-4. Genetic interactions involving<br />

these loci suggest that an AIR-2/ZEN-4 mitotic spindle pathway functions in parallel to a contractile ring<br />

pathway that includes CYK-1. We have identified temperature-sensitive alleles of both air-2 and zen-4. A<br />

temporal analysis of their function suggests that AIR-2 acts in metaphase or early anaphase, to localize<br />

ZEN-4 to the spindle interzone, while ZEN-4 acts in cytokinesis, during late anaphase or telophase.<br />

Intriguingly, ZEN-4 may also be required well after the apparent completion of cytokinesis, to maintain the<br />

separation of daughter cells. We are currently using the yeast two-hybrid system to determine if AIR-2<br />

and ZEN-4 interact directly. Additionally, we are collaborating with Dr. Jill Schumacher (University of<br />

Texas) to determine if AIR-2 and ZEN-4 associate in stable complexes that can be immunprecipitated.<br />

Our analysis provides genetic evidence that separable, parallel pathways coordinate microfilament and<br />

microtubule functions during cytokinesis in an animal embryo.<br />

17


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

THE L TYPE CYCLIN SAG-4 IS REQUIRED FOR HEAT-SHOCK<br />

INDUCED PROTEIN EXPRESSION<br />

Wen J. Chen, Yvonne M. Hajdu-Cronin, Paul W. Sternberg<br />

HHMI and Dept. of Biology, Caltech, Pasadena, CA91125, USA<br />

In a screen for suppressors of activated GOA-1 under the control of a heat shock promoter, we identified<br />

four genetic loci that affect heat-shock induction of GOA-1. sag-4 and sag-8 are wild type in appearance,<br />

while sag-3 and sag-5 are egg-laying defective. <strong>West</strong>ern analysis indicated that sag-4 or sag-8 mutations<br />

suppress activated Goa by decreasing heat-shock induced protein expression. Although endogenous<br />

GOA-1 expression is not affected, heat-shock induction of GOA-1 decreased in the suppressor strains.<br />

We cloned sag-4 locus, which encodes a cyclin most similar to cyclin L. The latter is a novel type of cyclin<br />

with unknown function, but also similar to cyclin T, K or C, which was identified as a subunit of TFIIH, part<br />

of RNA polymerase II complex and functions in basal transcription. Only transgenes with hsp16-2<br />

promoter can be affected by sag-4. These results suggest that sag-4 must suppress heat-shock GOA-1<br />

phenotypes by preventing heat- shock mediated transcription in C. <strong>elegans</strong>. We propose that cyclin L is<br />

the type of cyclin acting in TFIIH during heat-shock induced mRNA transcription, which carries function<br />

similar to cyclin T, K or C during basal transcription. sag-3, sag-5 and sag-8 might also be involved in<br />

similar processes.<br />

18


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

CDL-1 ENCODES A STEM- LOOP BINDING PROTEIN (SLBP)<br />

HOMOLOG AND MAY BE ESSENTIAL FOR CORE HISTONE<br />

EXPRESSION.<br />

Yuki Kodama 1 , Asako Sugimoto 1 , Joel Rothman 2 , Masayuki<br />

Yamamoto 1<br />

1Dept. of Biophys. & Biochem., Grad. School of Science, Univ. of Tokyo, Tokyo 113, JAPAN<br />

2Neuroscience Research Institute, Univ. of California-Santa Barbara, CA93106<br />

cdl-1 (cell death lethal) mutants show several embryonic defects: 1) delay in appearance of cell corpses<br />

and accumulation of cell corpses in late embryogenesis, 2) defects in elongation, 3) failure in attachment<br />

of the pharynx to the buccal cavity. To understand the cdl-1 function, we cloned the cdl-1 gene. By<br />

transformation assay, we found that a cosmid T19E10 could rescue the cdl-1 phenotypes. RNAi for ORFs<br />

on T19E10 revealed that some of R06F6.1(RNAi) F1s showed cdl-1-like phenotype, although most of<br />

them arrested at the early embryonic stage. We sequenced the corresponding region from cdl-1 mutants<br />

and identified mutations in two alleles, thus concluding that R06F6.1 is the cdl-1 gene.<br />

cdl-1 encodes a member of the stem-loop binding protein(SLBP) family, which binds to the 3’-stem-loop<br />

of core histone mRNAs. It has been described that metazoan core histone mRNAs have a stem-loop<br />

structure instead of a poly-A sequence, and SLBPs have been implicated in post-transcriptional regulation<br />

of core histone mRNAs. In C. <strong>elegans</strong>, 58 of core histone genes contain a conserved stem-loop sequence<br />

in their 3’-UTR sequence. We confirmed the interaction between CDL-1 protein and the stem-loop<br />

structure by yeast three-hybrid system. This result suggests that CDL-1 may also function in the<br />

post-transcriptional regulation of core histones.<br />

To examine the early embryonic phenotypes in cdl-1(RNAi) embryos, we observed them by DAPI staining<br />

and Nomarski optics. In these embryos, chromosomes were less condensed during mitosis, cytokinesis<br />

occurred before completion of the nuclear division, and nuclear fragments existed in some blastmeres.<br />

These observations suggest that the chromatin structure, especially its condensation, might be defective<br />

in cdl-1(RNAi) embryos. We then performed RNAi with core histone genes, the probable targets of<br />

CDL-1. Most RNAi embryos showed early arrest phenotype similar to cdl-1(RNAi) embryos, which<br />

supports the hypothesis that CDL-1 regulates the expression of core histones.<br />

We are currently trying to examine whether the original cdl-1 phenotypes are also caused by the defect of<br />

core histone expression.<br />

19


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

UNC-23 IS A MEMBER OF THE BAG FAMILY OF<br />

CHAPERONE REGULATORS<br />

Poupak Rahmani, Donald Moerman<br />

Department of Zoology. University of British Columbia, 6270 University Blvd. Vancouver, BC Canada.<br />

V6T 1Z4<br />

Mutations in the unc-23 gene result in detachment of the anterior body wall musculature and a bent-head<br />

phenotype (Waterston et al, 1980). This phenotype is not observed when animals are grown in liquid<br />

culture (Bullerjahn and Riddle, pers. comm.) Neither muscle cell positioning nor myofilament assembly is<br />

affected in liquid grown unc-23 animals; muscle cell attachment, however, is affected since a small<br />

amount of stress applied results in detachment of the muscle cells from the hypodermis.The result of<br />

immunological staining of unc-23 animals with antibodies to basement membrane and hypodermal<br />

components suggest that the primary defect in unc-23 animals is located within the hypodermis. We have<br />

recently cloned unc-23, and found it to be a protein most similar to a chaperone regulator known as<br />

BAG-2 (BCL2-associated athanogene-2). In humans, the BAG family of chaperone regulators contains a<br />

conserved 45 amino acid region near their C termini (the BAG domain) that binds Hsp70/Hsc70 and<br />

control their chaperone activity (Takayama et al. 1999). Human BAG-2 and UNC-23 share 40% amino<br />

acid identity and 62% similarity over the BAG domain and its upstream region. We are currently<br />

attempting to obtain a full-length unc-23::GFP transgenic line to study the spatial and temporal expression<br />

pattern of this gene<br />

20


MATERNAL UNC-45 PROTEIN CO-LOCALIZES WITH NMY-2,<br />

A NON-MUSCLE MYOSIN AT THE CLEAVAGE FURROW OF<br />

EARLY EMBRYOS<br />

Wanyuan Ao, Dave Pilgrim<br />

Department of Biological Sciences, University of Alberta, Edmonton, AB T6G 2E9, Canada.<br />

unc-45 is an essential gene for normal body wall muscle thick filament development and mutants show a<br />

partial maternal effect. The unc-45 protein product (UNC-45) contains tetratricopeptide (TPR) repeats and<br />

similarity to fungal proteins, but its biochemical function is still unknown. We have previously shown that<br />

UNC-45 is a component of muscle thick filaments and co-localizes with myosin heavy chain B but not<br />

myosin heavy chain A in the body wall muscles of adult worms [1] . Previous genetic evidence also<br />

suggests that UNC-45 may interact with myosin heavy chain isoforms in the muscle cells [2] . We show<br />

here that UNC-45 is also contributed maternally to the embryos and present in all cells of the early<br />

embryo. Zygotic UNC-45 is only detected in the developing muscle cells of the embryo.<br />

Moreover, our yeast two-hybrid screens show that UNC-45 interacts specifically with NMY-2, a<br />

non-muscle myosin. These two proteins are also co-localized at the cleavage furrow of the early embryos.<br />

The localization of UNC-45 at the cleavage furrow is dependent on the presence of NMY-2. NMY-2 has<br />

been previously shown to be required for embryonic polarity and cytokinesis [3,4] . Our results suggest that<br />

the maternal UNC-45 may have a function in the early embryo which is independent of muscle function.<br />

References<br />

<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

1. Ao, W., and Pilgrim, D. (2000). J. Cell Biol. 148, 375-384.<br />

2. Venolia, L., and R.H. Waterston. (1990). Genetics. 126, 345-354.<br />

3. Guo, S., and Kemphues, K. J. (1996). Nature, 382, 455-458.<br />

4. Shelton, C. A., Carter, J. C., Ellis, G. C., and Bowerman, B. (1999). J. Cell Biol. 146, 439-451.<br />

21


POLYUNSATURATED FATTY ACIDS REQUIREMENTS FOR<br />

PROPER FUNCTIONING OF THE NERVOUS SYSTEM<br />

Jenny Watts, John Browse<br />

<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

Institute of Biological Chemistry, Washington State University, Pullman, WA 99164-6340<br />

Polyunsaturated phospholipids are critical for the function of excitable membranes. Membrane-mediated<br />

information transfers are intimately related to the biochemical events that occur within the neuronal<br />

plasma membrane. Polyunsaturated fatty acid components of phospholipids are necessary to create a<br />

fluid environment as well as to provide precursors of second messenger signaling molecules. C. <strong>elegans</strong><br />

can synthesize a wide range of polyunsaturated fatty acids using only saturated and monounsaturated<br />

fatty acids from E. coli as precursors. In order to study the role of polyunsaturated fatty acids in the<br />

nervous system, we designed a unique biochemical screen which enabled us to isolate a number of<br />

mutant lines exhibiting a range of altered fatty acid compositions. We discovered that many of the<br />

mutations are in known desaturase genes that encode enzymes responsible for inserting double bonds<br />

into a fatty acid chain. The phenotypes of these strains range from no apparent defects in strains lacking<br />

specific classes of 20-carbon polyunsaturated fatty acids to severe locomotion defects and impaired<br />

defecation in strains with more extreme alterations in fatty acid composition. These more extreme strains<br />

also grow slowly and display temperature sensitive embryonic lethality. Providing the worms with dietary<br />

polyunsaturated fatty acids rescues these defects. We are currently performing assays of neurological<br />

function on the whole range of mutants, both unsupplemented and supplemented with various fatty acids.<br />

Comparison of the worm fatty acid composition with the severity of neurological defects will allow us to<br />

determine the polyunsaturated fatty acid requirements for proper movement and behavior.<br />

22


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

TESTING FUNCTIONS OF PHAGOCYTOSIS RECEPTOR<br />

HOMOLOGS IN CELL CORPSE ELIMINATION AND GONADAL<br />

OUTGROWTH<br />

Sambath Chung 1 , Monica Driscoll 2<br />

1UMDNJ-Graduate School of Biomedical Sciences, Piscataway, NJ 08855<br />

2Rutgers University<br />

Cell death can occur as a normal event in development or as a consequence of cell injury. Effective<br />

elimination of cell corpses is essential for maintaining tissue homeostasis, recycling cellular metabolites,<br />

and removing potentially harmful residual cellular contents. Both C. <strong>elegans</strong> programmed cell death<br />

corpses and necrotic-like corpses (such as those generated by mec-4(d), deg-3(d) and other stimuli) are<br />

removed via the action of seven engulfment ced genes--ced-1, ced-2, ced-5, ced-6, ced-7, ced-10, and<br />

ced-12. We have been interested in identifying genes that might specifically be involved in the<br />

recognition/elimination of necrotic cell corpses. Because the receptors that initially mediate recognition of<br />

the necrotic cells might be different from those recognizing the morphologically distinct programmed cell<br />

death corpses, we considered the hypothesis that a subset of nematode genes related to phagocytosis<br />

receptor genes in other organisms might be required for recognition of necrotic cell corpses.<br />

Mammalian CD36 and Drosophila Croquemort are related scavenger receptors that function in cell corpse<br />

removal. We searched the C. <strong>elegans</strong> genomic database and identified six homologs of the<br />

CD36/Croquemort family. We generated a deletion mutation affecting the gene most closely related to<br />

CD36/Croquemort. This allele harbors a deletion of approximately 1kb, starting about 200 bp upstream of<br />

the receptor open reading frame. We named this locus scr-1, for scavenger receptor-like. The scr-1<br />

deletion mutant does not exhibit necrotic or programmed cell death corpse persistence, nor does this<br />

mutation enhance corpse persistence when present in combination with any of the seven engulfment ced<br />

mutations. Interestingly, however, a significant percentage of scr-1 mutants arrest at the L1 larval stage<br />

and appear to have programmed cell death corpses throughout their bodies. scr-1 mutants do exhibit<br />

distinctive defects in distal tip cell migration, similar to that observed in ced-2, ced-5, ced-10, and ced-12<br />

engulfment mutants. This observation suggests that SCR-1 might function as a receptor important in<br />

gonadal outgrowth in the process involving CED-2, -5, -10, -12.<br />

We have also tested for effects of the other five scr homologs using RNAi.<br />

23


REGULATION OF CELL FUSION IN C. ELEGANS<br />

Scott Alper, Cynthia Kenyon<br />

University of California, San Francisco<br />

<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

Cell fusion is a common process in C. <strong>elegans</strong>. The C. <strong>elegans</strong> hypodermis consists of several<br />

multinucleate syncytia that are generated by the fusion of cells throughout development. The largest<br />

syncytium is hyp7, which spans most of the length of the worm and which contains more than 130 nuclei.<br />

We are carrying out two screens to identify mutations affecting cell fusion. First, we screened for<br />

mutations that prevent fusion of the Pn.p cells with hyp7 on the ventral surface of the worm. Mutations<br />

identified in this screen affect the decision of these cells to fuse. Second, in order to identify mutations in<br />

genes that carry out the cell fusion process, we are screening for mutations that affect fusion of the seam<br />

cells that line the lateral surface of the worm.<br />

We identified several mutations that affect the pattern of Pn.p cell fusion and have been characterizing<br />

two mutations that affect Pn.p cell fusion by altering Hox protein activity. The fusion decision of the 12<br />

Pn.p cells is controlled by two Hox genes, lin-39 and mab-5. lin-39 is expressed in the mid-body [P(3-8).p]<br />

and in hermaphrodites prevents fusion of these cells. mab-5 is expressed more posteriorly [in P(7-11).p]<br />

in both sexes, but is not active in hermaphrodite Pn.ps. In ref-1(mu220) (REgulator of Fusion)<br />

hermaphrodites, P9.p and P10.p fail to fuse with hyp7. This is due, in part, to inappropriate activation of<br />

MAB-5 in ref-1 hermaphrodites. ref-1 encodes a gene with two basic-helix-loop-helix DNA binding<br />

domains of the hairy/E(spl) family.<br />

In males, lin-39 and mab-5 each individually prevent Pn.p cell fusion in P(3-6).p and P(9-11).p,<br />

respectively. However, in P7.p and P8.p, where both Hox genes are expressed in the same cell, they<br />

somehow neutralize one another’s activities, so that P7.p and P8.p fuse with hyp7. In ref-2(mu218)<br />

males, P7.p and P8.p fail to fuse with hyp7, perhaps because LIN-39 and MAB-5 fail to cancel each<br />

others activities. ref-2 has been mapped to a 40 kb region on the center of the X chromosome and<br />

transformation rescue experiments are in progress.<br />

Descendants of the seam cells fuse with hyp7 at all larval stages and ultimately the seam cells fuse with<br />

each other during L4. We are currently using a membrane localized gfp fusion expressed in the seam<br />

cells to identify mutants in which seam cell fusions fail to occur.<br />

24


ETHANOL SENSITIVITY GENES IN CAENORHABDITIS<br />

ELEGANS<br />

MinGi Hong, JaeYoung Kwon, InYoung Lee, MinSung Choi, Junho Lee<br />

Department of Biology, Yonsei University<br />

<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

The mechanisms and sites of action of volatile anesthetics and ethanol are not fully understood. In the<br />

hope of understanding the mechanisms of ethanol, we first identified genes that control sensitivity to<br />

ethanol and anesthetics in the invertebrate system <strong>Caenorhabditis</strong> <strong>elegans</strong>. We identified 10 mutations<br />

that confer ethanol resistance either by EMS mutagenesis or transposon insertion mutagenesis. The<br />

genes are being cloned by positional cloning and analyses on the mutations are under way. In the next<br />

experiments, we used the cDNA microarray to identify genes that are either up-regulated or<br />

down-regulated by exposure of the animals to 7 % ethanol for 6 hours. Several gene families including<br />

heat-shock protein family, glutamate receptor family, and gene families with unknown function, were<br />

up-regulated by ethanol. Also, there are gene families down-regulated. We are now examining these<br />

candidate ethanol-affected genes by northen analysis and GFP reporter analysis. To establish an<br />

experimental system by which one can study Fetal Alcohol Syndrome using the nematode, we examined<br />

the effect of ethanol on embryogenesis. After incubating adult hermaphrodites in 7% EtOH for 12 hours,<br />

we observed egg-laying defects and abnormal embryogenesis. Based on this preliminary data, we will<br />

investigate and characterize the ethanol sensitivity genes involved in embryogenesis. In summary, we<br />

identified ethanol resistance genes by EMS or tensposon mutagenesis; we identified genes whose<br />

transcription levels are altered by ethanol in the microarray analysis; and we established an experimental<br />

model system to study Fetal Alcohol Syndrome.<br />

25


STATE-DEPENDENT LEARNING IN C. ELEGANS.<br />

Jill C. Bettinger, Steven L. McIntire<br />

Gallo Center and Program in Biological Sciences, Department of Neurology, UCSF<br />

The execution of learned behaviors may be triggered by contextual information, consisting of<br />

environmental cues or the internal state of the organism. State-dependent learning refers to the ability of<br />

an organism to more effectively execute learned behaviors if the organism experiences internal contextual<br />

influences similar to those experienced when the learning occurred. Such contexts can be<br />

pharmacologically manipulated by treating with cholinergic compounds, opiates, cocaine and<br />

amphetamines, and with neurodepressants such as ethanol and barbituates 1 . <strong>Worm</strong>s become<br />

intoxicated by ethanol in a manner similar to that of most other organisms tested (see abstracts by A.<br />

Davies and H. Kim, this meeting). We have sought to study the mechanisms of ethanol-induced<br />

state-dependent learning in worms.<br />

Olfactory adaptation in C. <strong>elegans</strong> is a decrease in the chemotaxis response to an odorant as a result of<br />

prior exposure to the odorant 2 . We demonstrate a form of state-dependent learning in worms by pairing<br />

olfactory adaptation and ethanol administration. Ethanol does not interfere with olfactory adaptation,<br />

however, worms exposed to an odorant while being treated with ethanol will only show subsequent<br />

adaptation to the odorant if ethanol is again administered during chemotaxis testing. If the odorant is<br />

presented without ethanol during testing, the animals behave as naïve animals and therefore fail to alter<br />

their behavior based on their previous experience or prior exposure to the odorant.<br />

Further, we demonstrate that the state-dependent effects of ethanol require normal dopaminergic<br />

function. The dopamine-defective cat-1 4 and cat-2 5 mutants are able to adapt to volatile odorants,<br />

however, they do not show state-dependency when they are adapted to volatile odorants while<br />

intoxicated by ethanol. These results suggest that C. <strong>elegans</strong> is capable of a form of learning and indicate<br />

a conserved role of dopamine in the modulation of behavioral responses to ethanol.<br />

1 Izquierdo. in Neurobiology of Learning and Memory (eds Lynch, McGaugh, and Wienberger) 333<br />

(Guilford, New York, 1984).<br />

2 Shulz, Sosnik, Ego, Haidarliu and Ahissar (2000) Nature 403, 549, and references therein.<br />

3 Colbert and Bargmann (1995) Neuron 14, 803.<br />

4 Duerr et al. (1999) J. Neuroscience 19, 72.<br />

<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

5 Lints and Emmons (1999) Development 126, 5819.<br />

26


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

THE UT236 MUTANT IN C. ELEGANS HAS DEFECTS IN THE<br />

INTERACTION OF TWO SENSORY SIGNALS AND AN<br />

ASSOCIATIVE LEARNING.<br />

Takeshi Ishihara 1 , Yuichi Iino 2 , Isao Katsura 1<br />

1Struct.Biol.Cent., Natl.Inst.Genet., Dept of Genet. Grad.Univ.Adv.Stud.<br />

2Mol. Genet. Res. Lab., Univ. of Tokyo<br />

Among many environmental stimuli, C. <strong>elegans</strong> seems to respond to only a few stimuli simultaneously. To<br />

elucidate how the neural circuit processes many sensory signals to respond properly, we have designed a<br />

new assay system to analyze interaction between two responses: chemotaxis to odorants and avoidance<br />

from Cu 2+ ion. In this assay, the behavior depends on the concentration of both stimuli, which are sensed<br />

by different neurons. Our observation suggests that these two responses repress each other. We can<br />

argue that this mutual interaction takes place in a defined part of the neural circuit, which consists of<br />

about 10 pairs of neurons.<br />

One mutant, ut236, has defects in this interaction. It prefers to avoid Cu 2+ ion rather than to go to<br />

odorants, while its dose responses to the odorants and Cu 2+ ion are indistinguishable from those of the<br />

wild type. Positional cloning revealed that the ut236 gene encodes a novel secretory protein (C36B7.7)<br />

with an LDL receptor ligand binding motif. Expression studies with a functional GFP fusion gene and by<br />

immunostaining show that it is expressed in a few sensory neurons and AIY neurons. By using cell type<br />

specific promoter, we found that the expression of C36B7.7 in AIY or ASE is sufficient for the rescue of<br />

this phenotype. Also, by using a heatshock promoter construct, it is shown that the expression of<br />

C36B7.7 in embryonic stage is not sufficient for the rescue but the expression in the late larval or adult<br />

stage is. These results suggest that C36B7.7 is necessary for the neuronal function of these neurons, but<br />

is not for the development of the nervous system.<br />

The ut236 mutant seems to have another defect in an associative learning by paired presentation of NaCl<br />

and starvation. This phenotype can be also rescued by the C36B7.7 transgene. Therefore, we postulate<br />

that this novel secretory protein is involved in the modification of sensory signals in the neural circuit to<br />

regulate their preference in chemical stimuli.<br />

27


MUTATION IN THE LIM HOMEOBOX GENE LIM-6 DISRUPTS<br />

ASYMMETRIC FUNCTION OF THE ASE CHEMOSENSORY<br />

NEURONS<br />

J.T. Pierce-Shimomura, M.R. Gaston, B.J. Pearson, S.R. Lockery<br />

Institute of Neurosci, Univ of Oregon, Eugene, OR 97405<br />

C. <strong>elegans</strong> detects chemicals with 11 pairs of bilaterally symmetric sensory neurons. All of the right-left<br />

members of these pairs share similarity in lineage, morphology, and synaptic connectivity. However, one<br />

of these pairs (ASE) expresses genes asymmetrically 1 . ASEL expresses two putative chemoreceptor<br />

guanylyl cyclases gcy-6&7 and the homeobox gene lim-6, while ASER expresses a different guanylyl<br />

cyclase gcy-5. The ASE neurons are important for chemotaxis to soluble attractants including salts 3 . We<br />

tested whether the asymmetry in expression pattern correlated with an asymmetry in function by ablating<br />

ASER, ASEL, or both neurons in individual worms and tracked each animal during chemotaxis in<br />

gradients of ammonium chloride, sodium acetate, potassium acetate, and ammonium acetate. To<br />

measure chance performance, we tracked worms (n=74) in the absence of a gradient, but analyzed them<br />

as if they were in a gradient. Most ASER- (n=29) and ASE- (n=27) worms failed to reach the peak of the<br />

NH 4Cl gradient, while most ASEL- worms (n=22) reached the peak similar to sham worms<br />

(anaesthetized and recovered but not ablated; n=35). Likewise, most ASER- (n=17) and ASE- (n=15)<br />

worms failed to reach the peak of the K-acetate gradient, while most ASEL- worms (n=20) reached the<br />

peak similar to sham worms. Surprisingly, most ASEL- (n=26) and ASE- (n=12) worms failed to reach the<br />

peak of the Na-acetate gradient, while most ASER- worms (n=25) reached the peak similar to sham<br />

worms. <strong>Worm</strong>s did not perform better than chance in the ammonium-acetate gradient (n=37). Thus,<br />

ASER controls chemotaxis to Cl - and K + , while ASEL controls chemotaxis to Na + . In lim-6 deletion<br />

mutants gcy-5 is ectopically expressed in ASEL, while gcy-6&7 expression is correctly restricted to<br />

ASEL 2 . To examine if LIM-6 specifies asymmetry in ASE function, we tested whether ASE neurons retain<br />

their asymmetric function in lim-6 mutants. In contrast to wildtype worms, many ASER- lim-6 worms<br />

(n=28) were able to reach the peak of the ammonium chloride gradient. Thus, a homeobox gene is<br />

involved in breaking symmetry in neuronal function.<br />

1. Yu et al (1997) PNAS 95:3384-7<br />

2. Hobert et al (1999) Dev 126:1547-62<br />

3. Bargmann & Horvitz (1991) Neuron 7:729-42<br />

<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

28


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

INFORMATION CODING IN THE C. ELEGANS OLFACTORY<br />

SYSTEM<br />

PD Wes, A Sagasti, G Jansen, RHA Plasterk, CI Bargmann<br />

HHMI and Department of Anatomy. University of California, San Francisco, CA 94143-0452<br />

In order to better understand the molecular and cellular basis of behavior, we have begun to genetically<br />

scrutinize odorant discrimination in C. <strong>elegans</strong>. C. <strong>elegans</strong> senses a broad array of attractive odors with<br />

just two pairs of sensory neurons, AWA and AWC. Each member of the pair are thought to be largely<br />

identical, except that the G protein-coupled receptor, STR-2, is stochastically expressed in either the left<br />

or right AWC neuron. A number of odorants activate each neuron. Isoamyl alcohol (iaa),<br />

2,3-pentanedione (pd), butanone (bu) and benzaldehyde (bz) all activate AWC. Each odorant utilizes the<br />

same cGMP signaling cascade, yet animals retain the ability to distinguish amongst some of these<br />

odorants. For instance, when placed in a uniform field of bu, animals will no longer chemotax toward a<br />

point source of bu (defined as saturation), yet will chemotax toward bz. A screen was conducted to<br />

identify mutants that failed to chemotax toward a point source of bz in a field of bu (defined as<br />

cross-saturation). One mutant, ky542, displayed total cross-saturation, as well as defects in pd<br />

chemotaxis and olfactory adaptation. ky542 was cloned and found to be an allele of nsy-1, a "neuronal<br />

symmetry" mutant in which STR-2 is expressed in both AWC neurons. Other nsy mutants, including<br />

nsy-2, nsy-3 and egl-2(gf), also showed cross-saturation phenotypes. One model proposes that odorant<br />

discrimination is achieved by expressing the bu receptor in the STR-2 "ON" cell, the pd receptor in the<br />

STR-2 "OFF", and the bz receptor in both cells. A variety of forward and reverse genetic studies and<br />

ablation experiments support this model. Nevertheless, neuronal asymmetry cannot fully account for<br />

odorant discrimination since the 2 AWC neurons can distinguish at least 4 classes of odorants. For<br />

instance, iaa is distinguished from bu, bz and pd. Therefore, informational processing must also occur<br />

intracellularly. Since at least 6 Ga proteins are expressed in AWC, we reasoned that this protein family<br />

may provide the requisite degree of diversity for signaling specificity. Indeed, gpa-5 is required for<br />

discrimination between iaa and bu in AWC, suggesting that different odorants may employ specific<br />

modulatory signaling pathways within single cells.<br />

29


ROLES OF OSM-9/CAPSAICIN RECEPTOR FAMILY<br />

MEMBERS IN SENSORY BEHAVIORS<br />

D. Tobin 1 , D. Madsen 2 , G. Moulder 3 , R. Barstead 3 , A.V. Maricq 2 , M.<br />

deBono 4 , C. Bargmann 1<br />

1University of California San Francisco, HHMI<br />

2University of Utah<br />

3Oklahoma Medical Research Foundation<br />

4MRC, Cambridge<br />

<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

The osm-9 gene is required for a wide range of sensory modalities in C. <strong>elegans</strong> including<br />

chemosensation, mechanosensation, osmosensation, and certain forms of olfactory adaptation. osm-9<br />

encodes a putative ion channel with striking homology to the vertebrate capsaicin receptor, a cation<br />

channel expressed in pain-sensing neurons and gated by the active component of chili peppers. In the<br />

amphid neuron AWA, OSM-9 localizes to sensory cilia, suggesting a direct role in sensory transduction.<br />

The vertebrate capsaicin receptor VR1 responds to heat, capsaicin, and low pH. Expression of<br />

mamamlian VR1 under an AWA-specific promoter partially rescued osm-9’s AWA defects. Thus, the<br />

homology between the two proteins may have functional relevance. However, OSM-9 is not sensitive to<br />

capsaicin. We found that expression of mammalian VR1 in C. <strong>elegans</strong> nociceptive neurons created a<br />

robust capsaicin-avoidance behavior. Heterologous expression of VR1 should be a useful tool for specific,<br />

drug-inducible neuronal activation.<br />

Four relatives of osm-9 are each expressed in subsets of osm-9-expressing cells. We call these genes<br />

ocr (osm-9/capsaicin receptor-related)genes and have generated mutations in two of them. ocr-1 is<br />

expressed primarily in AWA while ocr-2 is expressed in AWA, ASH, ADL, and ADF. Based on their<br />

expression patterns and similarity to osm-9 we hypothesized that the OCR channels might coassemble<br />

with OSM-9 to form heteromultimeric complexes. Mutations in ocr-2 recapitulate some of osm-9’s defects:<br />

there are dramatic defects in AWA-mediated chemotaxis and ASH-mediated avoidance behaviors. We<br />

propose that different combinations of subunits may account for the distinct functions of osm-9 in different<br />

sensory neurons.<br />

Interestingly, ocr-2 has a role in C. <strong>elegans</strong> social behavior. npr-1 encodes a putative neuropeptide<br />

receptor that regulates the choice between solitary and social foraging behavior. osm-9 and ocr-2 mutants<br />

suppress the clumping behavior of npr-1 mutants. We have performed single-cell rescue experiments to<br />

identify the neurons in which ocr-2 expression is required for social behavior. Identification of these<br />

neurons should help define the sensory signals and neuronal circuitry that mediate clumping.<br />

30


EXECUTION AND REGULATION OF MALE C. ELEGANS<br />

SPICULE MUSCLE CONTRACTIONS DURING MATING<br />

L. René García, Paul W. Sternberg<br />

<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

HHMI & California Institute of Technology, Division of Biology, Pasadena, CA 91125 U.S.A.<br />

As the C. <strong>elegans</strong> male attempts to penetrate the hermaphrodite vulva with his spicules, the protractor<br />

muscles that are attached to these copulatory structures contract and relax rapidly such that the spicules<br />

prod the vulva slit at a frequency of 7.2 ± 1.3 Hz. Phasic protractor muscle contractions persist until the<br />

vulva slit is partially penetrated. Once the vulva is breached, the protractor muscles contract tonically and<br />

the spicules extend completely into the hermaphrodite. The spicule muscles stay contracted for 75 ± 20<br />

seconds, sufficient time for sperm to transfer into the hermaphrodite. The male protractor muscles are<br />

innervated by the SPC motor neurons (2), and removal of these cells results in male impotency (1). The<br />

SPC neurons are essential for tonic, but not phasic spicule muscle contractions; the spicules of<br />

SPC-ablated males can not fully extend into the hermaphrodite, but can prod the vulva at a frequency of<br />

5.1 ± 1.1 Hz.<br />

The ACh agonists levamisole, nicotine, arecoline, and oxotremorine can stimulate the protractor muscles<br />

to contract; and the SPC motor neurons support the expression of the unc-17-encoded VAChT. Therefore<br />

ACh most likely regulates the contractile state of the spicule muscles. The ACh agonists differentially<br />

require egl-19 and unc-68-encoded calcium channels to mediate the behavioral output; thus suggesting<br />

that calcium mobilization from these channels might have non-overlapping roles during copulation.<br />

During mating the spicules of egl-19 males can prod the vulva at a frequency of 5.0 ± 1.5 Hz, but they can<br />

not breach the vulva lips. In contrast, the spicules of unc-68 males can insert into the hermaphrodite, but<br />

prior to penetration, the spicules prod the vulva at a frequency of 0.48 ± 1.4 Hz. Therefore, the UNC-68<br />

channel is utilized in phasic muscle contractions, and the EGL-19 channel is required for tonic muscle<br />

contractions.<br />

1. Liu, K.S., and Sternberg, P.W. (1995).Neuron 14, 79-89.<br />

2. Sulston, J.E., Albertson, D.G., and Thomson, J.N. (1980). Dev. Biol. 78, 542-576.<br />

31


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

GENETIC ANALYSIS OF NICOTINE ADAPTATION IN C.<br />

ELEGANS.<br />

Jinah Kim 1 , Laura E. Waggoner 2 , Kari A. Dickinson 2 , Daniel S.<br />

Poole 2 , William R. Schafer 3<br />

1Department of Neuroscience, University of California, San Diego, La Jolla, CA.<br />

2Department of Biology, University of California, San Diego, La Jolla, CA.<br />

3Departments of Biology and Neuroscience, University of California, San Diego, La Jolla, CA.<br />

In many organisms, prolonged exposure to nicotine causes long-lasting changes in the abundance and<br />

functional activity of nicotinic receptors, processes thought to underlie nicotine addiction in humans. At<br />

present, the molecular basis for nicotine adaptation is poorly understood. We have begun using a genetic<br />

approach to identify molecules required for nicotine adaptation in the egg-laying circuitry of C. <strong>elegans</strong>.<br />

Acute exposure to nicotine or the nicotinic agonist levamisole have dramatic effects on behavior, including<br />

stimulation of egg-laying. These acute effects of nicotine on egg-laying require the activity of a nicotinic<br />

receptor containing subunits unc-29, unc-38, and lev-1, as mutations in these genes confer resistance to<br />

nicotine and levamisole. This receptor appears to function in the vulval muscle, since expression of an<br />

unc-29 transgene under a vulval muscle-specific promoter in unc-29 mutant animals restored egg-laying<br />

sensitivity to levamisole. Upon long-term nicotine treatment, wildtype worms undergo adaptation, and<br />

acquire a long-lasting loss of egg-laying sensitivity. This effect is at least partially mediated at the level of<br />

receptor abundance, as the expression of an UNC-29:GFP chimera was drastically reduced upon<br />

overnight treatment with nicotine.<br />

In order to identify molecules required for the long-term effects of nicotine, we examined known mutants<br />

and screened for new mutants with defects in adaptation. From the known mutants, tpa-1, a PKC<br />

homologue, was shown to be necessary for the control of UNC-29 receptor abundance, since tpa-1<br />

mutants remained sensitive to levamisole and retained high UNC-29 receptor levels in the vulval muscles<br />

even after long-term nicotine treatment. We also identified a new gene, nic-1, in a screen for adaptation<br />

defective mutants. These mutants displayed hypersensitivity to nicotine and a defect in adaptation, as<br />

well as an unusual locomotive phenotype and a deficiency in male mating ability, all of which are<br />

consistent with a defect in cholinergic function in the neuromusculature. Currently, we are in the process<br />

of mapping nic-1 in order to clone it, and hope to discover the identity of the molecule encoded by this<br />

gene.<br />

32


NEURAL CONTROL OF LOCOMOTION IN C. ELEGANS<br />

Saleem Mukhtar 1 , Jane Mendel 2 , Jehoshua (Shuki) Bruck 1 , Paul W.<br />

Sternberg 2<br />

1Mail Code 136-93, Caltech, Pasadena. CA 91125.<br />

2Biology 156-29, Caltech, Pasadena. CA 91125.<br />

<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

Our long-term goal is to understand the functioning of the neural circuit responsible for movement in C.<br />

<strong>elegans</strong>. We hope to accomplish this by perturbing the nervous system and observing what effect this has<br />

on the dynamics of the worm.<br />

We have developed an automated worm tracking system that enables us to record moving worms. We<br />

have also developed an automated worm recognition system that enables us to extract quantitative data<br />

describing the worm’s movement from these videos. Using these tools we have been able to uncover<br />

important constraints on the dynamics of wildtype worms that are imposed by the underlying neural<br />

circuitry. Based on these constraints we have formulated a set of physically relevant metrics that can be<br />

used to quantitatively compare the dynamics of two sets of worms.<br />

By applying these tools to the full spectrum of perturbations - genetic, pharmacological and cell ablations,<br />

we hope to elucidate the functioning of the ventral cord circuit for locomotion.<br />

33


ANALYSIS OF GLUTAMATERGIC NEUROTRANSMISSION BY<br />

KNOCKOUT OF GLUTAMATE TRANSPORTER GENES.<br />

Itzhak Mano, Monica Driscoll<br />

<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

Department of Molecular Biology & Biochemistry, Rutgers University, Piscataway, NJ.<br />

The amino acid L-Glutamate (Glu) is a neurotransmitter that mediates most of the excitatory<br />

neurotransmission in the human brain and is thus central to development, basic physiology and higher<br />

brain functions. Exaggerated activation of glutamatergic transmission leads to neuronal cell death (in a<br />

process termed excitotoxicity) and is believed to be a cause or a contributing factor to many acute and<br />

chronic neurological disorders, including stroke and ALS. In order to ensure accurate response to rapid<br />

Glu signals and to avoid buildup of toxic levels of this transmitter, synaptic Glu is rigorously pumped out of<br />

the synapse by specialized Glu Transporters (GluTs). The molecular components of Glu synapses, and in<br />

particular GluTs, are highly conserved from nematodes to humans, suggesting that a detailed description<br />

of the molecules and the processes involved in normal and pathological Glu neurotransmission in C.<br />

<strong>elegans</strong> might be used to gain insight to these processes in higher animals.<br />

Since a key feature common to the initial steps of many neurodegenerative disorders is a decline in GluT<br />

efficacy, we decided to create synaptic Glu buildup by systematic knockout of C. <strong>elegans</strong> GluT genes. We<br />

have so far knocked out 3 of the 6 GluT genes in the worm, and we are continuously screening additional<br />

deletion libraries for more GluT mutants. Analysis of the phenotypes of the current GluT deletion mutants<br />

by themselves and in combination with other Glu-related mutants is underway. Initial observations<br />

indicate that GluTs are key regulators of pharyngeal pumping and of the responses to a range of<br />

chemical, osmotic and mechanical stimuli. Together with other studies of Glu neurotransmission in C.<br />

<strong>elegans</strong>, these observations serve as initial steps for a detailed molecular and cellular description of key<br />

processes of synaptic function and behavior. Furthermore, the phenotypes of these GluT knockout<br />

mutants can serve as the bases for genetic screens aimed at the identification of additional components<br />

of the pathways involved in normal and pathological Glu neurotransmission.<br />

34


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

ELECTROPHYSIOLOGICAL ANALYSIS OF C. ELEGANS<br />

IONOTROPIC GLUTAMATE RECEPTORS<br />

Jerry E. Mellem, Penelope J. Brockie, David M. Madsen, Andres V.<br />

Maricq<br />

Dept. of Biology University of Utah 257 S. 1400 E. Salt Lake City UT 84112<br />

A fundamental problem in neurobiology is to understand how neuronal circuits function to control<br />

behavior. A simple neuronal circuit that controls movement has been identified in the nematode<br />

<strong>Caenorhabditis</strong> <strong>elegans</strong>. In this circuit, the command interneurons AVA, AVB, AVD, AVE, and PVC are<br />

essential for coordinated movement and escape from tactile stimuli.<br />

Our lab has shown that six putative ionotropic glutamate receptor subunits are expressed in the command<br />

interneurons and that perturbation of the subunits nmr-1 and glr-1 lead to altered locomotion (see abstract<br />

by Brockie et. al.). To better understand how ionotropic glutamate receptors modulate the activity of the<br />

locomotory control circuit, we are undertaking an electrophysiological analysis of the receptors expressed<br />

in the command interneuron AVA.<br />

We have modified the slit worm dissection to isolate neurons of the locomotory control circuit. Using<br />

patch-clamp techniques, we have recorded glutamate-dependent currents from the neuron AVA. The<br />

current can be elicited by glutamate, kainate, and NMDA and can be partially blocked by selective<br />

pharmacological antagonists. The current rapidly desensitizes to glutamate and recovery from<br />

desensitization is slow. The current evoked by NMDA is voltage dependent and exhibits an outward<br />

rectification.<br />

We have also determined the electrophysiological defects underlying the behavioral phenotypes observed<br />

in the glr-1 and nmr-1 deletion mutants. Both mutants display a diminshed response to glutamate. The<br />

nmr-1 mutant, in addition, has an aberrant response to NMDA.<br />

We will describe the dissection used to isolate the command interneuron circuitry, our findings on how<br />

glutamate receptors function in wild type and mutant animals, and our initial insights into how glutamate<br />

receptors modulate the activity of the locomotory control circuit.<br />

35


ELECTROPHYSIOLOGICAL ANALYSIS OF UNC-18<br />

MUTANTS.<br />

J. E. Richmond, R. Weimer, W. S. Davis, E. M. Jorgensen<br />

University of Utah, Salt Lake City, Utah 84112<br />

<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

UNC-18 is a C. <strong>elegans</strong> neuron-specific protein belonging to a conserved protein family implicated in<br />

vesicle fusion. UNC-18 interacts with the SNARE protein syntaxin, which is believed to mediate fusion of<br />

synaptic vesicles with the plasma membrane. Mutations in unc-18 result in severe locomotory defects and<br />

acetylcholine accumulation, implying a critical role for UNC-18 in synaptic transmission. Three mutants<br />

were examined, unc-18(e81 and e234) which have premature stop codons and unc-18(b403) which<br />

disrupts binding to syntaxin. The mutant phenotype was not caused by developmental abnormalities since<br />

GFP-expression patterns revealed motor neurons had normal morphology and exhibited wild-type<br />

distributions of synapses. We have used electrophysiological techniques to analyze synaptic function in<br />

these unc-18 mutants. Evoked release at the C. <strong>elegans</strong> neuromuscular junction was reduced to 5% of<br />

wild-type amplitudes (from 1800pA to 100pA) in all three mutants. A similar reduction in endogenous<br />

synaptic event frequency (from 44Hz to 5 Hz) was observed. The mutant synaptic event amplitudes were<br />

unaffected demonstrating that the reduction in the evoked response was not due to a defect in vesicle<br />

neurotransmitter filling or in post-synaptic receptor sensitivity. Preliminary EM data showed an<br />

accumulation of vesicles at the neuromuscular synapses of unc-18 mutants consistent with a defect in<br />

exocytosis as opposed to defects in vesicle biogenesis or retrieval. In the absence of external Ca2+, the<br />

rates of spontaneous fusion events were also reduced, suggesting that either the docking or fusion<br />

competence of unc-18 mutants is defective. Given these data, how might UNC-18 function in exocytosis?<br />

UNC-18 forms a heterodimer with syntaxin, which is mutually exclusive of SNARE complex formation.<br />

Several studies suggest that the interaction of UNC-18 and syntaxin is an essential step in regulated<br />

release, and that this interaction may be required to promote a conformational change in syntaxin which<br />

facilitates SNARE complex formation. To test this hypothesis, we have engineered a double mutation in<br />

syntaxin, which, in vitro eliminates UNC-18 binding, and induces the syntaxin open configuration. We are<br />

currently examining the effects of this mutation in wild-type, unc-64 (syntaxin) and unc-18 mutants.<br />

36


ELECTROPHYSIOLOGICAL ANALYSIS OF SEROTONIN<br />

MODULATION OF BODY WALL NEUROMUSCULAR<br />

PHYSIOLOGY.<br />

Jon Madison, Joshua Kaplan<br />

361 LSA, Univ. of Cal., Berkeley, CA 94720<br />

The ability of neurons to alter their synaptic function underlies our ability to control behavior. We are<br />

interested in understanding how neurotransmitters like serotonin (5-HT) cause changes in neuron and<br />

synapse function to effect behavioral changes. In C. <strong>elegans</strong>, 5-HT modulates the rate of locomotion.<br />

Recent analysis has shown that 5-HT acts presynaptically on motor neurons through a G-protein Gao<br />

subunit, GOA-1, to reduce acetylcholine (ACh) release at body wall neuromuscular junctions (NMJs) [1].<br />

Using a recently developed dissection technique and whole cell voltage clamp recordings [2], we have<br />

recorded the effects of 5-HT on adult body wall NMJ physiology.<br />

We have recorded from body wall muscle both miniature excitatory post-synaptic currents (mEPSCs) and<br />

muscle responses to ACh application in the presence of 5-HT. We see two effects of 5-HT: a reduction in<br />

EPSC frequency and a reduction in ACh-activated current amplitude. The amplitude of ACh-activated<br />

current is reduced by 40% in the presence of 5-HT. Previous physiological analysis by Richmond and<br />

Jorgensen has shown there to be two classes of ACh receptors at the body wall NMJ; one ACh receptor<br />

class is sensitive to the cholinergic agonist levamisole and one receptor is insensitive [2]. Recordings of<br />

levamisole-activated current show that 5-HT reduces this current up to 90%. These data suggest that the<br />

levamisole activated ACh receptors may be specifically modulated by 5-HT. To resolve the contribution of<br />

post-synaptic modulation by 5-HT to presynaptic changes in mEPSC frequency and to further understand<br />

the mechanism of the post-synaptic response, we have recorded from mutants that might eliminate the<br />

post-synaptic ACh receptor modulation. Since 5-HT’s downstream effects are mediated by G-protein<br />

coupled pathways, we have tested a number of candidate signal transduction mutants expressed in<br />

muscle for defects in ACh receptor modulation. We have tested the G-protein Ga subunit mutants, gpa-14<br />

and gpa-7, and an adenylate cyclase, acy-1, and all are normal for ACh receptor modulation by 5-HT.<br />

Future physiologic and genetic experiments will help further our understanding of 5-HT’s modulation of<br />

synaptic function and behavior.<br />

1. Nurrish et al., Neuron 24: 231-242 1999.<br />

<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

2. Richmond and Jorgensen, Nat Neurosci 2: 791-797 1999.<br />

37


SEROTONIN SIGNALING IN THE PHARYNX<br />

Timothy Niacaris 1,2 , Leon Avery 1,3<br />

<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

1University of Texas Southwestern Medical Center, Department of Molecular Biology, Dallas, TX<br />

75390-9148<br />

2tim@eatworms.swmed.edu<br />

3leon@eatworms.swmed.edu<br />

Serotonin increases the rate of pharyngeal pumping. This effect occurs by at least two different<br />

mechanisms. Serotonin can indirectly stimulate pharyngeal muscle by increasing the activity of the<br />

pharyngeal motorneuron MC. Serotonin can also act independent of the pharyngeal nervous system to<br />

increase pharyngeal pumping rate, presumably by acting directly on pharyngeal muscle. Serotonin has<br />

two additional MC-independent effects on the pharynx. It decreases the duration of pharyngeal<br />

contractions and enhances the effect of the pharyngeal motorneuron M3. To identify the receptor that<br />

mediates these effects, we have searched the genomic sequence for candidate serotonin receptors and<br />

determined their expression patterns. One strong candidate for mediating the effects of serotonin on<br />

pharyngeal muscle is ser-1. ser-1 is expressed predominantly in pharyngeal muscle and has recently<br />

been determined to be responsive to serotonin in a heterologous expression system 1 . We have isolated<br />

a deletion that eliminates the 3’ end and UTR of ser-1. ser-1(ad1675) truncates the receptor and<br />

eliminates a large portion of the C-terminal intracellular tail, a region important for several aspects of<br />

receptor desensitization. ser-1(ad1675) mutants have an exaggerated response to exogenous serotonin,<br />

consistent with an inability to desensitize in the presence of serotonin. We are currently characterizing<br />

worms defective in other components of the G-protein signaling cascade to identify proteins responsible<br />

for mediating the effects of serotonin on pharyngeal muscle.<br />

1 Hamdan et al. (1999) J. Neurochemistry 72(4):1372-83<br />

38


A MUSCARINIC CONTRIBUTION TO THE REGULATION OF<br />

FEEDING<br />

Kate Steger 1,2 , Leon Avery 3<br />

<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

1Department of Biology, McGill University. 1205 Dr. Penfield Ave. Montreal, QC, Canada<br />

2kate@eatworms.swmed.edu<br />

3Department of Molecular Biology, UT Southwestern Medical Center. 6000 Harry Hines Blvd. Dallas, TX<br />

Acetylcholine (Ach) is essential for worms’ survival and can stimulate feeding through nicotinic receptors<br />

on the pharyngeal muscle. However, we suspect that Ach affects the pharynx through muscarinic<br />

receptors as well. <strong>Worm</strong>s that lack Ach (e.g. cha-1 mutants) have a more severe feeding defect than<br />

worms that lack pharyngeal nicotinic transmission (eat-2; eat-18). Atropine, a muscarinic antagonist,<br />

causes worms to appear starved, although it increases their pumping rate.<br />

Three ORFs in the worm genome closely resemble vertebrate muscarinic receptors: C15B12.5 (acm-1),<br />

F47D12.2 (acm-2) and C53A5.12 (acm-3). We have examined the expression of these three genes: all<br />

three are expressed in pharyngeal tissue (muscle, neurons or both) and in the extra-pharyngeal nerve<br />

ring.<br />

We have obtained mutant alleles of two putative muscarinic receptors: acm-1, and acm-2 (a gift from<br />

Stefan Eimer and Rolf Baumeister). The two mutations have very different effects on worm physiology.<br />

acm-2 worms are hypersensitive to aldicarb (an inhibitor of cholinesterase) and to nicotine. They pump<br />

more rapidly than wild type worms in the absence of drug, and decrease their pumping rate in the<br />

presence of atropine. We suspect that acm-2 mutants are defective in down-regulating nicotinic<br />

transmission. We hypothesize that acm-2, which is expressed in neurons both inside and outside the<br />

pharynx, acts as a negative regulator at nicotinic synapses.<br />

acm-1 mutants, in contrast, are resistant to aldicarb, and are not hypersensitive to nicotine. In an unc-17<br />

background of reduced cholinergic transmission, acm-1 worms are hypersensitive to atropine. We<br />

suspect that acm-1, probably in combination with acm-3, affects pumping efficiency or the adaptation of<br />

feeding behavior to particular circumstances. We are currently designing assays and appropriate genetic<br />

backgrounds to examine these possibilities<br />

39


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

WORMBASE: FROM ACEDB TO A MORE COMPLETE AND<br />

USABLE DATABASE<br />

Paul W. Sternberg, Erich Schwarz, Norma Foltz, <strong>Worm</strong>Base<br />

Consortium<br />

Division of Biology 156-29, Caltech, Pasadena, CA 91125<br />

We briefly describe the current status and plans for <strong>Worm</strong>Base, initially an extension of the existing<br />

ACeDB database with a new user interface. The <strong>Worm</strong>Base consortium includes the team that developed<br />

ACeDB (Richard Durbin and colleagues at the Sanger Centre; Jean Thierry-Mieg and colleagues at<br />

Montpellier); Lincoln Stein and colleagues at Cold Spring Harbor, who developed the current web<br />

interface for <strong>Worm</strong>Base; and John Spieth and colleagues at the Genome Sequencing Center at<br />

Washington University, who along with the Sanger Centre team, continue to annotate the genomic<br />

sequence. The Caltech group will curate new data including cell function in development, behavior and<br />

physiology, gene expression at a cellular level, and gene interactions. Data will be extracted from the<br />

literature, as well as by community submission. We look forward to providing the C. <strong>elegans</strong> and broader<br />

research community easy access to vast quantities of high quality data on C. <strong>elegans</strong>. Also, we welcome<br />

your suggestions and criticism at any time. <strong>Worm</strong>Base can be accessed at www.wormbase.org.<br />

40


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

THE C. ELEGANS ORFEOME PROJECT<br />

Jerome Reboul 1 , Philippe Vaglio 1 , Cindy Jackson 2 , Troy Moore 2 ,<br />

Jean Thierry-Mieg 3 , Danielle Thierry-Mieg 3 , Jim Hartley 4 , Gary<br />

Temple 4 , Mike Brasch 4 , Nia Tzellas 1 , Marc Vidal 1<br />

1Dana-Farber Cancer Institute and Department of Genetics, Harvard Medical School, Boston, MA, USA<br />

2Research Genetics, Huntsville, AL, USA<br />

3IGM-CNRS, Montpellier, France<br />

4Life Technologies Inc., Rockville, MD, USA<br />

In addition to gene-based functional genomics approaches such as large-scale gene knock-outs and<br />

microarray or chip analysis, it is also important to develop protein-based approaches, e.g. protein<br />

interaction mapping, protein localization mapping, and biochemical and structural genomics. Most of<br />

protein-based approaches rely upon the availability of near complete set of open reading frames<br />

("ORFeomes") cloned into various expression vectors (i.e., for each ORF: the sequence between the start<br />

and the stop codons, in the absence of 5’ and 3’ untranslated sequences and introns).<br />

To clone the C. <strong>elegans</strong> ORFeome into various expression vectors, we use a Recombination Cloning<br />

technique (RC) referred to as Gateway TM (Walhout et al., 2000, Science, 287, 166-122). RC allows both<br />

the initial cloning of ORFs and their subsequent transfer into different expression vectors by site-specific<br />

recombination in vitro. In addition, RC is amenable to automation in 96-well (or 384-) plate settings, which<br />

is crucial for large-scale ORFeome cloning. So far we have cloned 2,000 C. <strong>elegans</strong> ORFs. At the current<br />

throughput (~400 ORFs/week), ~70% of the C. <strong>elegans</strong> ORFeome should be cloned by the end of the<br />

year. We will present: i) the details of the method used, ii) illustrations of our current throughput, iii) a<br />

description of the cloning quality, iv) how this resource will be made available to the community, and v)<br />

how the ORFeome project will help the protein interaction mapping project (see abstract by Walhout et<br />

al).<br />

41


ANALYSIS OF SPLICING AND REGULATORY ELEMENTS<br />

USING THE INTRONERATOR<br />

W. James Kent, Alan M. Zahler<br />

University of California, Santa Cruz<br />

<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

The Intronerator (http://www.cse.ucsc.edu/~kent/intronerator/) is a set of web-based tools for exploring<br />

RNA splicing, gene structure, and cross-species alignments in C. <strong>elegans</strong>. It features a display of<br />

mRNA/DNA and cross-species DNA/DNA alignments. It also includes a flexible tool for extracting<br />

sequences, a database of introns, and a catalog of alternatively spliced genes. This catalog of 845<br />

alternatively spliced genes identified through comparisons of ESTs with genomic sequence is currently<br />

the largest database of alternatively spliced genes available for any organism. The use of the Intronerator<br />

will be demonstrated and we will explain in brief the algorithms behind the program, and the use of the<br />

program to explore regulatory regions. We will present observations made during the alignment of 8<br />

million bases of C briggsae genomic DNA with the C. <strong>elegans</strong> genome, including characteristics of introns<br />

present in one species but not the other, the ability to confirm the identification of genes for which no EST<br />

data is available, and the degree of rearrangement that has occurred in the ~50 million years since the<br />

two species diverged.<br />

42


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

A GLOBAL PROFILE OF GERM LINE GENE EXPRESSION<br />

USING MICROARRAYS REVEALS GERM LINE-SPECIFIC<br />

REGULATION OF THE X CHROMOSOME IN MALES AND<br />

HERMAPHRODITES<br />

Valerie Reinke 1 , Harold E. Smith 2 , Jeremy Nance 2 , Abby F.<br />

Dernburg 1 , Anne M. Villeneuve 1 , Samuel Ward 2 , Stuart K. Kim 1<br />

1Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305<br />

2Department of Molecular and Cellular Biology, University of Arizona, Tuscon, AZ 85721<br />

We have produced DNA microarrays containing genomic PCR products corresponding to 11,917 C.<br />

<strong>elegans</strong> genes. To identify genes expressed in the germ line, we compared wild-type gene expression<br />

levels to that of glp-4 mutants, in which the germ line precursor cells do not proliferate. We also compared<br />

a mutant strain making only sperm, fem-3(gf), to a mutant strain producing only oocytes, fem-1(lf), to<br />

identify both sperm-enriched and oocyte-enriched genes. Using a statistical criterion for significance,<br />

these experiments together define 1416 germ line-expressed genes that fall into three categories: 650<br />

sperm-enriched, 258 oocyte-enriched, and 508 germ line-intrinsic. We have determined the temporal<br />

expression pattern of each of these genes during development. These germ line genes comprise a<br />

molecular definition of germ line components, and also provide the framework to identify individual genes<br />

involved in specific germ line functions. The sperm-enriched group contains an unusually large number of<br />

protein kinases and phosphatases. The oocyte-enriched group includes potentially new components of<br />

embryonic signaling pathways. The germ line-intrinsic group, defined as genes expressed similarly in<br />

germ lines making only sperm or only oocytes, contains a family of piwi-related genes that may be<br />

important for stem cell proliferation. Surprisingly, we found evidence for germ line-specific regulation of<br />

the X chromosome. Sperm-enriched and germ line-intrinsic genes are nearly absent from the X<br />

chromosome, and X-linked oocyte-enriched genes are expressed at about three fold lower levels than<br />

autosomal genes. Further, a marker for active gene expression (acetylated histone H4) is detectable on<br />

autosomes but staining is strongly reduced on the X chromosomes in germ line nuclei.<br />

43


THE PROMISE AND PERIL OF GENOMICS: SPERM<br />

DEVELOPMENT AS MODEL SYSTEM<br />

Harold Smith, Marci Millhouse, Sam Ward<br />

University of Arizona, Tucson, AZ 85721<br />

<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

Microarray screening allows the investigator to profile expression patterns on a genome-wide scale.<br />

However, even a successful screen can generate a daunting amount of data. In our effort to identify<br />

genes involved in sperm development, we compared expression levels in fem-3(gf) mutants, which make<br />

only sperm, to fem-1(lf) mutants, which make only oocytes. We identified 650 sperm-enriched genes (as<br />

well as 258 oocyte-enriched genes) out of 11,917 genes screened (see Reinke et al.). Now, how do we<br />

use this information? We have focused our efforts in two areas: 1) obtaining deletion mutations in<br />

potentially interesting genes; and 2) identifying sperm promoter elements and their relevant transcription<br />

factors.<br />

Prior work with the calmodulin inhibitor trifluoperazine (TFP) had implicated Ca++ function in both sperm<br />

activation and motility; therefore, we generated a deletion allele of the sperm-enriched Ca++ channel<br />

gene K01A11.4 (since named spe-39). The spe-39 mutant exhibits reduced fertility and an aberrant<br />

sperm morphology that mimics TFP treatment. We can now examine the role of other spe mutants in<br />

Ca++ sensing, sperm activation, and motility.<br />

We are using an in silico approach to identify potential sperm promoter elements, and molecular and<br />

genetic analyses to confirm functional significance. The algorithm (written by John Anderson, NCBI)<br />

determines which sequences are over-represented in the 5’ upstream sequences of sperm genes<br />

compared to non-sperm genes. One of these sperm-enriched sequences contains two potential binding<br />

sites for the GATA transcription factor elt-1. Prior work has shown that elt-1 is required to specify<br />

hypodermal cell fates in the developing embryo; however, our microarray data also identified elt-1 as a<br />

sperm-enriched gene. Yeast one-hybrid screening demonstrates that elt-1 binds to and activates<br />

transcription from these putative promoter elements. In collaboration with Barbara Page, we are currently<br />

investigating the role of elt-1 in sperm development.<br />

44


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

FUNCTIONAL ANALYSIS OF CHROMOSOME I<br />

Andrew Fraser, Ravi Kamath, Peder Zipperlen, Maruxa<br />

Martinez-Campos, Julie Ahringer<br />

Wellcome-CRC Institute, Tennis Court Road, Cambridge, UK<br />

RNAi is a powerful and specific means of inhibiting gene function. However, the most widely used<br />

technique for RNAi, the injection of dsRNA into adult animals, is too labour-intensive to allow efficient<br />

genome-wide screening for gene function by RNAi. An alternative approach for RNAi is to feed bacteria<br />

expressing dsRNA to worms. We have determined conditions for which RNAi by bacterial feeding is as<br />

potent as RNAi by injection. We have constructed a library of dsRNA-expressing bacteria that can be<br />

used to target 90% of genes on chromosome I by RNAi. This reagent can be used for an unlimited<br />

number of low-cost screens for gene function. We have used this library to screen for all genes on<br />

chromosome I that give a clear phenotype when targeted by RNAi. We will present our results and<br />

discuss their implications for genome-wide functional analysis of C. <strong>elegans</strong> genes.<br />

45


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

OPTIMIZING THE MUTAGENIC PROPERTIES OF THE MOS1<br />

TRANSPOSON IN C. ELEGANS<br />

Daniel C. Williams, Jean-Louis Bessereau, Erik M. Jorgensen<br />

Department of Biology, University of Utah, Salt Lake City, UT, 84112<br />

Forward genetic analysis in C. <strong>elegans</strong> provides an unbiased method to determine the function of<br />

individual genes. However, identifying the physical location and molecular nature of a mutation that<br />

results in a specific phenotype is quite laborious. One way to avoid genetic mapping is to use<br />

transposable elements as mutagens, which provide a tag at the mutation site. The C. <strong>elegans</strong> genome<br />

contains multiple copies of endogenous transposons, which hinder efforts to isolate the insertion of<br />

interest. Recently, we demonstrated that Mos1 (a Tc1/mariner family member from D. mauritiana) can<br />

hop in C. <strong>elegans</strong>. Mos1 insertions in the germ-line result in a unique tag at the precise location of gene<br />

disruption. Unfortunately, these results also demonstrate that Mos1 is an inadequate mutagen for two<br />

reasons: (1) the frequency of transposition is too low for genetic screens and (2) Mos1 insertions may be<br />

phenotypically silent.<br />

We are taking a number of approaches to make Mos1 a more effective mutagen. In order to increase<br />

transposition frequency, we determined whether the local DNA structure around Mos1 substrate<br />

molecules affects hopping. We determined that transposition of Mos1 can occur if substrate molecules<br />

are located in repetitive or complex arrays. Current experiments address whether Mos1 copy number<br />

within an array correlates with transposition frequency<br />

Exonic Mos1 insertions may be phenotypically silent because they are spliced out during mRNA<br />

processing in the same manner as Tc1 elements (Rushforth et al. 1996). We generated recombinant<br />

Mos1 elements that contain a polyadenylation signal in the middle of the transposon that should prevent<br />

splicing of the insertion and result in a truncated transcript. These experiments also allow characterization<br />

of the cis factors present within Mos1 that are necessary for transposition. During these experiments we<br />

isolated an allele of dpy-17. Using inverse PCR the location of this insertion was determined to be 176<br />

base pairs upstream of the putative DPY-17 open reading frame F54D8.1. Determination of the physical<br />

location of this mutation was extremely rapid, suggesting that Mos1 will be a valuable tool for forward<br />

genetic analysis.<br />

Rushforth, A. M. and P. Anderson (1996). Mol Cell Biol 16(1): 422-9.<br />

46


SNAP-25, A PROTEIN IMPLICATED GENETICALLY IN C.<br />

ELEGANS ANESTHETIC MECHANISMS, BINDS THE<br />

GENERAL ANESTHETIC ISOFLURANE<br />

Jason Berilgen 1 , Mike Crowder 1,2<br />

<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

1Department of Anesthesiology, Washington University School of Medicine<br />

2Department Molecular Biology/Pharmacology, Washington University School of Medicine<br />

Mutations in the neuronal syntaxin gene unc-64 profoundly alter the volatile anesthetic (VA) sensitivity of<br />

C. <strong>elegans</strong>. Two hypomorphic unc-64 alleles confer hypersensitivity to the VAs isoflurane and halothane,<br />

but a third unc-64 hypomorph, md130, which produces a truncated syntaxin product, is VA resistant. The<br />

difference between the isoflurane EC 50s of the hypersensitive and resistant alleles is over 30-fold; this is<br />

by far the biggest allelic variation in VA sensitivity thusfar seen in any animal. Given that these allelic<br />

differences cannot be explained by indirect effects on synaptic transmission and can be rescued and<br />

knocked-in, we hypothesized that syntaxin or a syntaxin-binding protein binds VAs and that the truncated<br />

md130 product somehow interferes with that binding. We have looked for isoflurane binding to syntaxin<br />

and to two syntaxin-binding proteins, VAMP and SNAP-25, which form a ternary complex with syntaxin to<br />

mediate synaptic vesicle fusion. Recombinant expression proteins (C. <strong>elegans</strong> syntaxin and VAMP -<br />

coded for by snb-1, and rat SNAP-25 - plasmids all kindly provided by M. Nonet) without their<br />

transmembrane domains or lipid modifications were used. We used 19 F-NMR to measure binding of<br />

isoflurane to the purified proteins. The transverse relaxation time (T2) of a nucleus is known to decrease<br />

when its Brownian motion decreases; thus, binding of small molecules (eg. isoflurane) to larger ones (eg.,<br />

a protein) shortens T2s. We found that the T2 of the CF 3-moiety of isoflurane was significantly shorter in<br />

solutions containing SNAP-25 than in buffer alone and was protein and isoflurane concentration<br />

dependent, changing over the in vivo relevant isoflurane concentration range. Recombinant syntaxin had<br />

no effect on the T2. We are currently gathering data for VAMP and the ternary complex. Thus, our data<br />

show that SNAP-25 but not syntaxin binds isoflurane at relevant concentrations; SNAP-25 is the first<br />

neuronal protein shown to bind VAs. Given our genetic data in C. <strong>elegans</strong> and electrophysiologic data in<br />

vertebrates showing that neurotransmitter release is reduced by VAs, we think that SNAP-25 may be a<br />

VA target that mediates some aspects of general anesthesia.<br />

47


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

HAPPY WORMS: FURTHER CHARACTERIZATION OF<br />

FLUOXETINE (PROZAC) RESISTANT MUTANTS<br />

Robert K.M. Choy 1,2 , James H. Thomas 2,3<br />

1Program in Molecular and Cellular Biology, Box 357360, University of Washington, Seattle, WA<br />

98195-7360<br />

2rchoy@genetics.washington.edu<br />

3Department of Genetics, Box 357360, University of Washington, Seattle, WA 98195-7360<br />

Although over 20 million people have taken fluoxetine for a wide range of mental disorders, its molecular<br />

mechanism of action remains unproven. Fluoxetine is a member of the selective serotonin reuptake<br />

inhibitor (SSRI) class of antidepressants, which inhibit the presynaptic serotonin reuptake transporter.<br />

However, it is still unclear whether this inhibition is responsible for their antidepressant action.<br />

Furthermore, the targets responsible for the various side-effects of SSRIs are poorly characterized.<br />

We previously reported that in C. <strong>elegans</strong>, SSRIs induce contraction of nose and body-wall muscles by<br />

acting on a non-serotonergic target. We also reported the isolation and characterization of several<br />

mutants that are nose resistant to fluoxetine (Nrf). These mutants are cross-resistant to other SSRIs, but<br />

are fully sensitive to several other drugs that also induce nose muscle contraction. Therefore, these Nrf<br />

mutations may identify novel genes relevant to antidepressant action.<br />

Mutations in three of the Nrf genes (nrf-5, nrf-6 and ndg-4) have a common secondary phenotype of<br />

producing pale eggs (Peg). These mutants are defective in yolk transport and accumulate yolk in the<br />

pseudocoelomic space. nrf-6 and ndg-4 both encode homologous transmembrane proteins and define a<br />

novel gene family of several dozen members in C. <strong>elegans</strong> and Drosophila. By a combination of mosaic<br />

analysis and tissue specific promoters, we have found that nrf-6 function is required in the intestine for<br />

both fluoxetine-induced nose contraction and yolk transport. We have also cloned nrf-5 and found that it<br />

is homologous to the mammalian BPI/CETP family of secreted lipid binding proteins. One possibility is<br />

that these genes are involved in a novel aspect of antidepressant transport. Alternatively, they may<br />

mediate some aspect of lipid metabolism that is required for antidepressant-induced nose muscle<br />

contraction.<br />

48


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

UNC-43 CA2+/CALMODULIN-DEPENDENT KINASE II<br />

(CAMKII) MUTANT WORMS HAVE CONVULSIONS IN<br />

RESPONSE TO THE SEIZURE-INDUCING DRUG PTZ<br />

Elizabeth M. Newton, James H. Thomas<br />

Dept. of Genetics, University of Washington, Seattle WA 98195<br />

Epilepsy is one of the most common neurological disorders, effecting an estimated 50 million people<br />

worldwide. While most seizures are treatable with present anti-convulsant drugs, approximately 20-30%<br />

of patients remain refractory to drug therapy. Recent work in mouse models has begun to identify genes<br />

involved in seizure susceptibility, but much about the mechanism of seizure susceptibility and the<br />

mechanism of anti-epileptic drugs remains to be elucidated.<br />

Null mutations in C. <strong>elegans</strong> unc-43 CaM Kinase II cause worms to appear nervous and jerky with<br />

occasional spontaneous muscle contractions that resemble convulsions. To test if these contractions<br />

could be related to seizure-like convulsions, we put unc-43 null worms on plates containing<br />

pentylenetetrazole (PTZ), a seizure-inducing drug commonly used in rodent epilepsy models. The<br />

phenotype observed is dramatic: the worms begin to have fast, strong simultaneous contractions of the<br />

anterior and posterior body muscles in an uncontrolled fashion, completely disrupting normal locomotion.<br />

(A videotape of this phenotype will be shown.) Wild-type worms appear mildly hyperactive in response to<br />

PTZ, as expected for a CNS stimulant, and become slightly jerky and less coordinated over time but<br />

never have convulsions, even at doses 10 fold higher than used to induce convulsions in unc-43 mutants.<br />

Other drugs that induce muscle contraction but are not convulsive in mouse models, such as aldicarb and<br />

levamisole, do not induce convulsions in unc-43 mutants but instead induce the expected<br />

hypercontraction/paralysis phenotype. a-CaM Kinase II knock-out mice are epileptic, supporting the idea<br />

that worm convulsions may be related to seizures in rodent models. The unc-43 convulsion phenotype<br />

has a neuronal basis as it is rescued by a transgene expressing an unc-43 cDNA from the aex-3<br />

promoter, which drives expression in all neurons but not muscle.<br />

We tested a number of different mutant strains that might be expected to have hyper-excited neuronal<br />

activity, such as loss of function mutations in potassium channels and mutations in the goa-1 pathway,<br />

but none had convulsions in response to PTZ. We are currently screening for more seizure-susceptible<br />

mutants to determine if this phenotype is specific to unc-43 CaM kinase II. We have also begun screening<br />

for suppressors of the unc-43 PTZ-induced convulsion phenotype, and we have isolated several<br />

independent mutants, indicating that it is likely that there are many genes involved in this response.<br />

Studying suppressors of convulsions may identify new theraputic targets for the future.<br />

49


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

NEUROTOXIN SENSITIVITY OF DOPAMINERGIC NEURONS<br />

IN C. ELEGANS: ROLE OF THE DOPAMINE TRANSPORTER<br />

AND CELL DEATH PATHWAYS<br />

R. Nass 1,2 , J. Duerr 3 , , J. Rand 3 , D. M. Miller 2,4 , R. D. Blakely 1,2<br />

1Dept of Pharmacol, Vanderbilt U. Med. School, Nashville, TN<br />

2Ctr. for Molecular Neuroscience, Vanderbilt U. Med. School, Nashville, TN<br />

3Program in Molecular and Cell Biology, OMRF, Oklahoma City, OK<br />

4Dept of Cell Biology, Vanderbilt U. Med. School, Nashville, TN<br />

The dopamine transporter (DAT) constitutes the primary mechanism for the inactivation of dopamine (DA)<br />

neurotransmission in the brain. DATs are targets for many psychoactive drugs and the cellular gateway<br />

for the accumulation of the neurotoxin 6-hydroxydopamine (6-OHDA) which evokes neuronal death and<br />

Parkinson-like syndrome in animal models. We have previously cloned and tagged the C. <strong>elegans</strong> DAT<br />

(CeDAT), and have shown that it is functionally similar to mammalian DATs and expressed exclusively in<br />

the DA neurons (Jayanthi et al. 1998, Nass et al. 1999). We have also developed WT and DAT knockout<br />

transgenic lines which target a CeDAT promotor-GFP fusion to all 8 DA neurons in the hermaphrodite.<br />

Intense GFP expression is seen throughout the axons and dendrites in the live animals. We now show<br />

that a brief exposure to 6-OHDA results in the blebbing of DA neuronal processes, swelling of DA<br />

neuronal cell bodies, and the loss of CeDAT-GFP expression, while co-exposure with CeDAT substrates<br />

or antagonists significantly reduces the 6-OHDA induced response. We also show that this response is<br />

dependent on the expression of CeDAT, since the CeDAT knockout line is insensitive to the effects of the<br />

neurotoxin. Furthermore, the effect appears to be dependent on a necrotic cell death pathway, since the 6<br />

OHDA induced response occurs in a caspase-deficient ced-3 knockout background. These studies as<br />

well as our progress on toxin-based genetic screens for regulators of CeDAT function, localization, and<br />

the toxin mediated cell death will be presented. Supported by MH19732-07 (RN), GM38679 (JR),<br />

NS26115 (DMM), MH58921 (RDB)<br />

50


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

GENE EXPRESSION IN TRANSGENIC C. ELEGANS ANIMALS<br />

EXPRESSING THE HUMAN BETA AMYLOID PEPTIDE.<br />

Chris Link 1 , Carolyn Johnson 1 , Amy Fluet 1 , Kyle K. Duke 2 , Stuart K.<br />

Kim 2<br />

1Institute for Behavioral Genetics, University of Colorado, Boulder, CO 80309<br />

2Department of Developmental Biology, Stanford Medical School, Stanford, CA, 94305-5427<br />

We have used Andy Fire’s smg-1 dependent expression vectors to engineer worms with inducible<br />

muscle-specific expression of the human beta amyloid peptide. These animals appear wild type at the<br />

permissive temperature (16 degrees C), but become paralyzed approximately 24 hrs after upshift to the<br />

non-permissive temperature (23 degrees C). As expected, this upshift is accompanied by a large increase<br />

in the synthesis of the beta amyloid peptide. We have examined gene expression profiles in these<br />

animals using DNA microarrays containing probes for all predicted C. <strong>elegans</strong> genes. For our initial<br />

experiments, we compared staged L4 populations from uninduced animals and animals upshifted 22 hr<br />

(harvested before onset of paralysis) or 29 hr (all animals arrested), prepared independently in triplicate.<br />

Approximately 150 genes show an average expression increase > 2X at both induced time points, while<br />

approximately 100 genes show an average expression decrease >2X at both timepoints. We have not yet<br />

independently confirmed these putative gene expression changes, with the exception of HSP-16-2, which<br />

is clearly also upregulated at the protein level, as demonstrated by immunoblots. Perhaps the most<br />

obvious pattern of expression changes is observed in the down-regulated class, where there appears to<br />

be a coordinated decrease in expression of a number of genes involved in energy production and<br />

mitochondrial function (e.g., enolase, GPDH, citrate synthase, succinate dehydrogenase, etc.) (These<br />

changes precede any obvious changes in motility.) These results are consistent with reports of abnormal<br />

glucose metabolism in the brains of Alzheimer patients. to send it in.<br />

51


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

A NEMATODE MODEL FOR MITOCHONDRIAL DISEASES<br />

William Y. Tsang, Bernard D. Lemire<br />

Department of Biochemistry, University of Alberta, Edmonton, Alberta, T6G 2H7, Canada.<br />

The mitochondrial respiratory chain (MRC) is composed of 5 protein complexes capable of generating<br />

ATP for most tissues. Its biogenesis requires the coordinate expression of genes from both the nuclear<br />

and the mitochondrial genomes. Defective MRCs are often associated with myopathies, neuromuscular<br />

and heart diseases. We are developing <strong>Caenorhabditis</strong> <strong>elegans</strong> as our model system to investigate the<br />

biochemical, genetic, and phenotypic consequences of mutations affecting the MRC.<br />

We have identified and cloned 3 MRC mutations. The first two mutations are nuclear deletions in the<br />

nuo-1 (C09H10.3) and in the atp-2 (C34E10.6) genes encoding the active site subunits of Complex I and<br />

V, respectively. Both mutations are homozygous lethal and arrest at the L3 stage. atp2 homozygotes<br />

have a normal life span and cannot enter dauer. Furthermore, development of these animals to L3<br />

requires maternal contribution of atp-2 mRNA.<br />

The third mutation is a mitochondrial DNA (mtDNA) deletion that removes 4 MRC and 7 tRNA genes.<br />

These animals are heteroplasmic and aphenotypic despite the different proportions of mutant mtDNA<br />

(~20-80%). However, exposure of N2 gravid adults to ethidium bromide, an inhibitor of mtDNA replication,<br />

results in L3 arrest progeny with characteristics similar to the nuclear mutants. The arrested animals<br />

exhibit a progressive decrease in the steady-state level of mtDNA with age.<br />

We speculate that the L3 arrest phenotype is due the failure of a common energy-requiring step in<br />

development. In support of this hypothesis, we have determined that the passage from L3 to L4 is<br />

associated with a 3-fold increase in the mtDNA copy numbers, as well as a significant increase in the<br />

levels of ATP-2. In addition, we believe that mitochondrial energy metabolism is linked to the regulation of<br />

dauer formation.<br />

52


BACILLUS TOXIN (BT) SUSCEPTIBILITY AND RESISTANCE<br />

IN C. ELEGANS<br />

Lisa Marroquin 1 , Dino Elyassnia 1 , Joel Griffitts 1 , Johanna O’Dell 1 ,<br />

Jerald Feitelson 2 , Raffi Aroian 1<br />

1U.C. San Diego<br />

2Akkadix Corporation<br />

<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

The protein toxins produced by Bacillus thuringiensis (Bt) are the most widely used natural insecticides in<br />

agriculture and have been expressed in transgenic corn, potato, and cotton to provide organic crop<br />

protection against insect pests. Despite successful and extensive use of these toxins, little is known about<br />

toxicity and resistance pathways in target insects since these organisms are not ideal for molecular<br />

genetic studies. To address this limitation and to investigate the potential use of these toxins to control<br />

parasitic nematodes, we are studying Bt toxin action and resistance in <strong>Caenorhabditis</strong> <strong>elegans</strong>. We<br />

demonstrate for the first time that a single Bt toxin can target a nematode. When fed Bt toxin, C. <strong>elegans</strong><br />

hermaphrodites undergo extensive damage to the gut, a decrease in fertility, and death, consistent with<br />

toxin effects in insects. We have screened for and isolated ten recessive mutants that resist the toxin’s<br />

effects on the intestine, on fertility, and on viability. These mutants define five genes, indicating that more<br />

components are required for Bt toxicity than previously known. We find that a second, unrelated<br />

nematicidal Bt toxin may utilize a different toxicity pathway. Our data indicate that C. <strong>elegans</strong> can be<br />

used to undertake detailed molecular genetic analysis of Bt toxin pathways and that Bt toxins hold<br />

promise as nematicides. More recently, we have achieved rescue of one of these mutants with a<br />

subclone containing a single predicted open reading frame. Once confirmed by sequencing of mutant<br />

alleles, this would result in the first definitive identification of a gene required for Bt toxin action in any<br />

organism.<br />

53


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

NEW DAUER GENES AND PATHWAYS<br />

Michael Ailion 1 , James H. Thomas 2<br />

1Molecular and Cellular Biology Program, University of Washington, Seattle, WA 98195<br />

2Department of Genetics, University of Washington, Seattle, WA 98195<br />

We have isolated and characterized mutants that are strongly Daf-c at 27° but that are not Daf-c at 25°,<br />

a phenotype we call Hid (high temperature-induced dauer formation). Previously identified mutants with<br />

this phenotype include unc-64, unc-31, unc-3, egl-4, daf-3 and the dyf mutants. We screened for new Hid<br />

mutants at 27° and isolated 100 mutants, including twenty-three alleles of known Daf-c genes (many<br />

weak alleles), sixteen alleles of dyf or other Daf-d genes and five alleles of unc-31 or unc-3. We also<br />

isolated alleles of a number of new dauer genes. These include alleles of the genes pdk-1, akt-1, aex-6,<br />

kin-8 and hid-1.<br />

Many of the Hid mutants are fully suppressed by mutations in daf-16, suggesting that these genes act in<br />

the daf-2/age-1 (insulin-receptor /PI3 kinase) pathway. These include unc-64 and unc-31, which encode<br />

neurosecretory proteins that may be involved in insulin secretion, and pdk-1and akt-1 which encode<br />

PI3-dependent protein kinases that act downstream of age-1. The aex-6 and hid-1 genes may also act in<br />

this pathway. In addition to their Hid phenotype, the aex-6 and hid-1 mutants also have defects in<br />

movement and defecation, suggesting that they may function in a common process to regulate multiple<br />

behaviors. We cloned hid-1 and found that it encodes a novel protein with many transmembrane<br />

domains. The HID-1 protein is strongly conserved in Drosophila and humans, with each organism<br />

appearing to carry only one gene of this type.<br />

Reexamining the epistatic interactions of strong Daf-c genes at 27° leads to several new inferences.<br />

Mutations in the TGF-b pathway Daf-c genes and daf-2 are completely suppressed by daf-5 and daf-16,<br />

respectively, at 25° but only partially suppressed at 27°. This suggests that there are additional branches<br />

of the TGF-b and insulin pathways that are not detected at 25°. Furthermore, epistasis results based on<br />

pheromone response at 25° show qualitative differences from epistasis results at 27°, indicating that<br />

gene interactions inferred from epistasis experiments performed under one set of conditions may not be<br />

the same as those under a different set of environmental conditions.<br />

54


TEMPORAL REGULATION OF AGING IN THE NEMATODE C.<br />

ELEGANS<br />

Andrew Dillin, Cynthia Kenyon<br />

<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

Department of Biochemistry and Biophysics, University of California at San Francisco<br />

It has long been assumed that the aging process is stochastic, beginning early in life and eventually<br />

gaining enough activity to cause the eventual decline and death of an organism. In contrast, aging can be<br />

viewed as a developmental process, much like early embryonic development, involving signals, receptive<br />

cues and timely action of these processes during discrete periods of an animal’s life cycle. I have tested<br />

these opposing theories by determining when several genes required for lifespan extension actually<br />

function. Aging in C. <strong>elegans</strong> is regulated by a subset of genes that form a signal transduction pathway.<br />

daf-2 is an insulin-like receptor that acts upstream of a PI3-kinase, age-1. Both genes normally function to<br />

limit life span by down regulating the activity of daf-16, a Forkhead-like transcription factor. daf-16<br />

normally functions to increase life span. I will present data indicating that the aging function of the genes<br />

age-1, daf-2 and daf-16 is exerted during a specific period of the worm’s life cycle. These results suggest<br />

that aging, at least in C. <strong>elegans</strong>, is regulated during specific periods in life. In addition, I have also started<br />

work on a novel screen to identify other genes required for life span regulation. Once these genes are<br />

identified I will test when they function.<br />

55


A LONGITUDINAL ANALYSIS OF ADULT NEURONS IN C.<br />

ELEGANS<br />

Mark I. Snow, Pamela L. Larsen<br />

<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

Molecular Biology Program and Division of Biogerontology, University of Southern California, Los<br />

Angeles, CA 90089<br />

An intriguing question that remains to be answered is what happens to the cells of the nervous system in<br />

an individual as it ages? Previous studies following the neuron number in rodents and humans have used<br />

a cross sectional design and the correlative conclusions drawn from these experiments are controversial<br />

with regard to whether or not there is a decline in neuron number with advancing age. C. <strong>elegans</strong> show a<br />

progressive decline in neuronally regulated behaviors as they age, so we chose to follow specific neurons<br />

longitudinally in worms by using integrated transgenic GFP arrays.<br />

The analysis of GFP expression for the 26 GABA and the 8 dopaminergic neurons in hermaphrodites was<br />

performed at early adulthood and again just before death. We found that 84% of the old animals with a<br />

wild-type background do not lose GFP expression in their GABA neurons. For the 16% of animals that<br />

express GFP in less than 26 GABA neurons just before dying, the loss of expression appears to be<br />

cell-specific. In animals expressing GFP in the dopaminergic neurons, 94% do not lose GFP expression<br />

with age. The small number that did display changes were in specific neurons.<br />

Two daf-2 alleles, m41 and e1370, were introduced into the strains expressing GFP in either the GABA or<br />

dopaminergic neurons. The GFP expression patterns of the double mutant strains were not altered and<br />

the life spans of the animals were that of the daf-2 alleles. No change in the number of neurons<br />

expressing GFP was observed in any of the animals with the daf-2(m41) or daf-2(e1370) alleles. The<br />

daf-2 mutation appears to prevent loss of GFP expression in older animals.<br />

The experiments described here are the first to follow neurons in a longitudinal manner in individuals as<br />

they age. Assuming that the endogenous gene and the transgene are similarly regulated, absence of<br />

GFP expression may indicate functional loss because the unc-25 and cat-2 genes encode enzymes<br />

necessary for neurotransmitter biosynthesis. From this approach, we conclude that loss of GFP<br />

expression is not widespread and the daf-2 mutations are neural protective.<br />

56


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

GERM-LINE CELLS THAT REGULATE AGING IN C. ELEGANS<br />

Nuno Arantes-Oliveira, Javier Apfeld, Cynthia Kenyon<br />

Department of Biochemistry and Biophysics, University of California San Francisco<br />

The lifespan of C. <strong>elegans</strong> is regulated by a hormonal pathway that integrates signals from the nervous<br />

and reproductive systems of the animal. Ablation of the germ-line precursor cells produces a lifespan<br />

extension of approximately 60%. This extension is dependent on the presence of the somatic tissues of<br />

the gonad and thus is not likely to be caused by sterility. We have used a variety of genetic approaches,<br />

RNAi and laser ablations to ask which cells in the germ-line (sperm, oocytes, mitotic precursors) regulate<br />

lifespan. Our findings support the interpretation that mitotically-dividing germ-line cells produce the signals<br />

that regulate lifespan. These signals may act by down-regulating daf-16.<br />

57


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

A SCREEN FOR GENES THAT CONTROL PROGRAMMED<br />

CELL DEATH IN THE GERM LINE<br />

S Milstein 1,2,3 , A Gartner 4 , M Hengartner 5<br />

1Program in Genetics, SUNY Stony Brook<br />

2Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724 USA.<br />

3milstein@cshl.org<br />

4Max Planck Institut for Biochemie, D-82152 Martinsried, Germany<br />

5Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724 USA<br />

Although we know a great deal about the apoptotic program in C. <strong>elegans</strong>, we know very little about how<br />

the program is regulated. Since this cell fate is responsible for the demise of about 10% of the somatic<br />

cells, and as many as half of the germ cell, we are interested in defining the genes that may be involved<br />

in the decision of a cell to die.<br />

In order to find such genes, we developed a screen using the vital dye acridine orange, which specifically<br />

stains apoptotic cells in the germ line. Using this screen we have looked at approximately 40,000<br />

genomes and have identified 21 mutants that have increased levels of germ cell death. The ten mutants<br />

that have thus far been mapped represent eight complementation groups, only one of which has multiple<br />

alleles. Of these I am currently cloning two by single nucleotide polymorphism mapping, gla-1(op212) and<br />

gla-3(op234).<br />

In order to determine if these mutants are specifically defective in the regulation apoptosis, or if they are<br />

pathologically compromised, and thus have damage that causes the cells to undergo apoptosis, we made<br />

double mutations with ced-3 and rad-5. Most of the mutations examined are suppressed in the ced-3<br />

background, suggesting that the damage is non pathological (we would have expected necrosis or some<br />

other gross morphological consequence if this were the case). In the rad-5 background, a mutation that<br />

we have shown to render worms insensitive to DNA damage, these mutations are not suppressed. This<br />

again suggests specificity since mutations that somehow result in DNA damage would be suppressed.<br />

58


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

C. ELEGANS P53: REQUIREMENT FOR<br />

RADIATION-INDUCED PROGRAMMED CELL DEATH,<br />

STRESS RESISTANCE, AND NORMAL ADULT LIFESPAN<br />

FOLLOWING DIAPAUSE.<br />

W. Brent Derry, Aaron Putzke, Joel H. Rothman<br />

Department of Molecular, Cellular & Developmental Biology, University of California, Santa Barbara, CA<br />

93106 USA<br />

We have identified the apparent C. <strong>elegans</strong> homologue of the p53 tumor suppressor gene, cep-1, and<br />

have begun to characterize it genetically. CEP-1 appears to be the only p53 family member present in the<br />

completely sequenced C. <strong>elegans</strong> genome and includes all of the signature domains of p53, including 4<br />

of the 5 conserved arginine residues frequently mutated in human cancer. A cep-1::gfp reporter shows<br />

ubiquitous expression throughout embryonic development, which becomes restricted to a subset of<br />

pharyngeal muscle and neuronal cells postembryonically. Ectopic expression of cep-1 or of human p53<br />

fails to induce cell cycle arrest but potently promotes the rapid ced-3-independent necrotic death of all<br />

somatic cells, demonstrating that regulation of cep-1 expression in a critical range is essential for survival.<br />

We isolated a TMP-induced cep-1 mutation that removes the conserved DNA binding domains. Though<br />

cep-1(-) homozygotes show a low level of embryonic lethality, they are generally viable. However, they<br />

are highly resistant to radiation-induced apoptosis of germ cells. Cep-1 deletion mutants also show<br />

enhanced hypoxia-induced lethality, indicating that C. <strong>elegans</strong> p53 is essential for normal physiological<br />

stress response. Furthermore, cep-1 mutants display both reduced viability and reduced lifespan after<br />

release from starvation-induced L1 diapause. These results provide the first evidence linking p53 to<br />

longevity of an animal in response to physiological stress.<br />

59


IDENTIFICATION OF CELL-SPECIFIC REGULATORS OF<br />

PROGRAMMED CELL DEATH IN C. ELEGANS.<br />

Shai Shaham, Cori Bargmann<br />

Dept. of Anatomy, UCSF<br />

<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

Programmed cell death (PCD) is a common metazoan cell fate. In C. <strong>elegans</strong> 12% of somatic cells and<br />

half of germ cells born undergo PCD. Excess or insufficient PCD can lead to a variety of human diseases.<br />

In C. <strong>elegans</strong> PCD is regulated by egl-1 (a BH3 domain protein), ced-9 (a bcl-2 family member), ced-4<br />

(similar to human Apaf-1), and ced-3 (a caspase protease). Although much is known about the machinery<br />

that executes PCD, only in a few instances have the signal transduction pathways leading to activation of<br />

this machinery been elucidated.<br />

Several lines of evidence suggest that ced-9, ced-4, and ced-3 are expressed in most if not all cells. We<br />

are interested in defining molecular events that trigger PCD activation in specific cells. As a first step, we<br />

identified a novel gene (cip-1) whose protein product interacts with the CED-9 protein. CIP-1 (CED-9<br />

interacting protein) binds to CED-9 in a two-hybrid assay. Precipitation experiments using GST-CED-9<br />

and 35 S-labelled CIP-1 also indicate that these proteins may interact. Reminiscent of C. <strong>elegans</strong> EGL-1,<br />

CIP-1 may contain a BH3 domain (bcl-2 homology domain 3) mediating interaction with CED-9.<br />

To understand the function of CIP-1 we expressed the protein in worms using heat-shock promoters.<br />

Animals expressing HS-CIP-1 are dead. This inviability is rescued by ced-3, ced-4, or ced-9(gf)<br />

mutations. Thus, CIP-1 may act upstream of the global PCD machinery to regulate PCD activation.<br />

Preliminary data suggest that cip-1::GFP is expressed in a small number of cells, suggesting that this<br />

gene may well be a cell-specific PCD activator.<br />

In addition to characterizing cip-1, we are screening for genes controlling PCD of the male-specific CEM<br />

neurons, which normally die in hermaphrodites. Using a pkd-2::GFP transgene specifically expressed in<br />

the CEMs, we have isolated 25 independent mutants that abnormally express pkd-2::GFP in<br />

hermaphrodites. Of these, five are alleles of the PCD activators ced-3 and ced-4, and one is an allele of<br />

the tra-2 sex-determination gene. Seventeen mutations promote CEM survival and do not result in any<br />

other gross abnormalities. These mutations may affect either CEM-specific PCD regulators or CEM cell<br />

fate genes. Two allelic mutations define a novel gene required to suppress CEM-like development of<br />

other head neurons.<br />

60


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

BIOCHEMICAL, STRUCTURAL, AND GENETIC ANALYSES<br />

OF THE ACTIVATION OF PROGRAMMED CELL DEATH<br />

Jay Parrish 1 , Betsy Metters 1 , Lin Chen 2 , Ding Xue 1<br />

1Dept. of MCD Biology, University of Colorado, Boulder, Colorado 80309<br />

2Dept. of Chemistry and Biochemistry, University of Colorado, Boulder, Colorado 80309<br />

In the nematode C. <strong>elegans</strong>, three genes egl-1, ced-3, and ced-4, are required for activation of<br />

programmed cell death whereas one gene, ced-9, prevents programmed cell death. Molecular<br />

characterization of these genes has shown that egl-1 encodes a BH3-only (Bcl-2 homology domain 3) cell<br />

death activator, ced-3 encodes an aspartate-specific cysteine protease (caspase), ced-4 encodes an<br />

Apaf-1-like (apoptotic protease activating factor-1) protein, and ced-9 encodes a Bcl-2 homologue.<br />

Genetic studies have placed these genes in a negative regulatory pathway, with egl-1 antagonizing the<br />

activity of ced-9, which inhibits the activity o f ced-4 to activate CED-3 and eventually cell death. In<br />

combination with in vitro biochemical studies, these genetic findings suggest that a protein interaction<br />

cascade involving EGL-1, CED-9, CED-4, and CED-3 regulates the activation of programmed cell death<br />

in C. <strong>elegans</strong>. We have initiated biochemical, structural, and genetic analyses to study the mechanisms<br />

that activate the death protease CED-3, including the protein interactions involved in this activation<br />

process. Specifically, we are attempting to reconstitute the events necessary for activation of CED-3 in<br />

vitro using purified recombinant proteins. Furthermore, we are using a structural modeling approach to<br />

analyze these protein interactions and guide our efforts to demonstrate the importance of these protein<br />

interactions in vivo. So far, we have been able to recapitulate the early events of cell death activation in<br />

vitro, including the EGL-1 mediated release of CED-4 from CED-4/CED-9 complexes and are in the<br />

process of purifying additional factors that are required for the activation of CED-3. Structural modeling of<br />

the EGL-1/CED-9 complex and corresponding biochemical analyses have revealed important molecular<br />

interactions between EGL-1 and CED-9 and the nature of an unusual gain-of-function mutation in ced-9,<br />

facilitating successful design of its second-site suppressor mutations. Further studies of this kind should<br />

help address the central question of how cell death and the cell death protease CED-3 are activated in<br />

the appropriate cells and at the right time.<br />

61


ANALYSIS OF RNA ASSOCIATED WITH P GRANULES IN<br />

GERM CELLS OF C. ELEGANS ADULTS<br />

Jennifer A. Schisa 1 , Jason N. Pitt 2 , James R. Priess 2<br />

1Fred Hutchinson Cancer Research Center<br />

2Howard Hughes Medical Institute, Seattle, WA 98109<br />

The germ plasm of many species contains germline-specific cytoplasmic granules. These granules (called<br />

P granules in C. <strong>elegans</strong>) have been hypothesized to have some function in germline development; and<br />

several of the proteins that are required for germline specification in the embryo (PIE-1, POS-1, MEX-1)<br />

transiently associate with P granules during the first few cell divisions. Because P granules are associated<br />

with germ cell nuclei during most of development, we have begun a characterization of the<br />

nuclear-associated P granules in adult gonads. These studies have shown that P granules in the gonad<br />

are tightly associated with clusters of nuclear pores 1 .<br />

Several protein components of P granules have been identified and found to have potential RNA-binding<br />

motifs. We find that P granules in the gonad contain RNA using a probe for SL1, a transpliced leader<br />

found on many C. <strong>elegans</strong> RNAs. We have also examined sterile animals lacking glh-1 and glh-2; germ<br />

cells in these gonads do not appear to assemble P granules, as ascertained by immunostaining with<br />

several P granule antibodies 2 . Animals lacking glh-1 and glh-2 show hybridization of SL1 on perinuclear<br />

foci. We examined the germ nuclei in these animals by electron microscopy and found perinuclear foci of<br />

abnormal, electron dense material that appeared to be associated with nuclear pores. Thus, it appears<br />

that the putative RNA-binding proteins GLH-1 and GLH-2 are not essential for RNA to localize outside the<br />

nuclear envelope.<br />

We examined various classes of maternally-expressed RNAs by fluorescence in situ hybridization to<br />

determine which specific mRNAs are in P granules. While ribosomal RNAs and some abundant<br />

housekeeping mRNAs do not appear to be enriched in P granules above the general level in the<br />

cytoplasm, several maternal mRNAs that are translated only in the early embryo appear to accumulate on<br />

germ cell P granules.<br />

1 Pitt et al., 2000. Dev. Biol. 219: 315-333.<br />

2 Gruidl et al., 1996. PNAS 93, 13837-13842.<br />

<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

62


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

THE SPLICING SM PROTEINS COLOCALIZE WITH P<br />

GRANULES IN GERM CELLS AND PARTICIPATE IN P<br />

GRANULE LOCALIZATION IN THE EARLY EMBRYO<br />

Scott A. Barbee, Alex L. Lublin, Thomas C. Evans<br />

Department of Cellular and Structural Biology, University of Colorado Health Science Center, Denver,<br />

Colorado, 80262<br />

Early embryonic polarity in C. <strong>elegans</strong> is initiated by asymmetric cell division which leads to the<br />

localization of maternally contributed proteins and mRNAs. In turn, these factors control the temporal and<br />

spatial expression of gene products that determine the developmental fate of cells. Conventional genetic<br />

screening has identified more than 20 genes involved in this process. However, these screens may not<br />

uncover pleiotropic genes that have a necessary function at other stages. With this in mind, we have used<br />

RNA interference (RNAi) to screen a cDNA library for new genes involved in generating asymmetries in<br />

the early embryo.<br />

One gene identified in the RNAi screen is a homolog to human SNRPE (SmE). SmE is one of several Sm<br />

proteins which form a complex required for mRNA splicing and import of snRNAs (U1, U2, U4, and U5)<br />

into the nucleus. RNAi of SmE and other Sm proteins, which together form a subcomplex, resulted in<br />

disruption of P granule segregation in the embryo. P granules are cytoplasmic ribonucleoprotein particles<br />

that segregate to the posterior germline precursor cells. RNAi of several subunits of the Sm complex<br />

caused mislocalization of P granules at various stages. Other aspects of early polarity may also be<br />

disrupted. However, RNAi of other distinct Sm proteins had no effect. RNAi of the core splicing factors<br />

U170K and U2AF65, or of RNA polymerase II (ama-1), also had no effect on P granule localization.<br />

These data suggest that the P granule phenotype is not the result of a general splicing defect. Therefore,<br />

Sm proteins may have a role in generating early embryonic polarity that is independent of snRNA import<br />

and splicing. Interestingly, using a monoclonal antibody against mammalian Sm proteins, we found<br />

colocalization of the Sm’s with PGL-1 in P granules at all stages of development. Sm proteins, therefore,<br />

may be P granule components that participate in P-granule localization and/or other polarity functions.<br />

63


POD-2 DEFINES A NEW CLASS OF MUTANTS REQUIRED<br />

FOR ANTERO-POSTERIOR ASYMMETRY IN THE EARLY<br />

CAENORHABDITIS ELEGANS EMBRYO<br />

Akiko Tagawa, Raffi V. Aroian<br />

<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

University of California, San Diego. La Jolla, CA 92093-0349<br />

The generation of asymmetry in the one-cell C. <strong>elegans</strong> embryo is indispensable for a proper segregation<br />

of developmental determinants and thus development of the multicellular organism. We have isolated a<br />

mutation in a previously unidentified gene, pod-2 (for polarity and osmotic defect), through a screen for<br />

cold sensitive embryonic lethals. Many pod-2 mutant embryos resemble par mutant embryos and show a<br />

loss of physical and developmental asymmetry at the one and two-cell stage. More strikingly, pod-2<br />

resembles another mutant discovered in our lab, pod-1, in that mutants embryos are also osmotically<br />

sensitive. Double mutant analysis indicates these two genes work in the same pathway thus defining<br />

pod-1 and pod-2 as a new class of polarity genes and a new pathway important for early embryonic<br />

development. In addition to polarity and osmotic defects, pod-2 mutation also causes abnormal<br />

localization of germline components such that in 25% of mutant embryos, germline components are<br />

located to a single cell where EMS normally would be. By GFP analysis, we show that this defect can be<br />

traced back to abnormal localization of germline components in the one-cell embryo. Our data suggests<br />

that in the mutant, germline components remain bundled but do not move to the proper position in the<br />

one-cell embryo. Thus pod-2 appears to be required for proper positioning of developmental components<br />

and perhaps other asymmetries. Cloning of pod-2 is underway, and we have recently achieved rescue of<br />

the mutant with a single cosmid.<br />

64


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

OOC-5 ENCODES A PUTATIVE ATPASE REQUIRED FOR THE<br />

REESTABLISHMENT OF ASYMMETRIC PAR PROTEIN<br />

LOCALIZATION IN TWO-CELL EMBRYOS<br />

Stephen E. Basham, Lesilee S. Rose<br />

Section of Molecular and Cellular Biology, University of California, Davis, CA 95616 USA<br />

C. <strong>elegans</strong> embryos display two distinct patterns of spindle orientation. Divisions in the AB lineage occur<br />

in an orthogonal pattern while some division in the P lineage repeatedly occur on the same axis, due to a<br />

rotation of the nuclear-centrosome complex. Genetic and molecular analyses suggests that PAR-3 protein<br />

plays an important role in preventing nuclear rotation in the AB cell, while PAR-2 protein functions to allow<br />

nuclear rotation in P1 by restricting PAR-3 localization within this cell.<br />

We have screened maternal-effect lethal mutants for alterations in cleavage pattern. Hermaphrodites<br />

homozygous for mutations in the ooc-5 or ooc-3 gene produce reduced sized embryos that fail to undergo<br />

P1 nuclear rotation. Our phenotypic characterization of ooc-5 and ooc-3 mutants indicates that polarity is<br />

perturbed in the P1 cell of 2-cell mutant embryos. In particular, we have found that in most ooc-5 and<br />

ooc-3 mutant embryos, PAR-3 localization appears normal at the 1-cell stage but is no longer restricted to<br />

the anterior of the P1 cell in 2-cell embryos. PAR-2 is also normal in 1-cell mutant embryos, but is absent<br />

from the periphery of the P1 cell in most 2-cell ooc mutant embryos. Finally, P granules are localized<br />

normally in 1-cell mutant embryos, but often fail to localize normally to the posterior of the P1 cell in 2-cell<br />

mutant embryos. Thus, ooc-5 and ooc-3 may function (directly or indirectly) in polarizing the P1 cell. One<br />

model for OOC-5 and OOC-3 is that they function in P1 to maintain PAR-2 at the posterior of P1, thus<br />

restricting PAR-3 localization in this cell. We have cloned ooc-5 and it is predicted to encode a 350 amino<br />

acid protein containing an amino-terminal signal sequence, a transmembrane spanning region and an<br />

ATP binding domain. A BLAST search indicates that ooc-5 is related to two other predicted genes in the<br />

C. <strong>elegans</strong> genome and is a member of a family of predicted ATPases that have been identified in a<br />

variety of species. A mutation in one of the human orthologs of this gene results in a neuromuscular<br />

disease, but the biological basis for this disease is not well understood. We are currently generating GFP<br />

reporter constructs and anti-OOC-5 antibodies to examine OOC-5 expression and localization.<br />

65


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

RIC-8 (SYNEMBRYN): A NOVEL REGULATOR OF G PROTEIN<br />

SIGNALING<br />

Kenneth G. Miller, Melanie D. Emerson, John R. McManus, James B.<br />

Rand<br />

Program in Molecular and Cell Biology, Oklahoma Medical Research Foundation, Oklahoma City, OK<br />

73104<br />

Recent studies describe a network of signaling proteins centered around GOA-1 (G oa) and EGL-30<br />

(G qa) that regulates neurotransmitter secretion in C. <strong>elegans</strong> by controlling the production and<br />

consumption of diacylglycerol (DAG). We sought other components of the G oa-G qa signaling network by<br />

screening for aldicarb resistant mutants with phenotypes similar to egl-30 (G qa) mutants. In so doing we<br />

identified ric-8, which encodes a novel protein named RIC-8 (synembryn). Through cDNA analysis we<br />

show that RIC-8 is conserved in vertebrates. Through immunostaining we show that RIC-8 is<br />

concentrated in the cytoplasm of neurons. Exogenous application of phorbol esters or loss of DGK-1<br />

(diacylglycerol kinase) rescues ric-8 mutant phenotypes. A genetic analysis suggests that RIC-8 functions<br />

upstream of EGL-30 (G qa), or in a parallel intersecting pathway. Both ric-8 and goa-1 reduction of<br />

function mutants also exhibit partial embryonic lethality. Furthermore, the embryonic lethality of ric-8<br />

mutants is enhanced to 95-100% by a 50% reduction in maternal goa-1 gene dosage. In a separate study<br />

we investigated the roles of RIC-8 and GOA-1 in early embryos (pre 8-cell stage) and found that goa-1<br />

and ric-8 mutant embryos exhibit defects in the movements of centrosomes. Comparing the roles of<br />

RIC-8 and GOA-1 in the nervous system and the embryo reveals potentially informative differences<br />

between the two pathways. First, EGL-30 (G qa) does not appear to play a role in the embryonic pathway.<br />

Second, in the embryonic pathway, reduction of function mutations in goa-1 and ric-8 lead to similar<br />

phenotypes that are enhanced in goa-1; ric-8 double mutants, whereas in the adult neuronal pathway the<br />

same goa-1 and ric-8 mutants have opposite phenotypes and suppress each other. Based on these<br />

findings, we propose that RIC-8 is required, directly or indirectly, for proper activation of G proteins in the<br />

early embryo and in the nervous system.<br />

66


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

MED-1 AND -2 ACT AT THE CONVERGENCE POINT OF<br />

SKN-1 AND POS-1 TO SPECIFY MS AND E IDENTITY<br />

Morris F. Maduro, Regina Broitman-Maduro, Joel H. Rothman<br />

Department of MCD Biology, UC Santa Barbara, Santa Barbara, CA 93106<br />

EMS produces daughters with different fates: E produces the endoderm while MS makes mesoderm,<br />

including the posterior pharynx. Depletion of the zygotically expressed MED-1 and -2 GATA transcription<br />

factors by RNAi results in a penetrant MS ’ C transformation and an impenetrant E ’ C transformation,<br />

similar to that seen in maternal skn-1 mutants. However, unlike skn-1 mutants, ABa still makes anterior<br />

pharynx in med-1,2(RNAi) embryos, indicating that the med genes are not required for the<br />

GLP-1-mediated signal that induces ABa pharynx. pos-1 mutants also misspecify MS, yet retain<br />

GLP-1-dependent pharynx (Tabara et al. 1999). These observations suggest that POS-1 is also required<br />

for an activity downstream of SKN-1.<br />

The med genes appear to act at the convergence of SKN-1 and POS-1 function in the EMS lineage.<br />

While expression of a med-1::GFP::MED-1 reporter is detected in the early E and MS lineages, it is<br />

undetectable in both skn-1(RNAi) and pos-1(RNAi) embryos. Ectopic med expression is detected in<br />

embryos expressing SKN-1 ectopically. In addition, expression requires a cluster of upstream conserved<br />

SKN-1 binding sites, which bind bacterially expressed SKN-1 in vitro. We propose that POS-1, which is<br />

primarily cytoplasmic, modulates an activity required for transcriptional activation of the meds by SKN-1.<br />

end-1 and end-3 specify E fate, and are likely downstream targets of med-1,2 in the E lineage. Indeed,<br />

GFP-tagged MED-1 binds in vivo to an extrachromosomal array containing the end-3 promoter. This<br />

interaction also occurs in MS, where end-1 and end-3 are known to be repressed by POP-1. Thus, the<br />

mechanism by which POP-1 represses E fate in MS does not preclude binding of MED-1 to end-3.<br />

Our data indicate that SKN-1 directly activates the med genes through a POS-1-requiring mechanism. As<br />

the meds are required for both E and MS specification, their activity appears to be modulated by POP-1,<br />

the target of the Wnt/MAPK pathways in the E blastomere, perhaps at a step after their binding to target<br />

promoters.<br />

67


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

MASS SPECTROMETRIC IDENTIFICATION OF PLP-1 AND ITS<br />

ROLE IN MESENDODERM SPECIFICATION<br />

E. Witze 1 , E. Field 2 , D. Hunt 2 , J.H. Rothman 1<br />

1Department of MCD Biology, University of California, Santa Barbara, CA<br />

2Department of Chemistry, University of Virginia, Charlottesville, VA<br />

end-1 encodes a GATA type transcription factor that is sufficient to specify the endoderm progenitor, E.<br />

end-1 is activated in the E lineage by the combined action of SKN-1 and the Wnt/MAP kinase-activated<br />

form of POP-1 (see abstract by Kasmir et al.). It is repressed in MS, the sister of E, by POP-1 in the<br />

absence of the Wnt/MAPK signal.<br />

We are taking a biochemical approach to characterize additional factors that activate end-1 in the E<br />

lineage and repress it in MS. We developed procedures for isolating factors from early developing C.<br />

<strong>elegans</strong> embryos that bind key regulatory elements in end-1. Mass spectrometry of affinity-purified<br />

proteins identified several end-1 binding factors, one of which, encoded by the plp-1 gene, is an apparent<br />

homologue of a mammalian transcription factor called pur alpha, whose in vivo function is unknown.<br />

We performed RNAi to examine the role of PLP-1 in mesendoderm development. Less than 10% of<br />

plp-1(RNAi) embryos arrest before hatching. A fraction of these contain extra intestinal cells and show a<br />

concomitant loss of MS-derived (posterior) pharynx. This phenotype appears to result from an MS to E<br />

transformation, consistent with our finding that end-1 is sometimes expressed in MS and its descendants<br />

in plp-1(RNAi) embryos. Thus, plp-1 appears to function in part to repress end-1 expression in the MS<br />

lineage. As such, it may act as a corepressor with POP-1.<br />

Paradoxically, we also observe an impenetrant (


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

THE C. ELEGANS NEUROD HOMOLOG CND-1 FUNCTIONS<br />

IN MULTIPLE ASPECTS OF MOTOR NEURON FATE<br />

SPECIFICATION<br />

Steven Hallam 1 , Emily Singer 1 , David Waring 2 , Yishi Jin 1<br />

1Department of Biology, Sinsheimer Laboratories, University of California Santa Cruz, California 95064<br />

2Fred Hutchinson Cancer Research Center, Seattle, Washington 98109<br />

The basic Helix-Loop-Helix transcription factor NeuroD has been implicated in neuronal fate<br />

determination, differentiation and survival. Here we report the expression and phenotypic analysis of<br />

cnd-1, a C. <strong>elegans</strong> NeuroD homolog*. cnd-1 expression was first detected in neuroblasts of the AB<br />

lineage in 14 cell embryos and maintained in many neuronal descendents of the AB lineage during<br />

embryogenesis, diminishing in most terminally differentiated neurons prior to hatching. Specifically, cnd-1<br />

reporter genes were expressed in the precursors of embryonic ventral cord motor neurons including<br />

ABplpp, ABprpp and their progeny. A loss of function mutant, cnd-1(ju29), exhibited multiple defects in<br />

the ventral cord motor neurons. First, the number of motor neurons was reduced, apparently caused by<br />

the premature withdrawal of the precursors from mitotic cycles. Second, the strict correlation between the<br />

fate of a motor neuron with respect to its lineage and position in the ventral cord was disrupted, as<br />

manifested by the variable spatial and temporal expression patterns of motor neuron fate specific<br />

markers. Third, motor neurons also exhibited defects in terminal differentiation characteristics including<br />

axonal morphology and synaptic connectivity. Finally, the expression patterns of three neuronal<br />

type-specific transcription factors, unc-3, unc-4 and unc-30, were altered. Our data suggest that cnd-1<br />

may specify the identity of ventral cord motor neurons both by maintaining the mitotic competence of their<br />

precursors and by modulating the expression of neuronal type-specific determination factors. cnd-1<br />

appears to have combined the functions of several vertebrate neurogenic bHLH proteins, and may<br />

represent an ancestral form of this protein family.<br />

* Singer E et al., <strong>Worm</strong> Breeder’s Gazette 16(1): 52 (October 1, 1999)<br />

69


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

LEFT-RIGHT ASYMMETRY IN C. ELEGANS INTESTINAL<br />

ORGANOGENESIS INVOLVES A LIN-12/NOTCH SIGNALING<br />

PATHWAY<br />

Greg J. Hermann, Ben Leung, James R. Priess<br />

Fred Hutchinson Cancer Research Center, Seattle, Washington 98109 and Howard Hughes Medical<br />

Institute<br />

The C. <strong>elegans</strong> intestine is a simple tube consisting of a monolayer of polarized epithelial cells. During<br />

embryogenesis, cells in the anterior of the intestinal primordium undergo reproducible movements that<br />

create an invariant, asymmetrical "twist" in the intestine. We have analyzed the development of twist to<br />

determine how left-right and anterior-posterior asymmetries are generated within the intestinal<br />

primordium. The twist requires the LIN-12/Notch-like signaling pathway of C. <strong>elegans</strong>. All cells within the<br />

intestinal primordium initially express LIN-12, a receptor related to Notch. However, only cells in the left<br />

half of the intestinal primordium contact external, non-intestinal cells that express LAG-2, a ligand related<br />

to Delta. LIN-12 and LAG-2-mediated interactions result in the left primordial cells expressing lower levels<br />

of LIN-12 than the right primordial cells. We propose that this asymmetrical pattern of LIN-12 expression<br />

is the basis for asymmetry in later cell-cell interactions within the intestinal primordium that lead directly to<br />

intestinal twist. Like the interactions that initially establish LIN-12 asymmetry, the later interactions are<br />

mediated by LIN-12. However, the later interactions involve a different Delta-related ligand, called APX-1.<br />

We show that the anterior-posterior asymmetry in intestinal twist involves the kinase LIT-1, which is part<br />

of a signaling pathway in early embryogenesis that generates anterior-posterior differences between<br />

sister cells.<br />

70


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

THE PHO-1 GENE AND THREE KINDS OF GUT POLARITY<br />

Tetsunari Fukushige, James D. McGhee<br />

Department of Biochemistry and Molecular Biology, University of Calgary, 3330 Hospital Dr. NW, Calgary,<br />

Alberta, CANADA T2N 4N1<br />

Many years ago (Beh et al., 1991), we described an acid phosphatase activity (pho-1) that was expressed<br />

along the intestinal brush border in every gut cell except int-1 and int-2, beginning at the three fold stage<br />

of embryogenesis. Thus, compared to early gut differentiation markers such as the ges-1 esterase, pho-1<br />

should allow us to investigate three different polarities within the gut: anterior-posterior, apical-basal, and<br />

temporal. The pho-1 gene was cloned and its expression pattern is now being analyzed. First of all,<br />

fusions of the pho-1 promoter to GFP reporters reflect the endogenous expression pattern and do not<br />

express in int-1 and int-2; in other words, the anterior-posterior patterning is at the level of the pho-1<br />

promoter. We have previously shown that A/P patterned expression of a modified ges-1 reporter depends<br />

on zygotic pop-1 activity (Schroeder and McGhee, 1998). This patterning within the E-lineage is<br />

gut-autonomous and occurs after the Wnt-dependent P2-EMS contact of the four cell stage. We are now<br />

investigating whether pho-1 responds to the same A/P patterning cues but in an opposite manner, i.e. we<br />

would expect that high level of pop-1 within the gut cell nuclei would repress pho-1 expression, whereas<br />

high level of pop-1 in gut nuclei activates the modified ges-1 reporter. An experiment to test this model is<br />

to inject lit-1 doublestranded RNA into a pop-1(zu189) mother. The gut is still formed (zu189 results in low<br />

maternal pop-1 and hence a double E cell) but loss of lit-1 activity should prevent downregulation of<br />

zygotic pop-1 within the gut. Our prediction is that pho-1 should be repressed throughout the gut because<br />

of high pop-1 and (in preliminary experiments) this is indeed what we see. The availability of a<br />

temperature sensitive allele of lit-1 should allow us to investigate the gut patterning events in much finer<br />

detail. To investigate apical-basal asymmetry, we are attempting to find a GFP construct that will reflect<br />

the endogenous pho-1 distribution, in order to allow for genetic screens of polarity defects. At the<br />

moment, all we know is that mutations in lin-2, lin-7 and lin-10 (shown by Stuart Kim’s lab to be involved<br />

in epithelial polarity) have no apparent effect on the normal distribution of pho-1 activity. Finally, we are<br />

investigating why pho-1 is expressed 2-3 E-cell divisions later than ges-1. There are GATA sites in the<br />

promoter, that might be a target for elt-2. However, ectopic expression of either elt-2 or end-1 does not<br />

activate ectopic expression of either endogenous pho-1 gene or pho-1 reporter gene, suggesting other<br />

factors or events may be required. We are analyzing the pho-1 promoter to identify the site of action of<br />

these timing factors or timing events.<br />

71


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

A ROLE FOR DISHEVELLED IN ASYMMETRIC CELL<br />

DIVISION.<br />

Nancy Hawkins 1 , Gregory Ellis 2 , Bruce Bowerman 2 , Gian Garriga 1<br />

1Dept. of Mol. and Cell Biology, University of California, Berkeley, CA 94720<br />

2Institute of Mol. Biology, University of Oregon, Eugene, OR 97403<br />

Both intrinsic and extrinsic mechanisms can polarize asymmetrically dividing cells. Asymmetrically<br />

distributed intracellular molecules are known to segregate fate to daughter cells during mitosis. Wnt<br />

signaling also polarizes asymmetrically dividing cells by an unknown mechanism. We propose that in C.<br />

<strong>elegans</strong>, Wnt signaling may segregate intracellular molecules that distribute developmental potential.<br />

In the lineage that generates the HSN and PHB neurons, an HSN/PHB neuroblast divides asymmetrically<br />

to generate an anterior daughter cell that dies and a posterior daughter cell, the HSN/PHB precursor. This<br />

precursor then divides to produce the HSN and PHB neurons. In ham-1 mutants, the HSN/PHB<br />

neuroblast frequently divides symmetrically producing two HSN/PHB precursors. ham-1 encodes a novel<br />

protein that is expressed in a subset of cells during embryogenesis, and in mitotic cells, is often restricted<br />

to one side in a crescent shaped pattern. In the HSN/PHB neuroblast, HAM-1 is asymmetrically localized<br />

and is inherited by the HSN/PHB precursor.<br />

To determine if Wnt signaling is also involved in asymmetric division of the HSN/PHB neuroblast, we<br />

focused on the three C. <strong>elegans</strong> dishevelled (dsh) homologs. Upon Wnt stimulation, Dsh becomes<br />

hyperphosphorylated and recruited to the cell membrane. Several lines of evidence indicate that dsh-2<br />

(C27A2.6) is necessary for asymmetric division of the HSN/PHB neuroblast. First, both RNAi with dsh-2,<br />

as well as a deletion allele, result in a Ham-1 phenotype in the HSN/PHB lineage. Second,<br />

overexpression of dsh-2 produces weak defects in asymmetric cell division. Finally, DSH-2 appears to be<br />

primarily membrane associated and is expressed from approximately the 4-6 cell stage until the 1 1/2 fold<br />

stage. In cells expressing HAM-1, the two proteins co-localize and when HAM-1 is asymmetric, DSH-2<br />

localization also appears to be asymmetric. We are performing co-immunoprecipitation and in vitro<br />

binding experiments to determine if HAM-1 and DSH-2 physically interact.<br />

72


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

RHO-1, A TARGET OF THE EXCHANGE FACTOR UNC-73, IS<br />

REQUIRED FOR CELL MIGRATIONS DURING C. ELEGANS<br />

DEVELOPMENT<br />

Andrew G. Spencer, Christian J. Malone, Satoshi Orita, Min Han<br />

Howard Hughes Medical Institute, Department of Molecular, Cellular, and Developmental Biology,<br />

University of Colorado, Boulder, CO, 80309-0347<br />

Members of the rho family of small GTPases have been implicated and characterized in a variety of<br />

cytoskeletal regulatory events. However, the spatial and temporal regulation of the activities of such<br />

proteins in vivo is not well understood and represents an important aspect of the cell movements and<br />

morphogenesis required during animal development. By expressing dominant-negative and<br />

dominant-active versions of the C. <strong>elegans</strong> rhoA homologue rho-1 from a tissue specific promoter<br />

(col-10), we establish a role for rho-1 during P cell migration to the ventral cord during the first larval<br />

stage. Expression of rho-1 (dn) causes a strong P cell migration defect. This effect appears to be<br />

specifically the result of rho-1 inhibition since expression of the C3 exoenzyme causes a similar P cell<br />

phenotype. Genetic and biochemical analyses of unc-73, a GDP/GTP exchange factor, indicate that<br />

unc-73 acts as an exchange factor for rho-1 during P cell migration in vivo. Another rho family GTPase,<br />

mig-2, is not required for P cell migrations but may contribute to a common pathway with rho-1 in some<br />

manner. This is based on observations that (a) mig-2 null alleles exacerbate a weak P cell migration<br />

defect in unc-73 mutants (Zipkin, et al., 1997), which is partially rescued by rho-1 (gf) (Zipkin, Kindt, and<br />

Keynon, 1997 1 ), and (b) mig-2 gain-of-function alleles cause a P cell migration defect, presumably by<br />

interfering with members of the rho-1 pathway. In order to identify molecules acting downstream of rho-1<br />

during P cell migration, we have studied multiple alleles of let-502. Putative null alleles of let-502 and<br />

let-502 RNAi result in a weak P cell migration defect in worms surviving to L2. A temperature-sensitive<br />

hypomorphic allele causes a similar P cell phenotype. We propose that unc-73 and rho-1 act together,<br />

partially through let-502, to direct P cell migration in C. <strong>elegans</strong>.<br />

1 Zipkin, Ilan D., Rachel M. Kindt, and Cynthia J. Kenyon Cell 1997 90: 883-894.<br />

73


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

PTP-1, A LAR-LIKE RECEPTOR PROTEIN TYROSINE<br />

PHOSPHATASE, MAY ACT IN PARALLEL WITH C. ELEGANS<br />

EPH SIGNALING TO DIRECT MORPHOGENESIS<br />

Robert J. Harrington 1 , Michael Gutch 2 , Michael Hengartner 2 ,<br />

Nicholas Tonks 2 , Andrew Chisholm 1<br />

1Dept. of Biology, University of California, Santa Cruz CA 95064<br />

2Cold Spring Harbor Laboratory, Cold Spring Harbor, New York 11724<br />

Mutations in the C. <strong>elegans</strong> Eph receptor tyrosine kinase, VAB-1, and the ephrins, VAB-2 and MAB-26,<br />

cause defects in neural and epidermal morphogenesis. These defects are incompletely penetrant,<br />

suggesting that other pathways may function in parallel. To identify components of such parallel pathways<br />

we investigated whether vab-1 mutations displayed synthetic lethality with other morphogenetic mutants.<br />

A mutation in a C. <strong>elegans</strong> receptor tyrosine phosphatase, ptp-1(op147), causes mild morphogenetic<br />

defects similar to those of weak vab-1 alleles. We have found that ptp-1(op147) synergizes with vab-1<br />

mutations, in particular, kinase domain mutations. ptp-1(op147) also synergizes to some degree with<br />

mutations in vab-2 and mab-26, suggesting that PTP-1 may act in parallel with Eph signaling to regulate<br />

morphogenesis. We are testing the specificity of this interaction with other morphogenetic mutants that<br />

are not known Eph signaling components.<br />

PTP-1 is most similar to LAR-like receptor protein tyrosine phosphatases. We have found that the ptp-1<br />

locus encodes two isoforms, PTP-1A and PTP-1B. PTP-1A contains 3 Ig-like domains, 8 fibronectin type<br />

III repeats, and two tandem cytoplasmic phosphatase domains. PTP-1B contains only 5 FNIII repeats and<br />

the two phosphatase domains. Both isoforms are expressed in neurons.<br />

We have analyzed the phenotype of ptp-1 single mutants and vab-1 ptp-1 double mutants at the 4D<br />

microscopy level. We have found that ptp-1 mutants have gastrulation and ventral closure defects similar<br />

to Eph signaling mutants. vab-1 ptp-1 double mutants show the same defects, only at a higher<br />

occurrence. Our data suggests that PTP-1 and Eph signaling may function in parallel in neurons to direct<br />

morphogenesis.<br />

74


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

GEX-2 AND GEX-3 DEFINE A CONSERVED PROTEIN<br />

COMPLEX REQUIRED FOR TISSUE MORPHOGENESIS AND<br />

CELL MIGRATIONS IN C. ELEGANS<br />

Martha Soto 1 , Katsuhisa Kasuya 2 , Hiroshi Qadota 2 , Kozo Kaibuchi 2 ,<br />

Craig C. Mello 1<br />

1University of Massachusetts Medical Center, Worcester, MA 01605, USA<br />

2Division of Signal Transduction, Nara Institute of Science and Technology, 8916-5, Takayama, Ikoma,<br />

Nara, 630-0101, Japan<br />

Morphogenesis during C. <strong>elegans</strong> embryogenesis requires coordinated cell movements. We report the<br />

identification of two genes, gex-2 and gex-3, necessary for tissue morphogenesis and cell migrations<br />

during C. <strong>elegans</strong> embryogenesis and for cell migrations in adult worms. Inactivation of gex-2 or gex-3<br />

with the RNAi technique or a loss of function mutation in gex-3 caused a maternal effect embryonic<br />

lethality with failure of dorsal intercalation and of ventral migrations of the epidermis. The gex-3 mutant<br />

larva showed zygotic phenotypes including an egg laying defect (Egl) and defects in distal tip cell<br />

migration.<br />

gex-2 and gex-3 encode proteins with no known motifs that are conserved from nematodes to humans,<br />

suggesting conserved essential functions for GEX-2 and GEX-3. The mammalian homolog of GEX-2,<br />

Sra-1 was identified as a specific interactor of GTP-bound Rac. GEX-3 has a human homolog;<br />

HEM2/NAP1/NCKAP1 is implicated in Rac signaling and is reported to bind the SH2-SH3 adaptor protein<br />

NCK/DOCK. The GEX-3 fly homolog, HEM2/KETTE affects axonal cell migrations, actin distribution and<br />

interacts genetically with NCK/DOCK.<br />

Immunostaining with anti-GEX-2 and GEX-3 antibodies showed that GEX-2 and GEX-3 colocalize to<br />

cell-cell contact sites of all embryonic cells. GEX-2 interacts with GEX-3 in the two hybrid system, and<br />

GEX-2 and GEX-3 proteins can immunoprecipitate each other, suggesting GEX-2 and GEX-3 form a<br />

complex. Our findings suggest that a novel protein complex including GEX-2 and GEX-3 localizes to cell<br />

boundaries to regulate cell migrations and cell shape changes required for embryonic tissue<br />

morphogenesis and for adult cell migrations. This complex may reorganize the actin cytoskeleton to<br />

control cell shape changes necessary for dorsal intercalation, migration of the hypodermal cells in the<br />

embryo and migration of the distal tip cells post-embryonically.<br />

75


PHARYNGEAL EXTENSION: THE SHORT AND THE LONG OF<br />

IT<br />

MF Portereiko, SE Mango<br />

<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

Huntsman Cancer Institute Center for Children and Department of Oncological, Sciences, University of<br />

Utah, Salt Lake City, UT 84112<br />

Pharyngeal morphogenesis initiates during mid-embryogenesis, when 78 of the 80 pharyngeal cells have<br />

been born, and the embryo has begun to elongate. At this time, the pharyngeal precursors form a<br />

compact ball that is attached to the nascent midgut and surrounded by a basement membrane that<br />

encloses the entire primordium except for a gap at the anterior. Over the next 80 minutes, the pharyngeal<br />

precursors alter their morphology and position to form a linear tube that is linked to the buccal cavity<br />

anteriorly and the midgut posteriorly. We call this process pharyngeal extension. We have used<br />

time-lapse videomicroscopy and GFP reporter constructs to follow the behavior of the pharyngeal<br />

precursors during pharyngeal extension. Our studies demonstrate that pharyngeal extension can be<br />

loosely divided into three stages i) reorganization of cellular polarity within pharyngeal cells (specifically<br />

the pharyngeal epithelial precursors), ii) formation of an epithelium that mechanically couples the<br />

pharyngeal cells to arcade cells in the nascent buccal cavity, and iii) an apparent contraction that shifts<br />

the buccal cavity posteriorly and the pharynx anteriorly. We are considering a ’purse string’ model to<br />

explain the cellular behaviors we see during contraction. Our findings suggest that pharyngeal extension<br />

is the result of ’pulling’ by anterior pharyngeal cells rather than ’pushing’ by posterior pharyngeal cells. To<br />

test this idea, we eliminated the posterior pharynx via genetic and physical means and demonstrated that<br />

pharyngeal extension still occurs normally. Our model also predicts that tension between cells of the<br />

pharynx and buccal cavity is required for the movement we observe during contraction. We tested this<br />

idea by destroying the arcade cells. As expected, this manipulation blocked the anterior-directed<br />

movement of the pharyngeal cells and reduced the posterior-directed movement of the buccal cavity. Our<br />

current goal is to identify molecules required for pharyngeal extension using forward and reverse genetic<br />

approaches.<br />

76


A VAB-8/UNC-51/UNC-14 COMPLEX MEDIATES AXON<br />

OUTGROWTH<br />

Tina Lai, Gian Garriga<br />

Molecular and Cell Biology, University of California, Berkeley, CA 94720<br />

In C. <strong>elegans</strong> most posteriorly directed cell and growth cone migrations require vab-8, a gene that<br />

encodes at least two protein products known as VAB-8L and VAB-8S. VAB-8L is a 1066 amino acid<br />

protein that contains an N-terminal kinesin-like motor domain and functions in vab-8-dependent growth<br />

cone migrations. VAB-8S is colinear with the C-terminal half of VAB-8L, and lacks the kinesin-like motor<br />

domain. VAB-8S is necessary for certain vab-8-dependent cell migrations.<br />

To identify VAB-8-interacting proteins, we conducted a yeast two-hybrid screen using full length VAB-8L<br />

as bait. One protein identified was UNC-51, a serine/threonine kinase required for proper axon outgrowth<br />

(Ogura et al., 1994). In yeast and in vitro, a region between the kinesin and shared domains of VAB-8L<br />

interacts with the C-terminal half of UNC-51, which lacks the kinase domain.<br />

Ogura et al. (1997) have shown that the C-terminal half of UNC-51 also interacts with UNC-14. Using<br />

yeast two-hybrid, we found that a portion of VAB-8L interacts with UNC-14, but full length VAB-8L does<br />

not. These observations suggest that UNC-14 interaction sites are masked in the full length VAB-8<br />

protein.<br />

Several observations suggest that VAB-8, UNC-14 and UNC-51 also interact in C. <strong>elegans</strong>. First, vab-8,<br />

unc-14 and unc-51 mutants display axon outgrowth defects. Second, all three genes are expressed in<br />

neurons that require vab-8 function. Finally, misexpression of the UNC-51-binding domain of VAB-8L<br />

under control of the ceh-23 promoter results in highly penetrant CAN cell and growth cone migration<br />

defects, presumably by interfering with UNC-51 binding with wildtype VAB-8L. We are in the process of<br />

determining whether this misexpression phenotype could be suppressed by simultaneous misexpression<br />

of unc-51.<br />

Our results and those of Ogura et al. (1997) argue that VAB-8, UNC-14 and UNC-51 form a complex that<br />

functions in the outgrowth of certain axons. We have observed that overexpression of vab-8 can suppress<br />

axon outgrowth defects of an unc-51 mutant, suggesting a positive regulatory relationship between the<br />

two genes. Two possibilities we are testing are whether VAB-8L is an UNC-51 target and whether VAB-8L<br />

can regulate UNC-51 kinase activity.<br />

Ogura et al. (1994) Genes & Dev. 8: 2389-2400.<br />

Ogura et al. (1997) Genes & Dev. 11: 1801-1811.<br />

<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

77


CYTOSKELETAL SIGNALLING IN RESPONSE TO THE UNC-6<br />

AXONAL ATTRACTANT<br />

Zemer Gitai 1 , Erik Lundquist 2 , Marc Tessier-Lavigne 1 , Cori<br />

Bargmann 1<br />

1HHMI, UCSF, San Francisco, CA 94143-0452<br />

2Univeristy of Kansas, Lawrence, KS 66045<br />

<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

The netrin UNC-6 is involved in attraction and repulsion of circumferentially migrating cells and axons in<br />

C. <strong>elegans</strong>. Attraction to netrin/UNC-6 is mediated by the UNC-40 receptor, but the signalling events that<br />

occur upon UNC-40 activation remain unknown.<br />

In wild type animals UNC-6 is expressed ventrally, and several axons are directed ventrally via the<br />

UNC-40 receptor. This ventral migration is disrupted in unc-6 and unc-40 mutants. To probe the<br />

mechanism of UNC-40 activation by UNC-6, we deleted the extracellular domain of an unc-40 transgene<br />

and added a myristylation signal to the cytoplasmic domain, generating what we refer to as<br />

MYR::UNC-40.<br />

When expressed under the control of the mec-7 promoter, MYR::UNC-40 produced a variety of novel<br />

phenotypes in all mec-7-expressing touch cells. These phenotypes included exuberant axon outgrowth<br />

and branching, defective axon guidance, and enlarged and deformed cell bodies. The effects are<br />

independent of the endogenous UNC-6 ligand and UNC-40 receptor. These results lead us to believe that<br />

the MYR::UNC-40 represents a ligand-independent, constitutively active form of UNC-40.<br />

The generation of a gain-of-function UNC-40 molecule allowed us to search for downstream components<br />

of the UNC-40 signalling pathway by looking for mutations that suppressed the MYR::UNC-40-induced<br />

phenotype. One such mutation was in unc-115, a gene encoding a molecule with three LIM domains and<br />

an actin-binding villin headpiece. Both unc-40 and unc-115 mutants alone had defects in the ventral<br />

migration of the AVM axon. In the unc-40; unc-115 double mutant, the AVM axon guidance defect was<br />

not enhanced, again suggesting that UNC-115 might function in the UNC-40 pathway. The agreement of<br />

the loss-of-function double mutant phenotype with the MYR::UNC-40 suppression further supports the<br />

idea that MYR::UNC-40 represents a gain-of-function of UNC-40.<br />

Biochemical and cell biological experiments are currently underway to determine whether UNC-40 and<br />

UNC-115 physically associate. Structure/function experiments are also being conducted to determine the<br />

portion of the UNC-40 cytoplasmic domain responsible for signalling to UNC-115. Finally, we are<br />

continuing to search for new components of the UNC-40 signalling pathway by looking for additional<br />

suppressors of MYR::UNC-40.<br />

78


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

IDENTIFYING GENES INVOLVED IN AXONAL BRANCHING IN<br />

C. ELEGANS<br />

Joe C. Hao, Marc Tessier-Lavigne, Cornelia I. Bargmann<br />

Department of Anatomy and HHMI, UCSF, San Francisco, CA 94143-0452, USA<br />

In both the developing and mature nervous system, neurons can innervate multiple targets by sprouting<br />

secondary axon collaterals, or branches, from a primary axon shaft. Although positive and negative<br />

regulators of primary axonal growth cone guidance have been identified, little is known about the<br />

molecular mechanisms mediating axonal branching. In order to elucidate genes that may control axonal<br />

branching, we have performed a screen to identify mutants defective in branch formation by using a<br />

reporter expressed in the chemosensory neuron ADL. Unlike other amphid neurons, whose axons enter<br />

the nerve ring via the amphid commissures, the axon of ADL projects into the nerve ring laterally, where it<br />

then branches into both a dorsal and a ventral process.<br />

As a first step, we have examined the effects of known axon guidance genes on ADL axonal morphology.<br />

Interestingly, the unc-6/unc-40 pathway may mediate ADL ventral branch formation. In unc-6 or unc-40<br />

mutants, the majority of ADL axons lack the ventral axonal process. sax-3 mutants also exhibit defects in<br />

branch formation, ranging from the loss of both branches to the absence of either process.<br />

From our screen, we have isolated 36 mutants defective in ADL axonal branching that represent at least<br />

17 complementation groups. These fall into three classes: i. mutants defective in ventral branching; ii.<br />

mutants defective in dorsal branching; and iii. mutants with multiple branching defects. We have named<br />

these the branching of axon defective, or bad mutants. In addition to known axon outgrowth and guidance<br />

mutants, we have also isolated several novel mutants defective in axon guidance and/or branching.<br />

We are currently attempting to clone and further characterize bad-1, a mutant with identical ADL<br />

branching phenotypes as those observed in unc-6 and unc-40 mutants. These three mutants appear to<br />

act in the same genetic pathway and also affect AVM axon guidance and PLM axon branching. However,<br />

bad-1 animals are not uncoordinated and display normal motor neuron axon guidance, suggesting that<br />

unc-5-dependent repulsion is preserved in these mutants. These results suggest a role for bad-1 in a<br />

subset of unc-6/unc-40-mediated processes. We are also mapping and characterizing several other bad<br />

mutants.<br />

79


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

UNC-119 SUPPRESSES SUPERNUMERARY BRANCHING IN<br />

C. ELEGANS<br />

Karla Knobel, Warren Davis, Michael Bastiani, Erik Jorgensen<br />

Biology Dept., University of Utah, Salt Lake City, UT 84112<br />

We previously characterized the behavior of migrating GABA motorneuron growth cones in C. <strong>elegans</strong><br />

larvae using time-lapse confocal microscopy. VD growth cones exhibit specific behaviors that result from<br />

the interaction between growth cones and different cellular substrates encountered during migration . The<br />

most dramatic behavior exhibited by migrating VD motorneuron growth cones in vivo is their collapse at,<br />

and subsequent extension beyond the dorsal body wall muscle. To identify molecules required for specific<br />

growth cone behaviors such as collapse we characterized the motorneuron axon outgrowth phenotype of<br />

existing uncoordinated mutants. One mutant, unc-119(ed3), possessed abnormally branched axons, most<br />

of which were located at the body wall muscle. This suggested that unc-119(ed3) growth cones extend<br />

supernumerary branches between the muscle and epidermis. Analysis of the UNC-119 expression<br />

pattern indicated that UNC-119 is expressed in neurons. Expression of UNC-119 under heterologous<br />

promoters demonstrated that it functions cell-autonomously to suppress axon branching. However,<br />

time-lapse analysis of motor neuron development and axon outgrowth indicated that UNC-119 does not<br />

function during growth cone migration. Comparison of axon outgrowth patterns 1 hour and 48 hours after<br />

the completion of growth cone migration indicated that axon branching occurs after axon outgrowth is<br />

completed in unc-119(ed3) mutants. Time-lapse analysis of established axons demonstrated that<br />

secondary growth cones sprout from motorneuron axons and cell bodies. These secondary growth cones<br />

extend extra branches to the dorsal nerve cord. Concurrently axonal extensions along the dorsal nerve<br />

cord are retracted. As a result, unc-119(ed3) adults have many commissural branches as well as large<br />

gaps in the dorsal nerve cord. Synapse development occurs after outgrowth is completed. To determine if<br />

synaptogenesis is affected we characterized synapses in unc-119(ed3) mutants. Synapses form and are<br />

morphologically normal as determined by electron microscopy. However, synapses are localized to<br />

inappropriate locations of the axon, including branches and dendritic regions. These data suggest that<br />

UNC-119 is a member of a new class of cell intrinsic factors that preserve the architecture of the nervous<br />

system and regulate synapse localization.<br />

80


UNC-119 AND AXON OUTGROWTH: TOWARD A<br />

MECHANISM<br />

Wayne Materi, Dave Pilgrim<br />

<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

Department of Biological Sciences, University of Alberta, Edmonton, Alberta, T6G 2E9<br />

UNC-119 is crucial for the correct development of the nematode nervous system. <strong>Worm</strong>s mutant for the<br />

neuronally-expressed unc-119 gene show locomotory and sensory abnormalities that are explained by<br />

structural defects resulting from aberrant nervous system development.<br />

Direct examination of nervous system structure using GFP reporters shows that unc-119 mutants have a<br />

variety of neurite outgrowth defects. The axons of chemosensory amphids exhibit highly penetrant ventral<br />

elongation defects within the nerve ring while the axons of anterior mechanosensory neurons have similar<br />

elongation defects in the dorsal direction. These defects suggest a disruption of normal synaptic patterns<br />

that is sufficient to explain both the inability of unc-119 mutants to form dauer larvae and their inability to<br />

respond to touch. The ventral nerve cord is moderately to severely defasciculated in 50% of mutant<br />

worms, supporting a general role for UNC-119 in axon guidance.<br />

Defects in the anteroposterior position of dorsolateral turns suggests an aberrant response to choice<br />

points. However, there are no circumferential pathfinding defects in motor neurons, similar to those seen<br />

in unc-6 , unc-5 or unc-40 mutants. Indeed, although some cell bodies are misplaced in unc-119 mutants,<br />

their axons are often correctly targeted, implying that turning at choice points is somewhat independent of<br />

other pathfinding mechanisms.<br />

Expression and rescue experiments with functional UNC-119::GFP constructs suggest that it acts cell<br />

autonomously. Although sub-cellular localization of the rat homologue, RRG4, has been demonstrated<br />

within photoreceptor ribbon synapses, antibody studies in the worm are still in progress. Yeast two-hybrid<br />

experiments suggest that UNC-119 is involved in a novel neurite development signaling pathway that<br />

mediates interaction between basement membrane collagen’s and an uncharacterized zinc-finger protein.<br />

We have used PCR-based reverse genetics to isolate strains bearing deletions in the latter of these and<br />

have observed a lack of exploratory behavior but otherwise normal locomotion and response to touch<br />

stimuli.<br />

81


THREE DISTINCT FUNCTIONS OF BETA-SPECTRIN (UNC-70)<br />

Marc Hammarlund, Warren S. Davis, Erik M. Jorgensen<br />

University of Utah, SLC UT 84112<br />

We cloned unc-70 and found that it encodes the C. <strong>elegans</strong> homolog of beta-spectrin, an essential<br />

component of the membrane skeleton.1 The membrane skeleton is a protein mesh that binds to the<br />

plasma membrane and that interacts with a large number of membrane and cytosolic proteins. It has<br />

been proposed that the membrane skeleton functions to support the plasma membrane or to generate<br />

cell polarity. However, we found that null mutations in unc-70 do not result in general membrane or<br />

polarity defects. unc-70 null animals have a severe paralyzed, dumpy phenotype. By analyzing this<br />

phenotype we have identified at least three specific processes that are perturbed by loss of beta-spectrin.<br />

First, we found defects in axonal morphology, suggesting that the membrane skeleton plays an essential<br />

role in neuronal development. We suspect that loss of beta-spectrin may specifically affect growth cone<br />

motility and are conducting time-lapse studies of unc-70 growth cones to test this hypothesis. Second,<br />

we found that loss of beta-spectrin causes defects in the myofilament attachment structures of body wall<br />

muscle (dense bodies and M lines). We are using a panel of antibodies against components of these<br />

structures to understand how the membrane skeleton functions in muscle. Finally, the membrane<br />

skeleton appears to function in synaptic transmission. We found that rare dominant alleles of unc-70 do<br />

not exhibit the null phenotypes described above; rather, they have abnormally high levels of<br />

neurotransmission. Preliminary evidence suggests that synaptic vesicles are mislocalized within the<br />

nerve terminal of these mutants. We are analyzing the synaptic ultrastructure of these mutants, and<br />

using electrophysiology to relate structural defects to possible functional roles for beta-spectrin in nerve<br />

terminals.<br />

1. Hammarlund, M. et al., J. Cell Biol., 149(4), 2000.<br />

<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

82


RPM-1, A CONSERVED NOVEL PROTEIN, REGULATES<br />

PRESYNAPTIC TERMINAL FORMATION<br />

Xun Huang 1 , Mei Zhen 1 , Bruce Bamber 2 , Yishi Jin 1<br />

1Department of Biology, University of California, Santa Cruz, CA 95064<br />

2Department of Biology, University of Utah, Salt Lake City, UT 84112<br />

Presynaptic terminals contain highly specialized subcellular structures to facilitate neurotransmitter<br />

release. We used the synaptic vesicle tagged GFP marker under the unc-25 promoter to visualize the<br />

presynaptic varicosities of the GABAergic DD and VD motor neurons 1 . In wild-type animals GFP<br />

expression is observed as distinct fluorescent puncta, corresponding to individual neuromuscular<br />

junctions along the dorsal and ventral nerve cords. rpm-1 mutations were isolated in a screen for<br />

abnormal morphology of the GFP puncta.<br />

In rpm-1 mutants the total number of these GFP puncta is reduced to various extents depending on the<br />

strength of the mutation, and regions along the dorsal and ventral nerve cords often contained no GFP<br />

puncta. Moreover, the remaining GFP puncta were variable in size and shape. The overall axonal<br />

morphology of the DD and VD neurons appeared normal. At the ultrastructural level, we observed two<br />

defects at GABAergic NMJs in the rpm-1 mutants: 1) "over-developed" synapses that contained more<br />

than two individual electron-dense presynaptic active zones within the same varicosity, 2)<br />

"under-developed" synapses that had few synaptic vesicles and instead were filled with electron-dense<br />

debris-like material. In cholinergic NMJs, we observed the presynaptic terminals with longer active zones.<br />

rpm-1 encodes a large protein similar to Drosophila Highwire (Hiw) and mammalian Pam. All three<br />

proteins have a putative GEF domain and a highly conserved C-terminal with several zinc finger domains.<br />

RPM-1 is localized to the presynaptic region independently of synaptic vesicles. Mosaic analysis indicated<br />

that RPM-1 functions cell-autonomously. Using a temperature sensitive allele of rpm-1, we found that<br />

RPM-1 is required around the time of the formation of presynaptic terminals. Our results suggest that<br />

RPM-1 may regulate the distribution of presynaptic terminals, or function to prevent the formation of<br />

excessive presynaptic structures.<br />

1. Zhen, M and Jin, Y. Nature 1999<br />

<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

83


A C. ELEGANS INOSITOL 5- PHOSPHATASE HOMOLOGUE<br />

INVOLVED IN INOSITOL 1,4,5-TRIPHOSPHATE SIGNALING<br />

AND OVULATION.<br />

Yen Kim Bui, Paul W. Sternberg<br />

<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

Howard Hughes Medical Institute and Division of Biology, California Institute of Technology, Pasadena,<br />

CA, 91125 USA.<br />

Understanding the basis of positive and negative regulation of the inositol signaling pathway may<br />

increase our understanding of the control of diverse biological processes dependent on inositol<br />

1,4,5-triphosphate (IP3). J. McCarter in T. Schedl’s lab demonstrated that mutations in lin-3/EGF or let-23<br />

/Receptor Tyrosine Kinase (RTK) result in a sterile Emo phenotype whereby the oocytes fail to be<br />

ovulated and become trapped in the gonad arm and undergo multiple rounds of DNA synthesis. T.<br />

Clandinin in our lab, showed that either a loss of function mutation in thelfe-2 /IP-3 kinase or gain of<br />

function (gf) in the lfe-1/IP3 Receptor can bypass the requirement for RTK signaling indicating that<br />

ovulation in C. <strong>elegans</strong> is dependent on IP3 signaling. Inositol 5-phosphatase plays a role in<br />

dephosphorylating IP3 to inositol 1,4-phosphate. However the contribution of this step in terminating IP3<br />

signaling in vivo remains unclear. The C. <strong>elegans</strong> genome predicts a putative ortholog of the human Type<br />

I inositol 5-phosphatase (CO9B8.1).<br />

Using a PCR based strategy, we screened for a deletion (D) mutant of the type I C. <strong>elegans</strong> inositol<br />

5-phosphatase. In contrast to the lfe-2 null or lfe-1 (gf), the 5-Ptase D mutant shows a novel ovulation<br />

phenotype whereby two oocytes enter the spermetheca during a single ovulation cylce; thus suggesting a<br />

crucial role for the 5-Ptase in regulating IP3 mediated ovulation. In contrast to the lfe-2 overexpression<br />

phenotype whereby the oocyte remains stuck in the dilated spermetheca, overexpression of the 5-Ptase<br />

has no noticeable affect. We are characterizing the mechanism by which it regulates ovulation by genetic<br />

analysis. Our genetic data support the hypothesis that increased IP3 signaling causes more ovulation,<br />

and that either too much signaling or too little signaling prevents ovulation. Like the lfe-2 null mutant, the<br />

5-Ptase D mutant suppresses the sterile defect of lin-3(n1058). This suggests that lin-3 sterile mutants fail<br />

to generate sufficient IP3 to execute ovulation. Furthermore, the 5-Ptase D mutant synthetically interacts<br />

with others mutants that increase IP3 signaling to produce a sterile Emo phenotype.<br />

84


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

MECHANISMS REGULATING THE TIMING AND SPECIFICITY<br />

OF ANCHOR CELL ATTACHMENT TO THE VULVAL<br />

EPITHELIUM<br />

David R. Sherwood, Paul W. Sternberg<br />

Division of Biology and HHMI, California Institute of Technology, Pasadena CA<br />

We are interested in understanding the basic cellular and molecular mechanisms that guide<br />

morphogenetic processes during development. Towards this goal we are examining the initial steps<br />

involved in the connection of the uterus and vulva during C.<strong>elegans</strong> development. The initial contact<br />

between the developing uterus and the vulva is established by the anchor cell (AC), which crosses the<br />

basement membranes separating both tissues and specifically attaches to the progeny of the P6.p cell<br />

during the mid- to late L3 stage. Following attachment, the AC invades between the inner descendants of<br />

the P6.p. cell and becomes positioned at the apex of the developing vulva. Using mutants lacking vulval<br />

induction, we have found that the descendants of the underlying induced vulval precursor cell, P6.p, send<br />

a signal to trigger the attachment behavior in the AC. We have also discovered that the competence of<br />

the AC to respond to this signal is regulated: AC attachment is either absent or delayed in the<br />

heterochronic mutant lin-28, which causes precocious vulval development. To understand the molecular<br />

mechanisms that regulate AC attachment, we have examined many known mutants, as well as initiated a<br />

genetic screen to search for new mutants with defective attachment behavior. Through these studies we<br />

have found that the evl-5 mutant, originally isolated in a screen for sterile p-vul’s (Seydoux et al., (1993)<br />

Dev. Biol. 157(2): 423-36), has a defect in AC attachment to the vulval epithelium. In evl-5 animals vulval<br />

induction and AC positioning over the P6.p cell is normal; however, AC attachment either does not occur<br />

or is severely delayed. Visualization of AC behaviour in this mutant revealed the extension of cellular<br />

processes from the AC toward the vulva, indicating that the AC is still attracted to the developing vulva.<br />

However, in many cases the AC processes appeared to flatten or broaden out at the basement<br />

membrane of the developing gonad, suggesting that the AC may not be able to cross this basement<br />

membrane. We are currently molecularly cloning evl-5.<br />

85


MUTATIONS IN CYCLIN E REVEAL COORDINATION<br />

BETWEEN CELL-CYCLE CONTROL AND VULVAL<br />

DEVELOPMENT.<br />

David S. Fay, Min Han<br />

<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

Howard Hughes Medical Institute and Department of Molecular, Cellular, and Developmental Biology,<br />

University of Colorado, Boulder CO 80309-0347<br />

In screens for mutations affecting vulval development, we have identified strong loss-of-function<br />

mutations in the C. <strong>elegans</strong> cyclin E gene, cye-1. Mutations in cye-1 lead to the under-proliferation of<br />

many postembryonic blast lineages as well as defects in fertility and gut-cell endoreduplication. In<br />

addition, cye-1 is required maternally, but not zygotically for embryonic development. Our analysis of<br />

vulval development in cye-1 mutants suggests that a timing mechanism may control the onset of vulval<br />

cell terminal differentiation: once induced, these cells appear to differentiate after a set amount of time,<br />

rather than a specific number of division cycles. cye-1 mutants also show an increase in the percentage<br />

of vulval precursor cells (VPCs) that adopt vulval cell fates, indicating that cell-cycle length can play a role<br />

in the proper patterning of vulval cells. By analyzing cul-1 mutants, we further demonstrate that vulval cell<br />

terminal differentiation can be uncoupled from associated changes in vulval cell division planes.<br />

86


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

NOVEL CELL-CELL INTERACTIONS DURING VULVA<br />

DEVELOPMENT IN PRISTIONCHUS PACIFICUS<br />

Benno Jungblut 1,2 , Ralf J Sommer 1,3<br />

1Max-Planck Institut für Entwicklungsbiologie, Abt. Evolutionsbiologie, Spemannstrasse 35, 72076<br />

Tübingen, Germany<br />

2email: benno.jungblut@tuebingen.mpg.de<br />

3email: ralf.sommer@tuebingen.mpg.de<br />

Comparative analysis of vulva development revealed several differences between the two nematode<br />

species <strong>Caenorhabditis</strong> <strong>elegans</strong> and Pristionchus pacificus including changes in cell fates and inductive<br />

processes. For example, seven of 12 ventral epidermal cells in P. pacificus die of apoptosis, whereas<br />

homologous cells in C. <strong>elegans</strong> fuse with the hypodermal syncytium. In addition vulva induction by the<br />

anchor cell is a one-step process in C. <strong>elegans</strong>, but requires an interaction of both anchor cell and the<br />

gonad with the vulval precursor cells in P. pacificus. We have identified novel cell-cell interactions while<br />

carrying out combinatiorial cell ablation studies of the vulval precursor cells. In particular the ventral<br />

epidermal cell P8.p and the mesoblast M contribute to vulval pattern formation. In contrast to the<br />

homologous cell in C. <strong>elegans</strong>, P8.p in P. pacificus is incompetent to respond to inductive signaling from<br />

the gonad. Nonetheless it can be induced to adopt a vulval fate by lateral signaling from a neighboring<br />

vulval precursor cell. Immunofluorescence studies showed that P8.p fuses with the hypodermis a few<br />

hours before the birth of the anchor cell. Using cell ablation studies we show that P8.p provides an<br />

inhibitory signal that limits the developmental competence of P(5, 7).p. This lateral inhibition also requires<br />

the mesoblast M. In Ppa-mab-5 mutants, M is misspecified and provides inductive signaling to the vulval<br />

precursor cells, including P8.p. Therefore P8.p differentiates gonad independently in these mutants.<br />

Taken together, vulva development in P. pacificus displays novel cell-cell interactions involving the<br />

mesoblast M and P8.p. In particular, P8. represents a new ventral epidermal cell fate, which is<br />

characterized by novel interactions and a specific response to gonadal signaling.<br />

87


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

CELLULAR AND GENETIC ANALYSIS OF G Q MEDIATED<br />

SIGNALING PATHWAYS IN C. ELEGANS<br />

C. A. Bastiani, S. Gharib, P.W. Sternberg, M.I. Simon<br />

Division of Biology, California Institue of Technology, Pasadena, CA 91125<br />

egl-30 encodes the C. <strong>elegans</strong> homologue of the mammalian heterotrimeric G protein alpha subunit, Gq.<br />

Reduction-of-function and loss-of-function mutations in egl-30 affect diverse behaviors in C. <strong>elegans</strong>.<br />

These include: viability, egg-laying, pharyngeal pumping, movement, and spicule protraction. In a screen<br />

for suppressors of the reduction-of-function mutation, egl-30(md186), two intragenic suppressor mutations<br />

were recovered. One of the mutations lies in a region in close proximity to residues involved in guanine<br />

ring binding. The other mutation is in a region that might affect receptor interaction, and is not expected to<br />

make contacts with the region that contains the original egl-30(md186) mutation. These mutants<br />

phenotypically resemble worms that overexpress egl-30, and have been used as a tool to characterize<br />

genes that function in a pathway with egl-30. From these studies, it is clear that EGL-30 regulates egg<br />

laying via downstream signaling pathways distinct from downstream pathways so far defined that control<br />

movement (1), viability (2), and spicule protraction (3). A rescuing gfp::egl-30 transgene confirms that<br />

egl-30 is coexpressed in some cells with egl-8 and lfe-1/itr-1/dec-4, two genes that encode signaling<br />

molecules defined as downstream of Gq in mammalian systems, and that appear to be downstream of<br />

egl-30 with respect to movement and viability, respectively (1,2). However, egl-30 is also uniquely<br />

localized and expressed in some cells. To determine the basis for the differences in genetic interactions<br />

with respect to different behaviors, we are develooping a system to characterize cell-type-specific<br />

molecular interactions with EGL-30 in vivo .<br />

1. Lackner MR, Nurrish SJ, Kaplan JM; Neuron 24: 335-346 1999<br />

2. Hajdu-Cronin YM and Sternberg PW, personal communication<br />

3. Garcia LR and Sternberg PW, personal communication<br />

88


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

CALCIUM/CALMODULIN-DEPENDENT PROTEIN KINASE II<br />

REGULATES C. ELEGANS LOCOMOTION IN CONCERT WITH<br />

A G-PROTEIN SIGNALING NETWORK<br />

Merrilee Robatzek, James H. Thomas<br />

Department of Genetics, University of Washington, Seattle, WA 98195 USA<br />

Calcium/calmodulin-dependent protein kinase II (CaMKII) is an important regulator of synaptic strength<br />

based on studies in mollusks, Drosophila, and mouse. Although the phosphorylation of many gene<br />

products has been attributed to CaMKII from in vitro assays, the mechanism of CaMKII activity in vivo has<br />

not been fully defined. We are using a genetic approach to study the in vivo function of the C. <strong>elegans</strong><br />

CaMKII homolog unc-43. UNC-43 is 83% identical in the kinase domain to mammalian CaMKII, indicating<br />

that the targets of this kinase are probably conserved.<br />

unc-43 regulates several behaviors, including locomotion. A kinase-activated allele of unc-43, n498,<br />

causes severe lethargy, as well as body-wall muscle hypercontraction, reduced egg laying, and reduced<br />

defecation. Several of these effects are genetically separable, suggesting that unc-43 regulates<br />

defecation, body-wall muscle tone, and locomotion rate through different effectors. To understand how<br />

unc-43 controls locomotion rate in C. <strong>elegans</strong>, we performed a genetic suppressor screen with<br />

unc-43(n498) to identify genes that act with unc-43 to control locomotion rate. From a screen of 28,000<br />

EMS-mutagenized haploid genomes, we recovered 19 recessive extragenic suppressors.<br />

Complementation tests showed that we had recovered multiple alleles of the genes goa-1, dgk-1, and<br />

eat-16, all involved in the goa-1/egl-30 heterotrimeric G-protein network , , , and alleles of a fourth gene,<br />

eat-11, that probably affects this same pathway. In addition to suppressing the unc-43(n498) lethargy,<br />

mutations in these genes also suppress the unc-43(n498) egg-laying defect. Quantitative analysis of the<br />

suppression indicates that UNC-43 may control locomotion rate and egg-laying activity by regulating this<br />

G-protein signaling network.<br />

89


A NOVEL LATERAL SIGNALING PATHWAY DETERMINES<br />

ASYMMETRIC OLFACTORY NEURON FATES<br />

Alvaro Sagasti, Cori Bargmann<br />

UCSF and HHMI<br />

<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

The AWC olfactory neurons in C. <strong>elegans</strong> are a bilaterally symmetric pair of cells required for sensing<br />

certain attractive volatile odorants. Although bilaterally homologous neurons are usually thought to be<br />

identical, the candidate seven transmembrane olfactory receptor str-2 is expressed asymmetrically in the<br />

AWC neurons. About half of a population of worms expresses str-2 in the left homolog of the AWC pair,<br />

and half expresses it in the right homolog. The AWC cells are able to coordinate their fates by<br />

communicating with each other, probably through their axons which contact each other in the nerve ring.<br />

To study the signaling mechanisms by which this communication is accomplished we conducted a screen<br />

for mutant worms that express a str-2::GFP reporter in both AWC neurons. The screen yielded mutations<br />

in three novel neuronal symmetry genes (nsy-1, nsy-2, and nsy-3) and three known genes that affect<br />

calcium signaling: alleles of the voltage-gated calcium channel subunits unc-2 and unc-36 and an allele of<br />

the calcium/calmodulin-dependent protein kinase II homolog unc-43. This suggests that calcium signals<br />

are an important component of the AWC asymmetry signaling pathway. Genes in a cGMP signaling<br />

pathway necessary for olfaction have the phenotype opposite of the calcium signaling genes. They are<br />

required for any expression of str-2 in AWC, perhaps in an activity-dependent mechanism used for<br />

maintenance of receptor expression. Epistasis analysis is consistent with nsy-1 and nsy-2 acting after<br />

calcium signaling but before cGMP signaling. In contrast, the dominant mutant nsy-3 seems to act after<br />

axon outgrowth but before calcium signaling. We recently cloned the nsy-1 gene and found that it<br />

encodes a protein kinase. We are currently performing experiments to determine where nsy-1 is<br />

expressed, whether it is acting in the AWC cells, and how it is activated by CaMKII. We are also setting<br />

up a system that will permit us to identify easily AWC left/right mosaics, allowing us to determine which<br />

genes in this pathway act permissively and which act instructively<br />

90


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

THE SEARCH FOR DOSAGE COMPENSATION COMPLEX<br />

BINDING SITES ON X CHROMOSOMES<br />

Raymond C. Chan, Tammy F. Wu, Barbara J. Meyer<br />

HHMI and Dept. of Molecular and Cell Biology, University of California, Berkeley, CA 94720-3204<br />

Dosage compensation (DC) equalizes the level of X-linked gene products between XX hermaphrodites<br />

and XO males. A protein complex (DCC), with remarkable similarity to the Xenopus mitotic 13S<br />

condensin complex, is directed by the sex-determination and dosage compensation (SDC) proteins to the<br />

hermaphrodite X chromosomes, where it represses gene expression. A mechanistic link between dosage<br />

compensation and mitotic chromosome condensation is further indicated by the dual roles of many DCC<br />

subunits in mitosis or meiosis. This link raises an intriguing question of how the DCC specifically<br />

recognizes X chromosomes to effect global repression of gene expression. To study X recognition, we<br />

developed a chromatin immunoprecipitation (ChIP) protocol to isolate DNA bound by the DCC. We<br />

initially devised an affinity purification method to precipitate DC proteins using peptide antibodies against<br />

the DCC. The IP not only confirmed the association of DCC subunits, it also provided a means to isolate<br />

large quantities of DC and mitotic complexes for identifying novel co-purifying factors (see Hagstrom et<br />

al.). When combined with in vivo formaldehyde crosslinking, we can also recover DNA in the precipitate,<br />

thus enabling us to identify candidate X recognition sequences.<br />

To validate this ChIP assay, we tested the known interaction between the SDC proteins and the her-1<br />

promoter. Aside from DC, the SDC proteins bind and repress the promoter of her-1 to establish the<br />

female fate in young XX embryos. In the sdc-3(y52Tra) mutant strain, SDC proteins fail to bind the her-1<br />

promoter leading to the masculinization of XX animals. With our current protocol, we detected her-1<br />

promoter sequences by PCR in both SDC-2 and SDC-3 ChIP. Moreover, ChIP performed on extracts<br />

from sdc-3(Tra) embryos revealed drastically reduced levels of her-1 DNA consistent with the loss of<br />

her-1 binding in these mutants.<br />

We are now focused on identifying X sequences bound by the SDC and DCC proteins. We have<br />

examined 180 kb around myo-2, a known dosage compensated gene on X, and we found an<br />

approximately 4-kb region enriched in the SDC and DCC ChIP. Experiments are in progress to further<br />

define this region and to confirm SDC and DCC protein binding in vivo and by additional in vitro methods.<br />

91


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

RECOGNITION AND ASSEMBLY OF SDC PROTEIN<br />

COMPLEXES ONTO SPECIFIC DNA TARGET SITES<br />

Diana Chu 1 , Heather Dawes 1 , Jason Lieb 2 , Annie Kuo 1 , Barbara J.<br />

Meyer 1<br />

1HHMI and Department of Molecular and Cell Biology<br />

2HHMI and Department of Biochemistry, Stanford University Medical Center, Stanford, CA 94305<br />

SDC (Sex and Dosage Compensation) proteins trigger hermaphrodite development by activating dosage<br />

compensation and repressing the male sex determination gene her-1. We have biochemically defined an<br />

SDC protein complex including SDC-1, SDC-2, and SDC-3. This complex localizes to the her-1 promoter,<br />

where it represses transcription at least 20-fold. It also associates along the entire length of the<br />

hermaphrodite X chromosomes, where it recruits other dosage compensation proteins (DC) such as<br />

DPY-26, DPY-27, DPY-28 and MIX-1 to repress X-linked gene expression 2-fold. We are interested in<br />

how the SDC complex can recognize and act differentially at these separate sites.<br />

Despite the fact that DC proteins are not required for transcriptional repression of her-1, we have found<br />

that the SDC proteins recruit these DC proteins to the her-1 promoter. We showed this co-localization in<br />

transgenic animals that express a GFP-LacI fusion protein from extrachromosomal arrays that also<br />

contain both lac operator repeats (lacO) and the her-1 promoter. Analysis of SDC and DC protein<br />

localization to the her-1 promoter arrays in SDC and DC mutant backgrounds has revealed sequential<br />

requirements for recognition and assembly of the SDC and DC proteins onto target sequences.<br />

To define how SDC proteins recognize specific target sites, we mapped regions within the her-1 promoter<br />

that are important for SDC protein recognition using the assay above. To test the functional significance<br />

of the three regions we identified, we mutated them in the context of a full length her-1 rescuing construct.<br />

Stable transgenic XX strains transformed with a wild-type her-1 show little masculinization. In contrast,<br />

stable transgenic XX strains transformed with mutated her-1 show varying degrees of transformation to<br />

the male fate, including induction of male tail structures and decreased fertility. We interpret this<br />

masculinization as an indication of her-1 derepression. Interestingly, sequences in the regions important<br />

to support SDC-2 localization are not homologous to sequences on the X chromosome. Thus, recognition<br />

of the X chromosome and the her-1 promoter appears to be specified by distinct mechanisms.<br />

92


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

THE TBP-LIKE FACTOR CETLF IS REQUIRED TO ACTIVATE<br />

RNA POLYMERASE II TRANSCRIPTION IN C. ELEGANS<br />

EMBRYOS<br />

Linda S. Kaltenbach, Susan E. Mango<br />

Huntsman Cancer Institute and Department of Oncological Sciences, University of Utah, Salt Lake City,<br />

UT, 84112<br />

Modulation of transcription is crucial to establish differences between cells during development. While the<br />

importance of activators and repressors for regulating transcription is well known, recent data suggest<br />

coregulators and general transcription factors may provide an additional source of control. TATA-binding<br />

protein (TBP) is a component of the basal transcription machinery that is ubiquitously expressed in<br />

metazoans and essential for all transcription in yeast. Recently, two variants of TBP have been<br />

discovered: 1) TBP-Related Factor, known only in Drosophila, which may act as a cell-type specific TBP<br />

and 2) TBP-Like Factor (TLF), found in most, if not all, metazoans. Given the central role of TBP, what is<br />

the function of the TBP paralogs?<br />

On the basis of biochemical studies, four models have been proposed for the role of TLF in RNA<br />

polymerase II (pol II) transcription: i) TLF and TBP could function redundantly, ii) TLF could antagonize<br />

TBP, iii) TLF could be a tissue-specific TBP or iv) TLF and TBP could have unique activities. To<br />

distinguish between these models, we examined the function of C. <strong>elegans</strong> TLF (CeTLF) in vivo.<br />

We have found that CeTLF has an essential role to activate pol II transcription in early embryos. Loss of<br />

CeTLF activity by RNAi results in embryonic arrest at the 70-200 cell stage with no evidence of<br />

differentiation. This phenotype and global expression of CeTLF demonstrate that CeTLF is not a<br />

tissue-specific TBP. Embryos lacking CeTLF fail to gastrulate, a phenotype that resembles loss of ama-1,<br />

which encodes the large subunit of pol II. These embryos also express a marker of elongating pol II at a<br />

reduced level, suggesting that CeTLF is required for some but not all pol II transcription. To test this idea,<br />

we have examined whether CeTLF is necessary to express individual genes normally transcribed in the<br />

gastrula-stage embryo and find that CeTLF is required to activate two of four surveyed genes. Preliminary<br />

experiments indicate that CeTLF physically associates with at least one of these genes in vivo. These<br />

results suggest that CeTLF performs a unique function to activate pol II transcription at some promoters,<br />

and that this activity is distinct from that of CeTBP.<br />

93


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

THE INTRACELLULAR DOMAIN OF THE FEMINISING<br />

RECEPTOR TRA-2A INTERACTS DIRECTLY WITH THE<br />

TRANSCRIPTION FACTOR TRA-1A<br />

David H. Lum 1 , P. Kuwabara 2 , D. Zarkower 3 , A.M. Spence 1<br />

1Dept. of Molecular and Medical Genetics, University of Toronto, ON, M5S 1A8, CANADA.<br />

2MRC Laboratory of Molecular Biology, Hills Road, Cambridge , CB2 2QH.<br />

3Institute of Human Genetics, Minneapolis, MN 55455, U.S.A.<br />

In XX animals the tra-2 gene negatively regulates the activity of the three fem genes. The absence of fem<br />

activity in XX animals frees tra-1, the terminal regulator of somatic sex determination, to promote female<br />

development. The tra-2 gene encodes two proteins TRA-2A and TRA-2B. The larger of the two proteins,<br />

TRA-2A, is a predicted transmembrane protein with a large C-terminal intracellular domain. TRA-2B is<br />

expressed in the hermaphrodite germline and is predicted to be a soluble protein consisting of just the<br />

C-terminal domain of TRA-2A.<br />

The intracellular domain of TRA-2A negatively regulates the FEMs through a direct interaction with<br />

FEM-3. Overexpression of the C-terminal domain of TRA-2A (TRA-2Ac) is sufficient for FEM negative<br />

regulation and the transformation of XO animals into females. To investigate the mechanism of TRA-2Ac<br />

feminizing activity we overexpressed various TRA-2Ac fragments. Surprisingly, a C-terminal fragment of<br />

TRA-2Ac (TRA-2AcD 5), incapable of interacting with FEM-3 in the yeast 2-hybrid system, maintained<br />

weak feminizing activity. Both yeast 2-hybrid and biochemical data demonstrate that TRA-2AcD 5<br />

physically interacts with TRA-1A. tra-2(mx) mutations affect tra-2 activity in both the soma and germline.<br />

Three mx mutations all disrupted the TRA-2Ac/TRA-1A interaction and reduce or eliminate the feminising<br />

activity of TRA-2AcD 5 in overexpression assays.<br />

Our surprising results suggest that in the soma TRA-2A plays a role in regulating TRA-1A through a direct<br />

interaction. TRA-1A has been demonstrated to directly regulate transcription of several genes and is<br />

predicted to be nuclear localized. We observe strong nuclear localization of both GFP::TRA-1A and<br />

Myc::TRA-1A in transgenic animals. Consistent with a nuclear role for TRA-2A regulation of TRA-1A, we<br />

observe nuclear localization of a highly active GFP::TRA-2Ac fusion protein. Our data raise the intriguing<br />

possibility that in the soma TRA-2A is cleaved to allow the intracellular domain to enter the nucleus and<br />

interact with TRA-1A.<br />

94


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

CHW-1 ENCODES A NOVEL PROTEIN THAT INTERACTS<br />

WITH PHA-4<br />

Michael Horner, Linda Kaltenbach, Susan Mango<br />

Huntsman Cancer Institute and Department of Oncological Sciences, University of Utah, Salt Lake City,<br />

Utah 84112<br />

The winged-helix transcription factor PHA-4 specifies organ identity during pharynx and rectal<br />

development. Embryos lacking pha-4 activity produce no pharyngeal or rectal cells while embryos<br />

expressing ectopic pha-4 generate extra pharyngeal and rectal cells at the expense of other cell types.<br />

PHA-4 is also expressed in the gonad, but its function in this organ is unknown.<br />

Homologs of pha-4 are found in most if not all metazoans, where they also regulate gut development.<br />

Proteins belonging to this family are predicted to be transcription factors, with a well-characterized DNA<br />

binding domain. However, little is known about how these proteins regulate transcription to produce such<br />

profound effects on development. To begin to address this issue, we have initiated an analysis of PHA-4<br />

protein and PHA-4 cofactors. Our characterization of five pha-4 alleles demonstrates that the amino<br />

terminus is critical for PHA-4 function. Surprisingly, the carboxyl terminus, which is conserved with<br />

vertebrate HNF-3 proteins and Ce-DISTALLESS, is dispensable.<br />

Since transcription factors often function by recruiting other proteins to target genes, we used a yeast<br />

2-hybrid screen to identify proteins that bind the PHA-4 amino terminus. We isolated six potential<br />

cofactors from a screen of >1x106 clones and have chosen one of these proteins for further study (the<br />

chw genes for Components that Heterodimerize with a Winged helix protein). We have confirmed that<br />

CHW-1 can bind PHA-4 in vitro, suggesting these proteins contact each other directly.<br />

PHA-4 and CHW-1 may function together to regulate expression of a subset of PHA-4 target genes.<br />

CHW-1 is a novel, glutamine-rich protein that is expressed in the intestine, rectum and pm6 pharyngeal<br />

muscles, as is PHA-4. In addition, CHW-1::GFP is expressed in body wall muscles where PHA-4 is not<br />

found. We have used RNAi to show that loss of chw-1 activity affects cells that normally express PHA-4.<br />

In many chw-1(RNAi) animals, the pharyngeal grinder is deformed, larvae appear starved, and gonad<br />

migration is defective. Interestingly, these phenotypes are strongly enhanced when PHA-4 activity is<br />

compromised. We suggest that PHA-4 may bind distinct CHW factors to regulate different target genes in<br />

the pharynx, rectum or gonad.<br />

95


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

THE UNC-4 HOMEOPROTEIN AND ITS TRANSCRIPTIONAL<br />

CO-REPRESSOR UNC-37/GROUCHO REGULATE<br />

NEUROTRANSMITTER VESICLES IN CHOLINERGIC MOTOR<br />

NEURONS<br />

Kim Lickteig 1 , Janet Duerr 2 , Dennis Frisby 3 , David Hall 4 , Jim Rand 2 ,<br />

David Miller 1<br />

1Vanderbilt University, Nashville, TN<br />

2Oklahoma Medical Research Foundation, Oklahoma City, OK<br />

3Univ. of Central Oklahoma, Edmond, OK<br />

4Albert Einstein College of Medicine, Bronx, NY<br />

The UNC-4 homeoprotein is expressed in VA motor neurons to prevent the adoption of synaptic inputs<br />

normally reserved for their lineal sisters, the VB neurons. Work from this laboratory has shown that<br />

UNC-4 functions as a negative regulator of VB-specific genes and that this activity depends on physical<br />

interaction with the ubiquitously expressed UNC-37/Groucho co-repressor protein. UNC-4 is also<br />

expressed in other motor neurons (DA, SAB, VC) but these cells (e.g. DAs) are not miswired in unc-4<br />

mutants. Here we show that unc-4 and unc-37 mutations result in decreased levels of synaptic vesicle<br />

(SV) proteins in "unc-4-expressing motor neurons" and that this deficit impairs the function of these cells.<br />

Antibody staining reveals that five vesicular proteins (UNC-17, ChAT, synaptotagmin, synaptobrevin,<br />

RAB-3) are substantially reduced in unc-4 and unc-37 mutants whereas other non-vesicular presynaptic<br />

proteins (syntaxin, UNC-18, UNC-11) are not affected. This finding is consistent with ultrastructural<br />

analysis of VA motor neurons in unc-4(e120) which show a 40% reduction in SVs at presynaptic<br />

densities. Because the UNC-4/UNC-37 complex has been shown to mediate trancriptional repression, we<br />

believe that these effects must be executed through an intermediate gene. Furthermore, our results<br />

indicate that this intermediate gene ("Gene-X") functions as a negative regulator of SV biogenesis or<br />

stability. Experiments with a temperature sensitive unc-4 mutant indicate that the adult level of SV<br />

proteins strictly depends on unc-4 function during a critical early period of motor neuron differentiation.<br />

unc-4 activity during this sensitive larval stage (L2 to mid L3) is also required for the creation of proper<br />

synaptic inputs to the VA neurons. The temporal correlation of these events offers the intriguing possibility<br />

that a common regulatory mechanism involving unc-4 may account for the specificity of synaptic input as<br />

well as the strength of synaptic output for the VA motor neurons.<br />

96


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

THE COMPONENTS OF SENSORY CILIA IN C. ELEGANS<br />

Peter Swoboda, Kerry Bubb, James H. Thomas<br />

Department of Genetics, University of Washington, Seattle, WA 98195<br />

Cilia are an important cellular differentiation of sensory neurons for receiving information from the<br />

environment. 60 of the 302 C. <strong>elegans</strong> neurons have ciliated endings. Mutations in a large number of<br />

genes have been identified that affect the structure of these sensory cilia. Of these, daf-19 mutations are<br />

unique in completely lacking all sensory cilia. Aside from the absence of sensory cilia, the neurons seem<br />

to be morphologically normal. We previously reported that daf-19 encodes an RFX-type transcription<br />

factor that is expressed in ciliated sensory neurons.We also showed that DAF-19 regulates via its target<br />

site in promoter regions, the x-box, a number of effector genes involved in sensory cilium formation.<br />

These effector genes are expressed in essentially all ciliated sensory neurons and, when mutated, cause<br />

morphological defects in sensory cilia (e.g. che-2, osm-1, osm-6). Our results strongly suggest that<br />

DAF-19 regulates the differentiation of sensory cilia by activating the transcription of a battery of genes<br />

whose products form the sensory cilium.<br />

Cilia and flagella are structurally very similar subcellular organelles. Work done on the unicellular,<br />

flagellated green alga Chlamydomonas lead to an estimate of around 300 different protein components<br />

required for structure and function of flagella. We expect a similar number for cilium structure and<br />

function. We aim to initiate the identification of all ciliary components by analyzing genes that have an<br />

appropriately spaced x-box promoter element: so called xbx genes, hypothesized to be regulated by<br />

daf-19. In a first search through the C. <strong>elegans</strong> genome sequence, using an algorithm designed to find<br />

promoter elements, we uncovered more than 200 candidate xbx genes, several of which had C. briggsae<br />

homologues that also had an appropriately positioned x-box promoter element. Initial expression analyses<br />

of a subgroup of these xbx genes showed that, indeed, some of them were specifically expressed in<br />

ciliated sensory neurons in a daf-19 dependent manner. In another approach, using oligo-nucleotide<br />

arrays representing nearly all genes of C. <strong>elegans</strong> (1), we compared the gene expression profile of wild<br />

type and daf-19 mutants. A small group of genes was reproducibly down-regulated in a daf-19<br />

background, among them known cilium-structure genes and several xbx genes. In combination with<br />

previous data about sensory cilia, these approaches have the potential to reveal all the genes required for<br />

sensory cilium function and structure.<br />

(1) in collaboration with Allan Jones (Rosetta Inpharmatics, Kirkland, WA, U.S.A.)<br />

97


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

IN VIVO IMAGING OF HSN OUTGROWTH<br />

Carolyn E. Adler, Cornelia I. Bargmann<br />

Department of Anatomy, UCSF, San Francisco, CA 94143<br />

Axons in developing organisms must migrate and extend long distances in order to synapse onto their<br />

proper targets. Little is known about how extracellular guidance cues elicit the changes in cytoskeletal<br />

dynamics that drive axon outgrowth. In order to understand the factors that control the development of the<br />

HSN motorneuron, we are undertaking an observational approach. We are utilizing two-photon<br />

microscopy to analyze the trajectory of HSN as it grows ventrally in late L2/early L3 stage worms. Using<br />

worms expressing GFP under the unc-86 promoter, we are able to visualize HSN during the course of its<br />

outgrowth.<br />

Preliminary imaging indicates that HSN initially extends filopodia in all directions from its cell body, but<br />

that only those filopodia facing the ventral side persist. Eventually these filopodia develop into a large<br />

lamellopod that extends ventrally and stalls at the ventral muscle barrier. A few processes protrude<br />

through the body wall muscle attachment sites, as seen in VD and DD motorneuron growth through the<br />

dorsal muscle attachment sites.1 Although multiple processes extend toward the ventral nerve cord, only<br />

one is stabilized and develops into the axon.<br />

We plan to characterize HSN outgrowth over the course of its development. With this information, we can<br />

analyze the HSN axon in mutant worms known to have defects in ventral growth. In particular, we will<br />

examine worms containing mutations in the extracellular guidance cues to which HSN responds as well<br />

as in cytoplasmic proteins thought to participate in the regulation of cytoskeletal dynamics. In addition, we<br />

plan to label the actin and microtubule cytoskeletons and visualize their dynamics during HSN outgrowth.<br />

This approach will allow us to elucidate the roles of particular proteins in the process of HSN outgrowth.<br />

1. Knobel KM, Jorgensen EM, Bastiani MJ. Development. 1999 Oct;126(20):4489-98.<br />

98


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

TEMPORAL AND SPATIAL REQUIREMENT OF SENSORY<br />

CILIA IN THE REGULATION OF WORM LIFESPAN<br />

Joy Alcedo, Javier Apfeld, Bella Albinder, Jennifer Dorman, Honor<br />

Hsin, Bernadine Tsung, Cynthia Kenyon<br />

Department of Biochemistry and Biophysics, UCSF, San Francisco, CA 94143<br />

C. <strong>elegans</strong> is an excellent organism in which to study the regulation of lifespan. Genetic analyses have<br />

already shown that an insulin/IGF-like hormonal control system, the DAF-2 pathway, regulates lifespan in<br />

the worm. However, many components of the pathway, whose existence has been inferred from previous<br />

studies (Apfeld and Kenyon, (1999). Nature 402, 804-809; and Hsin and Kenyon, (1999) Nature 399,<br />

362-366) have not been identified. To search for more genes that would help to elucidate mechanisms<br />

that control lifespan in the worm, our group has done an EMS mutagenesis screen for long-lived worms<br />

and found 29 independent long-lived mutants.<br />

At least one of the mutants, mu377(IV), maps to and fails to complement the gene daf-10, which is<br />

required for the normal development of the worm’s sensory cilia. Apfeld and Kenyon (1999) have recently<br />

shown that worms defective in sensory cilia, including daf-10 mutants, live longer than normal worms,<br />

which suggests that the lifespan of C. <strong>elegans</strong> might be regulated by the perception of a signal(s) from<br />

the environment. mu377(IV) has a temperature-sensitive defect in its sensory cilia, which is linked to its<br />

temperature-sensitive lifespan phenotype. At the restrictive temperature, its lifespan is longer than<br />

wild-type and it exhibits a defect in its sensory cilia; whereas at the permissive temperature, its lifespan is<br />

the same as wild-type and it exhibits normal sensory cilia. Since the defect in the sensory cilia of<br />

mu377(IV) is reversible, we determined the temporal requirement for the gene product of mu377(IV) in<br />

regulating the lifespan of the worm. To gain insight into the spatial requirement for sensory perception in<br />

controlling the worm’s lifespan, we are also in the process of determining which sensory neurons are<br />

necessary to regulate its lifespan by expressing wild-type sensory genes in subsets of mutant sensory<br />

neurons.<br />

99


THE TTX-3 LIM HOMEOBOX GENE IS A CENTRAL<br />

REGULATOR OF INTERNEURON CELL FATE<br />

Z. Altun-Gultekin, O. Hobert<br />

Columbia University, College of Physicians & Surgeons, Center for Neurobiology and Behavior, New<br />

York, NY 10032<br />

The LIM homeobox (Lhx) gene ttx-3 is required for correct thermotactic behavior (1,2). In ttx-3 mutants,<br />

the AIY interneuron, a component of the thermoregulatory neural circuit, is structurally and functionally<br />

defective. ttx-3::gfp reporter gene fusions are expressed in AIY. We have set out to test what cellular role<br />

the ttx-3 gene plays and present here our analysis of the execution of the AIY cell fate in ttx-3 mutants.<br />

Previously we have shown that in ttx-3 mutants maintenance of postembryonic ttx-3 expression is lost in<br />

AIY due to autoregulation. Using GFP reporter gene constructs, we have now found that expression of<br />

other AIY cell fate markers including a 7-TM receptor, sra-11, a homeobox gene, ceh-23, and a secreted<br />

protein, C36B7.7 (kindly provided by T. Ishihara) is also downregulated in AIY in ttx-3 mutants. A GFP<br />

fusion to the octopamine/serotonin receptor ser-2 (kindly provided by T. Niacaris), which we identified as<br />

being expressed in AIY as well, is also downregulated. Additionally, AIY loses its cholinergic phenotype in<br />

ttx-3 mutants as detected by VAchT antibody staining (antibodies kindly provided by J.Duerr). These<br />

results suggest that AIY fails to differentiate correctly in ttx-3 mutants. So far it is unclear whether ttx-3<br />

directly controls the expression of the genes described above or functions through intermediary<br />

transcription factors. We are addressing this question by delineating and comparing the ttx-3 -dependent<br />

regulatory elements in the distinct AIY cell fate markers.<br />

Previously it was shown that mutations in the Lhx gene lim-4 lead to a switch of the fate of the AWB<br />

sensory neuron to AWC (3). Although many if not all aspects of the correct fate of AIY are lost in ttx-3<br />

mutants, our preliminary tests for a cell fate switch of AIY into any of its lineal, structural or functional<br />

homologs suggest that AIY has not taken over the identity of AIM, AIZ, ASE or AWC. In contrast to AWB<br />

in lim-4 and AIY in ttx-3 mutants, the DVB motor neuron maintains its correct fate in animals mutant for<br />

the Lhx gene lim-6 (O. H., unpubl.). Hence, in Lhx mutants the identity of a neuron may be maintained,<br />

lost or switched.<br />

References:<br />

(1) Hobert et al., 1997, Neuron 19, 345<br />

(2) Mori and Oshima, 1995, Nature 376, 344<br />

(3) Sagasti et al., 1999, Genes Dev 13, 1794<br />

<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

100


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

THE HETEROCHRONIC GENE PATHWAY: REGULATORY<br />

INTERACTIONS AND REGULATORY OUTPUTS.<br />

Victor Ambros 1 , Marta Hristova 1 , Rosalind Lee 1 , Eric Moss 2<br />

1Department of Biology, Dartmouth College, Hanover, NH 03755<br />

2Fox Chase Cancer Center, Philadelphia PA<br />

The heterochronic gene pathway controls the timing of larval developmental events. Central to the action<br />

of this pathway is the developmental down-regulation of lin-14 and lin-28, whereby high levels specify L1<br />

events, and lower levels specify L2 and L3 events. The decrease in lin-14 and lin-28 results from<br />

translational repression by the small RNA product of lin-4. lin-4 RNA is complementary to sequences in<br />

the 3’ UTR of its target mRNAs, and recent experiments suggest that lin-4 acts by gating access to the<br />

3’UTR by other positive and negative inputs. These other inputs include a mutual positive feedback<br />

between lin-14 and lin-28 and a daf-12-dependent negative regulatory input.<br />

The outputs of the heterochronic gene pathway are the expression of cell-type specific developmental<br />

programs. To seek downstream targets of lin-14 for L2 events, we employed a microarray of 12,000<br />

ORFs prepared by the Kim lab at Stanford University. Probes were prepared from mRNAs of<br />

synchronized mid-L1 populations of N2 and lin-14(n179ts) animals. Among the genes that displayed<br />

lin-14-dependent expression by microarray analysis, we chose four genes for follow-up by Northern and<br />

GFP-fusions. By Northern analysis, three of these four genes show precocious mRNA expression in<br />

lin-14(lf) animals, consistent with their temporal regulation by lin-14 in the wild type. The expression of<br />

GFP promoter fusions suggests that at least one of the genes, ins-33, appears to be developmentally<br />

regulated by lin-14 at the transcriptional level.<br />

For L3 events, the chief effectors downstream of lin-14 and lin-28 appear to be daf-12 and lin-46. lin-46<br />

encodes a member of a conserved family of proteins involved in protein-protein interactions, suggesting a<br />

complex of regulators controling L3 targets. In the vulva precursor cells, L3 events controled by the<br />

heterochronic genes are progress through the G1/S phase of the cell cycle and the acqusition of<br />

competence to express vulval cell fates. VPC G1/S is regulated by the transcriptional activation of a cyclin<br />

kinase inhibitor gene, cki-1. We are identifying regions of the cki-1 promoter which mediate the<br />

VPC-specific control of cki-1 transcription by the heterochronic gene pathway.<br />

101


GENETIC AND PHENOTYPIC CHARACTERIZATION OF<br />

EVL-14 AND EVL-20, GENES INVOLVED IN C. ELEGANS<br />

VULVA DEVELOPMENT<br />

Igor Antoshechkin, Min Han<br />

<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

Department of MCD Biology, University of Colorado, Boulder, CO 80309<br />

In an attempt to identify new genes that participate in C. <strong>elegans</strong> vulva development, we carried out a<br />

screen for sterile mutants with protruding vulva. The mutants were then examined using Nomarski optics<br />

for abnormal vulva morphology at the "Christmas tree" stage. Several of isolated mutations were alleles of<br />

evl genes previously identified by Geraldine Seydoux . Here we report genetic and phenotypic<br />

characterization of two such genes, evl-14 and evl-20. Both mutations are completely recessive and result<br />

in a 100 % sterile phenotype. Their vulva structures are variably abnormal and are missing anywhere<br />

from 0 to 10 cells suggesting either under induction or cell cycle/division defect. In order to distinguish<br />

between these possibilities we are making doubles with members of Ras signaling pathway and GFP<br />

reporters for vulval cell fates. We are also performing detailed lineage analysis of the mutants. evl-20 also<br />

displays gonad migration defect not seen in evl-14. evl-14 maps to linkage group III just left of pal-1.<br />

evl-20 is located on chromosome II next to rol-6. We have injected cosmids from these regions and<br />

identified cosmid pools that rescue both evl-14 and evl-20. We are in the process of injecting single<br />

cosmids and making subclones to identify single open reading frames that will rescue the mutants.<br />

102


CAENORHABDITIS ELEGANS T05H10.5, A HOMOLOGUE OF<br />

YEAST UBIQUITIN FUSION DEGRADATION PROTEIN<br />

(UDF-2), IS EXPRESSED THROUGHOUT THE NERVOUS<br />

SYSTEM AND IN THE GUT<br />

Wanyuan Ao, Dave Pilgrim<br />

<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

Department of Biological Sciences, University of Alberta, Edmonton, AB T6G 2E9, Canada<br />

C. <strong>elegans</strong> UNC-45 is a component of muscle thick filaments and co-localizes with myosin heavy chain B<br />

in body wall muscles. The N-terminal of UNC-45 contains three tetratricopeptide repeats (TPR) and its<br />

C-terminal shows limited similarity to fungal proteins. In our yeast two-hybrid screens using unc-45 cDNA<br />

as a bait, we detected that UNC-45 interacts with T05H10.5 and the TRP domain of UNC-45 is sufficient<br />

for that interaction. The predicted T05H10.5 gene encodes a 113.2 KD protein that is a homologue of<br />

yeast ubiquitin fusion degradation protein (UDF-2). In further characterization of T05H10.5, We found that<br />

the GFP reporter driven by T05H10.5 promoter is expressed throughout the nervous system and also<br />

strongly in the gut of the adult worm. The UDF-2-like proteins are highly conserved from yeast to human<br />

and its putative human homologue is expressed in brain tissues. Our results indicates that T05H10.5 is a<br />

nerve cell and gut-specific gene and may interact with TRP-containing proteins (like UNC-45).<br />

103


GENETIC ANALYSIS OF NEUROENDOCRINE CONTROLS OF<br />

FAT METABOLISM IN C. ELEGANS<br />

Kaveh Ashrafi, Gary Ruvkun<br />

<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

Dept. of Molecular Biology, Massachusetts General Hospital and Dept. of Genetics, Harvard Medical<br />

School, Boston, MA 02114<br />

Body fat regulation is a complex process that affects behavior, physiology, and metabolism of an<br />

organism in order to balance energy intake with energy expenditure. C. <strong>elegans</strong> is an attractive choice for<br />

the systematic identification of components of fat homeostasis and cell biology. As in mammals, an insulin<br />

signaling pathway in C. <strong>elegans</strong> couples nutritional status to the tempo and mode of metabolism, while<br />

neuronal outputs, e.g. serotonin, translate food sensation to various motor and endocrine outputs.<br />

We have used the phenoxazine dye Nile Red to visualize fat droplets in the intestinal and hypodermal<br />

cells of living C. <strong>elegans</strong>. The Nile Red staining pattern closely matches the pattern observed with the<br />

traditional methods of fat staining in C. <strong>elegans</strong> (e.g Sudan Black B). Moreover, the known and predicted<br />

patterns of fat accumulation arising from mutations in insulin-like (daf-2 / daf-16) and TGF-b like (daf-7 /<br />

daf-3) signaling pathways are well recapitulated by Nile Red staining. Thus, Nile Red staining of living<br />

animals provides a means for genetic screens based on fat metabolism and homeostasis. Two such<br />

screens are presented:<br />

(i) To identify genes involved in the biogenesis and cell biology of fat storage, we screened<br />

EMS-mutagenized N2 animals for mutant worms with abnormalities in lipid droplet size or number. In a<br />

screen of 15,000 mutagenized F2, we identified four mutants that display abnormally large droplet sizes.<br />

(ii) The C. <strong>elegans</strong> insulin signaling pathway determines whether larvae grow to a fast metabolizing adult<br />

stage or enter a slow metabolizing dauer stage. Reduced signaling through the DAF-2 insulin-like<br />

receptor promotes a shift in metabolism to fat storage and dauer arrest. The forkhead transcription factor<br />

DAF-16, is the key target of DAF-2 signaling pathway and is required for the shift in fat metabolism and<br />

dauer arrest. Mutant adult daf-2 animals have a diffuse pattern of Nile Red staining. In contrast, Nile Red<br />

appears in very discrete droplet spots in daf-16 animals. To identify downstream metabolic targets of<br />

DAF-16, we have begun screening EMS-mutagenized daf-16 worms to find mutants that recapitulate the<br />

daf-2 pattern of staining.<br />

104


ZIG GENES AND THE PVT GUIDEPOST NEURON<br />

Oscar Aurelio, Oliver Hobert<br />

Columbia University,New York, NY 10032<br />

<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

We have identified a novel family of putative cell adhesion and/or signaling molecules which are defined<br />

by the presence of a signal sequence and 2 Ig domains; none of these molecules contain a predicted<br />

transmembrane sequence, but several contain consensus sites for GPI anchorage. C.<strong>elegans</strong> contains 9<br />

members of this family, termed the zig gene family. We hypothesize that these genes may play a role in<br />

neural patterning. As part of receptor/ligand complexes, they may be involved in cell recognition or,<br />

alternatively, they may serve as secreted and diffusible signaling molecules.<br />

GFP fusion to the promoter region of these 9 genes reveals that 6 of these genes are expressed almost<br />

exclusively in very specific subdomains of the nervous system, while 2 of them are exclusively expressed<br />

in body wall muscles. One zig gene is expressed ubiquitously in the nervous system and in body wall<br />

muscles.<br />

The most intriguing aspect of zig gene expression in the nervous system relates to the PVT neuron: 5 of<br />

the 9 zig genes are expressed in PVT. Aside from the panneuronally expressed zig-1 reporter gene,<br />

zig-2, zig-4 and zig-9 are expressed in few other cells than PVT, while zig-5 is expressed in about one<br />

dozen additional head neurons. PVT acts during embryonic patterning as a guidepost cell for pioneer<br />

neurons of the ventral nerve cord (Durbin, 1987) and is a source of unc-6 expression (Wadsworth et al.,<br />

1996). Given the embryonic role of PVT, we were surprised to find that at least two of the five<br />

PVT-expressed zig genes, zig-2 and zig-4, are not expressed embryonically; instead their expression is<br />

turned on right after hatching and presists throughout adulthood. Additionally, we found that maintenance,<br />

but not initiation of zig-2 and zig-4 expression requires the lim-6 LIM homeobox gene, which is expressed<br />

in PVT.<br />

The postembryonic expression of zig-2 and zig-4 suggests that PVT subserves a postembryonic role in<br />

axonal patterning. What could this role be? In hermaphrodites relatively few neurons extend their axon<br />

into the ventral nerve cord postembryonically; however, in males, many neurons are born<br />

postembryonically in the tail and presumably require cues to extend their axon along the ventral nerve<br />

cord. We will test whether PVT plays a role in this process by driving expression of cytotoxic agents under<br />

control of the postembryonic zig-2 promoter.<br />

105


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

ANALYSIS OF GABA RECEPTOR PLASTICITY IN C.<br />

ELEGANS<br />

Bruce A. Bamber 1 , Janet E. Richmond 2 , Pierrette K. Danieu 1<br />

1 Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, UT, 84112<br />

2 Department of Biology, University of Utah, Salt Lake City, UT, 84112<br />

Neuronal activity depends on the balance of excitatory and inhibitory neurotransmission. One way for a<br />

neuron to regulate its activity level is to alter its sensitivity to inhibitory neurotransmitters. Most inhibitory<br />

neurotransmission in the mammalian brain is mediated by g-aminobutyric acid (GABA), which acts mainly<br />

through GABA A receptors (ligand-gated chloride channels). Neurons regulate their GABA sensitivity by<br />

regulating the density and GABA-responsiveness of synaptic GABA A receptors. This receptor plasticity is<br />

important for regulating neuronal excitability in both normal and diseased nervous systems. The<br />

mechanisms of GABA A receptor plasticity are not well understood, in part because it is difficult to apply<br />

genetic techniques to study mammalian synapse function. The C. <strong>elegans</strong> neuromuscular junction is a<br />

powerful model system to study GABA receptor plasticity. A GABA A receptor homolog, consisting of<br />

UNC-49B and UNC-49C subunits, functions in C. <strong>elegans</strong> muscle cells. This receptor exhibits<br />

mammalian-like plasticity: The GABA receptor agonist muscimol acutely paralyzes C. <strong>elegans</strong>, but<br />

chronic exposure results in adaptation (i.e. the appearance of muscimol resistance, and a GABA<br />

receptor-defective phenotype). Patch-clamp analysis of muscle cells in adapted animals revealed an 85%<br />

decrease in GABA-evoked currents compared to untreated animals, indicating that GABA receptor<br />

downregulation underlies adaptation. Muscimol exposure also caused GFP-tagged UNC-49 receptors to<br />

form clusters in extra-synaptic muscle membranes, which implicates the clathrin-mediated endocytosis<br />

pathway in downregulation. We are pursuing two strategies to study GABA receptor plasticity in C.<br />

<strong>elegans</strong>. First, we are identifying UNC-49 sequences required for synaptic localization and<br />

downregulation. Our results have demonstrated that the UNC-49B subunit is necessary and sufficient for<br />

both processes. Second, we are performing a yeast two-hybrid screen to identify GABA<br />

receptor-associated molecules in C. <strong>elegans</strong>. We have identified several interesting candidates, and we<br />

are using RNA interference to determine which of these proteins are important for GABA receptor<br />

regulation.<br />

106


ISOLATION OF SUPPRESSORS OF A DOMINANT SYNAPSE<br />

DEFECTIVE MUTANT, SYD-5(JU89)<br />

Renee Baran, Yishi Jin<br />

Department of Biology, Sinsheimer Labs, University of California, Santa Cruz, CA 95064<br />

syd-5 (ju89)was identified in a screen for mutations that alter the differentiation of GABAergic synapses in<br />

C. <strong>elegans</strong> using the presynaptic vesicle marker, Punc-25::SNB-1-GFP 1,2 . In adult wild-type worms,<br />

Punc-25-SNB-1-GFP is expressed as puncta of uniform size and spacing along the dorsal and ventral<br />

nerve cords, corresponding to neuromuscular junctions made by the GABAergic DD and VD neurons with<br />

dorsal and ventral body wall muscles, respectively. syd-5(ju89) mutant worms are uncoordinated and<br />

exhibit abnormally large and small SNB-1-GFP puncta dispersed in an irregular pattern along the nerve<br />

cords. This phenotype closely resembles the SNB-1 GFP phenotype of loss-of-function syd-3/rpm-1<br />

mutants, which have overdeveloped synapses with multiple active zones 3 . syd-5 maps to chromosome I<br />

between egl-33 and lin-11. syd-5( ju89) behaves genetically as a weak gain-of-function mutation: Df/+<br />

worms are wild-type, but heterozygous ju89/+ worms exhibit a locomotion and SNB-1 GFP phenotype<br />

that is intermediate between wild-type and homozygous ju89 worms. A suppressor screen was performed<br />

to identify loss-of-function syd-5 alleles and genes that may function in the same pathway. syd-5(ju89)<br />

worms were mutagenized with EMS, and 30,000 F1 progeny were screened for suppression of the<br />

syd-5(ju89) uncoordinated phenotype. Twenty-four mutants were isolated that suppress both the syd-5<br />

locomotion phenotype and the synaptic vesicle marker defect. Four suppressors are linked to ju89 and<br />

may represent loss-of-function syd-5 alleles. Preliminary characterization of the syd-5 suppressors will be<br />

presented, along with progress in the cloning and molecular characterization of syd-5.<br />

1 Zhen & Jin. 1999. Nature<br />

2 Nonet, M. 1999. Neurosci. Methods<br />

3 Zhen et al. 2000. Neuron<br />

<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

107


CARGO RECOGNITION BY SYNAPTIC VESICLE KINESIN<br />

Ewa Bednarek, Erik M. Jorgensen<br />

<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

Department of Biology, University of Utah, Salt Lake City, UT 84112, USA<br />

Synaptic vesicles are transported to the synapse by kinesin, an ATP-dependent motor protein that moves<br />

along axonal microtubules. In the cell body the kinesin must specifically bind vesicles destined for the<br />

nerve terminal. We would like to identify the molecular basis for vesicle cargo recognition by UNC-104, a<br />

<strong>Caenorhabditis</strong> <strong>elegans</strong> homolog of the motor protein kinesin. One theory suggests that UNC-104<br />

recognizes specific protein molecules associated with the vesicle membranes. Alternatively, we<br />

hypothesize that UNC-104 recognizes synaptic vesicles by binding specific lipids. This is supported by the<br />

fact that UNC-104 has a pleckstrin homology (PH) domain, which is a lipid binding moiety. If PH<br />

domain-lipid binding determines cargo recognition, the following predictions can be made: the PH domain<br />

should bind lipids specific to synaptic membranes, and the PH domain should be required for cargo<br />

recognition. To test these predictions we are first expressing the PH domain in bacteria and determining<br />

whether it binds phosphoinositides. Second, we are testing whether mutations in the PH domain disrupt<br />

synaptic vesicle trafficking.<br />

108


THE EXP-1 LOCUS MAY ENCODE A SUBUNIT OF AN<br />

EXCITATORY GABA RECEPTOR<br />

Asim A. Beg, Erik M. Jorgensen<br />

<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

Dept. of Biology, University of Utah 257 South 1400 East Salt Lake City, UT 84112<br />

Gamma-aminobutyric-acid (GABA) is a major neurotransmitter of inhibitory synapses. In the worm, as in<br />

vertebrates, GABA mediates inhibitory neurotransmission (VD/DD to body muscles). However, in the<br />

enteric muscles, excitatory neurotransmission seems to be mediated by GABA. The GABAergic neurons<br />

AVL and DVB directly synapse on the enteric muscles and are the only neurons required for excitation of<br />

the enteric muscles. We have cloned a potential excitatory GABA receptor subunit, exp-1. Of the six<br />

genes required for GABA neurotransmission, only exp-1 specifically eliminated the excitatory function of<br />

GABA while leaving all other functions intact. We hypothesize that EXP-1 is a subunit of a novel<br />

excitatory GABA receptor expressed in the enteric muscles.<br />

We cloned the exp-1 gene by microinjection rescue. Genetic map data showed that exp-1 is located on<br />

Chromosome II between lin-4 and lin-23. One fosmid in this region, H35N03, contains a gene related to<br />

GABA receptors. Transgenes bearing a subclone of this single open reading frame (ORF) completely<br />

rescued exp-1(sa6) mutants (gift of J. Thomas). In addition, we identified point mutations in this ORF in<br />

three alleles, which confirmed that exp-1 encodes this GABA receptor related protein. We isolated exp-1<br />

cDNAs which show that EXP-1 has a divergent transmembrane 2 (TM2) from all known GABA receptor<br />

subunits. TM2 has been shown to confer ion selectivity to the ligand-gated ion channel superfamily.<br />

Consistent with our hypothesis that EXP-1 is an excitatory GABA receptor subunit, TM2 has numerous<br />

negatively charged residues, suggesting this subunit might form a receptor that passes cations rather<br />

than the anion chloride.<br />

The majority of physiological GABA receptors are heteromultimers. By BLAST search we have identified<br />

a second putative subunit that is very similar to EXP-1. Specifically, TM2 of the predicted subunit is<br />

nearly identical to EXP-1, suggesting this might be the physiological partner of EXP-1. We are currently<br />

performing RT-PCR to isolate cDNAs for this subunit. We will coinject Xenopus laevis oocytes with<br />

cRNAs from both subunits and determine GABA gating and ion permeability of the channel using<br />

electrophysiological analysis.<br />

109


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

THE LIFE SPAN GENE CLK-2 IS ESSENTIAL FOR<br />

EMBRYONIC DEVELOPMENT<br />

Claire Bènard, Brent McCright, Yue Zhang, Stephanie Felkai, Siegfried<br />

Hekimi<br />

Department of Biology, McGill University, 1205 Dr Penfield Ave., Montreal, Quebec, Canada<br />

Mutations in the C. <strong>elegans</strong> maternal-effect genes clk-1, clk-2, clk-3, and gro-1 are highly pleiotropic<br />

resulting in an average slowing of development, adult rhythmic behaviors, reproduction, as well as in an<br />

extended life span. Here we will present the genetic and molecular characterization of the gene clk-2.<br />

This gene is defined by one recessive mutation, qm37, which was isolated in a screen for viable<br />

maternal-effect mutations (Hekimi et al., 1995). At 20C, the duration of embryonic and post-embryonic<br />

development of clk-2(qm37) mutants is lengthened and adult behaviors such as defecation, pharyngeal<br />

pumping, and egg laying are slower. In addition, they live long. In contrast, at 25C, clk-2(qm37) mutants<br />

die as embryos. Moreover, at 20C, clk-2(qm37) mutants can be fully rescued both zygotically and<br />

maternally, while at 25C there is a strict-maternal effect. We believe that clk-2(qm37) is a<br />

temperature-sensitive mutation that behaves as a hypomorph at 20C and as a null at 25C. As we have<br />

found that clk-2 is not required for the development of the germline per se at 25C, we have explored the<br />

origin of the embryonic lethality at 25C by carrying out temperature shift experiments. We have shown<br />

that clk-2 is required before the two-cell stage of embryogenesis. The temporal and spatial expression<br />

pattern of clk-2 deduced from northern and western analyses, and from the use of clk-2::gfp reporter<br />

fusions, is consistent with a maternal role of clk-2 early in development and throughout the life of the<br />

worm.<br />

110


DOES CEH-20, AN EXD/PBX HOMOLOG IN C. ELEGANS,<br />

PLAY A ROLE IN WORM EMBRYOGENESIS?<br />

Q.F. Boese, W.B. Wood<br />

Dept of MCD Biology, University of Colorado, Boulder, CO 80309<br />

Several studies have shown that the TALE (three-amino-acid-loop-extension) homeodomain (HD)<br />

proteins, Exd and Hth in Drosophila and their mammalian homologs, Pbx1-3 and Meis, complex with Hox<br />

proteins to enhance binding specificity for target promoter sites. Studies have further shown that Hth/Meis<br />

interacts with Exd/Pbx to promote nuclear localization facilitating their roles as co-factors in the nucleus.<br />

Recent work in our lab revealed that loss-of-function (lf) mutations in the C. <strong>elegans</strong> Hth/Meis homolog,<br />

unc-62, result in pleiotropic defects including defects in embryonic development. To investigate whether<br />

the Exd/Pbx homolog, CEH-20 might also be involved in embryonic patterning, we have begun<br />

characterizing the defects of two strong ceh-20(lf) alleles (gift from M. Stern) with respect to their potential<br />

interactions with unc-62 and other embryonically required genes (Hox or non-Hox). Both ceh-20 alleles<br />

result in low-penetrance larval lethality and no apparent embryonic defects suggesting that ceh-20 is not<br />

normally required for embryogenesis. Injection of double-stranded ceh-20 RNA into wild-type<br />

hermaphrodites [ceh-20 (RNAi)] results in an Unc Egl phenotype but no embryonic defects.<br />

To test for an interaction with the embryonically required posterior Hox paralog gene nob-1 (1), ceh-20<br />

(RNAi) was performed in the background of a weak allele, nob-1(ct230). No enhancement of the mutant<br />

phenotype was observed. Whether this reflects compensatory functions of two other Exd/Pbx homologs,<br />

ceh-40 and F22A3.x, in the absence of ceh-20, is under investigation.<br />

To test for interactions with unc-62, ceh-20 RNAi was performed in the background of the weak allele<br />

unc-62 (e644). The low penetrance e644 lethal phenotype was enhanced over the uninjected control,<br />

consistent with an interaction between these genes during embryogenesis. To test for interactions with<br />

other non-Hox HD proteins important for embryogenesis, ceh-20 RNAi was performed in the background<br />

of a weak pal-1 allele (e2091). The e2091 mutant phenotype was also enhanced suggesting a previously<br />

undescribed interaction between Exd/Pbx and Caudal/CDX homologs. Further tests for ceh-20<br />

interactions with other embryonically required genes are in progress.<br />

(1) Van Auken, K. et al., 2000, PNAS 91: 4499-503<br />

<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

111


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

A-DOMAIN-CONTAINING PROTEIN FAMILY IN C. ELEGANS.<br />

Michael Brannan, Joaquin Muriel, Kathryn Taylor, Gordon Lithgow,<br />

Danny Tuckwell<br />

School of Biological Sciences, University of Manchester, Stopford Building, Manchester M13 9PT, UK.<br />

The extracellular matrix (ECM) plays an important structural and functional role in metazoans and its<br />

major components include collagen, laminin, and proteoglycan. The cuticle and basement membrane are<br />

the major forms of ECM in C. <strong>elegans</strong>: the cuticle is an exoskeleton important for shape and motility, and<br />

it is also a barrier between the worm and its environment. The basement membrane provides structural<br />

support and tissue separation. Very little is known about the molecules that organise the ECM or the cell<br />

receptors for ECM molecules in C. <strong>elegans</strong>.<br />

A-domains are 200-amino acids modules identified in a range of mammalian ECM proteins. We have<br />

previously studied the A-domains of the integrin a subunits. Integrins a 1b 1 and a 2b 1 are the two major<br />

receptors in the human body for collagens. We have showed that it is the A-domains found within the a<br />

subunits that are responsible for collagen binding, and also for the interaction of these integrins with other<br />

ECM proteins such as laminin. Studies of a number of other A-domain-containing proteins indicate a<br />

collagen or ECM-binding function.<br />

Using bioinformatics approaches we have identified 8 novel A-domain-containing proteins in the C.<br />

<strong>elegans</strong> genome. For some of the novel proteins, regions flanking the A-domains were found to resemble<br />

mucin core protein-like sequence and cuticulin domains. Since mucins and cuticulins are ECM<br />

components, this suggests that a number of the novel proteins are likely to be ECM molecules. Genetical<br />

and biochemical characterisation of these molecules will give insights into the function of these proteins in<br />

ECM organisation and cell-ECM interactions.<br />

112


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

DISTRIBUTION AND REGULATION OF GLUTAMATE<br />

RECEPTORS IN THE LOCOMOTORY CONTROL CIRCUIT OF<br />

C. ELEGANS.<br />

Penelope J. Brockie, David M. Madsen, Yi Zheng, Jerry E. Mellem,<br />

Andres V. Maricq<br />

Department of Biology, University of Utah, Salt Lake City, UT 84112<br />

Ionotropic glutamate receptors are ligand gated ion channels that mediate rapid excitatory synaptic<br />

communication in most nervous systems. Ten putative ionotropic glutamate receptor subunits (glr-1 -<br />

glr-8, nmr-1 and nmr-2) have been identified in C. <strong>elegans</strong>. Analysis of the predicted amino acid<br />

sequence suggests that GLR-1 - GLR-8 are members of the non-NMDA class of glutamate receptors,<br />

where NMR-1 and NMR-2 are most similar to members of the NMDA class. Using GFP fusions we have<br />

shown that the subunits are differentially expressed in the nervous system with some found in only a<br />

single neuron.<br />

The NMDA subunits and four of the non-NMDA subunits (GLR-1 - GLR-4) are expressed in at least one<br />

of the five pairs of command interneurons that regulate worm locomotion (AVA, AVB, AVD, AVE and<br />

PVC). This provides insight into the possible combinations of receptor subunits that form ion channels in<br />

vivo, and suggests that a diversity of both NMDA and non-NMDA type glutamate receptors function in the<br />

locomotory control circuit. What regulates expression of the glutamate receptors in this circuit? It has<br />

recently been shown that the homeodomain protein UNC-42 regulates glr-1 expression in a number of<br />

neurons, including AVA, AVD and AVE (1). We show that unc-42 is also required for glr-3 and glr-4<br />

expression in these neurons, but not for the expression of glr-2, nmr-1 or nmr-2. This indicates that<br />

expression of glutamate receptors in the command interneurons is differentially regulated.<br />

Like glr-1 mutants, unc-42 mutants are nose touch defective and show additional mechanosensory and<br />

locomotory defects. Using the nmr-1::gfp fusion, we show that unc-42 mutants also have dramatic defects<br />

in axon outgrowth in the command interneurons. In wild-type worms, these neurons send their axons<br />

along the ventral cord. In unc-42 mutants, axons are dorsally, laterally and anteriorally misplaced. Using<br />

the nmr-1 promoter to express the unc-42 cDNA does not rescue the axon defects. This suggests that<br />

expressing UNC-42 in the command interneurons alone is not sufficient to direct proper axon outgrowth in<br />

these cells.<br />

1. Baran, et al., Development 126, 2241-2251 (1999)<br />

113


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

MUTATIONS THAT AFFECT SYNAPTIC LOCALIZATION OF<br />

GLR-1<br />

Michelle Burbea, Joshua M. Kaplan<br />

Department of Molecular and Cellular Biology, 361 Life Sciences Addition, UC Berkeley, Berkeley, CA<br />

94720-3200<br />

The orderly flow of information in the nervous system requires that proteins are specifically targeted to<br />

pre- and post-synaptic specializations. We are interested in identifying the molecules that target and<br />

maintain proteins to synapses. We have used the GLR-1 AMPA type glutamate receptor as a marker for<br />

post-synaptic localization. A wild type animal expressing a GLR-1::GFP fusion exhibits punctate spots<br />

along the ventral nerve chord representing clustering of the glutamate receptor at specific synaptic<br />

terminii. To identify genes involved in synaptogenesis, we are screening mutagenized GLR-1::GFP<br />

worms for defects in GLR-1 localization.<br />

We predict classes of mutants to represent trafficking, targeting, localization and stabilization molecules<br />

necessary to initiate and maintain synaptic architecture. Approximately 7000 genomes have been clonally<br />

screened so that sterile or inviable mutants may be recovered as heterozygously. Nine complementation<br />

groups have been isolated which define loci important for the proper localization of GLR-1 in the nervous<br />

system. Several of the loci are in genes whose identity is already known. These include mutations in<br />

unc-11, an AP180 homologue, and unc-101, a medium chain subunit of the AP-1 clathrin adaptin<br />

complex. The implication of these mutants on receptor trafficking and maintanance at the synapse will be<br />

discussed.<br />

114


REGULATION OF C. ELEGANS DAUER FORMATION BY AN<br />

RNA QUALITY CONTROL PATHWAY COMPONENT<br />

J Burgess 1 , JC Labbe 2 , S Hekimi 1<br />

<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

1Department of Biology, Mc Gill University, 1205 Dr. Penfield Ave, Montreal, Quebec, Canada H3A1B1<br />

2Department of Biology, University of North Carolina at Chapel Hill, 3280 Coker Hall, Chapel Hill, North<br />

Carolina. 27599-3280, USA<br />

Under conditions that do not favor developmental growth and reproduction, worms can enter an alternate<br />

third-larval stage termed the dauer stage. Dauer larvae are developmentally arrested and are adapted for<br />

long term survival and dispersal. In order to make this decision, animals monitor a variety of<br />

chemosensory cues including the concentration of a constitutively secreted pheromone, as well as food<br />

and temperature. It is currently unknown whether internal states, such as the amount of molecular<br />

damage accumulated, is also taken into account when making the decision to enter the dauer stage. The<br />

gene rop-1 encodes the worm homologue of the human Ro ribonucleoprotein 60-kDa constituent, which<br />

has previously been shown to participate in the quality control of 5S ribosomal RNA. Here, we present<br />

biochemical evidence that rop-1 is cleaved at the L2/L3 molt concommittant with entry into L3 but is not<br />

cleaved when animals enter the dauer stage. In addition, rop-1 is not cleaved in daf-2 mutants suggesting<br />

cleavage of rop-1 may require functional insulin-like signaling. Furthermore, we provide evidence that<br />

rop-1 genetically interacts with the insulin receptor-like kinase daf-2 in an allele-specific manner as well as<br />

with the TGF-b transforming growth factor daf-7 in a temperature dependent manner. Finally, we show<br />

that the enhancement of dauer formation by rop-1 in daf-2(e1370) is daf-16 dependent. However, the<br />

processing of rop-1 appears to be daf-16 independent. We suggest that an RNA quality control<br />

checkpoint may exist for dauer formation.<br />

115


SYNAPTIC VESICLE LOCALIZATION IS MISREGULATED IN<br />

UNC-16 MUTANTS<br />

DT Byrd, Y Jin<br />

University of California, Santa Cruz, Dept. Biol., Sinsheimer Labs, CA 95064<br />

The DD motoneurons provide a model in which to study synaptic development and remodeling. The DDs<br />

are born embryonically, innervate ventral body wall muscles in L1 animals, and remodel to innervate<br />

dorsal body wall muscles in L2 through adult animals. We visualize DD synapses and the process of DD<br />

synaptic remodeling in vivo with a synaptic vesicle marker expressed by the DDs and other GABAergic<br />

neurons1. In wild type L1s, the synaptic vesicle marker is strictly localized to the ventral processes of the<br />

DDs until just before the molt to L22. After the L1 molt, the synaptic vesicle marker is localized along the<br />

dorsal processes of the DDs, indicating that the DDs have remodeled.<br />

We isolated a partial loss-of-function mutation in unc-16 in a screen for dorsal localization of the synaptic<br />

vesicle marker in the DDs of early L1 animals. unc-16 mutant L1 animals exhibit full ventral localization of<br />

the synaptic vesicle marker as well as varying amounts of dorsal localization in the DDs. Seven alleles of<br />

unc-16 form an allelic series in which n730, e109, ju79, and s1072 are partial loss-of-function alleles of<br />

increasing severity, respectively, and s453, s529, and s222 are genetic null alleles. unc-16 encodes a<br />

pan-neurally expressed novel protein with interesting motifs that may interact with a number of signaling<br />

pathways. We are currently determining the molecular lesions in the unc-16 mutants.<br />

We hypothesize that the vesicle localization defect may be caused by disruptions in synaptic vesicle<br />

cargo selection, transport, or docking. Supporting this hypothesis, we found that weak unc-16 mutations<br />

partially suppress partial loss-of-function mutations in unc-104, a gene encoding a kinesin. This suggests<br />

that in the DDs, unc-16 may play a role in regulating synaptic vesicle transport.<br />

1. Nonet ML. 1999. J Neurosci Methods 89(1):33-40.<br />

2. Hallam and Jin. 1998. Nature 395:78-82.<br />

<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

116


THE EGL-21 GENE ENCODES A CARBOXYPEPTIDASE E,<br />

WHICH IS REQUIRED FOR PRO-NEUROPEPTIDE<br />

PROCESSING<br />

Tija Carey, Joshua M. Kaplan<br />

<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

UC Berkeley, MCB Dept , 361 LSA, Berkeley CA 94720<br />

We are interested in understanding the mechanisms for neurotransmitter and neuropeptide modulation of<br />

C. <strong>elegans</strong> behavior, including locomotion, defecation, and egg laying. We previously reported that the<br />

egl-3 gene encodes a prohormone convertase involved in proteolytic processing of pro-neuropeptides<br />

(Kass and Kaplan IWM99). We will present evidence that the egl-21 gene encodes a carboxypeptidase<br />

E(CPE), which mediates the next enzymatic step in peptide processing, removal of C-terminal basic<br />

residues.<br />

Mutations in egl-21 were first isolated in a screen for egg laying defective mutants, but also showed<br />

defective defecation and uncoordinated locomotion (Trent, Genetics 104: 619-647 1983). The various<br />

defects in egl-21 mutants show differing drug sensitivity: the egg laying defect is resistant to serotonin and<br />

imipramine, while the locomotion phenotype is sensitive to both.<br />

We found that egl-21 mutations map very close to mec-3. The genome sequence of this region indicated<br />

that a gene very similar to vertebrate CPE mapped in this region. We found that a cosmid containing this<br />

CPE rescued the egl-21 defecation defects. Next, we showed that two egl-21 alleles correspond to<br />

mutations in the CPE gene. The n576 allele alters an invariant G in a splice donor sequence whereas the<br />

n476 allele corresponds to a 122 bp in-frame deletion. Since egl-21 encodes the only apparent CPE in<br />

the worm genome, we would expect that the phenotype of egl-21 mutants corresponds to a neuropeptide<br />

null phenotype. We are currently studying the role of egl-21 CPE in modulation of several behaviors.<br />

117


HOW ARE ANTERIOR CELL MIGRATIONS GUIDED BY<br />

MIG-13?<br />

QueeLim Ch’ng, Cynthia Kenyon<br />

Department of Biochemistry, UC San Francisco, CA 94143-0448<br />

mig-13 was previously identified as a guidance factor specifically required for the anterior migrations of<br />

the QR descendants. mig-13 acts by promoting cell migrations in the anterior direction (Sym et. al., 1999).<br />

We are taking several approaches to understand further how the anterior migrations of the QR<br />

descendants are guided by mig-13. mig-13 is predicted to encode a novel transmembrane protein<br />

containing a CUB domain and LDL-receptor repeat in the extracellular region as well as a proline-rich<br />

domain in the intracellular region (Sym et. al., 1999). Our structure/function studies suggest that the<br />

extracellular domain of MIG-13 alone can confer partial function in guiding the QR descendants to the<br />

anterior.<br />

A rescuing mig-13::GFP fusion was previously found to be expressed in the anterior and mid-body ventral<br />

cord motor neurons, which cross the migratory track of the QR descendants. Consistent with this<br />

expression pattern, mosaic analysis revealed that mig-13 acts non-autonomously to direct the migrations<br />

of the QR lineage (Sym et. al., 1999). To determine where mig-13 expression is sufficient to guide the<br />

migrating cells, we are expressing mig-13 in broad sets of tissues, as well as in specific subsets of cells<br />

that express mig-13::GFP. We also intend to refine previous mosaic analysis to pinpoint the cells in which<br />

mig-13 acts.<br />

Reference<br />

<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

Sym M, Robinson N and Kenyon C. Cell, 1999 Jul 9, 98(1):25-36.<br />

118


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

NEW SCREENS FOR NEGATIVE REGULATORS OF LET-23<br />

Monica Chan, Marie Tiongsen, Romel C. Castro, Vanessa Lee, Gregg<br />

Jongeward<br />

University of the Pacific, Department of Biological Sciences, 3601 Pacific Avenue, Stockton, Cal 95211<br />

We are trying to identify additional genes that act to negatively regulate the Epidermal Growth Factor<br />

Receptor (EGF-receptor) during vulval induction in Caenorabditis <strong>elegans</strong>. The gene let-23 encodes the<br />

C. <strong>elegans</strong> homolog of the EGF-receptor. Mutations in let-23 generally result in a number of defects,<br />

including larval arrest, sterility, and defects in the vulva and the male tail. At 20 o C, animals homozygous<br />

for the allele let-23(n1045) display excess vulval differentiation. A similar phenotype is observed in<br />

animals homozygous for lin-2(n768). lin-2 activity is required to properly localize EGF-receptor on the<br />

vulval precursor cell. For as yet unknown reasons, animals homozygous for both let-23(n1045)and<br />

lin-2(n768) display a highly penetrant vulval defect unlike that seen in either homozygote. These doubly<br />

mutant animals fail to form any vulval tissue. We have begun to screen for new alleles that suppress the<br />

egg-laying defect associated with the inability to form vulval tissue in animals of this genotype. Thus far,<br />

we have recovered four new alleles. At least one of these alleles appears to be a new lin-1 mutation, as<br />

animals homozygous for the suppressor display excessive vulval differentiation even if there are no other<br />

mutations present. A second (independent) suppressor causes a similar phenotype. The other two alleles<br />

restore approximately normal vulval differentiation. Animals of the genotype let-23(n1045); lin-2(n768);<br />

new suppressor are generally able to lay eggs but lack prominent pseudovulvae. We are currently<br />

characterizing these new alleles to determine if they are mutations in previously identified genes.<br />

119


C. ELEGANS MRE-11 IS REQUIRED FOR MEIOTIC<br />

RECOMBINATION AND DNA REPAIR BUT NOT FOR THE<br />

MEIOTIC G2 DNA DAMAGE CHECKPOINT<br />

Gregory Chin, Anne Villeneuve<br />

<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

Department of Developmental Biology, Stanford University School of Medicine Stanford, CA 94305<br />

We have isolated and analyzed mutants defective in the nematode ortholog of yeast MRE11, a<br />

multifunctional protein with roles in diverse cellular processes required to maintain genome integrity,<br />

including meiotic recombination, DNA repair, and telomere length maintenance. While MRE11 is highly<br />

conserved among several species and has been linked to genetic disease in humans, exploration of its in<br />

vivo roles in metazoan systems has been hampered by the fact that vertebrate cells that lack MRE11 are<br />

inviable. We have found that worms homozygous for an mre-11 null mutation are viable, allowing us to<br />

demonstrate an in vivo requirement for MRE-11 in meiotic recombination and DNA repair. In mre-11<br />

mutants, crossovers are not detected and chromosomes lack chiasmata at diakinesis but appear<br />

otherwise intact. Irradiation of mre-11 mutants not only fails to induce chiasmata but also eliminates<br />

progeny survivorship and leads to cytologically visible chromosomal abnormalities including<br />

fragmentation. These results indicate a defect in the ability of mre-11 mutant germ cells to repair<br />

radiation-induced damage. While they are repair-deficient, we show that mre-11 mutant germ cells retain<br />

function of the meiotic G2 DNA damage checkpoint.<br />

Although mre-11 homozyogtes derived from heterozygous parents are fully viable and produce a normal<br />

number of embryos, there is a marked drop both in the number and in the survivorship of embryos<br />

produced by succeeding generations. As a result, the strain cannot be propagated as a homozygous<br />

stock. This progressive loss of fecundity and viability sets mre-11 mutants apart from many other meiotic<br />

recombination-defective mutants, and indicates that MRE-11 performs an additional essential function in<br />

maintaining reproductive capacity in the species.<br />

120


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

SUPPRESSOR ANALYSIS OF EPH/EPHRIN DEFECTIVE<br />

SIGNALING IN C. ELEGANS<br />

Ian Chin-Sang, Julie McCleery, Andrew Chisholm<br />

Department of Biology, Sinsheimer Laboratories, University of California, Santa Cruz, CA 95064, USA<br />

Mutations in the genes vab-1 and vab-2 cause similar defects in neural and epidermal morphogenesis.<br />

The phenotypes range from embryonic lethality, due to epidermal enclosure defects, and a ventral<br />

notched head in larvae and adults. vab-1 encodes an Eph receptor tyrosine kinase that is expressed in<br />

the developing nervous system, and is required in neurons for proper epidermal morphogenesis (George<br />

et al., Cell 92:633). vab-2/efn-1 encodes a GPI anchored ephrin that specifically interacts with VAB-1 in<br />

neurons to regulate neuronal and epidermal morphogenesis (Chin-Sang et al., Cell 99:781). What are the<br />

molecular consequences of the VAB-1/VAB-2 interaction? We have taken a genetic approach to identify<br />

genes that might act in the VAB-1/2 Eph/Ephrin signaling pathway or in parallel pathways. The incomplete<br />

penetrance and variability of phenotypes displayed by vab-1 and vab-2 mutants, made it difficult in the<br />

past to carry out genetic modifier screens. Therefore we sought to create strains that would make vab-1<br />

and vab-2 mutants more suitable for genetic modifier screens. A mutation in a LAR like receptor tyrosine<br />

phosphatase, ptp-1, results in a synthetic lethality with both vab-1and vab-2, suggesting that PTP-1 may<br />

function redundantly with Eph signaling (see abstract by Harrington et al.). The ptp-1(op147);vab-2(ju1)<br />

double mutant is temperature sensitive- viable at 15 0 C and 20 0 C, however, at 25 0 C almost completely<br />

inviable. We screened for suppressors of the ptp-1(op147);vab-2(ju1) lethality at 25 0 C and isolated at<br />

least 19 suppressors from a screen of over 67,000 F1 animals. The suppressors are of two classes: those<br />

that reduce the lethality and those that suppress both the lethality and the notched head phenotype of<br />

vab-2. Five suppressors of the second class map to LGIV and are dominant suppressors of vab-2.<br />

Intriguingly, these suppressors display a low penetrance notched head phenotype (about 2-5% of the<br />

animals), however, unlike vab-1 and vab-2 the notch is always on the dorsal side. We are currently<br />

determining the specificity of the suppression effect. We will present results from the characterization of<br />

these suppressors.<br />

121


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

RNAI SCREEN FOR COMPONENTS OF THE C. ELEGANS<br />

MEIOTIC MACHINERY<br />

Mónica Colaiácovo, Gillian Stanfield, Kirthi Reddy, Anne Villeneuve<br />

Dept. of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305<br />

Meiosis is the specialized cell division process by which diploid organisms generate haploid gametes. In<br />

preparation for the meiosis I division, homologous chromosomes condense, recognize each other,<br />

synapse, and undergo recombination. During this process a proteinaceous structure known as the<br />

synaptonemal complex (SC) forms between homologs maintaining them in a side-by-side alignment.<br />

Crossing over occurs in the context of the SC, leading to formation of chiasmata that connect the<br />

homologs and allow them to orient toward opposite poles of the meiosis I spindle.<br />

We are using a functional genomics strategy to identify genes required for these key meiotic prophase<br />

events, taking advantage of a genome-wide survey of germline gene expression conducted by V. Reinke<br />

and collaborators in the lab of S. Kim. We defined a subset of 206 germline-enriched genes whose<br />

expression profiles most closely match those of known meiosis genes, and designed a screen to identify<br />

those genes for which RNAi elicits defects in meiotic prophase. Defects in pairing, synapsis or<br />

recombination that lead to chromosome non-disjunction are initially identified by the production of inviable<br />

embryos and an increased frequency of male progeny. Defects that lead to arrest in meiotic progression<br />

are initially identified by a sterile phenotype. Candidates fitting either of these profiles are then subjected<br />

to cytological analysis to investigate possible meiotic defects. The efficacy of our screening procedure has<br />

been validated using genes encoding known meiotic recombination and synapsis components as positive<br />

controls.<br />

Our initial screening has already identified several distinct phenotypic classes with defects in<br />

chromosome behavior and/or morphology. These define genes with putative roles in meiotic chromosome<br />

synapsis (see accompanying poster by Reddy et al.), as well as genes that may have both mitotic and<br />

meiotic roles. A summary of the results of this ongoing screen will be presented.<br />

122


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

EXPLORING THE ROLE OF PINCH/UNC-97 IN MUSCLE<br />

DEVELOPMENT AND FOCAL ADHESION ASSEMBLY IN<br />

CAENORHABDITIS ELEGANS AND MAMMALIAN TISSUE<br />

CULTURE CELL LINES<br />

Shaun Cordes 1 , May Dang-Lawson 1 , Poupak Rahmani 1 , Linda<br />

Matsuuchi 1 , Donald G. Moerman 1,2<br />

1Department of Zoology, U.B.C., Vancouver, B.C., Canada<br />

2Biotechnology Laboratory, U.B.C., Vancouver, B.C., Canada<br />

PINCH/UNC-97 is a phylogenetically conserved adaptor protein consisting entirely of 5 LIM domains<br />

which has been implicated in the assembly of dense bodies, focal adhesion (FA)-like structures in worm<br />

muscle. An examination of the subcellular localization of UNC-97 in body wall muscles using a full length<br />

UNC-97::GFP reporter construct reveals that the protein co-localizes with integrin-containing attachment<br />

structures and that it is also in the nucleus. Studies of partial and complete loss of function mutations in<br />

unc-97 demonstrate that the protein is necessary for the integrity of FA-like attachment structures found in<br />

nematode body wall muscles (JCB 144: 45-57). We are using a combination of genetics in C. <strong>elegans</strong><br />

and expression studies in mammalian tissue culture cells to further investigate the role of PINCH in<br />

muscle development and FA assembly. Moreover, we are investigating the functional importance of<br />

nuclear-localized UNC-97 in the regulation of these processes.<br />

To identify the regions of PINCH responsible for its localization, we have designed altered UNC-97::GFP<br />

reporter constructs which delete one or multiple UNC-97 LIM domains. Preliminary results in worms<br />

suggest that LIM2 and/or LIM3 are involved in nuclear localization. When a full length UNC-97::GFP<br />

reporter construct is transiently transfected into COS fibroblasts, the pattern of expression in the<br />

cytoplasm is similar to that of the FA associated protein alpha-actinin. Preliminary results in COS cells<br />

suggest that UNC-97 requires LIM1 for cytoplasmic localization to punctate cytoplasmic structures. Our<br />

results suggest that the interaction between PINCH and its LIM1 binding partners (such as the<br />

integrin-linked kinase; Mol. Cell Biol. 19: 2425-34), is important for the recruitment of UNC-97 to specific<br />

locations in the cytosol. Future experiments will explore the localization of our reporter constructs in the<br />

mouse myoblast cell line C2C12.<br />

123


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

THE SAD-1 KINASE REGULATES PRESYNAPTIC VESICLE<br />

CLUSTERING IN C. ELEGANS<br />

Justin Gage Crump 1 , Mei Zhen 2 , Kang Shen 1 , Yishi Jin 2 , Cornelia I.<br />

Bargmann 1<br />

1Howard Hughes Medical Institute and Department of Anatomy, University of California, San Francisco,<br />

CA 94143-0452 USA<br />

2Department of Biology, Sinsheimer Laboratories, University of California, Santa Cruz 95064, USA<br />

Synaptic development is a multistep process that includes the presynaptic clustering of vesicles and<br />

active zone proteins, the postsynaptic clustering of neurotransmitter receptors and regulatory proteins,<br />

and the termination of axon outgrowth as neurons recognize their targets. We report here the<br />

identification of a novel serine/threonine kinase, SAD-1, that regulates several aspects of presynaptic<br />

differentiation in C. <strong>elegans</strong>. In sad-1 mutant animals presynaptic vesicle clusters in sensory neurons and<br />

motor neurons are disorganized and more diffuse. Sensory axons fail to terminate in sad-1 mutants,<br />

whereas overexpression of SAD-1 causes axons to terminate prematurely. SAD-1 is related to PAR-1, a<br />

kinase that regulates cell polarity during asymmetric cell division. SAD-1 is expressed in the nervous<br />

system at the time of synaptogenesis and localizes to synapse-rich regions of the axons, where it can<br />

function cell-autonomously in presynaptic neurons. Strikingly, overexpression of SAD-1 can induce the<br />

formation of well-organized, evenly spaced vesicle clusters in dendrites, which are normally devoid of<br />

synaptic vesicles. These results reveal a potent function for sad-1 in specifying synaptic regions in<br />

polarized neurons. We have also started to study the molecular events that coordinate the presynaptic<br />

and postsynaptic specialization.<br />

124


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

MUTANTS WITH ALTERED SENSITIVITY TO THE EFFECTS<br />

OF ETHANOL ON LOCOMOTION<br />

Andrew G. Davies, Tod R. Thiele, Catharine Eastman, Steven L.<br />

McIntire<br />

Gallo Center and Program in Biological Sciences, Dept. of Neurology, UCSF<br />

Ethanol is reported to have multiple targets in the nervous system, a factor that has complicated efforts to<br />

understand the actions of ethanol that bring about behavioral effects in mammalian systems. We are<br />

using C. <strong>elegans</strong> to confirm suspected targets of ethanol and identify new targets and pathways that can<br />

be affected by ethanol to produce behavioral changes. We have concentrated on the effects of ethanol on<br />

locomotion. With increasing doses of ethanol in a plate assay, wild-type animals display a decrease in<br />

activity, velocity and amplitude of the body waveform. Non-saturating screens for mutants with altered<br />

sensitivity to ethanol for locomotion have produced eight mutants with increased resistance to ethanol<br />

and four mutants with increased sensitivity to ethanol. We have characterized the locomotion, egg laying<br />

and pharyngeal pumping behaviors of these mutants in the presence and absence of ethanol and<br />

examined nervous system function by pharmacological analysis.<br />

Mapping and cloning of the genes mutated in these strains is underway using positioned Tc1<br />

polymorphisms or single nucleotide polymorphisms that result in alterations to restriction enzyme sites<br />

(thanks to Stephen Wicks for unpublished data). We have identified one of the resistance genes as the<br />

calcium-activated potassium channel gene slo-1. Potassium channels of the slo class are thought to<br />

regulate the excitability of neurons. There is in vitro evidence for a direct activating interaction of ethanol<br />

on channels of this class derived from mammalian systems (reviewed by Dopico et al. 1999, Neurochem.<br />

Int. 35:103-106) raising the possibility that we may have identified a conserved direct target of ethanol in<br />

the nervous system of the worm.<br />

125


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

A SCREEN FOR DD/DV AXONAL MORPHOLOGY DEFECTS<br />

M. Wayne Davis, Erik M. Jorgensen<br />

Department of Biology, University of Utah, 257 South 1400 East Salt Lake City, UT 84112-0840<br />

Growth cones undergo a series of morphological changes as they migrate from the neuronal cell body to<br />

their target. VD motor neuron growth cones slow down and change their shape when they encounter the<br />

lateral nerve cord and body wall muscle (Knobel et al. Development 1999). In order to pass through the<br />

narrow spaces between the body wall muscles and the hypodermis, the growth cone stops and projects<br />

filopodia through these spaces. When the forward tip of one filopodium reaches the other side of the<br />

muscle the growth cone behind the muscle collapses and re-forms at the tip of this projection. The axon<br />

then branches at the dorsal nerve cord and synapses onto the dorsal muscles.<br />

Using an integrated unc-47::GFP construct that specifically labels the DD and VD axons, I screened for<br />

mutations that disrupt DD/VD axon morphology. In this screen, I identified two main classes of mutations.<br />

The mutations in the first class cause wandering of the axons; mutations in the second class cause<br />

premature termination and branching, either at the lateral nerve cord or at the dorsal muscle boundary.<br />

Most of the wandering axon mutations cause an Unc phenotype, and many of these are likely to<br />

represent alleles of previously known genes. However, the premature branching mutants have no other<br />

obvious phenotype. These mutations may identify new genes necessary for migrating growth cones to<br />

deal with obstacles or changes in substrate.<br />

I have done this screen in the CB4856 Hawaiian background. This should allow the use of<br />

single-nucleotide polymorphisms between this strain and N2 to facilitate mapping of the mutations with<br />

subtle phenotypes.<br />

126


SPN-2 AND SPN-3 FUNCTION TO ORIENT THE SPINDLE<br />

DURING EARLY CLEAVAGES<br />

Leah R. DeBella, Lesilee S. Rose<br />

<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

Section of Molecular and Cellular Biology, University of California, Davis CA 95616 USA<br />

Orientation of cell division is critical for partitioning cytoplasmic factors and setting up cellular interactions<br />

necessary for proper development. In C. <strong>elegans</strong>, the first cleavage bisects the long axis of the embryo,<br />

generating the AB and P1 cells. Subsequent cleavages in the AB lineage are perpendicular to the<br />

previous axis of division. Cleavages in the P1 lineage occur repeatedly on the same axis, due to a nuclear<br />

rotation event. Previous studies have shown actin, microtubules, dynein, and components of the dynactin<br />

complex to be required for proper spindle orientation in some cells. However, the precise mechanism by<br />

which spindles align is not understood.<br />

We are studying two genes, spn-2 and spn-3, which are required for proper spindle orientation in<br />

embryos. spn-2 embryos show defects in spindle orientation at the 2 and 4-cell stage in both AB and P1<br />

lineages, while spn-2/sDf130 worms show defects in the generation of fertilized embryos. Together these<br />

phenotypes suggest a continuing role for spn-2 in spindle orientation during embryogenesis, as well as a<br />

role prior to embryonic division. Some spn-3 embryos fail to rotate at the 2-cell stage while others have<br />

ectopic rotation at the 4-cell stage. However, spn-3/sDf127 embryos are defective in positioning the first<br />

mitotic spindle. This data suggests spn-3 plays a role in spindle orientation during the first three divisions<br />

after fertilization.<br />

An examination of polarity markers in homozygous spn mutants indicates these markers are localized<br />

normally prior to abnormal spindle alignment. In addition, cell divisions are asymmetric and cell cycle<br />

timing is normal, suggesting that overall polarity is not affected in these embryos.<br />

Our working hypothesis is that the spn genes play a role specifically in spindle orientation by regulating<br />

interactions between the microtubules and actin cytoskeleton. As a first step towards testing our model of<br />

spn gene function, we are isolating these genes using a positional cloning approach. spn-2 has been<br />

placed within 220 kb and spn-3 within 11 kb. Examination of the spn genes, combined with the analysis of<br />

other genes in our lab, will provide a basis for understanding the mechanism behind spindle orientation in<br />

all blastomeres of the early embryo.<br />

127


MOLECULES ACTING IN PARALLEL WITH UNC-34 TO<br />

CONTROL CELL MIGRATION<br />

Megan Dell 1 , N Chugh 1 , N Hawkins 1 , E Kong 1 , J Hardin 2 , G Garriga 1<br />

1MCB, UC Berkeley<br />

2Dept Zoology, U Wisconsin-Madison<br />

<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

Guidance cues and their receptors direct cell and growth cone migrations by regulating cytoskeletal<br />

dynamics. Several signaling molecules that function downstream of guidance receptors to regulate actin<br />

have been described. We have found that three signaling molecules, UNC-34, the C. <strong>elegans</strong> homolog of<br />

Enabled, WAS-1, a homolog of the Wiskott-Aldrich Syndrome Protein (WASP), and DAB-1, a homolog of<br />

Disabled, act in parallel to regulate cell migrations in C. <strong>elegans</strong>.<br />

Drosophila Enabled (Ena) and its mouse homolog Mena function in axon outgrowth and fasciculation. The<br />

phenotypes of unc-34 mutants show that this gene functions not only in these processes but also in cell<br />

migrations. unc-34 null mutants are viable and display only partially penetrant defects in cell and growth<br />

cone migrations, suggesting the existance of parallel pathways in C. <strong>elegans</strong> that can compensate for the<br />

absence of UNC-34. By conducting RNAi experiments in an unc-34 background, we have found that<br />

WAS-1 and DAB-1 act in parallel to UNC-34.<br />

Because both WASP and Ena have an EVH-1 domain at their N-termini and regulate actin assembly, we<br />

injected was-1 RNA into wild-type and unc-34 mutant animals. While was-1(RNAi) animals appeared<br />

normal, was-1(RNAi); unc-34(null) animals died as embryos due to ventral enclosure defects. Injection of<br />

was-1 RNA into a temperature-sensitive allele of unc-34 resulted in temperature-dependent lethality.<br />

Injection into worms raised at 25° , the restrictive temperature, resulted in embryonic lethality, while<br />

injected worms raised at 20° showed synthetic cell migration defects and partial lethality. These results<br />

show that UNC-34 and WAS-1 act in parallel pathways to control ventral enclosure and cell migration. A<br />

screen for mutations that result in synthetic lethal and migration defects in the unc-34(ts) background has<br />

yielded several potential mutants.<br />

Disabled is a negative regulator of Ena in flies. dab-1(RNAi) animals show defects in embryonic neuronal<br />

migrations, and injection of dab-1 RNA into unc-34(null) mutants results in enhanced migration defects.<br />

This result is different from the genetic evidence in Drosophila in which Disabled mutations suppress the<br />

Ena phenotype and suggests that unc-34 and dab-1 act in parallel pathways to regulate neuronal<br />

migrations in C. <strong>elegans</strong>.<br />

128


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

INSIGHTS INTO THE ROLE OF C. ELEGANS PROTEIN<br />

UNC-119 IN AXONOGENESIS<br />

Chantal Denholm 1 , Wayne Materi 1 , Daniel Gietz 2 , David Pilgrim 1<br />

1Department of Biological Sciences, University of Alberta, Edmonton, Alberta, Canada, T6G 2E9<br />

2Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, Manitoba,<br />

Canada, R3E 0W3<br />

UNC-119 has widespread effects on the development and maintenance of the C. <strong>elegans</strong> nervous<br />

system. Unc-119 mutants have several movement, sensory, and behavioral abnormalities due to<br />

structural and functional defects in the nervous system. Homologues have also been identified in humans,<br />

rat, Drosophila, and zebrafish, and their functional conservation suggests that this protein plays a<br />

fundamental role in neurogenesis.<br />

Human protein HRG4 has 57% identity with UNC-119 and is a novel photoreceptor enriched protein<br />

which begins to be expressed at the time of photoreceptor differentiation in the ribbon synapse. It is<br />

thought to be involved in visual neural signal transmission, however it is also expressed at lower levels in<br />

other tisues including fetal brain.<br />

Although the phenotype of unc-119 mutants has been well described, the biological role of UNC-119 and<br />

HRG4 remains to be determined because their sequences give no clues to their function(s). To gain<br />

insights about how this protein exerts its effects in vivo, yeast-two hybrid screens have been conducted<br />

using UNC-119 and its functional homologue HRG4 to determine with what proteins they interact. Both<br />

screens have yielded good candidate interactions which are consistent with these proteins’ involvement in<br />

neurite outgrowth, and these results, along with possible models of UNC-119/HRG4 function will be<br />

discussed.<br />

129


THE DEFECATION GENE AEX-1 MAY REGULATE A<br />

RETROGRADE SIGNALING PATHWAY AT<br />

NEUROMUSCLULAR JUNCTIONS.<br />

Motomichi Doi, Kouichi Iwasaki<br />

National Institute of Bioscience, Japan<br />

Communication between pre- and postsynaptic cells is crucial for synaptic transmission regulation. In one<br />

regulatory mechanism, retrograde signaling, signals from postsynaptic cells control synaptic connectivity<br />

and transmission of presynaptic cells. Although there is evidence that retrograde signaling occurs at<br />

neuromuscular junctions in C. <strong>elegans</strong> 1) , its mechanism is unknown. Here we report that the defecation<br />

gene aex-1 may be a regulatory component of C. <strong>elegans</strong> retrograde signaling.<br />

aex-1 mutants show phenotypes similar to those in aex-3 mutants, which were consistent with<br />

presynaptic defects. These phenotypes include defecation defects, reduced male mating, and resistance<br />

to the acetylcholinesterase inhibitor aldicarb. Based on these observations we initially hypothesized that<br />

aex-1 encodes a component which functions at presynaptic terminals.<br />

To determine the molecular function of AEX-1, we cloned and characterized aex-1. aex-1 encodes a<br />

novel protein of 1027 amino acids whose C-terminus is similar to the second C2 domain of the mouse<br />

protein Munc-13-4 and the human protein BAP-3. Using GFP fusion constructs we unexpectedly found<br />

that aex-1 was expressed primarily in body wall muscles. Furthermore, aex-1 expression driven by the<br />

muscle-specific unc-54 promoter conferred hypersensitivity to aldicarb but did not change sensitivity to<br />

the acetylcholine agonist levamisole. This suggests that AEX-1 affects presynaptic activities at<br />

neuromuscular junctions.<br />

To investigate whether AEX-1 regulates retrograde signaling, we generated egl-30(gf);aex-1 double<br />

mutants and tested whether or not hyperexcitation of ventral cord motor neurons suppresses the<br />

aldicarb-resistance phenotype of aex-1 mutants. egl-30(gf) expression in ventral cord motor neurons<br />

confers hypersentivity to aldicarb 2) . Our prediction was that if aex-1 functions downstream of ventral cord<br />

motor neurons then the egl-30(gf) mutation would not suppress the aex-1 phenotype. However, the<br />

egl-30(gf) mutation could suppress the aldicarb-resistance phenotype of aex-1 mutants, suggesting that<br />

aex-1 acts upstream of the egl-30(gf) mutation. This observation is consistent with the hypothesis that<br />

AEX-1 regulates retrograde signaling at neuromuscular junctions.<br />

1) Zhao, H. and Nonet, M. L. Development 127 1253-1266 (2000)<br />

2) Lackner, M.R. et al., Neuron 24 335-346 (1999)<br />

@<br />

<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

130


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

COSUPPRESSION IN THE GERMLINE: SILENCING IS<br />

GOLDEN<br />

Abby F. Dernburg 1 , Mónica P. Colaiácovo 1 , Jonathan Zalevsky 2 ,<br />

Anne M. Villeneuve 1<br />

1 Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA<br />

94305-5329<br />

2 Current address: Department of Biochemistry and Biophysics, University of California, San Francisco,<br />

San Francisco, CA 94143-0448<br />

When genes required in the nematode germline are introduced as trangenes, they show notoriously poor<br />

expression. Paradoxically, the presence of such an array in a wild-type worm can have a bizarre<br />

outcome: it can produce specific phenocopy of loss-of-function mutations in the homologous gene. Similar<br />

"cosuppression" or "quelling" phenomena, in which transgenes induce functional silencing of endogenous<br />

genes, have been reported in diverse organisms, from plants to flies to fungi. We have demonstrated the<br />

generality of this phenomenon and characterized its rules. We show that functional repression is not a<br />

consequence of persistent physical association between transgenes and endogenous genes, or of<br />

mutations in affected genes. Constructing transgenes with different regulatory and coding information, we<br />

obtained evidence that cosuppression is likely to require transcription from the array to generate an RNA<br />

mediator that dictates its target specificity. Because this potential RNA involvement is reminiscent of<br />

RNAi, we asked whether mutations that abrogate RNAi have any effect on cosuppression. This analysis<br />

demonstrated that the genetic requirements for these two silencing phenomena do overlap but show key<br />

differences. Specifically, we find that both rde-2 and mut-7 are essential for cosuppression, but that the<br />

function of rde-1 is dispensable.<br />

Transgene-mediated cosuppression provides a valuable alternative to RNAi as a technique for probing<br />

gene function. Targeting interesting candidate genes, we have elicited diverse germline phenotypes,<br />

including defective chromosome synapsis, failures in spindle assembly, or altered meiotic progression.<br />

Because the effects of cosuppression appear to be largely restricted to the germline, this approach can<br />

reveal germline-specific functions for essential genes for which mutation or RNAi would result in lethality.<br />

131


SUR-9 A SUPPRESSOR OF ACTIVATED LET-60(N1046) IN<br />

THE C.ELEGANS VULVA.<br />

Dennis Eastburn, Min Han<br />

<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

HHMI, Department of Molecular, Cellular and Developmental Biology, University of Colorado at Boulder<br />

The adult C. <strong>elegans</strong> vulva is induced by an EGF like signal which activates the Ras/MAPK pathway.<br />

Constitutively active alleles of ras result in a multivulva (Muv) phenotype where numerous pseudovulvae<br />

are formed from vulval precursor cells (VPCs) that are normally not induced in a wild-type background. By<br />

initiating suppressor screens of activated let-60 ras, many previously unknown components of this<br />

pathway have been identified. One gene, sur-9, has been isolated from such a screen and is currently<br />

being cloned and characterized. sur-9(ku258) has been mapped to chromosome III and is currently being<br />

fine mapped. The ku258 allele can suppress the Muv phenotype of let-60(n1046) from 80% to<br />

approximately 1%. In addition, ku258 is dominant with respect to its suppression. Cloning and elucidation<br />

of sur-9’s molecular identity could contribute to our knowledge of a highly conserved and important<br />

biological pathway.<br />

132


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

KNOCKOUTS IN C. ELEGANS: MADNESS AND<br />

METHODOLOGY<br />

Mark Edgley 1 , Erin Gilchrist 1 , Greg Mullen 1 , Bin Shen 1 , Margaret<br />

Kotarska 1 , Don Moerman 1,2 , Steven Jones 3 , Anil Dsouza 4 , Gary<br />

Moulder 4 , Malini Viswanathan 4 , Martin Lansdale 4 , Robert Barstead 4<br />

1Biotechnology Laboratory,U.B.C., Vancoouver, B.C., Canada<br />

2Department of Zoology, U.B.C., Vancouver, B.C., Canada<br />

3Genome Sequence Centre, Vancouver, B.C., Canada.<br />

4Department of Molecular and Cell Biology, OMRF, Oklahoma City, Oklahoma<br />

As the first multicellular metazoan to have its genome sequenced the nematode C. <strong>elegans</strong> offers an<br />

unprecedented opportunity to investigate larger scale developmental problems including tissue formation<br />

and organogenesis. The C. <strong>elegans</strong> genome contains approximately 19,000 ORF’s, of which only a<br />

fraction have been characterized through genetic mutational analysis. Functional analysis of the complete<br />

genome may be feasible using reverse genetic methodology. Random chemical mutagenesis with<br />

trimethylpsoralen and UV irradiation, coupled to PCR amplification using primers to a specific gene, can<br />

allow for targeted gene disruption. Potential deletions are detected using agarose gel electrophoresis.<br />

The approach is sensitive enough to detect deletions in single animals in complex populations. Through a<br />

process of "sib-selection" one can derive a clone of animals bearing a specific deletion. The mutations are<br />

then sequenced and mutations conferring lethality are balanced using (where possible) a GFP-balancer<br />

chromosome.<br />

Our laboratories are part of the C. <strong>elegans</strong> Gene Knockout Consortium, an international group of labs that<br />

do gene knockouts by request from the worm community [see web site:<br />

http://www.cigenomics.bc.ca/<strong>elegans</strong>]. In the past year the consortium has received 662 requests for<br />

targeted gene disruptions. From this request list consortium labs have eliminated the function of<br />

approximately 80 genes using the chemical mutagenesis approach [as of January, 2000]. All mutants and<br />

data provided by consortium laboratories are in the public domain and freely available to all researchers.<br />

133


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

USING DNA MICROARRAYS TO IDENTIFY TARGETS OF<br />

HOMEOBOX GENES IN C. ELEGANS<br />

Andreas Eizinger 1 , Tibor Vellai 2 , Fritz Müller 2 , Stuart K. Kim 1<br />

1Stanford University Medical Center, Developmental Biology, 279 Campus Drive B369, Stanford, CA<br />

94305, USA<br />

2University of Fribourg, Institute of Zoology, Perolles, Fribourg CH-1700, Switzerland<br />

Homeobox genes are necessary to specify anterioposterior identity in metazoa. Despite their importance,<br />

targets of homeobox genes are poorly understood. Using our DNA microarrays, which contain nearly<br />

every gene of C. <strong>elegans</strong>, we wish to identify a comprehensive list of homeobox target genes in C.<br />

<strong>elegans</strong>. Currently we are focusing on the four major HOX genes ceh-13, lin-39, mab-5 and egl-5.<br />

Preliminary data showed that heat shock expression of homeobox genes in embryos cause strong<br />

lethality and malformation of the hatched larvae. This shows that embryonic cells are responsive to<br />

excessive or ectopic HOX gene expression. We will use our DNA microarrays to identify the earliest gene<br />

expression changes following induction of HOX gene expression by heat shock. By comparing the gene<br />

expression profiles from heat shock treatment of HOX transgenic embryos to control strains, we will be<br />

able to separate the heat shock and HOX responses. We are also interested in the specificity of HOX<br />

genes. Elucidation of the targets of each of the four major HOX genes will show which targets are<br />

common between all HOX genes and which are specific to individual HOX genes.<br />

134


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

DED GENES DISRUPT CELL DIVISION TIMING AND<br />

PATTERNING IN C. ELEGANS EMBRYOS<br />

Sandra Encalada, Paula Martin, Jennifer Phillips, Rebecca Lyzcak,<br />

Danielle Hamill, Kathryn Swan, Bruce Bowerman<br />

Institute of Molecular Biology, University of Oregon, Eugene, OR 97403<br />

In early C. <strong>elegans</strong> embryos, asymmetric cell divisions produce descendants with characteristic cell cycle<br />

times. To understand how these differences in cell cycle timing arise, and to investigate their role in<br />

pattern formation, we are characterizing ded (delayed division) mutants, in which embryonic cell divisions<br />

are delayed and cell fate patterning is abnormal.<br />

In genetic screens for both non-conditional and temperature-sensitive embryonic lethal mutations, we<br />

identified 25 ded alleles. Twelve mutations define nine genes (ded-1 through ded-9), based on genetic<br />

mapping and complementation tests. A common phenotype of these mutants is a prolonged three cell<br />

stage due to a delay in the division of P1. The terminal phenotype of some ded mutants resembles skn-1<br />

mutants: they produce differentiated cells but lack pharynx and gut and instead make extra hypodermis.<br />

The skn-1 gene encodes a transcription factor that specifies mesoderm and endoderm fate in the EMS<br />

blastomere in a wild-type 4-cell embryo. The protein PIE-1 normally blocks SKN-1 function in the<br />

germiline precursor P2. Analysis of a ded-1;pie-1::GFP strain shows that PIE-1 accumulates to<br />

abnormally high levels in EMS and C in ded mutant embryos. These observations suggest that PIE-1 is<br />

blocking SKN-1 function in both P2 and EMS. Consistent with this conclusion, ded-1;pie-1 double mutants<br />

resemble pie-1 mutants, producing extra phrynx and intestine. Finally, the delayed cell divisions of ded-1<br />

mutants also result in a loss of asymmetry in the size of the daughters of P1, and in the mislocalization of<br />

P-granules.<br />

We have cloned ded-1, and it encodes the B subunit of the DNA polymerase-primase complex. Based on<br />

RNA interference experiments with other DNA replication genes, the other loci we have identified likely<br />

define a set of DNA replication genes. Based on these results, we conclude that (1) interference with DNA<br />

replication delays the onset of mitosis, suggesting that a checkpoint monitors the completion of DNA<br />

replication, and (2) proper control of cell division timing is important for multiple aspects of asymmetric cell<br />

division in early C. <strong>elegans</strong> embryos.<br />

135


VOLTAGE-DEPENDENT CURRENTS IN HOMOLOGOUS<br />

CHEMOSENSORY NEURONS WITH DIFFERENT FUNCTIONS<br />

IN C. ELEGANS<br />

S Faumont, S.R. Lockery<br />

<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

Institute of Neurosci, Univ of Oregon, Eugene, OR 97405<br />

In C. <strong>elegans</strong>, chemotaxis to the attractant NaCl is largely controlled by a pair of left-right homologous<br />

chemosensory neurons ASER and ASEL. Although the two neurons are bilaterally symetric in their<br />

morphology and pattern of connectivity , recent work argues for functional asymetry between these<br />

neurons in chemotaxis (Pierce-Shinomura et al., abstr. WCWM). Laser ablation of ASER greatly reduces<br />

chemotaxis to Cl- but not Na+; conversely, ablation of ASEL greatly reduces chemotaxis to Na+ but not<br />

Cl-. To determine if this functional asymetry reflects a difference in the ionic currents expressed in the two<br />

neurons, we made whole-cell voltage clamp recordings fron ASEL (n=20) for comparison with previous<br />

recording from ASER (Goodman et al., Neuron 20: 763-72, 1998).<br />

We found that ASEL is similar to ASER in three main respects. First, ASEL exhibits an outward current<br />

activated by depolarisation (-30 to 100 mV) and an inward current activated by hyperpolarisation below<br />

-70 mV. Little or no current is activated in the region between -70 and -30 mV. Second, the outward<br />

current comprises inactivating and sustained components which are probably carried by K+ ions, because<br />

both components are eliminated by substitution of N-methyl-glutamine (NMG) for K+ in the recording<br />

pipette. Third, outward current inactivation is voltage-dependent, because prepulses more positive than<br />

-70mV decreases peak amplitude of the outward current. These results suggest that the functional<br />

asymetry reflect differences in the response to chemical stimuli rather than differences in voltage<br />

dependent ionic current. Support: NIMH 51383 and NSF IBN-9458102.<br />

136


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

VAV IS REQUIRED FOR PHARYNGEAL MUSCLE<br />

CONTRACTION IN C. ELEGANS<br />

R.T. Fazzio, J.E. Mellem, M.C. Beckerle, A.V. Maricq<br />

Dept. of Biology and Huntsman Cancer Institute, Univ. of Utah, SLC, UT 84112<br />

In vertebrates, VAV, VAV-2 and VAV-3 are guanine nucleotide exchange factors for Rho-family GTPases.<br />

Nucleotide exchange is a key mechanism by which Rho GTPases are regulated during cytoskeletal<br />

remodeling. Disruption of this regulation by specific mutations in VAV results in the constituative activation<br />

of Rho-family members and oncogenesis. To better understand the role of VAV in cytoskeletal control, we<br />

have undertaken a genetic and molecular analysis of VAV in C. <strong>elegans</strong>.<br />

We have cloned the C. <strong>elegans</strong> homolog of vav, vav-1, from first-strand cDNA. vav-1 encodes a 975<br />

amino acid protein that shares 34% overall identity with human VAV and contains the predicted exchange<br />

factor domain and other important functional motifs. We have previously reported vav-1 expression in<br />

pharyngeal tissue, body wall muscle, vulval tissue and somatic gonad. In the pharynx, VAV-1 is found in<br />

muscle, in some neuronal cells and in the g1 gland cells. Subcellular localization of VAV-1 will be studied<br />

using a mouse monoclonal antibody generated to this protein.<br />

We have engineered a deletion mutation in vav-1 that removes 85% of the mature protein product. This<br />

disruption yields a pharyngeal contraction defect that ultimately results in early larval lethality. Mutant<br />

animals display asynchronous pharyngeal muscle contraction or, in some cases, complete pharyngeal<br />

paralysis. Electrophysiological analysis of mutant pharyngeal muscle confirms the importance of VAV-1<br />

for synchronous contraction. We can rescue this defect by the transgenic expression of a genomic DNA<br />

fragment that contains the vav-1 open reading frame.<br />

We are currently pursuing experiments designed to provide a better understanding of the vav-1 null<br />

phenotype: Through tissue-specific expression of vav-1 in the null background, and possibly mosaic<br />

analysis, we aim to identify the pharyngeal cells (and other cells) that require VAV-1. In addition, we aim<br />

to complete a mutational analysis of the vav-1 gene to determine the domains/residues of VAV-1 that are<br />

important for function. The generation of multiple alleles in vav-1 will provide the reagents necessary for a<br />

genetic screen designed to identify proteins that function in the VAV signal transduction pathway.<br />

137


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

REGULATION OF C. ELEGANS BODY SIZE BY SENSORY<br />

CUES<br />

Manabi Fujiwara, Hoan Phan, Steven L. McIntire<br />

Gallo Center and Program in Biological Sciences, Dept. of Neurology, UCSF, San Francisco, CA94608<br />

In C. <strong>elegans</strong>, body size may be regulated by the nervous system. Lewis and Hodgkin initially reported<br />

that cilium-defective mutants such as che-2 and che-3 are smaller than wild-type animals. A smaller body<br />

size is also observed in other cilium-defective mutants and in the tax-4 mutant (tax-4 encodes a<br />

cGMP-gated channel that is necessary for chemosensation). Furthermore, DBL-1/CET-1 TGF-b, which is<br />

involved in body size regulation, is expressed in neurons. These observations suggest that if an animal<br />

cannot sense an environmental cue such as food, body size may be reduced through altered neural<br />

activity. Such regulation may be useful if a smaller body is economical. Recently Apfeld and Kenyon<br />

reported that sensory cues also regulate aging of C. <strong>elegans</strong>.<br />

In order to analyze the putative neural regulation of body size, we isolated suppressor mutants of the<br />

che-2 small body size phenotype (chb). 15,000 haploid genomes were screened, yielding 28 candidates.<br />

These suppressor mutants do not suppress the dye-filling defect of che-2, but suppress the small body<br />

size of che-2. Some of these suppressors show the same body size with or without the che-2 mutation in<br />

the background. Such mutants may result from defects downstream of sensory signals. Nine strains of<br />

this category are being mapped using the snipSNP method (Wicks and Plasterk). One of these, chb-1,<br />

maps to the left arm of chromosome IV and may be allelic with odr-9/egl-4. odr-9/egl-4 maps to the same<br />

region. chb-1 has the chemotaxis defect to diacetyl that is found in odr-9/egl-4. chb-1 and odr-9/egl-4 both<br />

have a slightly larger body than wild type. We have mapped chb-1 to a reigion that is less than 100kb and<br />

are attempting to rescue with cosmids covering this region.<br />

138


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

REGULATION OF INTRACELLULAR DYNAMICS OF<br />

MAPKAPK2 IN LIVING C.ELEGANS<br />

Makoto Fukuda, Yves Bobinnec, Eisuke Nishida<br />

Department of Cell and Developmental Biology, Graduate School of Biostudies, Kyoto University,<br />

Sakyo-ku, Kyoto 606-8502, Japan<br />

MAPKAPK2, a target of p38 MAPK, is shown to be exported from the nucleus to the cytoplasm upon its<br />

activation in mammalian cells. This nuclear export is believed to be dependent on its nuclear export signal<br />

(NES). Here, we have examined intracellular dynamics of C. <strong>elegans</strong> MAPKAPK2 (C44C8.6) during<br />

whole processes of development. To visualize subcellular localization of MAPKAPK2 in living C. <strong>elegans</strong>,<br />

we constructed a fusion between GFP and MAPKAPK2 under the control of its own promoter. This<br />

construct was expressed in late embryo, all larval stages and adults. In adults, MAPKAPK2-GFP was<br />

expressed mainly in neuron, neuroblast and vulva. MAPKAPK2-GFP was present mainly in the nucleus of<br />

those cells. However, in response to extracellular stresses that induce activation of P38 MAPK and<br />

MAPKAPK2, MAPKAPK2-GFP was completely exported from the nucleus to the cytoplasm. An<br />

NES-disrupted form of MAPKAPK2 was not exported from the nucleus under any conditions. In addition,<br />

RNAi with C. <strong>elegans</strong> CRM1, which is known as an NES receptor, resulted in the inhibition of nuclear<br />

export of MAPKAPK2. These results define the requirement of the NES of MAPKAPK2 for its nuclear<br />

export. Moreover, we found that MAPKAPK2 was exclusively localized in the cytoplasm in some neuronal<br />

cells under normal condition, suggesting that MAPKAPK2 is activated in these cells. It is possible that<br />

nuclear export (= cytoplasmic localization) of MAPKAPK2 is tightly associated with activation of p38<br />

MAPK and MAPKAPK2. We are currently examining when and where the p38 MAPK/MAPKAPK2<br />

pathway is functioning in living C. <strong>elegans</strong> by this method.<br />

139


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

ROLE OF CKI-1 IN TERMINAL EMBRYONIC<br />

DIFFERENTIATION AND CELL-CYCLE ARREST<br />

Masamitsu Fukuyama, W. Brent Derry, Joel H. Rothman<br />

Department of MCD Biology, UC Santa Barbara, Santa Barbara, CA 93106<br />

The CIP/KIP family of Cyclin-dependent Kinase Inhibitors (CKIs) function to arrest the cell cycle at the<br />

appropriate time during metazoan development. In C. <strong>elegans</strong>, two apparent CIP/KIP CKIs are encoded<br />

by the adjacent cki-1 and cki-2 genes (1). We previously reported that deficiencies that remove both cki<br />

genes cause extra embryonic cell divisions in multiple tissues and accumulation of excess cell corpses,<br />

and that a cosmid containing these genes can rescue these deficiency phenotypes (2). We recently found<br />

that the cki-1 transgene alone can rescue the hyperplastic phenotype of homozygous deficiency embryos,<br />

while the cki-2 transgene shows only very weak rescuing activity, suggesting that cki-1 is essential for<br />

embryonic cell cycle arrest. In support of this hypothesis, cki-1(RNAi) but not cki-2(RNAi) leads to<br />

embryonic hyperplasia in most tissues and the appearance of many cell corpses. Interestingly, embryonic<br />

germ cells arrest the cell cycle in a CKI-1-independent manner.<br />

The CIP/KIP CKIs in vertebrates have been implicated in terminal differentiation as well as exit from the<br />

cell cycle. We previously demonstrated that cki-1(RNAi) causes loss of a differentiation marker in the<br />

adult somatic gonad (3). We recently found that cki-1(RNAi) can eliminate expression of two late<br />

differentiation markers during embryogenesis, one in seam cells and one in the gut, suggesting that cki-1<br />

is required to promote terminal stages of differentiation. We are currently attempting to determine (1)<br />

which stage in differentiation is blocked by loss of cki-1 activity, (2) whether terminal differentiation in the<br />

other lineages requires cki-1 activity, and (3) whether CKI-1 promotes differentiation through a<br />

cyclin-dependent kinase activity.<br />

1. Hong, Y et al. (1998). Development 125, 3585-3597. Feng, H et al. (1999). Nature Cell. Biol. 1,<br />

486-492.<br />

2. Gendreau, SB and Rothman, JH. (1997). 11th International <strong>Worm</strong> <strong>Meeting</strong>.<br />

3. Fukuyama, M et al (1998). 1998 <strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong>.<br />

140


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

SAX-1 AND SAX-2 ACT IN PARALLEL WITH UNC-34 TO<br />

MAINTAIN NEURON POLARITY.<br />

Maria E. Gallegos 1 , Jennifer A. Zallen 2 , Cori Bargmann 1<br />

1HHMI, Dept. of Anatomy, UCSF, San Francisco, CA 94143<br />

2Dept of Molecular Biology, Princeton University, Princeton, New Jersey, 08544<br />

During development each neuron in C. <strong>elegans</strong> extends a precise number of processes from its cell body.<br />

This polarity is established normally in sax-1 and sax-2 mutants, but later in development ectopic neurites<br />

often emerge from the soma of select neurons. In addition, sax-1 and sax-2 mutants have neurons with<br />

enlarged, irregularly shaped cell bodies (Zallen et al. 1999). Whereas similar phenotypes have been seen<br />

in a variety of mutants known to disrupt neuron function, neurons in sax-1 and sax-2 mutants function<br />

normally suggesting that sax-1 and sax-2 may act more directly with the actin cytoskeleton to maintain<br />

neuron polarity.<br />

To ask if the late-emerging neurites in sax-1 and sax-2 mutants result from a deregulated axon guidance<br />

pathway, I made double mutants of sax-1 and sax-2 with various axon guidance mutants including<br />

unc-34, a homolog of Enabled (M. Dell and G. Garriga, pers. comm.). unc-34 lf mutants have axon<br />

guidance and early axon termination defects suggesting that unc-34 functions to direct axon guidance<br />

and promote axon outgrowth during development. Surprisingly, unc-34;sax-1 and unc-34;sax-2 double<br />

mutants are enhanced for the ectopic neurite outgrowth defect. Furthermore, these ectopic neurites<br />

emerge after initial polarity has been established. These results suggest that sax-1 and sax-2 act in<br />

parallel with unc-34 to inhibit ectopic neurite outgrowth. Since the early axon termination defects of<br />

unc-34 suggest that it functions to promote axon outgrowth one interesting possibility is that unc-34 is<br />

bifunctional. These results also demonstrate that unc-34 plays a role during the establishment and<br />

maintenance of neuron polarity.<br />

sax-1 encodes a S/T kinase related to the Ndr protein kinase in humans (62% id.) and flies (60 % id.)<br />

(Zallen and Bargmann submitted). While the function of the Ndr kinases is unknown, other closely related<br />

kinases have been shown to play a role in cell shape and polarity in nonneuronal cells. Since double<br />

mutant analysis places sax-1 and sax-2 in the same genetic pathway, I am in the process of cloning sax-2<br />

in order to identify other components of this kinase pathway. Progress in cloning and additional<br />

phenotypic characterization will be presented.<br />

141


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

IDENTIFYING PHARYNGEAL TARGETS OF PHA-4 USING<br />

DNA MICROARRAYS<br />

Jeb Gaudet 1 , Michael Horner 1 , Stuart Kim 2 , Susan E. Mango 1<br />

1Huntsman Cancer Institute and Department of Oncological Sciences, University of Utah, Salt Lake City,<br />

UT 84112<br />

2Department of Developmental Biology, Stanford University Medical School, Stanford, CA 94305-5427<br />

Development of the C. <strong>elegans</strong> pharynx requires the activity of the gene pha-4. Animals that lack pha-4<br />

activity fail to make a pharynx, while ectopic expression of pha-4 leads to the ectopic production of<br />

pharyngeal markers at the expense of other cell types. These data indicate that pha-4 specifies<br />

pharyngeal organ identity.<br />

The PHA-4 product is a member of the family of winged helix transcription factor and is expressed in all<br />

pharyngeal cells. Several genes are known to be specifically expressed in the pharynx, and at least two of<br />

these (ceh-22 and myo-2) require PHA-4 for their expression. Therefore, pha-4 probably functions by<br />

activating pharynx-specific gene expression.<br />

For pha-4 to direct development of the entire pharynx, it must act either directly or indirectly on all<br />

pharyngeal genes. One possibility is that pha-4 directly activates a small subset of pharyngeal genes,<br />

which then activate other downstream targets. This model proposes a regulatory hierarchy with PHA-4<br />

directly activating those genes immediately downstream of it. Another hypothesis is that pha-4 directly<br />

regulates all pharyngeal genes, whether they act early or late in pharyngeal development. A third<br />

possibility is that pha-4 directly activates a subset of genes at multiple levels in the hierarchy and<br />

indirectly activates others.<br />

To understand how pha-4 directs organogenesis we have identified potential targets of PHA-4 using<br />

microarray experiments. Probes were made from RNA from skn-1 embryos, which produce no pharyngeal<br />

cells, and from par-1 embryos, which produce excess pharyngeal cells.<br />

The results of these experiments have identified ~500 genes whose transcripts are more abundant in<br />

par-1 versus skn-1 embryos. Among these genes are known targets of PHA-4 (such as myo-2) as well as<br />

other genes that are involved in pharyngeal development, including pha-4 itself and ceh-22. We are<br />

currently analyzing the other positives to determine whether they are expressed in the pharynx, and<br />

whether they are direct or indirect targets of PHA-4.<br />

142


AN OVERVIEW OF PREDICTED CYTOCHROME P450 GENES<br />

IN C. ELEGANS<br />

Erin Gilchrist<br />

<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

C. <strong>elegans</strong> Reverse Genetics Core Facility, Biotechnology Laboratory, U.B.C., Vancouver, B.C., Canada<br />

P450 heme-thiolate genes (more commonly known as cytochromes P450) are found in all eukaryotes and<br />

most bacteria and Archaea. They encode a family of heme proteins that are responsible for metabolizing<br />

a wide range of endogenous and exogenous compounds. These enzymes have been implicated in<br />

carcinogenesis, and in the bioactivation or detoxification of many drugs and other xenobiotics.<br />

The <strong>Worm</strong>pep database has identified 75 predicted proteins that encode P450 enzymes in C. <strong>elegans</strong>.<br />

These can be sorted into 16 different classes based on their sequence similarity to each other and to<br />

P450s in other systems. The genes in different classes are likely to be involved in different metabolic<br />

pathways, therefore, I have compared the sequences of the C. <strong>elegans</strong> P450 genes with the known<br />

substrate-binding domains of P450s in other systems. This indicates which processes the different<br />

classes of C. <strong>elegans</strong> proteins may be involved in. Future biochemical and genetic analysis of the C.<br />

<strong>elegans</strong> loci will be used to resolve which amino acids in the substrate-binding domain are essential for<br />

the binding of that substrate.<br />

Several classes of P450 proteins in the worm are believed to be specific to C. <strong>elegans</strong>, or at least to<br />

nematodes, while others have orthologues in vertebrate or other invertebrate systems. Some of the<br />

worm’s P450 genes are more than 90% identical to each other, and several of them are clustered,<br />

suggesting that they evolved by gene duplication of some ancestral heme-thiolate locus. In addition, the<br />

polycistronic nature of some of these clusters indicates that their expression is likely to be coordinated<br />

and that their function may be redundant. I am endeavoring to generate a knockout of one of these<br />

clusters of P450-encoding genes: the C. <strong>elegans</strong>-specific CYP13 cluster on cosmid T10B9. I am also<br />

comparing the ORFs and flanking DNA of the C. <strong>elegans</strong> P450 sequences with sequences from C.<br />

briggsae to predict sequences that are important for regulating the expression of the genes in this family.<br />

143


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

CLUES TOWARD UNDERSTANDING EGF/ WNT SIGNAL<br />

INTEGRATION IN THE SPECIFICATION OF P12 FATE:<br />

ANALYSIS OF THE EGL-5 PROMOTER<br />

Lisa Girard 1 , Henrique B. Ferreira 2 , Scott Emmons 2 , Paul Sternberg 1<br />

1Howard Hughes Medical Institute and California Institute of Technology Pasadena, CA 91125<br />

2Department of Molecular Genetics, Albert Einstein College of Medicine, Bronx, New York 10461<br />

The C. <strong>elegans</strong> Hox gene egl-5 is most similar, based on sequence analysis, to Abdominal-B. Consistent<br />

with its assignment into this paralog group, egl-5 is expressed in the posterior region of the worm.<br />

Immuno-staining results have shown that in the hermaphrodite, egl-5 is expressed in the hermaphrodite<br />

specific neuron, body wall muscle, posterior lateral microtubule neuron, PVC interneuron, M, V6, the<br />

rectal epithelial cells K, F, B, U and the P12 neuroectoblast cell.<br />

The two most posterior P cells are P11 and P12. The anterior products of their first division are both<br />

neuroblasts. P11.p fuses with the epidermal syncytium and P12.p divides again during late L1. The<br />

anterior division products, the epidermal cells P12.pa and P11.p are distinguishable by their distinct<br />

nuclear morphologies and positions.<br />

Earlier ablation experiments have shown that before interdigitating at the ventral midline during early L1,<br />

either of the two most posterior P cells can adopt the P12 fate. Previous genetic analysis indicates that<br />

P12 fate specification requires the synergistic action of the EGF and the Wnt signaling pathways.<br />

Reduction - or loss of function mutations in components of the EGF or the Wnt pathway result in partially<br />

penetrant P12 to P11 or P11 to P12 transformations. Double mutants of EGF and Wnt pathway<br />

components significantly enhance the frequency of transformation. P12 is not specified in an egl-5(lf)<br />

mutant and overexpression of egl-5 can rescue the loss of P12 specification phenotype of let-23 mutants.<br />

In order to understand how information from the EGF and Wnt pathway are integrated at a cis-regulatory<br />

level, we have undertaken an analysis of the egl-5 promoter to determine elements required for egl-5<br />

expression in P12. Do the two pathways converge on the egl-5 promoter or upstream of it? We have<br />

identified an approximately 1.3 kb. fragment of egl-5 promoter sufficient to drive expression of a<br />

heterologous promoter in the cells K, F, B, U, body wall muscle and P12. We are further dissecting this<br />

region in an attempt to identify P12 specific elements. Additionally, we hope to utilize this P12 enhancer<br />

as a tool to identify additional players involved in egl-5 activation in P12.<br />

144


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

SPN-4: A GENE REQUIRED FOR MITOTIC SPINDLE<br />

ORIENTATION IN THE 2-CELL STAGE C. ELEGANS EMBRYO<br />

José E. Gomes, Kathryn A. Swan, Christopher A. Shelton, Bruce<br />

Bowerman<br />

Institute of Molecular Biology, University of Oregon, 1390 Franklin Blvd, Eugene OR 97403<br />

Mitotic spindle orientation is crucial during development for proper segregation of cytoplasmic factors<br />

upon asymmetric cell division. Nevertheless the mechanism underlying the specification of spindle<br />

orientation is not well understood. In the C. <strong>elegans</strong> embryo the P1 blastomere divides asymmetrically<br />

with the mitotic spindle oriented along the anterior-posterior (a-p) axis. The centrosomes, after duplication<br />

and migration, are initially positioned transversely to a-p axis and rotate subsequently, setting up the<br />

spindle longitudinally. The remnant site, a structure derived from the completion of cytokinesis, has been<br />

proposed to be necessary for this centrosome-nucleus complex rotation, by capturing the astral<br />

microtubules, in a process involving the actin cytoskeleton. In addition mutations in the par genes exhibit<br />

defects on spindle orientation in the 2-cell stage suggesting that these genes also play a role in P1<br />

spindle orientation. In genetic screens for non-conditional and for temperature sensitive embryonic lethal<br />

mutations we found three mutant alleles (or25, or80 and or191ts) of a gene, spn-4, exhibiting defects on<br />

mitotic spindle orientation in the P1 blastomere. In spn-4 mutant embryos the P1 spindle fails to set up<br />

along the a-p axis due to a deficiency in centrosome-nucleus complex rotation. In contrast to par mutants,<br />

other aspects of polarity in the 1-cell stage are not affected. Like in wild type embryos the spindle in the<br />

first division is oriented along the a-p axis, AB and P1 blastomeres are unequal in size and divide<br />

asynchronously. Thus spn-4 may be more specifically required for the process of mitotic spindle<br />

alignment in P1. spn-4 maps to linkage group V, position +0.1 map units. We are using a combination of<br />

RNA mediated interference and cosmid rescue to determine its molecular identity. Using the or191ts<br />

allele we have identified set of 8 cosmids that is able to rescue the mutant phenotype.<br />

145


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

CA 2+ -SIGNALLING VIA THE NEURON-SPECIFIC CA 2+<br />

SENSOR NCS-1 IS ESSENTIAL FOR THERMOTAXIS, A FORM<br />

OF ASSOCIATIVE LEARNING AND MEMORY IN C. ELEGANS<br />

Marie Gomez 1 , Edouard De Castro 2 , Ernesto Guarin 1 , Patrick Nef 1<br />

1Department of Central Nervous System, F. Hoffmann-La Roche, Basel, Switzerland<br />

2Department of Institute for Behavioral Genetics, University of Colorado at Boulder, USA<br />

A form of associative memory in <strong>Caenorhabditis</strong> <strong>elegans</strong> is the pairing of the temperature of growth with<br />

food. After conditioning, worms seek food at the temperature of growth in a very precise circular fashion<br />

on a radial gradient of temperature. This phenotype is called the isothermal behavior. Here we show that<br />

NCS-1, a neuron-specific calcium sensor highly conserved throughout evolution, is essential for proper<br />

isothermal behavior. ncs-1 knockout animals are unable to perform isothermal track behavior, although<br />

their loco motor and thermal avoidance behaviors are normal. The knockout phenotype is rescued by<br />

re-introducing wild-type NCS-1, but is not rescued by a loss-of-function form of NCS-1 unable to bind<br />

calcium. Calcium signaling via NCS-1 is therefore essential for proper thermotaxis, a pavlovian form of<br />

associative learning and memory in C. <strong>elegans</strong>.<br />

146


CHARACTERIZING THE NEURAL CIRCUITRY OF<br />

CHEMOTAXIS TO VOLATILE ODORANTS<br />

Jesse Gray, Maria Gallegos, Tim Yu, Cori Bargmann<br />

UCSF and HHMI, San Francisco, CA 94143<br />

The C. <strong>elegans</strong> chemotaxis circuit offers the possibility of applying genetics to understand a functionally<br />

complex neural circuit. Further characterization of this circuit, as well as an in vivo physiological assay,<br />

would help relate new results to knowledge of circuitry in other systems. Anatomically, the chemotaxis<br />

circuit can be defined as having three main layers: sensory neurons, interneurons, and motorneurons.<br />

The sensory input to the chemotaxis circuit is relatively well-understood. As a first step in characterizing<br />

the remainder of the circuit, we are using laser ablations to identify functionally important interneurons,<br />

motorneurons, and muscles.<br />

We are also developing new behavioral assays to more specifically control chemotaxis circuit input and<br />

more directly assay circuit output. By flowing odorants over an airtight plate, we can create a temporal<br />

gradient of volatile odorant. It has recently been shown that ASE-mediated (water-soluble) chemotaxis is<br />

accomplished by modulating the frequency of pirouettes (i.e., omega bends and reversals) (3). Using a<br />

video camera to record worm movement during odorant application and subsequently counting omega<br />

bends and reversals, we have begun to characterize the kinetics of the pirouette response to volatile<br />

odorants.<br />

Finally, we will apply imaging techniques to assay physiology. We have constructed a myo3::cameleon<br />

(1) that expresses in body wall muscles. We will use myo3::cameleon worms to identify muscles<br />

important for omega bends and will follow up by ablating the corresponding muscles and motorneurons.<br />

We hope to extend this analysis to neurons in order to characterize circuit activity during olfactory<br />

stimulation and adaptation. We are also exploring other imaging technologies.<br />

1. Miyawaki et al. (1997) Nature. 388, 882-887.<br />

<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

2. Pierce-Shimomura et al. (1999) J Neurosci. 19(21), 9557-69.<br />

147


STUDIES ON THE NEMATICIDAL BACILLUS THURINGIENSIS<br />

TOXINS<br />

Joel S. Griffitts, Raffi V. Aroian<br />

UC San Diego, Department of Biology<br />

<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

The insecticidal crystal (Cry) proteins produced by the soil bacterium Bacillus thuringiensis have been<br />

used for decades to control herbivorous pests. The ability to produce these toxins in transgenic crop<br />

plants has given rise to more extensive application in recent years. Little has been accomplished in<br />

elucidating the mode of action of Cry toxins, and even less in determining how pests generate resistance<br />

to them. One reason for this is that agricultural pests are generally not amenable to molecular genetic<br />

analysis. That C. <strong>elegans</strong> is susceptible to a subset of the toxins in this family makes it a promising<br />

system in which to approach these questions genetically. We present findings based on two toxins, Cry5B<br />

and Cry21. These toxins are similar in their amino acid sequence (42% identical) and in their destructive<br />

effects on the worm gut. Previously, our laboratory has characterized the susceptibility of C. <strong>elegans</strong> to<br />

Cry5B, and isolated mutations in five complementation groups that confer resistance (see abstract by<br />

Marroquin et al). Here, we discuss the cloning of one of these resistance genes, bre-5. Mutations in the<br />

bre-5 gene result in strong resistance to Cry5B toxin. We have mapped the bre-5 gene to a small interval<br />

on chromosome IV. Based on homology, we have identified a candidate gene in this region. We are<br />

currently working to determine whether this gene, or some other nearby gene, is the bre-5 gene. We have<br />

started to characterize the toxicity of Cry21. It appears to be a more potent nematicide than Cry5B,<br />

inducing remarkable destruction of the gut. Additionally, we have not found animals in large mutagenized<br />

populations which escape the toxicity of Cry21, even over a series of dilutions. The results outlined incite<br />

fundamental questions about what molecules in the worm mediate the toxicity response and the<br />

generation of resistance, and about what element or elements in the two toxins under study account for<br />

the aforementioned differences in spite of overall high sequence similarity. The experimental approach we<br />

have developed will undoubtedly shed light on these questions, and perhaps on issues of normal gut<br />

development and maintenance in C. <strong>elegans</strong>.<br />

148


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

SYNAPTIC LOCALIZATION OF THE GLUTAMATE-GATED<br />

CHLORIDE CHANNEL GBR-2<br />

Maria E. Grunwald, Joshua M. Kaplan<br />

Department of Molecular and Cell Biology, University of California, Berkeley, CA 94720<br />

In C. <strong>elegans</strong> and other invertebrates, glutamate receptors are likely to play a dual role at excitatory, as<br />

well as at inhibitory synapses. Recently, an AMPA-type glutamate receptor (GLR-1) has been shown to<br />

be localized to excitatory synapses (Rongo et al., 1998), and glutamate-gated chloride channels (GluCls)<br />

have been implicated in the formation of inhibitory synapses (Lee et al., 1999). We are interested whether<br />

these different types of glutamate receptors are in fact localized to functionally diverse synapses and if so,<br />

how this differential targeting is achieved.<br />

We are comparing the synaptic targeting of GLR-1 receptors and the glutamate-gated chloride channel<br />

GBR-2, encoded by the avr-14 gene (Dent and Avery, WCWM 1998). Using a GFP-tagged version of<br />

GBR-2, we found that these receptors are expressed in interneurons and localized to distinct synapses<br />

along the ventral cord. In addition, GBR-2 is expressed in motorneurons and in the sensory neurons ALM<br />

and PVD. Thus, compared to GLR-1, which is mainly expressed in interneurons, GBR-2 is more broadly<br />

expressed. We have also compared the subcellular localization of GBR-2 to that of GLR-1. Interestingly,<br />

GBR-2 and GLR-1 receptors are targeted to different synapses. GBR-2 does not co-localize with GLR-1<br />

and only a minor fraction of GBR-2 was found at touch neuron-interneuron synapses, where GLR-1 is<br />

found. However, a significant fraction of GBR-2 was identified at interneuron-interneuron synapses in the<br />

ventral cord. We speculate that these GBR-2-containing synapses correspond to the reciprocal synapses<br />

between forward and backward command interneurons, and that these putatively inhibitory synapses<br />

coordinate transitions between forward and reverse locomotion. We are presently investigating this<br />

hypothesis. Furthermore, we are interested in what causes the differential targeting of GBR-2 molecularly.<br />

Recently, we were able to show that the PDZ protein LIN-10 is responsible for GLR-1 localization (Rongo<br />

et al., 1998). GBR-2 is likely to use alternative pathways for its localization. We are presently searching<br />

for proteins that are responsible for its clustering using EMS mutagenesis, yeast-two-hybrid screening<br />

and co-localization assays with candidate proteins.<br />

149


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

REGULATION AND FUNCTION OF LIN-11 IN C. ELEGANS<br />

VULVAL DEVELOPMENT<br />

Bhagwati P Gupta, Paul W. Sternberg<br />

HHMI and Division of Biology, California Institute of Technology, Pasadena, California, USA<br />

The C. <strong>elegans</strong> hermaphrodite vulva has been a well-established model system to address the<br />

mechanisms of cell fate specification. The highly invariant lineage of the vulval precursor cells (VPCs)<br />

provides an opportunity to study the molecular interactions of genes that control cell types so precisely.<br />

We are interested in understanding the mechanism of the nuclear factors that function to specify the<br />

terminal fate of the vulval cells. Towards this we have been analyzing the role of lin-11 that encodes a<br />

LIM Homeobox family of transcription factor. Mutations in lin-11 gene cause egg-laying defective (Egl)<br />

phenotype. Cell lineage analysis and reporter gene expression experiments suggest its possible function<br />

in the specification of a subset of 2 o vulval cells. In addition, the gene is also required for the normal<br />

development of uterine p precursors that ultimately form vulva-uterus connection, and a subset of<br />

neurons. In order to understand the function and regulation of lin-11, we have characterized its regulatory<br />

region and identified distinct cis-elements for the vulva and uterine - specific function. We have also<br />

re-examined its expression pattern using multiple reporter constructs and the gene function in egg-laying<br />

behavior. Our results show that lin-11 is likely to function in many more cells and at multiple times than<br />

previously reported. In an effort to further understand its function, we have searched the C. <strong>elegans</strong><br />

genome database to identify a putative binding partner of LIN-11 - LBP-1 (LIM Binding Protein-1) - and<br />

currently investigating its function in vulval morphogenesis. We plan to carry out genetic screens using<br />

different genetic backgrounds to identify the upstream and downstream genes of the lin-11 pathway.<br />

These experiments are likely to provide a better understanding of the function of LIM Homeobox family<br />

members that act to specify diverse cell types in multiple organisms.<br />

150


SUR-7, A GENE THAT SUPPRESSES ACTIVATED RAS<br />

Eric Hague 1 , Min Han 2<br />

<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

1Department of MCD Biology, University of Colorado at Boulder<br />

2HHMI, Department of MCD Biology, University of Colorado at Boulder<br />

Suppressors of let-60 (n1046) ras have proven to be important in the regulation of the ras signal<br />

transduction pathway. The sur (suppressor of activated ras) genes include sur-1/mpk-1, sur-2<br />

(downstream of mpk-1), sur-3/ksr-1, sur-6 (a PP2A regulatory subunit) and sur-8 (a leucine rich repeat<br />

containing protein). ksr-1, sur-6 and sur-8 appear to act positively between ras and raf.<br />

I am attempting to clone sur-7using single nucleotide polymorphisms (SNP’s). sur-7 has no phenotype<br />

alone, but suppresses n1046 better than 99%. sur-7 maps between sdc-1 and sup-10 on LGX.<br />

151


CHARACTERIZATION AND SUPPRESSION OF EAT-16;<br />

SAG-1/DGK-1 LETHALITY<br />

Yvonne M. Hajdu-Cronin, Wen J. Chen, Paul W. Sternberg<br />

HHMI and Division of Biology, Cal Tech, Pasadena CA 91125<br />

goa-1 (Ga o) modulates many behaviors in C. <strong>elegans</strong>, including locomotion, egg-laying, and feeding.<br />

Reducing function in goa-1 causes hyperactive locomotion and constitutive egg laying (1, 2); in contrast,<br />

overexpressing the constitutively activated Q205L mutation causes lethargy and cessation of egg laying<br />

(1). Previously, we screened for suppressors of the paralysis of syIs17, an integrated transgene<br />

containing goa-1(Q205L) under control of a heat shock promoter. Hyperactive mutants were isolated in<br />

two genes, eat-16 and sag-1/dgk-1. We found that in addition to suppressing the phenotype of activated<br />

Ga o, eat-16 and sag-1/dgk-1 mutations also partially suppressed the phenotype of several reduction of<br />

function mutations in egl-30 ( C. <strong>elegans</strong> Ga q). eat-16 encodes a regulator of G protein signaling, which<br />

we believe functions as a GAP for EGL-30, and sag-1/dgk-1 encodes a diacyl glycerol kinase (3) which<br />

likely functions to reduce the levels of diacyl glycerol (DAG), a second messenger produced upon<br />

stimulation of PLCb by activated EGL-30.<br />

eat-16(sy438) and sag-1/dgk-1(sy428) double mutants arrest during larval development. This lethal<br />

phenotype is highly penetrant (>99%). We hypothesized that the lethality of eat-16; sag-1/dgk-1 is caused<br />

by excessive levels of second messengers produced downstream of Ga q, such as DAG. In support of<br />

this hypothesis, the triple mutant egl-30(md186) eat-16(sy438); sag-1/dgk-1(sy428) is viable to adulthood<br />

and fertile. To further understand the cause of death and identify more components in the pathway, we<br />

are seeking other suppressors of eat-16; sag/dgk-1 lethality besides egl-30, both by testing mutations in<br />

genes that have already been described, and by screening for new suppressors. To date we have<br />

screened about 7000 genomes and have backcrossed 7 suppressor mutants. We are beginning to<br />

characterize the lethal phenotype by Nomarski optics and plan to determine the site of action.<br />

References:<br />

1. Mendel et al., 1995. Science 267: 1652-1655.<br />

2. Segalat et al., 1995. Science 267: 1648-1651.<br />

3. Nurrish et al., 1999. Neuron 24: 231-242<br />

<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

152


IMPROVED TISSUE PRESERVATION USING METAL MIRROR<br />

FREEZING OR HIGH PRESSURE FREEZING FOR TEM<br />

David H. Hall 1 , Frank Macaluso 2 , Gloria Stepheney 1 , Marie-Christine<br />

Paupard 1<br />

1Center for C. <strong>elegans</strong> Anatomy, Albert Einstein College of Medicine, Bronx NY 10461<br />

2Analytical Imaging Facility, Albert Einstein College of Medicine, Bronx NY 10461<br />

Several recent reports (see below) have demonstrated that C. <strong>elegans</strong> tissues can be very well preserved<br />

for electron microscopy by high pressure freezing (HPF) followed by freeze substitution, perhaps<br />

substantially better than by standard chemical immersion fixation. HPF shows the potential to capture a<br />

more "life-like" view of the worm’s ultrastructure. We have been testing both HPF and a related technique,<br />

rapid freezing on a metal mirror (MMF) followed by freeze substitution. Both methods obtain similar high<br />

quality fixation, although there are some freezing artifacts using the metal mirror device that are<br />

eliminated in HPF. For MMF, live animals are concentrated on a small piece of filter paper and plunged<br />

against a metal mirror at liquid nitrogen temperature. While freezing damage often occurs about 5-15<br />

microns into the worms, some animals are very well frozen throughout. The frozen samples are held at<br />

low temperature and freeze substituted into 1% osmium tetroxide in acetone, then embedded into plastic<br />

resin and cured for thin sectioning. For HPF, we have tried two methods to concentrate live animals into<br />

small metal planchette, either holding the animals within fine strands of dialysis tubing (C. Lavin, pers.<br />

comm.), or mixing them into a slurry of yeast paste to form a space-filling solid support (McDonald, 1999).<br />

Examination of fast-frozen specimens by TEM reveals excellent views of membrane events and<br />

organelles. For instance, we see many omega figures on coelomocytes which are indicative of active<br />

endocytosis, events which are not commonly captured by chemical fixation. Synaptic active zones and<br />

vesicles are well preserved, as are their relationships to microtubules. A network of microtubules can also<br />

been seen extending to the periphery of hypodermis. Basal laminae look strikingly different, much looser<br />

and more mesh-like when compared to chemical fixation. Sample images are shown on our website<br />

[www.aecom.yu.edu/wormem/new.html].<br />

These two preparation methods, HPF and MMF, also hold great promise for high resolution immuno-EM.<br />

By reducing the osmium content and adding a dilute aldheyde fixation to the freeze substitution medium,<br />

we can obtain better resolution than is currently possible by our microwave technique. We have<br />

successfully localized epitopes in thin sections from HPF samples. MMF equipment is available here at<br />

Einstein campus. We are conducting HPF trials with the help of Stan Erlandson and Ya Chen at the<br />

University of Minnesota. As our skills improve, we will be happy to offer such services to the C. <strong>elegans</strong><br />

community.<br />

For further information on HPF, we recommend the following sources:<br />

Colleen Lavin’s website at www.geology.wisc.edu/~uwmr/caoting.html<br />

Martin Muller’s website at www.em.bio.ethz.ch/<br />

<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

Kent McDonald, Methods in Molecular Biology, vol 117, pp. 77-97 (Human Press) 1999.<br />

In the U.S., there are HPF machines open to the outside users in Madison, Berkeley, Minneapolis and<br />

Albany.<br />

153


ROLE OF THE CAENORHABDITIS ELEGANS HOMOLOGS OF<br />

CDK5 AND P35 IN MIGRATION AND AXON OUTGROWTH<br />

Thomas Harbaugh, Gian Garriga<br />

Department of Molecular and Cell Biology, 401 Barker Hall, University of California, Berkeley, CA 94720<br />

While cdk5 was originally identified in vertebrates based on its homology to the cell cycle regulated cdc2,<br />

further study has demonstrated a function not in the cell cycle but instead in post-mitotic neurons. Mouse<br />

knockouts of cdk5 and its activator, p35, result in defects in the pattern of neuronal migration in the cortex<br />

(1,2). Studies in cultured rat neurons demonstrated that the cdk5/p35 kinase activity can promote neurite<br />

outgrowth (3).<br />

The C. <strong>elegans</strong> homolog of cdk5 is 74% identical to mouse, and the p35 homolog contains a 159 AA<br />

region that is 54% identical. Translational GFP reporter constructs for cdk5 and p35 are expressed in the<br />

cytoplasm of most neurons beginning around the two-fold stage of embryogenesis and continuing into<br />

adulthood.<br />

Simultaneous overexpression of both cdk5 and p35 produces a strongly uncoordinated phenotype, with<br />

defects in fasciculation and axon pathfinding. Along with the expression pattern, these phenotypes<br />

suggest a role for cdk5 and p35 in neuronal development.<br />

The mig-18(k140) mutation was identified based on defects in distal tip cell migration (4) and may<br />

represent a Ce-cdk5 allele. Extrachromosomal arrays containing the Ce-cdk5 genomic region can<br />

partially rescue the distal tip cell migration defect of mig-18(k140). Furthermore, sequencing of the<br />

Ce-cdk5 genomic region revealed a point mutation predicted to change a conserved histidine into a<br />

tyrosine. To confirm that k140 is a Ce-cdk5 allele, we plan to isolate additional mig-18 mutants and<br />

determine whether they contain Ce-cdk5 molecular lesions. Also, we will screen a deletion library for<br />

alleles of Ce-cdk5 and its activator Ce-p35.<br />

(1) Gilmore EC, Ohshima T, Goffinet AM, Kulkarni AB, Herrup K, J Neurosci, 18:6370-7 (1998)<br />

(2) Chae T, Kwon YT, Bronson R, Dikkes P, Li E, Tsai LH, Neuron, 18:29-42 (1997)<br />

(3) Nikolic M, Dudek H, Kwon YT, Ramos YF, Tsai LH, Genes Dev, 10:816-25 (1996)<br />

(4) Nishiwaki K, Genetics, 152:985-97 (1999)<br />

<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

154


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

REGULATION OF EGG-LAYING BY SENSORY CUES<br />

Laura Anne Hardaker, William R. Schafer<br />

Dept. of Biology, University of California at San Diego, La Jolla, CA 92093-0349<br />

Egg-laying in C. <strong>elegans</strong> is regulated by sensory cues. For instance, it has been found that food<br />

deprivation increases the length of the inactive phase of egg-laying. However, the exact mechanisms by<br />

which this occurs are not known. We are investigating this phenomenon by focussing on two pathways,<br />

the flp-1 pathway and the type II TGF-beta dauer pathway.<br />

The gene flp-1 encodes a precursor of FMRFamide related neuropeptides, and it was shown by Chris Li<br />

to be expressed in a specific subset of head neurons. She found that flp-1 knockouts affect a variety of<br />

behaviors, including egg-laying. When we tested flp-1 worms, we found that they had a lengthened<br />

inactive phase (similar to the food-deprived worms). In addition, their egg-laying rate did not change<br />

depending on the presence or absence of food. Ablation experiments have shown that the effects of flp-1<br />

encoded peptides on egg-laying are independent of the presence of the HSN. Thus, we believe that the<br />

effects of FLP-1 may be mediated, at least in part, by a hormonal mechanism.<br />

Jim Thomas’ lab showed that the type II TGF-beta daf (dauer formation) mutants also displayed defects in<br />

egg-laying. As the Ruvkun and Riddle labs found that the TGF-beta pathway functions in sensory<br />

neurons, it seems to be a reasonable candidate to be involved in the regulation of egg-laying by sensory<br />

cues. Interestingly, we found that the egg-laying patterns of the Daf-c mutants (daf-1, daf-4, daf-7, and<br />

daf-8) are similar to that of flp-1: all have a lengthened inactive period of egg-laying. In addition, the<br />

egg-laying of daf-4 and daf-8 (but not daf-1 or daf-7) is unable to be modulated by the presence or<br />

absence of food. Furthermore, we found that the Daf-d mutation in daf-3 does not cause a defect in food<br />

modulation, and in fact has the ability to suppress the food insensitivity of daf-4.<br />

These results suggest that there may be two hormonal pathways involved with relaying sensory cues to<br />

the egg-laying circuitry, one involving flp-1 and the other involving the Daf-c genes. We are currently<br />

investigating connections between the flp-1 and daf pathways by constructing double mutants, and the<br />

results will be presented.<br />

155


CHARACTERIZATION OF THE C. ELEGANS<br />

SEROTONIN-SYNTHETIC AROMATIC AMINO ACID<br />

DECARBOXYLASE GENE BAS-1<br />

Emily Hare, Curtis M. Loer<br />

<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

Dept. of Biology, Univ. of San Diego, 5998 Alcala Park, San Diego, CA 92110<br />

We are attempting to identify mechanisms by which neurons choose a neurotransmitter during<br />

development. We are currently characterizing genes used by the serotonergic neurons in C. <strong>elegans</strong> to<br />

learn how they are regulated. <strong>Worm</strong>s with mutations in the bas-1 gene (biogenic amine synthesis<br />

abnormal) are serotonin- and dopamine-deficient. Wildtype serotonin can be restored by the application of<br />

exogenous serotonin, but not its immediate precursor 5-HTP; this phenotype is consistent with the loss of<br />

serotonin-synthetic aromatic amino acid decarboxylase (AAADC) activity. We have previously shown that<br />

wildtype serotonin immunofluroescence can be rescued in bas-1 mutants by injection of a 15.1 kb<br />

genomic clone (C05D2XN) containing two adjacent AAADC homologous sequences. The genomic<br />

structure of the region suggests the two AAADC sequences (called C05D2.4 and C05D2.3) will be<br />

transcribed together as an operon (e.g., the two coding regions are less than 400 bp apart). The pair of<br />

sequences must be from an ancient gene duplication since the predicted amino acid sequences are only<br />

66% identical. Interestingly, the second sequence (C05D2.3) is missing a highly conserved segment of<br />

protein, suggesting it may not be able to function as an AAADC. The existence of a partial cDNA for<br />

C05D2.3, however, does demonstrate the gene is expressed. We are currently attempting to determine<br />

which of the two predicted sequences is necessary for serotonin synthesis and how the AAADC gene is<br />

expressed and regulated. Two bas-1 mutant alleles sequenced to date contain point mutations in<br />

C05D2.4 coding sequence resulting in premature stop codons. Furthermore, a construct in which<br />

C05D2.4 is mutated does not rescue bas-1 mutants whereas a construct mutated in C05D2.3 does<br />

rescue bas-1 mutants. Our preliminary results strongly suggest that C05D2.4 constitutes the bas-1 gene;<br />

we are still attempting to determine what role C05D2.3 might play. This work is supported by the Fletcher<br />

Jones Foundation and an NIH (AREA) Grant.<br />

156


XOL-1 FILES<br />

<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

Christian A. Hassig, Barbara J. Meyer<br />

HHMI and Department of Molecular and Cell Biology, University of California, Berkeley, CA 94720<br />

Sexual differentiation in C. <strong>elegans</strong> is mediated by the developmental switch gene xol-1. XOL-1 represses<br />

one or more of the hermaphrodite-specific sdc genes or products, thereby directing the male modes of<br />

sex-determination and dosage compensation. XOL-1 has no known homologs and the primary structure<br />

offers no clues towards understanding its molecular function. Experiments are under way to determine the<br />

precise molecular mechanism(s) of this biological alien.<br />

The primary sex-determination signal (X:A ratio) is transduced by a set of signal elements, including sex-1<br />

and fox-1, that determine the level of XOL-1 protein. This X chromosome counting mechanism results in<br />

10-fold higher XOL-1 levels in XO versus XX embryos. Genetic studies suggest that both sex-1 and fox-1<br />

function as xol-1 repressors even in XO embryos. Consistent with these results, our antibodies detect<br />

XOL-1 in XO embryos carrying reduced copies of sex-1 (and 2 extra copies of xol-1), but not in wildtype<br />

XO embryos. Staining occurs in nearly all nuclei of embryos between the 100-500 cell stage. Similar<br />

experiments are being performed with fox-1 mutants. These results will establish a direct correlation<br />

between X-signal element dose and XOL-1 levels.<br />

Experiments are consistent with a role for XOL-1 as a repressor of sdc-2. Several features distinguish<br />

SDC-2 from other SDC proteins and make it a likely target for XOL-1 regulation: 1) SDC-2 directs the<br />

assembly of the dosage compensation complex on the hermaphrodite X chromosome, 2) SDC-2 is<br />

undetectable in wildtype XO embryos, 3) overexpression of SDC-2 causes male-specific lethality. These<br />

results suggest that XOL-1 could interfere with the expression of sdc-2 and thereby inhibit the formation of<br />

the dosage compensation complex in XO embryos.<br />

Our experiments using transgenic strains further support a role for XOL-1 in repressing sdc-2.<br />

Hermaphrodite lethality caused by overexpression of XOL-1 is partially suppressed by SDC-2 expressed<br />

from a heterologous promoter. This suppression is not seen with overexpression of SDC-3 and suggests<br />

that XOL-1 might regulate sdc-2 at the transcriptional level. Transgenic lines will be used to determine if<br />

XOL-1 associates with the sdc-2 locus in vivo. Results from these experiments will direct future inquiries<br />

into the XOL-1 Files.<br />

157


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

Y41G9A.1, THE C. ELEGANS HOMOLOGUE OF TG737, IS<br />

EXPRESSED IN CILIATED NEURONS<br />

Courtney J. Haycraft 1 , Patrick D. Taulman 1 , Stephen M. Krum 1 ,<br />

Bradley K. Yoder 2<br />

1MCLM 639, 1530 3rd Ave. South, Birmingham, AL 35294-0005<br />

2MCLM 656, 1530 3rd Ave. South, Birmingham, AL 35294-0005<br />

The C. <strong>elegans</strong> gene Y41G9a.1 encodes the homologue of the mouse gene Tg737 which was identified<br />

through a BLAST search of the C. <strong>elegans</strong> genome database. Y41G9a.1 (CeTg737) was isolated from a<br />

mixed stage C. <strong>elegans</strong> cDNA library. CeTg737 encodes a predicted 823 amino acid protein with 45%<br />

identity to the mouse protein (Polaris). As determined for Polaris, CeTg737 contains two blocks of<br />

tetratricopeptide motifs (TPR) thought to be involved in protein-protein interactions. Northern blot analysis<br />

reveals a 2.6 kb transcript with the highest level of expression during embryogenesis and early larval<br />

stages. Transcriptional Y41G9a.1::GFP fusions using 400 bp upstream of the predicted ATG show<br />

expression in ciliated neurons including the amphids and phasmids. Analysis of the promoter region<br />

identified a potential X-box sequence approximately 100 bp upstream of the ATG. X-boxes are targets for<br />

the DAF-19 transcription factor and have been identified in the promoters of several genes involved in<br />

sensory cilia formation or maintenance. The role of DAF-19 in regulation of CeTg737 is currently being<br />

explored. Consistent with GFP expression analysis, monoclonal antibodies raised against recombinant<br />

CeTg737 detect a protein in the dendrites and cilia of sensory neurons and the sensory rays of the male<br />

tail. Overall the results in the worm are in agreement with our analysis in the mouse where Polaris<br />

localizes to the base of cilia and its disruption results in a loss of cilia.<br />

158


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

LET-381 IS A FORKHEAD GENE<br />

Marika Hellqvist-Greberg 1 , Ann M Rose 2 , David L Baillie 1<br />

1IMBB, Simon Fraser University, Burnaby, B.C, V5A 1S6, Canada<br />

2Dep.of Medical Genetics, UBC, 6174 University Blvd., Vancouver, B.C V6T 1Z3 Canada<br />

Forkhead genes encode transcription factors characterized by a highly conserved region of 100 amino<br />

acids necessary for DNA binding. Members of the forkhead gene family have been identified in eukaryotic<br />

organisms ranging from yeast to mammals. They have proved to be an important gene family involved in<br />

development and tumorigenesis.<br />

We are studying this gene family in C.<strong>elegans</strong>. We have identified at least 15 members of the forkhead<br />

gene family in the finished sequence of C.<strong>elegans</strong>. 5 of them have known mutants, daf-16, pha-4, pes-1,<br />

unc-130 and lin-31.<br />

RNAi was used for three of the forkhead genes not yet described, T14G12.4, F26B1.7 and K03C7.2.<br />

F26B1.7 had a lethal phenotype that resembled the phenotype of let-381 identified in Ann Roses lab.<br />

Therefore we sequenced one of the alleles of let-381, h107, and found a point mutation in the splice site<br />

of exon 3 in the F26B1.7 gene. We are now analyzing the phenotype and the expression pattern in order<br />

to understand the function and identify target genes for this forkhead protein.<br />

159


GENETIC ANALYSIS OF DYNAMIC SEARCH BEHAVIOR IN C.<br />

ELEGANS<br />

T.T. Hills, F. Adler, A. V. Maricq<br />

<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

Dept. of Biology, University of Utah, Salt Lake City, UT 84112<br />

We are interested in identifying genes that control dynamic behavior modification underlying exploratory<br />

search in C. <strong>elegans</strong>. Immediately following removal from food, worms engage in an area-restricted<br />

search (ARS), characterized by an initial high frequency of sharp turns that then decays over time. This<br />

causes the search path to shift over time from local and overlapping to globally directed with infrequent<br />

overlap. ARS is found in a wide variety of animals and is theoretically supported as an efficient search<br />

strategy for finding resources that are distributed in patches. Using motion-sensitive computer analysis,<br />

we observed worms immediately after removal from food and quantified the temporal dynamics of their<br />

search paths including such features as velocity, radial displacement, and turning angle distribution.<br />

These features change continuously with time, but stabilize by approximately 40 minutes. This search<br />

behavior is experience dependent, changing significantly in a predictable way across worms reared in<br />

different resource distributions. <strong>Worm</strong>s grown in liquid also display ARS, suggesting a genetic<br />

predisposition for the behavior.<br />

To identify genes controlling this dynamic behavior modification, we quantified ARS in worms with<br />

mutations in candidate genes. Mutations in the glutamate receptor glr-1 and the amino acid<br />

decarboxylase bas-1 cause significant changes in the dynamic aspects of ARS, suggesting that the<br />

dynamics of ARS are modified by mutations in a single gene. To further identify genes controlling this<br />

behavior, we are developing a genetic screen to isolate mutants that lack the local search following<br />

removal from food. We are isolating worms that disperse to a point significantly farther than wild type<br />

following removal from food. bas-1 mutants, which lack the local search, are being used to calibrate the<br />

distance and time that provides the most efficient separation between wild type and potential mutants. In<br />

conclusion, the area-restricted search phenotype has been quantified in C. <strong>elegans</strong>, shows predictable<br />

interactions with the environment, and appears to be under the control of specific genes which we are<br />

currently in the process of identifying.<br />

160


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

MULTIPLE ROLES FOR THE RAS-MAPK SIGNAL<br />

TRANSDUCTION PATHWAY IN CHEMOTAXIS TO<br />

ODORANTS?<br />

Takaaki Hirotsu 1 , Satoshi Saeki 2 , Yuichi Iino 1<br />

1Molecular Genetics Research Laboratory, The University of Tokyo, Tokyo 113-0033, JAPAN<br />

2Present address: Tokyo Research Laboratories, Kyowahakkokogyo Co. Ltd., Machida 194-8533,<br />

JAPAN<br />

We have previously reported that mutants affected in the Ras-MAPK pathway show defects in chemotaxis<br />

to volatile odorants. Experiments in which let-60 ras was expressed using a heat shock promoter and a<br />

cell-specific promoter indicated that the normal activity of LET-60 Ras is required in mature olfactory<br />

neurons.<br />

To determine how the Ras-MAPK pathway is activated in olfactory neurons, we observed accumulation of<br />

activated MAPK by immunofluorescence. The activation of MAPK in the AWC neurons was detected after<br />

10 seconds of application of isoamylalcohol (which is sensed by AWC) at 10 -4 dilution. This activation of<br />

MAPK was dependent on the function of the G protein alpha subunit ODR-3, cyclic-nucleotide gated<br />

channel TAX-2/TAX-4 and the voltage-activated calcium channel subunit UNC-2. These results suggest<br />

that odorant-induced neuronal activity is essential for the activation of the Ras-MAPK pathway.<br />

When animals were exposed to higher concentration (10 -2 dilution) of isoamylalcohol for 10 seconds and<br />

stained for activated MAPK, the AWB neurons, as well as AWC, were stained. Interestingly, the activation<br />

of MAPK in AWC was shut off by longer exposure (for 5min) to 10 -2 isoamylalcohol, while the activation in<br />

AWB was sustained. On the other hands, the application of 10 -4 isoamylalcohol induced activation of<br />

MAPK in AWC, but not in AWB.<br />

AWB neurons are known to mediate avoidance of volatile odorants. Our immunofluorescence analysis<br />

indicated the possibility that high concentration of isoamylalcohol might induce avoidance behavior.<br />

Indeed, worms show avoidance behavior when large amount of undiluted isoamylalcohol is presented.<br />

The let-60(gf), lin-45(lf) and mek-2(0) mutants exhibited defects in this behavior, suggesting that the<br />

pathway is also important for avoidance of odorants.<br />

When wild-type worms are pre-exposed to isoamylalcohol at 10 -4 dilution for 5min, they show avoidance<br />

of isoamylalcohol at the normally attractive concentration. The let-60(lf) mutants showed defects in this<br />

behavior. The Ras-MAPK pathway may also play roles in this behavioral plasticity.<br />

161


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

MAB-26 ENCODES THE C. ELEGANS EPHRIN EFN-4<br />

Thomas Holcomb, Sean E. George, Ian Chin-Sang, Mei Ding, Andrew<br />

Chisholm<br />

Department of Biology, University of California, Santa Cruz, California 95064<br />

In C. <strong>elegans</strong>, Eph signaling functions in embryonic neuroblast movements after gastrulation, and in<br />

epidermal enclosure. The C. <strong>elegans</strong> genome encodes one Eph receptor, VAB-1, and four ephrins:<br />

VAB-2/EFN-1, EFN-2, EFN-3, and EFN-4. mab-26 mutants have defects in male tail morphogenesis<br />

(Chow and Emmons 1994). The predicted ephrin gene efn-4 (aka F56A11.3), was located in the same<br />

region as mab-26. A genomic clone containing F56A11.3 rescued Mab-26 phenotypes in transgenic<br />

arrays. These data combined with identification of mutant alleles demonstrate that mab-26 and efn-4 are<br />

the same gene.<br />

The EFN-4 protein encodes a divergent ephrin. EFN-4 overall is most similar to murine Ephrin-B2 (27%<br />

identity and 45% similarity). Like other worm ephrins, EFN-4 is predicted to be GPI-anchored. Unlike all<br />

other ephrins EFN-4 contains a 24 amino acid insert in the putative receptor-binding domain.<br />

The mutation bx80 is a deletion of second exon of efn-4, and thus is likely a null mutation. Three other<br />

alleles from the Cambridge collection cause alterations in efn-4: e36 causes an early nonsense mutation,<br />

and the weak alleles e660 and e1746 (provided by Jonathan Hodgkin) cause missense alterations in the<br />

receptor binding domain.<br />

The phenotypes of efn-4 mutants indicate that like other eph signaling proteins it is required for embryonic<br />

morphogenesis. Genetic interactions with the other ephrins and vab-1 are inconsistent with EFN-4 acting<br />

only in the VAB-1 pathway (see abstract by Moseley and Chisholm). These observations have led us to<br />

examine the the EFN-4/VAB-1 binding interaction using cell culture experiments. Previously the EFN-1<br />

ephrin was shown to bind VAB-1::AP fusion proteins with high affinity in mammalian cell culture assays<br />

(Chin-Sang et al 1999). We are using parallel approaches to test EFN-4/VAB-1 interactions.<br />

An EFN-4::GFP reporter gene is expressed in a subset of neurons. This expression pattern suggests that<br />

EFN-4/VAB-1 signaling in the embryo occurs between neurons and is thus required non-autonomously for<br />

epidermal development. We will test this idea using genetic mosaic analysis and tissue-specific rescue<br />

experiments.<br />

162


SYD-8, A NEW PLAYER IN AXON GUIDANCE.<br />

Xun Huang, Mei Zhen, Yishi Jin<br />

Department of Biology, University of California, Santa Cruz, CA95064<br />

Growth cones sense and respond to various extracellular cues to be guided to their targets. Many such<br />

cues and their receptors have been identified in recent years, including netrins, slits, and ephrins.<br />

However, the signal transduction pathways of these cues are not well understood. We describe here a<br />

gene that may function in the downstream signaling of axon guidance.<br />

syd-8 (ju39) was isolated in a genetic screen for abnormal synapse mutants 1 . syd-8 (ju39) animals have<br />

wild-type mating, moving and egg-laying behaviors. In syd-8 (ju39) animals, the synaptic GFP puncta are<br />

discontinuous in the dorsal cord. Analysis of axonal morphology in syd-8 mutant revealed several axonal<br />

defects in DD and VD neurons: 1) commissures terminate prematurely before reaching the dorsal cord, 2)<br />

commissures branch or wander to the lateral region, 3) axons defasciculate in both dorsal and ventral<br />

cords. Similar defects are also observed in the DA and DB axons, but to lesser extents. We also observed<br />

defects in pathfinding by the HSNs. In wild type animals, HSNs send out processes along the ventral<br />

nerve cord to the nerve ring . Normally, HSNL extends along the left small bundle of ventral cord. HSNR<br />

extends along the large right bundle of ventral cord. In syd-8(ju39) animals, HSNs have two types of<br />

defects: 1) HSN, mostly the HSNL, wander in the lateral region, and 2) HSNs fasciculate on the ventral<br />

cord. In 20% of syd-8 (ju39) animals, the HSNs are in the large right bundle of ventral cord. The touch<br />

neurons appear to be normal as judged by the Pmec-7-GFP marker.<br />

ju39 is a recessive, partial loss-of-function mutation. We have mapped syd-8 on Chromosome V between<br />

lin-25 and him-5. arDf1, itDf1 and yDf8 complement syd-8; yDf12 and nDf42 fail to complement it. Cloning<br />

of syd-8 is underway and we will report further molecular analysis of syd-8 at the meeting.<br />

1. Zhen, M and Jin, Y. Nature 1999<br />

<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

163


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

ANALYSIS OF GCY-31, A PUTATIVE SOLUBLE GUANYLYL<br />

CYCLASE GENE IN CAENORHABDITIS ELEGANS<br />

Martin L Hudson 1,2 , David S. Karow 1,2 , Michael A. Marletta 1,2 , David<br />

B. Morton 1,2<br />

1 Department of Biological Structure and Function, Oregon Health Sciences University, 611 SW Campus<br />

Drive, Portland, OR. 97201<br />

2 Howard Hughes Medical Institute and Department of Cell & Molecular Biology, University of Michigan,<br />

Ann Arbor, MI 48109<br />

Soluble guanylyl cyclases (sGCs) are involved in many cellular and physiological roles including neuronal<br />

signaling, axon guidance and maintenance of vascular tone. Many of these functions are carried out<br />

through activation of sGCs by the gaseous signaling molecule, nitric oxide (NO). Recently, a non-NO<br />

activated sGC (MsGCb-3) has been isolated from the developing larval nervous system of the tobacco<br />

hawk moth, Manduca sexta. By BLAST analysis, its closest homologue in C. <strong>elegans</strong> is the putative sGC,<br />

gcy-31. Previous work by Yu et al. failed to demonstrate any expression data for this gene using<br />

GFP-reporter constructs. In our hands, gcy-31::GFP constructs show expression in a bilaterally<br />

symmetrical pair of cells in the C. <strong>elegans</strong> head. These have processes that lead anteriorly to the tip of<br />

the pharynx, as well as distinctive circular projections posteriorly into the nerve ring. From this<br />

morphology, we have tentatively identified these as either OLL or IL1 neurons.<br />

cDNAs corresponding to gcy-31 have been isolated and show multiple splice variants. However,<br />

heterologous expression of the cDNAs in COS-7 cells has so far failed to show any cyclase activity. Apart<br />

from MsGCb3, all sGCs previously isolated have been obligate heterodimers. These are composed of an<br />

a and b subunit, hence an absent subunit partner may explain the lack of activity in this assay. To answer<br />

this, we are cloning and mapping the expression patterns of the remaining sGCs in C. <strong>elegans</strong>. Inverted<br />

repeat constructs under the control of heat-shock promoters are also being made to generate in-situ<br />

dsRNA corresponding to gcy-31, allowing us to generate and analyze a null mutation in this gene.<br />

164


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

USING C. ELEGANS TO DETERMINE THE MECHANISM OF<br />

ACTION OF PHARMACEUTICALS AND PESTICIDES<br />

Tak Hung, Ben Burley, Emery Dora, Dan Elkes, Steve Gendreau,<br />

Denise Jacobus, Rachel Kindt, Mark Lackner, Lisa Moore, Scott Ogg,<br />

Dianne Parry, Roxanna Peng, Ellyn Pham, Jenny Kopczynski<br />

Exelixis, Inc., 170 Harbor Way, P.O. Box 511, South San Francisco, CA 94083-0511 (www.exelixis.com)<br />

The primary goal of the mechanism of action program at Exelixis is to identify the cognate targets (binding<br />

partners) of compounds with unknown mechanisms of action. We start the process by identifying<br />

mutations in genes whose activity modulates the response of a model organism -- C. <strong>elegans</strong>, Drosophila,<br />

yeast, algae or plants -- to the drug, insecticide, fungicide or herbicide. We then clone these genes and<br />

their homologs in human or pest species, and use biochemical approaches to distinguish the cognate<br />

targets from the other genes in the response pathways. Once a cognate target has been identified, new<br />

compounds with increased activity, less toxicity and better delivery properties can be developed.<br />

Secondary goals of the program include identifying other genes in the response pathways that may also<br />

be drug or pesticide targets, and identifying genes that mediate undesirable side-effects of drugs.<br />

We have corporate partnerships with Bayer, Pharmacia and Bristol-Myers Squibb to identify the pathways<br />

and cognate targets of pharmaceuticals and agrochemicals. These partnerships typically start with a<br />

feasibility study to establish that a particular model system can be used to determine the mechanism of<br />

action for a particular compound. Roughly half of tested compounds cause genetically tractable<br />

phenotypes in C. <strong>elegans</strong>. Based on the observed phenotypes, mutations and RNAi of candidate genes<br />

in the response pathway are tested for resistance or hypersensitivity to the compound. Also, forward and<br />

reverse genetic screens are carried out to identify target pathways and candidate cognate targets.<br />

In addition to our corporate partnerships, we have an internally funded program to identify the cognate<br />

targets of pharmaceuticals. A significant number of drugs, with annual sales over $100 million, act by<br />

unknown mechanisms. Many more compounds- with unknown mechanisms of action- have demonstrated<br />

promising activities in clinical or pre-clinical studies. The development of second generation drugs with<br />

improved efficacy, fewer side effects, and lower production costs is often hindered by a lack of<br />

understanding of how these compounds act. One advantage of starting with compounds rather than<br />

mutations for target discovery is that we know in advance that at least the cognate targets will be<br />

druggable and therapeutic. We will present our analysis of C. <strong>elegans</strong> phenotypes caused by<br />

oncology-related compounds.<br />

165


IN VIVO CHARACTERIZATION OF THE EFFECTS OF THE<br />

UNC-64(MD130) MUTATION ON ANESTHETIC SENSITIVITY.<br />

Hunt S.J., Mike Crowder<br />

<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

Department of Anesthesiology, Washington University School of Medicine<br />

We have previously shown that mutations in the neuronal syntaxin gene unc-64 profoundly alter the<br />

volatile anesthetic (VA) sensitivity of C. <strong>elegans</strong>. Two hypomorphic unc-64 alleles confer hypersensitivity<br />

to the VAs isoflurane and halothane but a third unc-64 hypomorph is VA resistant. The difference<br />

between the isoflurane EC 50s (the concentration where the effect is half maximal) of the hypersensitive<br />

and resistant alleles is over 30-fold. The high-level resistance produced by the unc-64(md130) mutation<br />

indicates that VAs act specifically through a single major mechanism to disrupt coordinated locomotion in<br />

C. <strong>elegans</strong>. The md130 mutation is a single base change at the splice-donor site of the sixth intron,<br />

causing some aberrant splicing. By RT-PCR, md130 mutants produce truncated RNAs, in addition to<br />

wild-type unc-64 mRNAs. The predicted truncated forms of syntaxin are believed nonfunctional as they<br />

lack the transmembrane and part of the H3 domains proven vital for syntaxin’s ability to function in<br />

neurotransmitter release. The behavioral phenotype of md130 is recessive and similar to other weak<br />

reduction-of-function unc-64 alleles. However, the VA-resistance phenotype of md130 is semidominant<br />

and behaves as a gain-of-function mutation, consistent with the truncated forms acting dominantly to<br />

block VA action. The current studies attempt to address in vivo the molecular mechanism(s) by which the<br />

md130 mutation confers this resistance. A plasmid was constructed containing the full-length genomic<br />

unc-64(md130) sequence and used to transform wild-type and unc-64(null) animals. These animals are<br />

significantly VA resistant. Next we addressed if a truncated product alone could produce the VA<br />

phenotype. We constructed a plasmid to produce solely truncated and not wild-type unc-64 product by<br />

introducing mutations at both the donor and acceptor splice consensus sequence of the sixth intron. This<br />

plasmid was unable to rescue null syntaxin mutants, consistent with no wild-type product being produced;<br />

however it successfully knocked-in VA resistance in both the wild-type and null/wild-type backgrounds.<br />

Additional plasmids for expressing various syntaxin fragments are currently being constructed to define<br />

the specific region(s) of syntaxin that are regulating VA action.<br />

166


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

REGULATION OF THE C. ELEGANS EPIDERMAL GROWTH<br />

FACTOR HOMOLOG LIN-3<br />

Byung Joon Hwang, Paul W. Sternberg<br />

Division of Biology, California Institute of Technology, Pasadena, California 91125<br />

The C. <strong>elegans</strong> lin-3 gene, a homolog of the epidermal growth factors (EGFs), is required for multiple<br />

aspects of C. <strong>elegans</strong> development. LIN-3 is required for vulval induction, ovulation, and cell fate<br />

specification of the P12 neuroblast of hermaphrodites. It is also required for cell fate specification of B cell<br />

lineage in males, and for viability of males and hermaphrodites.<br />

LIN-3 has an extracellular domain with one EGF motif, a transmembrane domain, and a cytoplasmic<br />

domain that is longer than that of most other growth factors containing an EGF motif. Little is known about<br />

the mechanisms that regulate LIN-3.<br />

We are taking two approaches to understand the mechanisms regulating lin-3 expression. First, deletion<br />

analysis of lin-3 regulatory region has been performed to identify cis-acting elements for its tissue-specific<br />

expression. Second, a cis-acting element of lin-3 which is mutated in e1417 allele of lin-3 has been<br />

identified by sequencing the lin-3 regulatory regions. The wild type sequence of e1417 element inserted<br />

into an enhancer assay vector supports expression of lin-3::gfp in anchor cell, but the e1417 mutation<br />

abolishes enhancer activity in this assay. Currently, we are looking for proteins that bind to this<br />

AC-specific element of lin-3.<br />

167


CHARACTERIZATION OF THE REGULATORY ELEMENTS<br />

REQUIRED FOR NEURON-SPECIFIC EXPRESSION OF<br />

SNAP-25 IN THE NEMATODE<br />

Soon Baek Hwang, Junho Lee<br />

<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

Department of Biology, Yonsei University, Seoul, Korea 120-749<br />

SNAP-25 is a presynaptic protein exclusively expressed in the nervous system of the nematode. We<br />

wanted to characterize the regulatory elements that are required for this tissue-specific expression. We<br />

determined the whole sequence of the C. <strong>elegans</strong> SNAP-25 gene including the 5’ upstream region and<br />

the relatively large first intron. In order to obtain a C. briggsae SNAP-25 homolog, we screened a genomic<br />

library and obtained a fosmid clone named G01P23 that contained the full genomic sequence of the C.<br />

briggsae homolog. We obtained the entire sequence of the fosmid from the genome center. As<br />

transformation of G01P23 into the C. <strong>elegans</strong> SNAP-25 mutants complements the mutant phenotypes,<br />

specific transcription factors of C. <strong>elegans</strong> may be able to bind the cis-acting elements in the C. briggsae<br />

SNAP-25 gene, and the cis-acting elements may be conserved in these two species. By both examining<br />

serially-deleted 5’ upstream regions and the first intron of SNAP-25 and comparing the sequences<br />

conserved both in the C. <strong>elegans</strong> and C. briggsae SNAP-25 genes, we were able to identify the<br />

regulatory elements required for neuron-specific expression of SNAP-25. We defined two sequence<br />

motifs in the promoter region and two cis-acting regulatory motifs in the first intron. The -1096-1058 region<br />

of 5’ upstream was required in motor neurons of the body region and the -207-190 region was required in<br />

amphid and phasmid neurons. A 1.3Kb patch of the first intron may act in pharyngeal neurons and<br />

another 1.6Kb patch may act in mechanosensory neurons. We expect that each motif confers binding site<br />

for transcription activator(s) in different subsets of neuron cells, which may differ in cell lineage and<br />

function.<br />

168


ANALYSIS OF 2&DEG; VULVAL LINEAGE EXECUTION<br />

Takao Inoue, Paul W. Sternberg<br />

<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

California Institute of Technology, Division of Biology MC 156-29, Pasadena, CA 91125<br />

During the development of the hermaphrodite vulva, P6.p adopts the 1° cell fate and P5.p and P7.p adopt<br />

the 2° cell fate. Cells that adopt the 2° cell fate divide twice longitudinally, and then in a characteristic<br />

LLTN (P5.p) or NTLL (P7.p) pattern. Five distinct cell types are generated from this lineage; vulA (from<br />

outer L), vulB1 and vulB2 (inner L) vulC (T) and vulD (N). Our goal is to understand the mechanism of 2°<br />

lineage execution; specifically how these cell types are specified through a network of genetic interations.<br />

To facilitate the identification of different vulval cell types, we are assembling a panel of chromosomally<br />

integrated gfp reporter constructs which express in subsets of vulval cells. These markers will be<br />

examined under various conditions (e.g. mutant background and/or laser ablation of key cells) to<br />

determine how they are regulated. Furthermore, to identify genes involved, screens for mutations which<br />

alter the expression pattern will be carried out. Previously, egl-29 mutations were shown to affect the<br />

expression of egl-17::gfp in the 2° lineage (M. Wang and P.W.S). Since this gene may be involved in 2°<br />

lineage patterning, we are cloning it.<br />

169


DEVELOPING A C. BRIGGSAE GENETIC MAP<br />

B. Johnsen 1 , S. Gharib 2 , A. Mah 1 , K. Brown 2 , D. Baillie 1 , P.<br />

Sternberg 2<br />

1Simon Fraser University<br />

2Caltech<br />

<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

We are developing a genetic map of <strong>Caenorhabditis</strong> briggsae in order to facilitate comparative studies<br />

with <strong>Caenorhabditis</strong> <strong>elegans</strong>. The two nematode species are morphologically similar but are<br />

approximately 25 - 40 million years apart, which is about one-half the 60 - 90 million year evolutionary<br />

distance between mice and humans. Insights gained by the studies of the two nematode species should<br />

help in the studies of the two mammals. The 100 million base pair C. <strong>elegans</strong> genome is effectively<br />

completely sequenced revealing approximately 19,000 genes. Over 9.4 million base pairs (about 12%) of<br />

the C. briggsae genome has been sequenced and roughly another 4 million bases pair should be<br />

sequenced over the next year (M. Marra pers. comm.). The two nematodes show 30-50% divergence of<br />

unselected nucleotide sequence. Blocks of unaltered sequence are, presumably, under selective<br />

pressure to remain unchanged. The comparisons of the genetic maps of the two species should reveal<br />

how selective pressure constrains evolution of the linkage groups. For example, in C. <strong>elegans</strong>’<br />

chromosomes there are regions of low and high rates of recombination. Highly conserved genes might<br />

more likely be in regions of low recombination whereas rapidly evolving genes more likely reside in<br />

regions of high recombination. If this were true we would expect a similar gene distribution in C. briggsae.<br />

Although the two species are similar there are some differences. We have noted that C. briggsae matures<br />

quicker and lays its first eggs at about age three days, about one-half day sooner than C. <strong>elegans</strong>. C.<br />

briggsae also lives longer then C. <strong>elegans</strong> (averaging 29.7 days versus 18.6 days @ 20° for our lab’s<br />

strains). We have over 100 C. briggsae visible mutations. So far we have identified 15 cby, 7mip, 1 rot<br />

and 1 sml genes (the C. briggsae versions of dpy, unc, rol and sma respectively). Five of the cby genes,<br />

three of the mips and the rot are on the X-chromosome while the other 10 cbys, 4 mips and sml map to<br />

autosomes. We hypothesize that mip-1 is C. briggsae unc-22 and cby-4 is dpy-1. We have also shown<br />

that mip-1, mip-3, mip-6, cby-7 and cby-8 are linked.<br />

170


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

COENZYME Q AND AGING IN THE NEMATODE<br />

CAENORHABDITIS ELEGANS.<br />

Tanya Jonassen 1 , Pamela L. Larsen 2 , Catherine F. Clarke 1<br />

1UCLA Department of Chemistry and Biochemistry, Los Angeles, CA 90095.<br />

2The USC Andrus Gerontology Center, Los Angeles, CA 90089.<br />

The nematode <strong>Caenorhabditis</strong> <strong>elegans</strong> has been used as a model for the genetic studies of aging.<br />

Mutations in the clk-1 gene of C. <strong>elegans</strong> result in slowed development and rhythmic behaviors, and an<br />

extended life span. The C. <strong>elegans</strong> clk-1 gene is a homologue of COQ7, a gene in Saccharomyces<br />

cerevisiae required for the biosynthesis of ubiquinone (coenzyme Q). Coenzyme Q is a redox active lipid<br />

that functions in the electron transport chains of mitochondria and plasma membranes, and plays an<br />

important role as an antioxidant. The nematode, rat and human homologues of clk-1/COQ7 all function to<br />

restore coenzyme Q biosynthesis in the yeast coq7 null mutant. Given the functional conservation of<br />

yeast rat, human and C. <strong>elegans</strong> CLK-1/Coq7 polypeptides, it is crucial to test whether changes in the<br />

level of coenzyme Q may be responsible for the slowed development, behavior and rate of aging in the<br />

nematode model. We have examined whether mutations in the clk-1 gene effect the level of coenzyme Q<br />

in C. <strong>elegans</strong>. We have found conditions where the level of coenzyme Q affects developmental timing,<br />

behavior and lifespan. The studies to be presented show that the clk-1 mutations do impact the level of<br />

coenzyme Q in the nematode system. This system provides a model that is ideal for evaluating the<br />

relationship between coenzyme Q and aging. These studies also indicate that C. <strong>elegans</strong> provides a<br />

metazoan model uniquely suited to address questions regarding Q uptake, metabolism and redistribution.<br />

171


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

OSM-9 SIGNALING: WHO’S INVOLVED?<br />

Amanda H. Kahn, David Tobin, Cornelia I. Bargmann<br />

UCSF, 513 Parnassus Ave, San Francisco, CA 94143-0452<br />

osm-9 encodes a predicted cation channel that is involved in multiple C. <strong>elegans</strong> sensory modalities. Loss<br />

of osm-9 function eliminates chemotaxis to the AWA-sensed odorant diacetyl, compromises adaptation to<br />

a subset of AWC-sensed odorants, and diminishes avoidance of ASH-sensed noxious stimuli. osm-9<br />

mutants also exhibit reduced AWA expression of a Green Fluorescent Protein (GFP) transgene. OSM-9 is<br />

similar to sensory channels in other species, including the Drosophila TRP/TRPL phototransduction<br />

channels and a novel fly homolog. OSM-9 bears closest homology to the mammalian nociceptive<br />

receptors VR1/VRL-1. Although signaling through the TRP/TRPL channels has been well-studied,<br />

relatively little is understood about the mechanisms of osm-9 signaling. Thus, we are using forward and<br />

candidate genetic approaches to elucidate osm-9 signaling pathways.<br />

We employed the osm-9 AWA GFP phenotype in a visual screen for molecules that interact genetically<br />

with osm-9. The screen utilized an allele of osm-9 containing a charge substitution in a conserved residue<br />

of an ankyrin protein-protein interaction motif. In anticipation of identifying gain-of-function mutations, we<br />

screened for dominant suppressors of this allele -- that is, F1 worms with restored expression of the AWA<br />

GFP transgene. At present, we are mapping and characterizing three highly penetrant dominant<br />

suppressors of osm-9. Interestingly, one mutant (ky440) is suppressed for both the AWA GFP and ASH<br />

avoidance phenotypes of osm-9. We will present recent progress in characterizing and mapping these<br />

mutants.<br />

We have also taken a candidate approach to studying osm-9 signaling, using existing C. <strong>elegans</strong><br />

signaling mutants. The osm-9 AWA GFP phenotype can be suppressed by overexpression of the G_<br />

protein odr-3 and by a gain-of-function allele of the calcium-calmodulin dependent kinase unc-43. Our<br />

provisional model is that ODR-3 is an upstream activator of OSM-9, which allows Ca2+ entry to regulate<br />

UNC-43 and gene expression. We are presently characterizing the effects of these suppressors on osm-9<br />

mediated behaviors.<br />

These studies will intertwine novel and well-studied transduction components in sensory signaling<br />

pathways, with the eventual goal of understanding how C. <strong>elegans</strong> employs similar -- or different -elements<br />

in individual sensory neurons.<br />

172


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

LOOKING FOR SYNERGY WITH PHA-4 ON THE MYO-2<br />

PROMOTER<br />

John Kalb 1 , Pete Okkema 2 , Jim McGhee 1<br />

1Department of Biochemistry & Molecular Biology, University of Calgary, Calgary, Alberta, CANADA<br />

2Department of Biological Sciences, University of Illinois at Chicago, Chicago, Illinois<br />

PHA-4 is a fork head transcription factor that is essential for pharyngeal development. Forced expression<br />

of PHA-4 outside of the pharynx can activate pharynx-specific genes. For example, it has been previously<br />

shown that PHA-4 activates myo-2, pharyngeal myosin, through the C-subelement in the myo-2 promoter.<br />

Ectopic PHA-4 expression leads to strong ectopic expression of a C-subelement regulated reporter gene.<br />

However, it only weakly activates endogenous myo-2; with ectopic expression of PHA-4 throughout the<br />

embryo, ectopic expression of myo-2 is only detected in body wall muscles, showing factors in addition to<br />

PHA-4 are necessary for myo-2 expression. Thus, we suggest that PHA-4 works with co-factors to<br />

activate gene transcription. One likely candidate to be a co-factor with PHA-4 is PEB-1. PEB-1 is a novel<br />

DNA binding protein expressed in the pharynx and was identified by its ability to bind to the C-subelement<br />

in the myo-2 promoter (Thatcher, Fernandez, Beaster-Jones, Haun and Okkema). The binding sites for<br />

PHA-4 and PEB-1 overlap in the C-subelement. To test if PHA-4 and PEB-1 act synergistically via the<br />

C-subelement to activate gene expression, we have been assembling the transcriptional regulatory<br />

network in yeast. We find that both PHA-4 and PEB-1 are (rather weak) transcriptional activators on their<br />

own. However, when both PHA-4 and PEB-1 are expressed together, we find no synergism, either<br />

positive or negative. To continue our investigation into the biochemical basis of PHA-4 action, we have<br />

produced all three forms of PHA-4 in baculovirus to define preferred binding sites.<br />

173


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

INITIAL CHARACTERIZATION OF SOLUBLE GUANYLATE<br />

CYCLASES IN C. ELEGANS<br />

David Karow 1 , Jennifer Chang 2 , Scott Nicholls 2 , Ronald Ellis 3 ,<br />

Martin Hudson 4 , David Morton 4 , Michael Marletta 5<br />

1Cellular and Molecular Biology, University of Michigan, Ann Arbor, MI 48109<br />

2Howard Hughes Medical Institute, University of Michigan, Ann Arbor, MI 48109<br />

3Department of Biology, University of Michigan, Ann Arbor, MI 48109<br />

4Department of Biological Structure and Function, Oregon Health Sciences University, Portland, OR<br />

97201<br />

5Cellular and Molecular Biology, Howard Hughes Medical Institute, University of Michigan, Ann Arbor, MI<br />

48109<br />

Soluble guanylate cyclases (sGCs) catalyze the conversion of GTP to cGMP. In organisms other than C.<br />

<strong>elegans</strong>, biochemically characterized sGCs are heterodimers composed of a1 and b1 subunits. The b1<br />

subunit is responsible for binding heme, which is ligated to a histidine residue (His 105). Nitric oxide (NO)<br />

binds to the heme and stimulates the enzyme 400-fold, increasing cGMP levels.<br />

In contrast to the previously characterized heterodimers described above, analysis of the completed C.<br />

<strong>elegans</strong> genome revealed seven putative sGC b subunits but no a subunits. Each of the putative b<br />

subunits contains the residues necessary for catalysis, binding GTP and binding heme (His 105).<br />

However, no open reading frame for nitric oxide synthase was found. This finding raises the possibility<br />

that these putative b subunits might function as novel NO-insensitive isoforms.<br />

Our preliminary data are consistent with this hypothesis. We have identified NO-insensitive sGC activity in<br />

C. <strong>elegans</strong> lysates. For further analyses, we have begun by focusing on one subunit -- T04D3.4.<br />

Preliminary characterization of the over-produced putative heme domain from T04D3.4 suggests that it<br />

does bind heme. In addition, promoter::GFP fusion studies suggest that this subunit is expressed in<br />

specific ciliated sensory neurons and possibly some mechanosensory neurons.<br />

These cyclases are conserved in Drosophila. They are also more homologous to the relatively<br />

uncharacterized mammalian b2 subunit than to the b1 subunit. If our preliminary evidence proves correct,<br />

these sGCs would define the first class of heme-binding cyclases that are insensitive to NO.<br />

174


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

MULTIPLE REGULATORY ELEMENTS ACTIVATE END-1<br />

EXPRESSION IN THE E LINEAGE<br />

Jodie J. Kasmir, Morris Maduro, Joel H. Rothman<br />

University of California-Santa Barbara, Santa Barbara, CA 93106<br />

end-1 encodes a GATA factor that is sufficient to specify the identity of the sole endoderm precursor, the<br />

E blastomere. By dissecting the end-1 regulatory region, we have identified elements that control the<br />

levels and specificity of end-1 expression.<br />

end-1 is activated by at least one maternal factor, SKN-1. Heat-shock-driven expression of SKN-1 results<br />

in widespread end-1 expression. In addition, end-1 appears to be activated by zygotically expressed<br />

GATA factors: ectopic expression of either END-3, the genetically redundant partner of END-1, or MED-1,<br />

another zygotic GATA factor (see abstract by Maduro and Rothman), also result in widespread end-1<br />

expression. We are testing whether conserved SKN-1 and GATA consensus binding sites are directly<br />

responsive to SKN-1, END-3 and MED-1 action.<br />

While embryos lacking SKN-1 do not express detectable MED-1, end-1 is still expressed, albeit at low<br />

levels, and gut is still made 30% of the time. This observation suggests the existence of yet another<br />

activator of end-1. Previously, we reported that POP-1, a Lef-1 like protein that represses end-1<br />

expression in MS, appears to activate end-1 and gut differentiation in the E lineage in response to the<br />

MAPK/Wnt signal that induces endoderm. However, we have been unable to identify a single region in<br />

the end-1 promoter that is necessary for this POP-1-dependent activation of end-1. Indeed, our genetic<br />

and biochemical data indicate that multiple sites may be necessary for activation of end-1 in the E lineage<br />

by Wnt/MAPK-activated POP-1.<br />

Our further analysis has uncovered several important regulatory elements that are necessary to establish<br />

normal levels of end-1 expression. Some of these elements contain no consensus sites for known<br />

regulators, further indicating the existence of multiple positive inputs.<br />

175


MUTATIONS THAT PERTURB THE EFFECT OF<br />

OCTOPAMINE/SEROTONIN ON PHARYNGEAL ACTIVITY.<br />

John Keane, Leon Avery<br />

University of Texas Southwestern Medical Center, Department of Molecular Biology, Dallas, TX<br />

75390-9148, USA<br />

We have identified some mutants which increase the rate of pharyngeal pumping in C. <strong>elegans</strong>, most<br />

notably during the dauer larval stage. Pharynxes of these mutants (unc-9, unc-7 and goa-1) also respond<br />

abnormally to the application of exogenous octopamine. The neuromodulators serotonin and octopamine<br />

have previously been reported to act antagonistically (1). It is possible that these mutants are either<br />

hypersensitive to serotonin (normally stimulates pumping) or resistant to octopamine (inhibits pumping) or<br />

both. To address this we are examining the effect on pharyngeal activity of mutations which are reported<br />

to reduce octopamine (osm-3, che-3) and serotonin (tph-1) levels.<br />

Efforts to identify where unc-7, unc-9 and goa-1 mediate the observed effect have involved combining<br />

these mutants with eat-2, a mutant that knocks out excitatory synaptic communication between motor<br />

neuron MC and pharyngeal muscle. The double mutants reveal that MC is necessary for the observed<br />

increase in pumping rates. As yet, we have not determined whether this means that the motor neuron is<br />

more active in these mutants or whether the muscle has become more receptive to neuronal stimulation.<br />

As the increase in dauer pumping rate in these mutants is small compared to wild type, it is unlikely that<br />

octopamine has a major role in inhibiting pharyngeal activity during this developmental stage.<br />

(1) Horvitz R. et al. Science 216: 1012-1014 (1982)<br />

<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

176


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

PHEROMONE REGULATION OF NEUROENDOCRINE<br />

OUTPUTS IN C. ELEGANS<br />

Scott Kennedy, Gabriel Hayes, Gary Ruvkun<br />

Dept. of Molecular Biology, Massachusetts General Hospital and Dept. of Genetics, Harvard Medical<br />

School, Boston MA 02114<br />

A major regulator of the decision to enter the dauer stage is the environmental concentration of a<br />

constitutively secreted dauer promoting pheromone. One mechanism by which dauer pheromone controls<br />

dauer formation is via the regulation of the expression levels of the TGF-b like ligand DAF-7. To<br />

determine the mechanism(s) by which pheromone regulates the expression of DAF-7 we undertook a<br />

genetic screen to identify genes that when mutated would lead to both a loss of DAF-7 expression and a<br />

Daf-c (dauer constitutive) phenotype. In order to monitor DAF-7 expression we have utilized a daf-7p::gfp<br />

reporter construct, which is expressed predominantly in the ASI amphid neuron. From an initial screen of<br />

20,000 haploid genomes we identified one mutation, mg295. Mapping data indicates that mg295 maps to<br />

LGV, very near the daf-11 locus. In addition, mg295 fails to complement daf-11 (m47). Consequently we<br />

conclude that mg295 is very likely to be a mutation in daf-11. Daf-11 encodes a guanylyl cyclase , an<br />

enzyme that converts GTP to the intracellular second messenger cGMP. This suggests the possibility that<br />

pheromone signaling in C. <strong>elegans</strong> involves the second messenger cGMP, a molecule previously<br />

identified as essential for mammalian photoreceptor signal transduction.<br />

To further our understanding of the mechanism(s) by which pheromone and daf-11 control daf-7<br />

expression we have begun two additional genetic screens. The first screen seeks to identify mutants that<br />

lead to both a loss of daf-7p::GFP expression and a Daf-c phenotype under more stringent environmental<br />

conditions (27É). We anticipate that this screen will identify additional genes involved in the regulation of<br />

dauer formation and daf-7 expression. Progress on this screen will be reported. We have also begun a<br />

genetic screen to identify mutations that both suppress the daf-11 Daf-c phenotype and lead to a<br />

reversion in daf-7p::GFP expression. From an initial screen of 250,000 haploid genomes we have<br />

identified two mutations; tentatively named PollyAnna-1 (mg296), and PollyAnna-2 (mg297), that satisfy<br />

these criteria. Progress of this screen and on the characterization and mapping of mg296 and mg297 will<br />

be reported.<br />

177


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

CALCIUM IMAGING IN EXCITABLE CELLS OF C. ELEGANS.<br />

Rex Kerr 1 , Varda Lev-Ram 2 , Roger Y. Tsien 2 , William R. Schafer 1<br />

1Dept. of Biology, UCSD, La Jolla, CA 92093-0349<br />

2Dept. of Pharmacology, UCSD, La Jolla, CA 92093-0617<br />

It is desirable in behavioral studies to record the activity of neurons and muscles, but this has traditionally<br />

been difficult in C. <strong>elegans</strong>. We are using a transfectable calcium indicator, cameleon [Miyawaki et al.,<br />

Nature 388:882], to overcome this barrier.<br />

To develop the technique, we focused on the pharyngeal muscle. The pharynx has been previously<br />

characterized behaviorally and electrically, primarily by the Avery lab, and is large enough to simplify<br />

imaging. In addition, calcium influx in muscles causes contraction, which provides an independent<br />

measure of activity. Using the myo-2 promoter, we expressed cameleon in the pharynx and recorded<br />

distinctive calcium transients coupled to muscle contraction in intact animals. The duration of these<br />

transients varied in mutants of egl-19, a voltage-gated calcium channel, consistent with previous electrical<br />

recordings [Lee et al., EMBO 16:1066]. In mutants of unc-36, a channel-associated a2 subunit, we found<br />

that transients were substantially increased in magnitude, suggesting an inhibitory role for the wild-type<br />

UNC-36 protein. This was surprising as coexpression studies of vertebrate homologues in Xenopus<br />

oocytes had found that the a2 had many relatively subtle effects, including increasing expression levels of<br />

the pore-forming subunit. We are currently extending our study of muscular calcium transients to<br />

investigate the effects of neurotransmitters and mutations on the vulval muscles and their role in<br />

egg-laying behavior.<br />

In addition, we expressed cameleon using the pan-neuronal promoter unc-119. We provided direct<br />

electrical stimulation through an extracellular electrode inserted through the cuticle and positioned near<br />

the nerve ring. Transients were observed coupled to the stimulation, indicating that calcium influx through<br />

voltage-gated calcium channels can be observed in neurons using this method. This raises the possibility<br />

of constructing a functional map of neuronal connectivity by electrically stimulating single neurons and<br />

recording the activity of their synaptic partners using cameleon.<br />

178


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

A GENETIC ANALYSIS OF THE EFFECTS OF ETHANOL ON<br />

EGG LAYING<br />

Hongkyun Kim, M. Christina Yu, James Kim, Steven L. McIntire<br />

Gallo Center and Program in Biological Sciences, Department of Neurology, UCSF, CA 94608<br />

Ethanol causes dose-dependent suppression of egg laying in C. <strong>elegans</strong>. To elucidate the neuronal<br />

targets of ethanol, we screened for mutants showing ethanol-resistant or -inducible egg-laying behavior in<br />

a plate assay. F2 progeny of mutagenized animals were exposed to a dose of ethanol that strongly<br />

suppresses egg laying of wild type animals. Eggs were collected and allowed to hatch in the absence of<br />

ethanol. Once the F3 animals had matured to adulthood, the process was repeated for 2 cycles. By using<br />

this enrichment strategy, we isolated four different classes of mutants. Class I mutants are hyperactive<br />

and exhibit hyperforaging behavior and are aldicarb-hypersensitive. At least 4 mutants of class I belong to<br />

the same complementation group and are alleles of slo-1. Class II mutants show high amplitude<br />

sinusoidal tracks and slightly exaggerated head movement in the direction of the forward movement. This<br />

class exhibits variable aldicarb responses and normal foraging behavior. This class includes at least three<br />

complementation groups. The finding of the same behavioral defects in the different mutants of this class<br />

suggests that the same, ethanol-sensitive pathway may be affected by each of the mutations. Class III<br />

mutants have egg-laying defects in the absence of ethanol. Class IV mutants do not have any obvious<br />

phenotype. Egg laying of class I and II mutants is induced at relatively low ethanol concentrations that<br />

moderately suppress egg laying of N2. More eggs are laid in the presence than in the absence of ethanol<br />

in these mutants. Further mapping and characterization of these mutants may lead to the identification of<br />

the molecular targets of ethanol in C. <strong>elegans</strong>.<br />

179


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

GENES AFFECTING THE ACTIVITY OF NICOTINIC<br />

RECEPTORS INVOLVED IN EGG-LAYING BEHAVIOR<br />

Jinah Kim 1 , Daniel S. Poole 2 , Laura E. Waggoner 2 , Alexandra<br />

Treschow 2 , William R. Schafer 2<br />

1Group in Neuroscience, University of California at San Diego, La Jolla, CA 92093-0349<br />

2Dept. of Biology, University of California at San Diego, La Jolla, CA 92093-0349<br />

Egg-laying behavior in C. <strong>elegans</strong> is regulated by multiple neurotransmitters, including acetylcholine and<br />

serotonin. Agonists of nicotinic acetylcholine receptors such as nicotine and levamisole stimulate<br />

egg-laying; however, the genetic and molecular basis for cholinergic neurotransmission in the egg-laying<br />

circuitry is not well understood. We have examined the egg-laying phenotypes of eight known levamisole<br />

resistance genes which may mediate or regulate nicotinic receptor activity in the egg-laying<br />

neuromusculature. Five "strong" levamisole resistance genes, including unc-63, unc-74, and the nicotinic<br />

receptor subunit genes unc-29, unc-38, and lev-1, were essential for the stimulation of egg-laying by<br />

levamisole. In the absence of drug these mutants retained the characteristic biphasic pattern of<br />

egg-laying and caused only subtle shifts in the timing of egg-laying behavior. <strong>Worm</strong>s mutant for two or<br />

three of these nicotinic receptor genes also had a generally wild-type pattern of egg-laying. These genes<br />

appear to encode components of a levamisole-sensitive nicotinic receptor that promotes egg-laying but is<br />

not necessary for egg-laying muscle contraction. unc-29 and unc-38 mutants were also hypersensitive to<br />

the stimulation of egg-laying by serotonin, suggesting that nicotinic receptors might negatively regulate<br />

serotonin response pathways in the egg-laying muscles or neurons. Three "weak" levamisole resistance<br />

genes, lev-8, lev-9, and lev-10, had effects on egg-laying that were distinct from those of the levamisole<br />

receptor genes. Phenotypic analysis indicated that lev-8, lev-9, and lev-10 may encode regulatory<br />

molecules that control the functional activity of nicotinic receptors, since their loss of function attenuates<br />

nicotinic receptor function without seeming to eliminate the receptors themselves.<br />

180


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

SENSORY AXON GUIDANCE DEFECTS IN C. ELEGANS<br />

Susan Kirch, Gage Crump, Cori Bargmann<br />

HHMI and Department of Anatomy; UCSF; San Francisco, CA 94131<br />

<strong>Worm</strong>s depend on taste, touch and smell to sense and explore their environment. Appropriate responses<br />

to information received through these different modalities depends on precise connectivity between<br />

sensory neurons and downstream effector neurons in the nerve ring. Little is known about how axons<br />

migrate through this nerve ring neuropil during development to find their partners.<br />

How is positional information interpreted by an extending axon so that desired synaptic partners are<br />

found? We focused on the guidance of the ASI chemosensory neurons to their appropriate positions in<br />

the nerve ring. A mutant screen led to the isolation of 16 candidate mutant strains that appeared to have<br />

ASI axon guidance and termination phenotypes. These mutations define at least five new genes which<br />

we have named sax-10-14 (sensory axon guidance). We have isolated five alleles of sax-10. The<br />

canonical allele, ky297, has axon defects in several axons in the nerve ring including the chemosensory<br />

neurons AWB, ASI and ASH and the interneuron PVQ, as well as an altered expression pattern of the<br />

AWC- specific gene str-2. ky297 is normal for the VD, DD and HSN motoneurons, the chemosensory<br />

neuron ADL and the phasmids. sax-12 suffers from defects in ASI and ADL, but is normal for AWC<br />

structure. sax-13 displays abnormal axon structures when we examine ASI, ADL, AWB, ASH and PVQ,<br />

but appears wild-type for AWC. Mutants display either one or a combination of phenotypes (including:<br />

premature termination, branching, thickening, wandering and inappropriate pathfinding) with a penetrance<br />

that ranges from 13% to 100%. Dye filling of sensory neurons with the fluorescent dye DiI, and<br />

examination of neurons with other cell-specific GFP constructs in these mutants reveals that the overall<br />

morphology of the nerve ring is normal in many sax-10, sax-12, and sax-13 animals, and that the<br />

mutations disrupting ASI are probably in genes specifically required by ASI or a subset of neurons.<br />

sax-14, however, has more severe dye filling phenotypes that are being characterized further. To facilitate<br />

genetic analysis and cloning of these new sax genes, we are mapping the genes and currently trying to<br />

rescue the sax-10 phenotype by cosmid injections and germline transformation.<br />

181


ISOLATION OF A THIRD LIN-4 ALLELE FROM A LIN-3A<br />

OVEREXPRESSION LINE<br />

Martha Kirouac, Paul Sternberg<br />

<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

HHMI and Division of Biology, 156-29, Caltech, Pasadena, CA 91125<br />

In order to investigate the role of LIN-3 in cell fate specification, we conducted a F2 screen using a<br />

reduction of function lin-3(n378) strain which contains an integrated transgene that carries a mutation that<br />

presumably eliminates lin-3B splicing and should, therefore, only produce lin-3A. The line carrying the<br />

integrated transgene is fertile and 100% multivulva. Animals that lack lin-3A are sterile (Jing Liu & PWS,<br />

unpubl.). To potentiate rescuing any lines that are sterile because they lack lin-3A, lfe-1 was incorporated<br />

into the strain background. lfe-1(sy290) is a gain of function allele that partially rescues the sterility<br />

caused by let-23 loss of function animals. The strain is fertile and 100% multivulva. From these screens,<br />

we looked for animals which were non-Muv and isolated two mutations.<br />

One of these mutations still displayed some induction, but displayed neither invagination nor<br />

morphogenesis. Since these animals were 100% vulvaless, we artifically created a hole connecting the<br />

outside to the uterus using a typical injection scope and needle. After obtaining cross progeny and<br />

outcrossing this mutant, it displayed characteristic heterochronic defects; extra molts, late divisions, and<br />

no adult cuticle. Having preliminarily mapped this mutation to linkage group II, we tested the hypothesis<br />

that it was a lin-4 allele by performing single worm PCR on the homozygous animals using primers to<br />

lin-4. Indeed, there was a mutation in the lin-4 gene, C516T. Though there are only two other alleles of<br />

lin-4 ,this is the second time that this mutation has been isolated (the other allele was isolated in Victor<br />

Ambros lab and is known as ma161). Lin-4 is a heterochronic retarded mutant which is critical to the<br />

controlled coordination of cell division in postembryonic divisions. What is most interesting about lin-4 is<br />

that it encodes a 22 nt. RNA form to block translation of other proteins such as lin-14 and lin-28. The fact<br />

that this allele has been isolated twice is indicative of the importance of this site in the function of the lin-4<br />

RNA. It has been hypothesized (Ha, I. and colleagues) that this C forms a bulge which is recognized by<br />

another, as of yet unknown, factor. When this C is replaced with a T, this bulge does not form, and does<br />

not inhibit the translation of lin-14 and lin-28.<br />

182


ELT-5 AND ELT-6 ARE ESSENTIAL FOR DEVELOPMENT OF<br />

SEAM CELLS, THE VULVA, AND THE MALE TAIL.<br />

Kyunghee Koh, Joel H. Rothman<br />

<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

Neuroscience Research Institute, University of California, Santa Barbara, CA 93106<br />

Patterning of the embryonic epidermis into the major cell types (syncytial, P, and seam cells), and<br />

regulation of their post-embryonic development, involve a complex series of regulatory events. We<br />

previously described the role of elt-5 and -6, adjacent genes encoding GATA transcription factors, in<br />

embryonic seam cell development (WCWM, 1998). We since found that they perform a variety of<br />

essential functions in embryonic and post-embryonic ectodermal development.<br />

elt-5 and -6 show complex and dynamic expression patterns. GFP reporters express 1) in many anterior<br />

and ventral cells during embryogenesis, 2) in seam cells, 3) in a subset of sheath and socket cells, 4) at<br />

low levels in the P cells and higher levels in the descendants of the vulval precursor cells, and 5) in the<br />

developing male tail. Expression patterns of elt-5:gfp and elt-6:gfp overlap, but are not identical. ELT-5<br />

and -6 antibodies corroborate some of these observations.<br />

High levels of elt-5 dsRNA result in penetrant late embryonic or L1 lethality, in which the affected animals<br />

are lumpy, dumpy, uncoordinated, and show fusion of seam cells with epidermal syncytia. Anterior seam<br />

cells fuse more frequently than those in the posterior, suggesting that an A/P patterning system in the<br />

seam or surrounding syncytia regulates their propensity for fusion. Many, but not all seam-specific<br />

markers are eliminated in the affected animals, and elt-3, normally expressed in non-seam epidermis, is<br />

occasionally expressed in unfused seam cells.<br />

At lower doses of dsRNA, surviving vulvaless adults are observed. Examination of the vulval precursor<br />

cells shows that some inappropriately fuse with hyp7. In addition, migration of the gonad is aberrant, and<br />

rays in the male tail are often missing.<br />

Ectopic expression of elt-5 or -6 during mid-embryogenesis results in an excess of seam cells, while<br />

ectopic expression in L1s and L2s often results in adult males with missing rays and hermaphrodites that<br />

herniate through a malformed vulva.<br />

Collectively, these findings suggest that ELT-5 and -6 may collaborate with A/P patterning systems and<br />

inductive processes to regulate development of the embryonic ectoderm and post-embryonic genitalia.<br />

183


A GENETIC SCREEN FOR GENES INVOLVED IN GUT<br />

DEVELOPMENT AND DIFFERENTIATION IN<br />

CAENORHABDITIS ELEGANS<br />

Jay D. Kormish, James D. McGhee<br />

<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, Alberta T2N 4N1<br />

Canada<br />

Our lab is currently interested in studying genes, in particular transcription factors, that direct the<br />

development and differentiation of these cells into a functioning organ. I have developed a genetic screen<br />

to isolate zygotically expressed genes that when mutated result in a gut obstructed "gob" defect. When<br />

mutagenized worms are fed a mixture of bacteria and polystyrene fluorescent microspheres, mutant<br />

worms with an obstructed gut can be identified under the dissecting microscope because of the highly<br />

fluorescent beads that collect in the terminal bulb of the pharynx and in the anterior intestine. The<br />

indication that screening for such a phenotype could isolate genes specifically involved in gut<br />

development came from our observation that mutations in a gut specific GATA transcription factor gene<br />

elt-2 resulted in the gob phenotype. I am currently in the middle of a saturation screen; five thousand<br />

genomes have already been screened and the remaining five thousand are expected to be completed in<br />

the near future. During the characterization of my initial round of candidates, I have found three strains<br />

that appear to give gut specific defects. The strain with the highest degree of penetrance, 26-3(8), has<br />

been chosen for mapping and cloning of the gene. 26-3(8) has been mapped to the X chromosome and is<br />

being tested for complementation with elt-2. Electron microscopy of lumen cross sections and gut<br />

morphology markers are being used to describe the lumen morphology of the mutants.<br />

184


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

AN E1-LIKE ACTIVATING ENZYME IS INVOLVED IN CELL<br />

DIVISION PROCESSES IN THE EARLY C. ELEGANS<br />

EMBRYO.<br />

Thimo K. Kurz, Danielle R. Hamill, Bruce Bowerman<br />

Institute of Molecular Biology, University of Oregon, Eugene, OR 97403<br />

In a screen for temperature-sensitive embryonic lethal mutations, we isolated a mutant called or198ts that<br />

has several defects in early C. <strong>elegans</strong> embryos. In or198ts mutant embryos, the first mitotic spindle often<br />

is delayed in aligning along the anterior-posterior axis, and shortly after the spindle begins to elongate, it<br />

is no longer clearly visible by DIC videomicroscopy. In about half of or198ts mutant embryos, the first<br />

attempt at cytokinesis fails. In embryos in which cytokinesis succeeds, we observe penetrant defects in<br />

nuclear positioning and in mitotic spindle orientation at the two cell stage. In addition, in interphase partial<br />

ectopic cleavage furrows may form; there is extra pinching and blebbing at the cell membranes, and the<br />

cells are misshapen.<br />

or198ts is on LGIII at approximately --0.8 m.u., and corresponds to the Genefinder locus F11H8.1. We<br />

have identified a single base lesion in the mutant that converts a valine to alanine at amino acid position<br />

269. F11H8.1 encodes an E1-like activating enzyme with around 30% amino acid identity to its yeast and<br />

human homologs. E1-like activating enzymes are involved in conjugating ubiquitin-related proteins (e.g.<br />

SUMO-1/Smt3p) to a small subset of other cellular proteins in a manner similar, although not identical to<br />

ubiquitination. In contrast to ubiquitin, which targets proteins for degradation, post-translational<br />

modification by ubiquitin-like proteins may regulate the activity and intracellular localization of the proteins<br />

they modify. In both yeast and mammals, Smt3p/SUMO modifies multiple proteins. Perhaps the<br />

pleiotropic defects observed in or198ts embryos reflect multiple targets of this post-translational<br />

modification. To our knowledge, this is the first example of a ubiquitin-like modification being important for<br />

cell division processes in multicellular eukaryotes.<br />

185


OLFACTORY ADAPTATION<br />

Noelle L’Etoile, Cori Bargmann<br />

<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

HHMI/Dept. Anatomy, UCSF, San Francisco, CA 94143<br />

To extract information from a complex environment, an organism continuously adjusts its sensitivity to<br />

different stimuli. One strategy for integrating olfactory information over time is to down-regulate odorant<br />

perception at the level of the sensory neuron. Another strategy requires a circuit in which interneurons<br />

provide feedback inhibition. I have been focusing my efforts on examining adaptation to odorants sensed<br />

by the olfactory neuron AWC, a process that may take place within the sensory neuron itself.<br />

The pair of AWC neurons allows the animal to respond to at least five different odorants. The signal<br />

transduction pathway within the AWCs includes putative seven transmembrane G-protein coupled<br />

odorant receptors, at least two heterotrimeric G-proteins, at least two guanylyl cyclases and a<br />

cGMP-gated cation channel that may open in response to cyclase stimulation. Intriguingly, adaptation to<br />

each odorant is independent of the others sensed by this pair. I have identified a mutant, pkg-1, that fails<br />

to adapt to all AWC-sensed odorants tested. I mapped the mutation and was able to obtain rescue of the<br />

phenotype with an extrachromosomal array of a cosmid containing one of the two C. <strong>elegans</strong> genes<br />

encoding cGMP-dependent protein kinases (PKGs) present in the genome.<br />

I am attempting to identify the molecular lesion by sequencing the locus that encodes the PKG in our<br />

existing allele. I am also generating additional alleles using a non-complementation screen.<br />

What is the target of PKG-1 phosphorylation in olfactory adaptation? I am taking a candidate approach to<br />

this question by mutating potential PKG phosphorylated residues within members of the signal<br />

transduction pathway. Animals that express only the mutated form of the protein should be adaptation<br />

defective if that protein’s function is regulated by phosphorylation. I hope to examine the phosphorylation<br />

state of these candidates in extracts made from both N2 and pkg-1 worms. An important conundrum<br />

remains: how does the worm utilize a shared adaptation component to specifically down regulate its<br />

response to one of five odorants sensed by the same pair of neurons? This will be addressed by<br />

identifying PKG-1’s targets, binding partners and examining their localization within the pair of AWC<br />

neurons during adaptation.<br />

186


YOU CAN’T GET THERE FROM HERE: A GENE REQUIRED<br />

FOR PHARYNGEAL EXTENSION.<br />

SK Lange, JR Saam, SE Mango<br />

<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

Huntsman Cancer Institute Center for Children and Department of Oncological Sciences, University of<br />

Utah, Salt Lake City, UT 84112<br />

During pharyngeal extension, a ball of pharyngeal precursors is converted into a linear tube connected to<br />

the buccal cavity anteriorly and the midgut posteriorly. Time-lapse videomicroscopy has shown that this<br />

process depends on the reorientation of the pharyngeal epithelial cells and their adhesion to neighboring<br />

arcade cells in the buccal cavity (see abstract by Portereiko and Mango). Intriguingly, mutants that disrupt<br />

cell-cell or cell-substratum adhesion do not affect pharyngeal extension.<br />

In a survey of homozygous deficiencies, we discovered one locus, mnDf90, with defective pharyngeal<br />

extension. Nevertheless, homozygous mutant embryos form a pharyngeal primordium, differentiate<br />

normally and produce the correct number of PHA-4+ cells. This phenotype suggests that cell fate<br />

specification is unaffected and that the defect is specific for morphogenesis. We have identified three<br />

EMS-generated alleles that phenocopy the pharyngeal extension phenotype and map near mnDf90. The<br />

map position and phenotype resemble cdl-1 (Cell Death Lethal), discovered by the Yamamoto lab, and so<br />

we have provisionally called our locus cdl-1.<br />

Homozygous cdl-1 embryos arrest at the 3-fold or L1 stage with an unattached pharynx. This phenotype<br />

suggests that the locus is not required for at least two other morphogenetic processes: ventral closure<br />

and embryonic elongation. Surprisingly, two of our alleles also accumulate cell corpses. This phenotype is<br />

likely to reflect a defect in cell corpse engulfment, a process thought to depend on cytoskeletal<br />

reorganization to extend filopodia around the corpse. Since mnDf90 homozygotes do not show the cell<br />

corpse defect, one possibility is that cdl-1 is not required for engulfment of corpses, but that our cdl-1<br />

alleles produce altered proteins that interfere with normal engulfment. We suggest that cdl-1 is involved in<br />

regulation or function of the cytoskeleton.<br />

Our current goal is to determine what aspect of pharyngeal extension is disrupted by cdl-1 and whether<br />

other morphogenetic processes (e.g. migration) are also compromised. In addition, we are using standard<br />

approaches to clone cdl-1 and initiate a molecular analysis of pharyngeal extension.<br />

187


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

SIGNALING BY THE VAB-1 EPH RECEPTOR<br />

INTRACELLULAR DOMAIN<br />

Kristoffer Larsen,, Sean George, Andrew Chisholm<br />

Department of Biology, University of California, Santa Cruz, CA 95064<br />

The VAB-1 Eph receptor tyrosine kinase functions in embryonic morphogenesis and axon guidance. The<br />

VAB-1 intracellular domain (ICD), like those of other Eph receptors, includes a conserved juxtamembrane<br />

motif containing phosphotyrosines, a tyrosine kinase domain, and a C-terminal region with weak similarity<br />

to SAM domains. Genetic analysis suggests that VAB-1 has both kinase-dependent and<br />

kinase-independent functions. We are using two-hybrid and genetic screens to identify components of the<br />

VAB-1 kinase-dependent pathway.<br />

The full-length VAB-1 ICD could not be used as bait in two-hybrid screens because it caused reporter<br />

gene activation in the absence of prey. After extensive subcloning we identified fragments of the VAB-1<br />

ICD that did not cause self-activation. One of these contains the complete kinase domain and C-terminal<br />

tail, and has been the basis for a pilot screen from which we identified several positive clones. We will<br />

present our analysis of these potential interactors.<br />

Several proteins have been previously identified as binding to activated Eph receptor ICDs. These include<br />

SH2-domain containing adaptor proteins (Nck, etc), and other proteins whose mode of interaction is less<br />

clear. We have begun testing C. <strong>elegans</strong> homologs to ask if these interactions have been conserved.<br />

Finally, we are using genetics to ask whether the VAB-1 kinase-dependent pathway shares components<br />

with other RTK signaling pathways characterized in C. <strong>elegans</strong>. We have found that activating mutations<br />

in LET-60 Ras do not significantly suppress vab-1 loss-of-function phenotypes, suggesting that VAB-1<br />

does not signal via Ras activation. We will present results of additional double mutant experiments.<br />

188


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

MDF-1 SUPPRESSORS THAT MAY PLAY A ROLE IN THE<br />

METAPHASE TO ANAPHASE CHECKPOINT<br />

Elaine Law, Risa Kitagawa, Ann M. Rose<br />

Department of Medical Genetics, University of British Columbia, Vancouver V6T 1Z3<br />

The spindle assembly checkpoint is an evolutionarily conserved mechanism that ensures accurate<br />

chromosome segregation. This mechanism inhibits cell-cycle progression in response to a signal<br />

generated by mitotic spindle damage or by kinetochores that have not attached to microtubules prior to<br />

sister-chromatid separation. Such arrest allows cells to complete essential events for the maintenance of<br />

genetic fidelity. The spindle checkpoint mechanism has been studied in yeast and mammalian tissue<br />

culture systems, but little is known about it in multi-cellular organisms. We have identified mdf-1 and<br />

mdf-2 (mitotic arrest defective) as homologues of yeast spindle checkpoint genes MAD1 and MAD2 in<br />

<strong>Caenorhabditis</strong> <strong>elegans</strong>. Both genes are essential for the long-term survival and fertility of C. <strong>elegans</strong>.<br />

Loss of function of either gene leads to the accumulation of a variety of defects, which ultimately results in<br />

genetic lethality. To better understand how the checkpoint functions in C. <strong>elegans</strong>, and to identify<br />

additional components of the checkpoint, we screened for suppressors capable of rescuing the mdf-1<br />

(gk2) loss of function phenotype. Sixteen suppressors were recovered, which increased the number of<br />

fertile progeny and the survival of the genetic strain. The suppressors are being genetically mapped. One<br />

dominant suppressor was recovered, h1983. We have positioned this mutation and are currently<br />

examining the molecular basis for the suppression. One hypothesis, which we are now testing, is that the<br />

suppressor gene product is a downstream target of MDF-1 and acts compensatory to the spindle<br />

checkpoint defect to reduce chromosome missegregation.<br />

189


CHARACTERIZATION OF A C. ELEGANS DEFECATION<br />

MUTANT<br />

Anne Lehtela, Garry Wong<br />

<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

A.I. Virtanen Institute, Kuopio University, PL 1627, Kuopio 70211, FINLAND<br />

Defecation in C. <strong>elegans</strong> is a tightly regulated and complex behavior. The defecation behavior is<br />

generated from neuronal signals and is transmitted into movement of multiple body muscles in a highly<br />

coordinated fashion. Defecation occurs in 3 steps: contraction of posterior body muscles (pBoc);<br />

contraction of anterior body muscles (aBoc); expulsion of intestinal contents (exp). The defecation cycle<br />

occurs at regular 50-60 second intervals when wildtype Bristol N2 worms are feeding.<br />

A C. <strong>elegans</strong> defecation mutant was identified in a mutagenesis screen. Six-thousand genomes were<br />

mutagenized with 50 mM ethyl methanesulfonate for 4 h. F2’s were selected for resistance to 0.7 mM<br />

aldicarb, an acetylcholine esterase inhibitor. A single mutant, which appeared to be constipated, was<br />

cloned and then outcrossed to N2.<br />

The defecation mutant, in comparison to N2 per 30 min, had fewer pBOC (15.3 ± 2 vs. 32.8 ± 1), fewer<br />

exp (6.5 ± 1 vs. 31.8 ± 0), and fewer successful complete defecation motor program events (32 ± 9% vs.<br />

97 ± 2%), respectively. When pBOC was followed by an exp event, the latency appeared longer in the<br />

mutant (8-10 sec) compared to N2 (2-3 sec). On the few occasions when the complete defecation motor<br />

program was intact in mutants, the interval was longer (80-90 sec) than N2 (55-60 sec). Genetic analysis<br />

has tentatively identified the location of the mutation on chromosome III.<br />

These results, describing a derangement of the defecation motor program, combined with identification of<br />

the gene mutation responsible for this phenotype, should provide insights into the control of complex<br />

neuronal and neuromuscular controlled behaviors.<br />

190


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

ORGANOGENESIS OF THE C. ELEGANS INTESTINE<br />

Benjamin Leung 1 , Greg J. Hermann 2 , James R. Priess 3<br />

1MCB Program, University of Washington and Fred Hutchinson Cancer Research Center, Seattle, WA<br />

98109.<br />

2Fred Hutchinson Cancer Research Center, Seattle, WA 98109<br />

3MCB Program, University of Washington and Fred Hutchinson Cancer Research Center, HHMI, Seattle,<br />

WA 98109.<br />

The C. <strong>elegans</strong> intestine is a bilaterally symmetric tube of 20 polarized epithelial cells derived from a<br />

single early blastomere called E. Cell polarity in the intestine begins with reorganization of the microtubule<br />

cytoskeleton, followed by migration of the intestinal nuclei apically, and other organelles basally. Small<br />

apical membrane separations coalesce into a continuous compartment to form the lumen of the intestine.<br />

The early primordium consists of two tiers of polarized cells; specific cell intercalations rearrange the<br />

primordium into a single elongated tier.<br />

An E blastomere cultured in isolation produces a cyst of polarized cells, but the aggregate lacks the<br />

bilateral symmetry seen in the normal intestine. Thus the ability to polarize appears to be an intrinsic<br />

property of intestinal cells, but bilateral symmetry requires interactions with cells that normally surround<br />

the intestine. Cell killing experiments suggest these interactions must occur at an early stage in intestinal<br />

morphogenesis. We are currently using the technique of RNAi to determine whether genes implicated in<br />

epithelial polarity in other systems are required for proper development of the C. <strong>elegans</strong> intestine.<br />

191


EXPRESSION AND REGULATION OF DAF-16::GFP<br />

CONSTRUCTS<br />

Kui Lin, Cynthia Kenyon<br />

<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

Department of Biochemistry & Biophysics, University of California at San Francisco, San Francisco, CA<br />

94143-0448<br />

Wild-type C.<strong>elegans</strong> ages rapidly, undergoing development, senescence, and death in less than 3 weeks.<br />

In contrast, mutants with reduced activity of daf-2, a homolog of the insulin and insulin-like growth factor<br />

(IGF-1) receptors (Kimura et al., 1997), age more slowly and live more than twice as long (Kenyon et al.,<br />

1993). Wild-type DAF-2 activity also promotes growth to adulthood and prevents dauer formation, since<br />

severe loss of daf-2 function causes the animals to become dauers in the presence of food. Both the<br />

lifespan extension and dauer-constitutive phenotypes caused by daf-2 mutations are dependent on the<br />

activity of daf-16, which encodes an HNF-3/forkhead family member (Ogg et al., 1997; Lin et al., 1997).<br />

We have created GFP-tagged daf-16 constructs and used them to study the expression and regulation of<br />

daf-16. We will report our progress on this study.<br />

192


IDENTIFICATION OF NOVEL UNC-64 (SYNTAXIN) ALLELES<br />

Christine Liu, C. Michael Crowder<br />

<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

Department of Anesthesiology, Washington University in St. Louis, St. Louis, MO 63110-1093<br />

unc-64 encodes a homolog of vertebrate syntaxin 1A. Syntaxin is expressed ubiquitously in the nervous<br />

system of the nematode. unc-64 contains a high degree of homology with human and Drosophila<br />

syntaxin, suggesting that this molecule is conserved across species and performs similar functions.<br />

Syntaxin is involved in membrane fusion of synaptic vesicles and controls neurotransmitter release, in<br />

part through its H3 helical domain, which interacts with SNAP-25 and synaptobrevin. Very few viable<br />

syntaxin alleles have been identified in any animal, including C. <strong>elegans</strong>. In C. <strong>elegans</strong>, the null allele,<br />

js115, confers larval lethality: the nematode completes embryogenesis, but dies as L1 larva. Currently,<br />

only four viable syntaxin alleles have been identified in C. <strong>elegans</strong>: e246, md1259, js21, md130. js21 and<br />

e246 contain different C to T missense mutations in exon 7; md130 and md1259 contain G to A splice<br />

site mutations at the splice donors of intron 6 and intron 3, respectively. All these previously identified<br />

mutations affect the H3 domain of syntaxin or cause reduced expression. While all viable syntaxin alleles<br />

appear to reduce neurotransmitter release, large allelic differences are seen in their sensitivities to volatile<br />

general anesthetics: md130 is resistant to volatile anesthetics while js21 and md1259 are hypersensitive.<br />

In addition to the H3 domain, syntaxin also contains N-terminal H ABC domains and a hinge region that are<br />

also thought to regulate membrane fusion events. However, no mutations have been identified in these<br />

regions. To better define these portions of syntaxin that regulate transmitter release and anesthetic<br />

sensitivity, a non-complementation screen using EMS as the mutagen was conducted using the unc-64<br />

(e246) allele. Three potentially new unc-64 alleles have been isolated out of approximately 4800 F1<br />

genomes screened. These animals exhibit phenotypes consistent with syntaxin hypomorphic alleles,<br />

including being uncoordinated and failure on re-testing to complement e246 allele. Other experiments to<br />

characterize these putative new viable syntaxin alleles are currently in progress.<br />

193


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

MUTATIONS THAT CAUSE NEURITE SPROUTING OF THE<br />

DVB MOTOR NEURON<br />

Loria, P., Boulin, T., Conte, S., Hobert, O.<br />

Columbia University, College of Physicians & Surgeons, Center for Neurobiology and Behavior, New<br />

York, NY 10032<br />

Neuroanatomical studies in vertebrates long ago revealed that certain forms of insult such as axotomy or<br />

activity blockade result in the outgrowth of additional neurites. These observations suggest that<br />

postmitotic neurons contain an intrinsic capacity to assess their integrity and to modulate their anatomy<br />

accordingly. In C.<strong>elegans</strong>, several genetic lesions have been described that lead to the outgrowth of<br />

additional neurites ("neurite sprouting")(1,2). We have found that a null mutation in the lim-6 LIM<br />

homeobox gene causes neurite sprouting in the DVB motor neuron (3). Using this observation as a<br />

starting point, we have set out to understand the molecular components of neurite sprouting and to further<br />

dissect the role of lim-6 in this process. First, we used a candidate gene approach to test mutations in<br />

proteins involved in different aspects of neuron and muscle function. Second, we performed an unbiased<br />

genetic screen for mutants that phenocopy the lim-6 sprouting defect in DVB.<br />

Since activity blockade at the neuromuscular junction in vertebrates and flies causes neurite sprouting,<br />

we tested whether loss of the neurotransmitter GABA causes sprouting of the GABAergic DVB<br />

motorneuron. We indeed find that mutations in unc-25 cause neurite sprouting of DVB. These defects can<br />

be enhanced by mutations in unc-31, which presumably abolishes peptinergic neurotransmission.<br />

Furthermore, blocking the activity of the enteric muscle targets of DVB with an egl-2(gf) mutation, which<br />

activates a K-channel (4), leads to neurite sprouting of DVB, as does the complete removal of the enteric<br />

muscles in hlh-8/twist mutants. We also find that a gain-of-function mutation in CamKII/unc-43, a gene<br />

whose role in neurite outgrowth has been described extensively in vertebrates, causes neurite sprouting<br />

in DVB.<br />

Using a clonal, fluorescence microscope-based screen of 3300 haploid genomes, we identified nine<br />

mutants, ot1 through ot9, that affect neurite sprouting of DVB. These mutants fall into at least 4<br />

complementation groups and are not allelic to any of the genes described above. The mutants do not<br />

cause sprouting in other defined sensory or interneurons tested. We also find that the as yet uncloned<br />

unc-122 gene causes specific neurite sprouting of DVB. We are in the process of mapping and cloning<br />

these genes.<br />

194


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

A B -TUBULIN GENE, TBB-2, FUNCTIONS AS AN ACTIVATOR<br />

OF MEI-1 AND MEI-2 IN FEMALE MEIOTIC SPINDLE<br />

FORMATION IN CAENORHABDITIS ELEGANS.<br />

Chenggang Lu, Martin Srayko, Paul E. Mains<br />

Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, Alberta, Canada T2N<br />

2T9<br />

C. <strong>elegans</strong> female meiosis requires two meiotic-specific genes, mei-1 and mei-2. Loss of mei-1 or mei-2<br />

function blocks female meiotic spindle formation while the subsequent mitotic cleavages are normal. In a<br />

mei-1 gain-of-function mutant, however, the mitotic cleavages are disrupted after normal meiotic divisions.<br />

Wild-type MEI-1 and MEI-2 localize to the meiotic, but not mitotic spindle. 1,2 However, MEI-1(gf) and<br />

MEI-2 ectopically localize to mitotic spindles in mei-1(gf) embryos. mei-1 and mei-2 encode homologs of<br />

p60 and p80 subunits of the sea urchin microtubule severing protein katanin, respectively, and MEI-1 and<br />

MEI-2 together disassemble interphase microtubules in a HeLa cell system. 2 We propose that MEI-1 and<br />

MEI-2 form a complex in meiosis and regulate meiotic spindle formation by katanin-like activities.<br />

In a mei-1(gf) suppressor screen, we recovered three extragenic suppressors. One of them, sb26, was<br />

found to be a missense mutation in a b -tubulin gene, tbb-2. tbb-2(sb26) also enhances a weak mei-2(lf)<br />

allele. Together, these results suggest that tbb-2 is required for the function of mei-1 and mei-2. Antibody<br />

staining shows that TBB-2 is widely expressed during worm development. Neither tbb-2 nor tbb-1<br />

(another b -tubulin highly similar to tbb-2) RNAi has severe effects during early development. However,<br />

tbb-2 and tbb-1 double RNAi results in 100% dead eggs indicating that they act redundantly during<br />

embryogenesis. We are doing experiments to examine the interactions of MEI-1/MEI-2 with tbb-2 and<br />

tbb-1.<br />

1. Clark-Maguire, S. and Mains, P.E. (1994). J. Cell Biol. 126, 199-209.<br />

2. Srayko, M., Buster, D.W., Bazirgan O.A., McNally, F.J., and Mains, P.E. (2000). Genes Dev. 14 (9).<br />

195


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

GLOBAL PROFILE OF GENE EXPRESSION DURING AGING<br />

James Lund 1 , Pamela Larsen 2 , Pat Tedesco 3 , Thomas Johnson 3 ,<br />

Stuart Kim 1<br />

1Stanford University<br />

2University of California Los Angeles<br />

3Institute for Behavioral Genetics, University of Colorado, Boulder<br />

Aging is a physiological phenomenon characteristic of metazoan life. As animals age, they suffer a broad<br />

functional decline and an exponentially increasing probability of death. Many aspects of the aging<br />

phenotype are present in animals from C. <strong>elegans</strong> to mammals, including a build-up of lipofuscin<br />

deposits, mitochondrial DNA deletions, and degradation of organ function. The life span of an animal is<br />

thought to be set by the balance between deleterious events and repair mechanisms. A number of C.<br />

<strong>elegans</strong> mutants live significantly longer than wildtype, making C. <strong>elegans</strong> an attractive model for studies<br />

of aging.<br />

DNA microarrays can be used to profile the expression levels of a large number of genes in parallel.<br />

Using our array, which contains a PCR product representing every C. <strong>elegans</strong> gene, we are profiling<br />

expression patterns during the normal aging process. We isolated RNA from synchronized cultures of<br />

sterile animals at various ages from young adult to old age. We hybridized this RNA to DNA microarrays,<br />

and are now analyzing the data to identify groups of genes with expression levels which change as<br />

reproduction ceases and the worms age. These expression profiles with be compared with the partial<br />

profiles of aging mice and humans that have been published. The expression profile of normal aging will<br />

be used as a reference to understand the expression changes underlying the extended life spans in<br />

longevity mutants.<br />

196


CONDITIONAL MUTATIONS AFFECTING MITOTIC SPINDLE<br />

POSITIONING AND POLARITY IN THE C. ELEGANS EMBRYO<br />

Rebecca Lyczak, Bruce Bowerman<br />

<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

Institute of Molecular Biology, University of Oregon, Eugene, OR 97403<br />

Early polarity in the C. <strong>elegans</strong> embryo requires proper regulation of the cytoskeleton.<br />

Cytoskeletal-dependent processes such as pronuclear migration, centrosome rotation, and spindle<br />

positioning must be properly accomplished to ensure an asymmetric first cleavage. To investigate the role<br />

of the cytoskeleton in regulating asymmetric cell division, we have undertaken a screen for temperature<br />

sensitive embryonic lethal mutations affecting spindle positioning in the early embryo. We have identified<br />

mutations in two classes that affect P0 spindle positioning: 1) misorientation of the spindle axis 2)<br />

mispositioning of the spindle along the A-P axis.<br />

We have identified several mutants that show a defect in pronuclear migration and P0 spindle orientation.<br />

These include alleles of mel-26 and zyg-9 as well as at least three novel loci. In or346ts embryos, the<br />

oocyte pronucleus does not always migrate to the posterior of the embryo before initiation of the first<br />

mitotic spindle. The centrosome/nuclear complex remains in the posterior of the embryo and does not<br />

rotate. Thus, the first mitotic spindle sets up transverse to the A-P axis. The misoriented spindle results in<br />

a missegregation of P-granules and daughter cells of aberrant size and developmental potentials.<br />

Terminally differentiated or346ts embryos also show striking patterning defects, often making extra<br />

intestinal cells.<br />

We have also identified at least two mutants in which the P0 spindle is mispositioned along the A-P axis.<br />

In or282ts mutants, both pronuclei migrate toward the center of the embryo where they meet before<br />

setting up a spindle along the A-P axis. This spindle varies in its position along the A-P axis resulting in a<br />

randomization of cell size in the daughters. This mispositioning of the spindle is coupled with<br />

mislocalization of P-granules and a loss of polarity in the daughter cells. Terminally differentiated or282ts<br />

embryos show patterning defects, often making extra pharyngeal cells and lacking intestine. In or358ts<br />

embryos, the P0 spindle is displaced too far posterior resulting in an excessively asymmetric first division.<br />

Analysis of spindle positioning mutants should provide valuable insights into the link between<br />

establishment of polarity and the cytoskeleton.<br />

197


ROLE OF PDZ DOMAIN PROTEINS IN ESTABLISHING GUT<br />

EPITHELIAL POLARITY<br />

Kathleen E. Mach, Stuart K. Kim<br />

<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

Department of Developmental Biology, Stanford University, Stanford, CA 94305<br />

Epithelial cells are polarized cells that set up and maintain distinct basolateral and apical membrane<br />

domains. The PDZ protein motif, originally recognized in the postsynaptic density protein PSD-95, the<br />

Drosophila discs-large (Dlg), and the tight junction protein ZO-1, mediates protein-protein interactions and<br />

is found in many proteins having a central role in localizing proteins to the basolateral membrane domain<br />

of epithelial cells or to neuronal synapses. For example, our work has shown that three proteins with PDZ<br />

domains (LIN-2, LIN-7 and LIN-10) are involved in basolateral localization of the LET-23 EGF receptor in<br />

the vulval precursor cells. We are currently testing the hypothesis that other PDZ proteins may also be<br />

involved in epithelial cell polarity. Analysis of the C. <strong>elegans</strong> genome revealed 58 open reading frames<br />

predicted to encode PDZ domains, 51 of which represent genes that have not yet been characterized. To<br />

examine the role of each of the PDZ proteins, the loss of function phenotype each gene is being<br />

determined by RNAi. To date, the RNAi phenotype of over half the PDZ genes has been determined and<br />

several of these genes have a defects in the polarity of the gut epithelia. These phenotypes fall into 2<br />

distinct classes: 1. those where epithelial cell junctions form but cell organization is disrupted and 2. those<br />

where cell junctions fail to form or are disrupted. Two genes in the latter group are the C. <strong>elegans</strong><br />

homologues of Drosophila Dlg (dlg-1) and scribble (scb-1). Early embryonic cell divisions are normal in<br />

dlg-1(RNAi) and scb-1(RNAi) but embryos arrest at the two fold stage. MH27 antibody staining indicated<br />

that the epithelial cell junctions begin to form in these animals but are not completed leading to<br />

disconnected segments of junctional material.<br />

198


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

GENETIC ANALYSIS OF NMDA RECEPTOR EXPRESSION IN<br />

C. ELEGANS<br />

David M. Madsen, Chingju Lin, Penelope J. Brockie, Andres V. Maricq<br />

Dept. of Biology, University of Utah, Salt Lake City, UT 84112<br />

NMDA-type glutamate receptors are of great interest because of their roles in synaptic plasticity and<br />

neuronal excitotoxicity. We have identified two genes, nmr-1 and nmr-2, that encode NMDA-type subunits<br />

(see abstract by J. Mellem). Using the reporter molecule GFP, we have shown that nmr-1 and nmr-2 are<br />

co-expressed in a small subset of interneurons that contribute to the locomotory control circuit (AVA,<br />

AVD, AVE and PVC). Analysis of nmr-1 deletion mutants reveals defects in the timing of locomotion.<br />

Despite their important roles in most nervous systems, we do not understand which genes are required<br />

for the expression of NMDA receptors.<br />

We performed a visual-based genetic screen for mutations that affect the development and differentiation<br />

of NMDA expressing interneurons. We mutagenized a transgenic strain that expresses GFP under the<br />

control of the nmr-1 promoter and screened ~20,000 haploid genomes for aberrant GFP expression. One<br />

class of mutants showed a marked decrease of GFP expression in the PVC neuron. This neuron is still<br />

present, suggesting that the mutation affects steps late in differentiation.<br />

We identified this mutation as a nonsense mutation in ceh-14, a LIM-homeobox gene. Based on GFP<br />

reporter constructs, ceh-14 expression is observed in several neurons, including PVC. In the ceh-14<br />

mutant, expression of other glutamate receptor subunits (glr-1, glr-2, and nmr-2) is also decreased or<br />

absent in PVC. We are currently testing for cell autonomous rescue using PVC specific promoters to drive<br />

expression of ceh-14. Besides the defects in glutamate receptor expression, ceh-14 mutants exhibit a<br />

mechanosensory defect. We hypothesize that this is due to diminished or loss of PVC function in the<br />

locomotory control circuit. In addition, ceh-14 animals also exhibit a thermal avoidance defect (Tav) and a<br />

phasmid dye-filling defect. Introduction of a wild-type copy of ceh-14 into transgenic worms rescued the<br />

mutant phenotypes.<br />

199


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

LARGE SCALE REVERSE GENETIC APPROACH USING RNAI<br />

Sarah Mahoney, Alex Phan, Mark Maxwell, Candace Swimmer,<br />

Jonathan Heller, Brett Milash, Kate McKusick, Monique Nicoll<br />

Exelixis, Inc. 170 Harbor Way, S. San Francisco, CA 94083-0511<br />

One of our primary missions at Exelixis is the discovery of new drug targets for human pharmaceutical or<br />

agrochemical research through the use of model organism genetics. To rapidly survey for genes that<br />

function in disease-related pathways, we have undertaken several reverse genetic strategies, including<br />

development of a genome wide RNAi library. First we data mine, which involves compiling predicted<br />

cDNAs from GeneFinder in a database and sorting and categorizing them by predicted protein motifs.<br />

Next, we employ an automated primer picking program created at Exelixis to generate primer pairs<br />

against portions of the predicted coding sequence. PCR is then preformed on cDNA and the products are<br />

cloned into a modified version of pCCM113 (kindly provided by C. Mello). Individual clones are<br />

sequenced in an automated fashion and clones are validated by automatic blast via an inhouse database.<br />

This has allowed us to correlate GeneFinder predictions with expression data. In our initial examination of<br />

more than 1000 PCR reactions using cDNA as the template, we have observed that approximately 85 %<br />

of the predicted genes are expressed.<br />

In parallel to automating the cloning process, we are assessing the effectiveness of dsRNA delivery<br />

methods. We initiated these experiments with a set of 30 genes and have compared injection, soaking,<br />

and feeding. In addition, we have tested several parameters, including concentration dependence,<br />

pooling effectiveness, and tissue specificity of RNAi. Our results indicate that RNAi is concentration<br />

dependent, titratable, and that pooling of up to three dsRNAs gives us phenotypic results that are<br />

comparable to RNAi of each gene individually. Initial RNAi soaking results from more than 100 predicted<br />

genes suggests that a quarter of predicted genes have highly penetrant RNAi phenotypes in wild type<br />

worms by dissection scope analysis.<br />

200


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

SEQUENCE CONFIRMATION OF 182 SNPS BETWEEN C.<br />

ELEGANS N2 AND CB4856 STRAINS AND PLANS FOR<br />

GENERATION OF 1000 NEW SNPS.<br />

Penny Mapa, Kathryn Swan, Mike Ellis<br />

Exelixis Inc., 170 Harbor Way, South San Francisco, CA 94080<br />

Exelixis Inc. is a leader in model systems genetics and comparative genomics. As part of our C. <strong>elegans</strong><br />

mapping and cloning effort we have sequence confirmed 182 snps, evenly spaced throughout the<br />

genome, between the N2 and CB4856 (Hawaiian) strains. These snps are a subset of the 1416 potential<br />

snps initially identified by the Genome Sequencing center at the Washington University School of<br />

Medicine (http://genome.wustl.edu/gsc/CEpolymorph/snp_chrom.shtml). We have successfully used<br />

these genome wide, confirmed snps to map mutants. Additionally, to aid in fine scale mapping, we have<br />

begun efforts aimed at identifying another 1000 new genome wide snps between these same strains. As<br />

a service to the worm community we plan to release this information to the public.<br />

201


BUILDING A DICTIONARY FOR C. ELEGANS PROMOTER<br />

SEQUENCES<br />

Steven McCarroll, Hao Li, Cori Bargmann<br />

U.C. San Francisco<br />

<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

We would like to understand how regulatory sequences encode the expression patterns of genes. The<br />

availability of complete genomic sequence allows us to try to identify candidate transcriptional control<br />

sequences based on statistical criteria. We have used a dictionary algorithm (Bussemaker, Li, and Siggia,<br />

manuscript in preparation) to partition C. <strong>elegans</strong> promoter sequences into "words" -- discrete sequences<br />

that are distributed statistically as if they represent a coherent functional unit. We have built a dictionary<br />

for the upstream sequences of 850 C. <strong>elegans</strong> G-protein-coupled-receptor genes. When this dictionary is<br />

used to partition these promoter sequences according to maximum-likelihood criteria, 2.1% of the<br />

sequence is covered by words of length 8 or greater. Many of these words appear interesting according to<br />

multiple criteria: (i) non-Poisson distribution across genes, suggesting a tendency to appear in clusters;<br />

(ii) non-random distribution across positions, suggesting a preference for particular locations relative to<br />

the transcriptional start site, and (iii) appearance in genes that have similar expression patterns. We are<br />

using this dictionary in a number of ways:<br />

(i) We have correlated the expression patterns of several dozen chemoreceptor genes to the distributions<br />

of words across these genes, generating testable hypotheses about sequences that may confer<br />

cell-specific gene regulation;<br />

(ii) We are correlating gene-expression microarray data with the distribution of words across genes, to<br />

identify words that correspond to genes whose expression is modulated in particular experiments;<br />

(iii) We are testing candidate control sequences (words) in promoter-gfp fusion experiments, in which a<br />

particular word is deleted from a promoter (to test the necessity of this sequence for wild-type gene<br />

regulation) or added to a promoter (to test its sufficiency for conferring gene regulation).<br />

202


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

HIGH PRESSURE FREEZING METHODS FOR C. ELEGANS<br />

EMBRYO ULTRASTRUCTURE AND EM IMMUNOLABELING<br />

Kent L. McDonald 1 , Thomas Mueller-Reichert 2 , Akiko Tagawa 3 , Chad<br />

A. Rappleye 3 , Raffi Aroian 3<br />

1Electron Microscope Lab, UC Berkeley<br />

2Max-Planck-Institute of Molecular Cell Biology and Genetics, Dresden, Germany<br />

3Dept. of Biology, UC San Diego<br />

Electron microscopic analysis of C. <strong>elegans</strong> has a long and distinguished history, from the early studies<br />

reconstructing the nervous system to recent work by David Hall and his colleagues. However, attempts at<br />

more routine EM studies of C. <strong>elegans</strong> have been hampered by the impermeability of the cuticle of the<br />

worm and of the eggshell of embryos. These structures act as diffusion barriers, preventing the rapid<br />

exchange of fixatives and other solutions that are used in typical EM processing protocols.<br />

Our EM facility specializes in an alternative to conventional EM, high pressure freezing (HPF). In this<br />

technique, 50 or more nematodes at a time are frozen under 2100 bars pressure in 10-20 milliseconds.<br />

This method is useful for excellent preservation of ultrastructure to depths of 200 microns or more,<br />

compared to 10 microns or so for most other freezing methods. Furthermore, this preservation is<br />

independent of the impermeability of the eggshell or cuticle. HPF is particularly exciting for studying early<br />

embryogenesis since the embryos inside hermaphrodites are well preserved. Thus, within each gravid<br />

adult are a row of early embryos of various stages making it much easier than before to fix, find, orient,<br />

and work with embryos. We are currently using this technique for studying early embryogenesis. Recent<br />

work has suggested a new class of embryonic mutants important for early polarity, the polarity osmotic<br />

mutants (see abstracts by Tagawa et al and by Rappleye et al). Our models predict that these genes are<br />

involved in membrane trafficking. We are in the process of testing this at the ultrastructural level by<br />

looking at the general morphology of the mutants and by studying the immunolocalization of one of the<br />

proteins. We will present information of our technique of preservation, fixation, and embedding and some<br />

preliminary results in the early embryo.<br />

203


MOLECULAR IDENTIFICATION OF TRANSCRIPTIONAL<br />

TARGETS OF THE DAF-16 WINGED HELIX TRANSCRIPTION<br />

FACTOR<br />

Joshua J. McElwee 1 , James H. Thomas 1,2<br />

1Program in Molecular and Cellular Biology<br />

2Department of Genetics, University of Washington<br />

<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

Dauer arrest and longevity in C. <strong>elegans</strong> are controlled by an insulin-like signaling pathway transduced by<br />

the winged helix transcription factor DAF-16. Mutations in several genes within this pathway (daf-2, age-1,<br />

and pdk-1) result in constitutive dauer formation (Daf-c) and increased lifespan (Age) phenotypes.<br />

Invariably, both the Daf-c and Age phenotypes of these mutants are suppressed by loss of function<br />

mutations in daf-16. This suggests DAF-16 acts as the principal transcriptional output controlling both<br />

diapause and lifespan governed by this pathway. We are attempting to identify both direct and indirect<br />

transcriptional targets of DAF-16 utilizing several complementary molecular approaches. First, we are<br />

performing in vitro binding site selection to isolate the DAF-16 DNA consensus sequence. Utilizing this<br />

sequence, we will identify putative DAF-16 targets by genomic analysis and a candidate gene approach.<br />

Second, we are performing direct selection for genomic sequences that are bound by this transcription<br />

factor. Third, we are examining DAF-16 transcriptional outputs in vivo. Using cDNA arrays, we will assess<br />

global RNA expression in various Daf mutants and double mutants. Presumably, these experiments will<br />

allow us to begin to identify genes downstream of daf-16 involved in both dauer formation and regulating<br />

lifespan.<br />

204


FUNCTIONAL CONSERVATION OF C. ELEGANS UNC-30 AND<br />

MOUSE PITX2 IN GABAERGIC NEURON SPECIFICATION<br />

Jason McEwen, Yishi Jin<br />

Department of Biology, University of California, Santa Cruz, CA. 95064<br />

The C. <strong>elegans</strong> GABAergic nervous system contains 26 neurons of 5 types. The homeodomain protein<br />

UNC-30 is required for the specification and function of the type-D motor neurons. In unc-30 mutants the<br />

D neurons fail to express GABA, have axon guidance errors, form improper synapses and shrink on<br />

themselves while trying to move backwards. It has previously been shown that UNC-30 directly activates<br />

the expression of unc-25, the glutamic acid decarboxylase enzyme (GAD) and unc-47, the GABA<br />

vesicular transporter, in these neurons (1).<br />

The UNC-30 homeodomain is over 80% identical to those of the newly identified vertebrate Pitx family<br />

and dPtx in Drosophila (2). Pitx proteins are expressed in many tissues and have been implicated in a<br />

variety of developmental functions (2). Pitx2 is also widely expressed in the developing and adult central<br />

nervous system, but its function there is not known. UNC-30 and Pitx2 can activate mammalian GAD67<br />

expression in cultured neurons (Condie B, Pers. Com.), suggesting that Pitx2 and UNC-30 may have an<br />

evolutionarily conserved role in GABAergic neuron specification.<br />

We have shown that mouse Pitx2 can functionally replace UNC-30 in unc-30 mutant worms. Site-directed<br />

mutagenesis experiments on the UNC-30 binding sites in the unc-25 promoter and the Pitx2<br />

homeodomain support that Pitx2 mediated unc-25 activation depends on a homeodomain/UNC-30<br />

binding site interaction. Like UNC-30, ectopic expression of Pitx2 under the control of heat shock<br />

promoters activates unc-25 expression in other tissues (3). Our data supports that UNC-30 and Pitx2 are<br />

likely functional homologues.<br />

UNC-30 also regulates the expression of genes required for axon guidance and synapse formation but<br />

these target genes have yet to be found. To address this we have generated modified UNC-30 proteins<br />

using the activation domain of VP16 and the repressor domain of Engrailed. The modified UNC-30<br />

proteins have shown corresponding changes in transgenic studies using the Punc-25::GFP reporter.<br />

These constructs will be used to sensitize future screens for UNC-30 target genes.<br />

References:<br />

<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

1.<br />

2. Eastman et al. (1999) J. Neuroscience 19(15), 6225-6234<br />

3. Gage et al.(1999) Mamm. Gen. 10, 197-200<br />

4. Jin et al. (1994) Nature Vol 372, 780-783<br />

205


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

GENES INVOLVED IN NICOTINIC NEUROTRANSMISSION IN<br />

THE PHARYNX<br />

Jim McKay 1,2 , David Raizen 3,4 , Leon Avery 1,5<br />

1Department of Molecular Biology. University of Texas Southwestern Medical Center. Dallas, Texas<br />

75390-6148<br />

2jim@eatworms.swmed.edu<br />

3Department of Neurology. University of Pennsylvania School of Medicine. Philadelphia, Pennsylvania<br />

19104<br />

4raizen@mail.med.upenn.edu<br />

5leon@eatworms.swmed.edu<br />

MC is the main excitatory motorneuron of the pharynx and is required for the large increase in pumping<br />

rate in response to food. We identified eat-2 and eat-18 in genetic screens for worms incapable of rapid<br />

pharyngeal pumping. Both eat-2 and eat-18 lack MC neurotransmission but have no other obvious<br />

defects. eat-2 encodes a non-alpha nicotinic receptor subunit. It is expressed in pharyngeal muscle and is<br />

localized to a region where we think the MC synapse is. We conclude that MC is cholinergic and acts<br />

directly on pharyngeal muscle to stimulate pumping. We observed allele specific genetic interaction<br />

between eat-2 and eat-18 indicating that the gene products physically interact. In an eat-18 mutant<br />

background, EAT-2 is expressed and correctly localized but the channel is not functional. We are<br />

attempting to clone eat-18 by transformation rescue. We have narrowed down to a 3 kb piece of genomic<br />

DNA that can rescue eat-18 mutants and we are sequencing this region from two eat-18 mutant alleles to<br />

help identify the coding region.<br />

206


GENETIC ANALYSIS OF THE FUNCTIONS OF A<br />

GSK-3&SZLIG; HOMOLOG CALLED SGG-1 AND A<br />

&SZLIG;-TRCP/SLIMB HOMOLOG DURING C. ELEGANS<br />

EMBRYOGENESIS<br />

Marc Meneghini 1 , Greg Ellis 1 , Ann Schlesinger 2 , Bruce Bowerman 1<br />

1Institute of Molecular Biology, University of Oregon, Eugene OR, 97403<br />

2Whitehead Institute, Cambridge MA<br />

During C. <strong>elegans</strong> embryogenesis, a four cell stage blastomere called P2 uses Wnt signaling to induce<br />

anterior-posterior polarity in its sister blastomere EMS. In embryos defective for the function of Wnt<br />

pathway components the posterior EMS daughter E, which normally produces endoderm, develops like<br />

its anterior sister MS. We have previously reported the characterization of a C. <strong>elegans</strong> GSK-3ß homolog<br />

called sgg-11, which shares this phenotype suggesting that sgg-1 acts positively with the Wnt pathway to<br />

regulate a-p polarity in EMS. This was somewhat surprising because in other systems, GSK-3ß functions<br />

to repress Wnt pathway outputs and is negatively regulated by Wnt signaling to relieve this repression.<br />

We also reported that sgg-1 mutant embryos produce an ectopic E-like cell that is derived from C, a<br />

daughter of P2. Further analysis has shown that the source of this ectopic endoderm is Cp, the posterior<br />

daughter of C. Double mutant analysis shows that Wnt pathway components also are required for the<br />

production of Cp-derived endoderm in sgg-1 embryos. This result suggests that Wnt signaling functions to<br />

make Cp different from Ca and that sgg-1 is required in Cp to prevent it from adopting an E-like fate.<br />

Furthermore, sgg-1 synergizes with the Wnt pathway in EMS indicating that sgg-1 and the Wnt pathway<br />

have some non-overlapping functions. Rather than functioning with the Wnt pathway during polarity<br />

induction, perhaps sgg-1 functions at the level of blastomere identity to make Cp and E, or perhaps EMS<br />

and C, different from each other. Like the Wnt pathway, sgg-1 is required for the orientation of the mitotic<br />

spindle in EMS1 suggesting an overlap with the Wnt pathway during induced polarity. We are also<br />

studying the function of a ß-TRCP/slimb related gene which we call slm-1. ß-TRCP/slimb related proteins<br />

are known to function with GSK-3ß to target proteins for proteolytic degradation. Interestingly, slm-1<br />

embryos have many phenotypic similarities with sgg-1 embryos. We are performing more phenotypic<br />

analyses to better understand the functions of sgg-1 and slm-1 during C. <strong>elegans</strong> embryogenesis.<br />

1 Schlesinger et al., Genes & Dev. (1999)<br />

<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

207


THE EFFECT OF NONIMMOBILIZERS ON C. ELEGANS<br />

Laura B. Metz 1 , Mike Crowder 1,2<br />

<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

1Department of Anesthesiology Washington University, St. Louis, MO<br />

2Department of Molecular Biology/Pharmacology, Washington University, St. Louis, MO<br />

The mechanism of action of volatile anesthetics (VAs) is still uncertain. The Meyer - Overton hypothesis<br />

tries to correlate anesthetic potency directly to lipid solubility. Yet this theory does not explain why some<br />

lipophilic compounds in many homologous series of anesthetics are devoid of any anesthetic potency. F6<br />

and F8, so called nonimmobilizers, are two such compounds that should have anesthetic action but don’t.<br />

Here, we test whether C. <strong>elegans</strong> is also unaffected by nonimmobilizers and whether mutants<br />

hypersensitive to VAs are sensitized to nonimmobilizers. Behavioral assays including locomotion and<br />

mating assays were all performed with concentrations of F6 and F8 at 3-4 times the EC 50 (concentration<br />

when effect should be half-maximal) predicted by the Meyer-Overton hypothesis. The nonimmobilizers did<br />

not significantly effect N2 in any behavioral assay, even after 24-hr exposure. Strains extremely<br />

hypersensitive to bona fide VAs were tested with F6 and F8. At 4 times the EC 50, F8 had a slight effect<br />

on ric-4(js20) while F6 showed none. Both F6 and F8 affected unc-64(js21) at those same high<br />

concentrations. These data suggest that reduced neurotransmission somehow sensitize animals to these<br />

drugs. To test whether F6 and F8 alter cholinergic neurotransmission as has been shown for true VAs, we<br />

measured the effect of F6 and F8 on aldicarb sensitivity (which correlates with the level of cholinergic<br />

neurotransmission). Neither F6 nor F8 had an effect on aldicarb sensitivity, and neither antagonized<br />

VA-induced aldicarb resistance. Thus, as in vertebrate models, nonimmobilizers do not affect the<br />

behavior of the wildtype C. <strong>elegans</strong>; however, the drugs do have some effect when synaptic transmitter<br />

release is reduced.<br />

208


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

ISOLATION AND CHARACTERIZATION OF MUTATIONS<br />

THAT ENHANCE LET-23(SA62GF) DURING VULVAL<br />

DEVELOPMENT<br />

Nadeem Moghal, Paul W. Sternberg<br />

Dept. of Biology, California Institute of Technology. Pasadena, CA 91125<br />

In C. <strong>elegans</strong> hermaphrodites, adoption of vulval fates by P5.p-P7.p is dependent on activation of the<br />

LIN-3/LET-23 signaling pathway. This pathway is structurally and functionally related to EGF receptor/Ras<br />

signaling pathways described in other systems. Although the major positive effectors of this pathway<br />

including EGF(lin-3), the EGFR(let-23), GRB2(sem-5), SOS(let-341), Raf(lin-45), MEK(mek-2), and<br />

MAPK(sur-1/mpk-1) have been identified, little is known regarding modulation or negative regulation of<br />

this pathway in C. <strong>elegans</strong> or in any system.<br />

To identify modulators and negative regulators of this pathway, a genetic screen was undertaken using<br />

an allele of let-23, sa62, that encodes a ligand-independent activated receptor. In wildtype<br />

hermaphrodites or animals harboring one copy of sa62, three Pn.p cells adopt vulval fates. However, in<br />

the presence of two copies of sa62, on average, four cells acquire vulval fates. sa62 heterozygous<br />

animals were mutagenized and screened for mutants with enhanced vulval induction. To date, at least<br />

seven different loci have been isolated, and six have been assigned linkage to specific chromosomes.<br />

The best characterized mutation, sy598, also enhances the activity of a reduction in function allele of<br />

let-23, sy1, but has no effect on its own or in sensitized backgrounds harboring loss of function mutations<br />

in the negative regulators sli-1 or gap-1, or a gain of function mutation in let-60. Laser microsurgery<br />

experiments indicate sy598 does not regulate the production of LIN-3(EGF) in the somatic gonad. These<br />

data suggest that sy598 may control LET-23 levels in the Pn.p cells. sy598 has been mapped to linkage<br />

group IV, and its identity is being sought.<br />

209


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

THE TRAMPOLINE ASSAY: A NEW METHOD FOR<br />

MEASURING THE STEP RESPONSE OF THE CHEMOTAXIS<br />

MECHANISM IN C. ELEGANS.<br />

Moravec, M.L, Cervantes, J., Lockery, S.R.<br />

Institute of Neurosci., Univ of Oregon, Eugene, OR 97403<br />

Chemotaxis in C. <strong>elegans</strong> involves a series of abrupt turns (pirouettes) triggered by movement down a<br />

gradient of chemical attractant (Pierce-Shimomura, J.T., et al.,J. Neurosci. 19:9557-9569, 1999). Analysis<br />

of the time series of concentration change experienced by a chemotaxing worm, together with its<br />

pirouette record, suggests a three-stage model in which instantaneous attractant concentration is<br />

differentiated, smoothed by low-pass filter, and thresholded by a sigmoidal function relating filter output to<br />

pirouette probability. This model predicts that a sudden decrease in attractant concentration will produce<br />

a sudden increase in pirouette probability. Moreover, the increase in probability should decay<br />

approximately exponentially with a time constant that reflects the worm’s memory for concentration<br />

changes in the recent past. To test these predictions, we have devised an apparatus that allows us to<br />

stimulate an unteathered worm with a nearly instantaneous (step-wise) change in the concentration of<br />

soluable attractants such as NaCl. The apparatus consists of a thin (10 mm) agarose film suspended over<br />

a buffer-filled chamber, resembling trampoline placed over a swimming pool. The underside of the<br />

agarose film contacts the surface of the buffer solution, while the top side of the film contacts the air. The<br />

worm is placed on the top of the film and allowed to adapt to a buffer containing a high concentration of<br />

attractant for 5 min. The chamber is then drained and quickly refilled with buffer containing a low<br />

concentration of attractant. Preliminary results indicate that a step-wise decrease in attractant<br />

concentration causes an immediate increase in pirouette probability, as predicted by the model. We plan<br />

to use the step response to investigate the time course of the worm’s memory for concentration changes,<br />

and how this memory is affected by mutations and neuronal ablations. Supported by NIMH MH51383,<br />

and NSF IBN9458102,<br />

210


IDENTIFICATION OF GENES REGULATING BODY LENGTH IN<br />

THE DBL-1 PATHWAY BY DIFFERENTIAL HYBRIDIZATION<br />

OF ARRAYED CDNAS<br />

Kiyokazu Morita 1 , Makoto Mochii 1 , Yukiko Sugihara 1 , Satoru<br />

Yoshida 1 , Yo Suzuki 2 , William B. Wood 2 , Yuji Kohara 3 , Naoto Ueno 1<br />

1 Division of Morphogenesis, Department of Developmental Biology, National Institute for Basic Biology,<br />

Okazaki 444-8585, JAPAN<br />

2 Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, USA<br />

3 National Institute of Genetics, Mishima 411-8540, JAPAN<br />

dbl-1 has been shown to regulate C. <strong>elegans</strong> body length and male tail ray patterning 1, 2). To identify<br />

downstream target genes of DBL-1 signaling, we screened arrayed cDNAs using differential hybridization<br />

(HDF) analysis. C. <strong>elegans</strong> cDNAs representing 7,584 independent genes were arrayed on a nylon<br />

membrane at a high density and hybridized with 33P-labeled DNA probes synthesized from mRNAs,<br />

which were isolated from L3-stage worms. We compared signals from dbl-1(-) worms with those from N2<br />

worms3), dbl-1(++) worms that carry multiple copies of a dbl-1 genomic fragment, or dbl-1(-); HSP::dbl-1<br />

worms that express DBL-1 protein upon heat shock. Many genes positively or negatively regulated by the<br />

dbl-1 signal were identified. dsRNAi experiments were performed with all the clones identified. Several<br />

clones caused Sma or Lon phenotypes by dsRNAi. These results demonstrate that analysis with arrayed<br />

cDNA combined with dsRNAi gene inactivation is a powerful approach to identify genes that are directly<br />

or indirectly regulated by an extracellular signal.<br />

1)K. Morita et al., (1999) Development 126, 1337-1347<br />

2)Y. Suzuki et al., (1999) Development 126, 241-250<br />

<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

3)M. Mochii et al., (1999) Proc. Natl. Acad. Sci. USA 96, 15020-15025<br />

211


MUTATIONS IN THE EPHRIN MAB-26/EFN-4 CAUSE<br />

DEFECTS IN CLOSURE OF THE GASTRULATION CLEFT AND<br />

IN EPIDERMAL ENCLOSURE<br />

Sarah L. Moseley, Andrew Chisholm<br />

Department of Biology, UC Santa Cruz, CA 95064<br />

<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

Signaling involving the Eph receptor tyrosine kinase VAB-1 and its ephrin ligand VAB-2/EFN-1 is required<br />

for normal epidermal and neuronal morphogenesis. Our lab has shown that mab-26, identified by virtue of<br />

its role in male tail morphogenesis, corresponds to another C. <strong>elegans</strong> ephrin ligand, efn-4 (see abstract<br />

by Holcomb et al.).<br />

mab-26/efn-4 null mutants display incompletely penetrant defects in epidermal morphogenesis.<br />

Approximately 10% arrest during embryogenesis, and about 20% arrest as larvae. Surviving mab-26<br />

adults do not display the head morphology defects (’Notched head’) characteristic of vab-1 or vab-2<br />

mutants, but are instead typically defective in morphogenesis of the posterior body and tail epidermis.<br />

Using 4-D microscopy we have found that mab-26 embryos, like vab-1 and vab-2 mutants, are defective<br />

in the closure of the ventral gastrulation cleft by neuroblast movements, and in later enclosure of the<br />

embryo by epidermal cells. Interestingly, almost all mab-26 embryos show delays in gastrulation cleft<br />

closure, although in only some cases does this result in later morphogenetic defects. Thus, the defects of<br />

mab-26 mutants are different from (although partly overlapping with) those of vab-1 and vab-2 mutants.<br />

mab-26 mutations also display unexpected synthetic-lethal interactions with all vab-1 and vab-2 mutations<br />

tested. Double mutants show completely penetrant morphogenetic defects, in striking contrast to the<br />

incompletely penetrant vab-1 null phenotype. We are currently analyzing the phenotypes of these double<br />

mutants.<br />

The differences in mutant phenotypes and the synthetic lethal interactions are inconsistent with EFN-4<br />

acting only as a ligand for VAB-1. If EFN-4 is not signaling through the VAB-1 receptor, how is it<br />

functioning? To identify other genes required for mab-26 function we are screening for new mutations that<br />

are synthetic-lethal with a weak vab-1 allele. Results of a pilot screen will be presented.<br />

212


CELLULAR AND DEVELOPMENTAL EVENTS REQUIRED TO<br />

GENERATE FUNCTIONAL MUSCLE IN C. ELEGANS.<br />

K. Norman, S. Cordes, G. Mullen, P. Rahmani, T. Rogalski, D.<br />

Moerman<br />

Dept of Zoology, UBC<br />

<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

In C. <strong>elegans</strong> a physical linkage between the myofilament lattice of the body wall muscle and the cuticle is<br />

required to allow myofilament contraction to result in the movement of the animal. This complex linkage<br />

system is composed of integrin containing adhesion-complexes within the body wall muscle cells that<br />

anchor the myofibrils to the muscle cell membrane, a specialized basement membrane underlying the<br />

muscle quadrant, and hemidesmosomal-like structures present in the epidermis. The epidermis has<br />

specialized faces, one to anchor the epidermis to the basement membrane and the other to secrete and<br />

anchor the cuticle. The nematode provides a very elegant system to study the interaction between the<br />

body wall muscle and the epidermis during development. Through genetic and molecular approaches we<br />

have identified (1) several components involved in assembly, localization, and maintenance of adhesion<br />

complexes within muscle (the unc-52, unc-97, unc-112, dim-1 and spc-1 gene products), (2) components<br />

that are involved in structuring the underlying basement membranes (the let-268 gene product), and (3)<br />

others that function in regulating epidermal attachment structures (the unc-23 gene product). The role of<br />

these molecules in muscle and epidermal development during C. <strong>elegans</strong> morphogenesis will be<br />

described. For more information on unc-23 and unc-97 see posters by P. Rahmani and S. Cordes,<br />

respectively.<br />

213


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

IS THE DAG KINASE DGK-1 AN EFFECTOR OF GO ALPHA<br />

(GOA-1)?<br />

Stephen Nurrish, Michael Dybbs, Joshua Kaplan<br />

Molecular and Cell Biology, University of California, Berkeley, CA 94720<br />

We (and others) have previously described two competing G-protein pathways acting in motor neurons to<br />

either facilitate or inhibit synaptic transmission at neuromuscular junctions (NMJs) [1-4]. Facilitation of<br />

release occurs via a pathway composed of a Gq alpha (EGL-30), a phospholipase C beta (EGL-8), and<br />

the diacylglycerol (DAG) binding protein UNC-13 [2,4]. The Gq alpha pathway can be activated by<br />

muscarinic agonists leading to the formation of the membrane bound second messenger DAG by EGL-8<br />

and the subsequent recruitment of UNC-13 to sites of Acetylcholine release [2]. UNC-13 is essential for<br />

neurotransmitter release and it’s enrichment at release sites correlates with an increase in<br />

neurotransmitter release. Addition of serotonin agonists inhibits acetylcholine release at NMJs. Inhibition<br />

of synaptic transmission by serotonin requires Go alpha (GOA-1) and a diacylglycerol kinase (DGK-1)<br />

which phosphorylates DAG to Phosphatididic Acid which is unable to bind UNC-13. Serotonin antagonists<br />

and goa-1 mutations result in the recruitment of UNC-13 to NMJs suggesting that the Gq and Go<br />

pathways converge to regulate UNC-13 localization, most likely by regulating levels of DAG [1,2]. As yet<br />

the Go signaling pathway is not well defined. Go may reduce DAG levels by activating DGK-1, by<br />

negatively regulating the Gq signaling pathway, or by an as yet undefined pathway. We are currently<br />

testing the first of these models. Co-expression of DGK-1 and activated Go alpha in human tissue culture<br />

cells (HEK cells) has no effect on the kinase activity of DGK-1. This suggests that there is no direct<br />

interaction between Go alpha and DGK-1. However, it is possible that Go alpha indirectly regulates<br />

DGK-1 via intermediates that are not conserved in HEK cells. Therefore we have generated a rescuing<br />

MYC tagged GFP::DGK-1 construct. The myc-DGK-1 protein can be immunoprecipitated from adult<br />

animals and possesses DAG kinase activity. We are currently testing whether the DGK-1 DAG kinase<br />

activity changes in response to levels of Go alpha signaling. We are also testing whether Go regulates the<br />

subcellular localization of DGK-1.<br />

1.<br />

2. Nurrish et al.,(1999) Neuron:24 p231-242<br />

3. Lackner et al.,(1999) Neuron:24 p335-346<br />

4. Hajdu-Cronin et al.,(1999) Genes Dev 13: 1780-93<br />

5. Miller et al.,(1999) Neuron 24: 323-33<br />

214


TRANSFORMING NEMATODES INTO INSECTS:<br />

UNDERSTANDING BT-RESISTANCE<br />

Johanna O’Dell, Raffi Aroian<br />

<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

University of California, San Diego, La Jolla, CA 92093<br />

The utilization of transgenic crops expressing Bacillus thuringiensis (Bt) toxins is becoming a prevalent<br />

alternative to chemical pesticides. Bt toxins have enormous potential as they are not harmful to<br />

vertebrates, demonstrate a high degree of species-specific toxicity, and are a more environmentally<br />

friendly pest control agent. Currently genetically engineered corn and cotton are being extensively used in<br />

agriculture and express highly similar toxins. It is predicted that resistance to these toxins will develop<br />

within the next 10 years.<br />

We are interested in identifying the cellular machinery underlying Bt-toxicity and resistance and are using<br />

the nematode <strong>Caenorhabditis</strong> <strong>elegans</strong> as a model system. The physiological mode of Bt action has been<br />

elucidated from insect studies. Following ingestion by a susceptible animal, Bt toxin is solubilized and<br />

activated by proteolytic processing. Active toxin interacts with receptors in the gut and becomes inserted<br />

into the apical membrane creating a channel, ultimately leading to death. Despite our knowledge of this<br />

process, we know very little about the molecular components required for toxin action.<br />

Our laboratory has shown that C. <strong>elegans</strong> is susceptible to some Bt toxins and has successfully isolated<br />

mutants resistant to one Bt toxin (see abstract by Marroquin et al). One of these mutants, called bre-2 for<br />

Bacillus toxin resistant mutant, maps to the right arm of chromosome III. Three-factor mapping<br />

experiments position bre-2 very close to dpy-18. We are performing transgenic rescue experiments and<br />

additional mapping by polymorphisms to ascertain the molecular identity of this gene.<br />

To determine whether our studies are directly applicable to understanding pest resistance to transgenic<br />

crops we are attempting to make a C. <strong>elegans</strong> strain susceptible to Cry1Ac, the major Bt toxin used in<br />

transgenic cotton. As toxin specificity is thought to result from binding to species-specific receptors, our<br />

strategy involves the creation of transgenic worms that express the known Cry1Ac receptor,<br />

aminopeptidase N (APN). If APN-expressing worms prove to be sensitive to Cry1Ac toxin, we will<br />

determine whether the bre genes are required for a common pathway of Bt toxicity or for a Cry5B-specific<br />

pathway.<br />

215


THE CYTOSKELETAL PROTEIN ZK370.3 MAY CONTRIBUTE<br />

TO OOCYTE DEVELOPMENT AND FERTILIZATION<br />

Alex Parker, Ann M. Rose<br />

<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

Department of Medical Genetics, University of British Columbia, 6174 University Blvd., Vancouver, B.C.,<br />

V6T 1Z3, Canada<br />

The cytoskeleton is a dynamic structure that facilitates many events within a cell. We are using the model<br />

organism <strong>Caenorhabditis</strong> <strong>elegans</strong> to study a cytoskeletal gene in the context of animal development. The<br />

C. <strong>elegans</strong> gene product, zk370.3, is conserved through evolution, from yeast to humans. Work in yeast<br />

has determined a cellular role for this gene in the maintenance of the actin cytoskeleton, as well as in<br />

endocytosis. However, very little is known about the role of this gene in the development of higher<br />

organisms. We have used RNA interference to investigate the loss of function phenotype for this gene<br />

product (protocol after Tavernarakis et al. 2000). Treated animals lay significantly more unfertilized<br />

oocytes than controls. This experiment suggests that silencing of the endogenous zk370.3 gene product<br />

results in developmental arrest prior to oocyte fertilization. We are working to determine where the defect<br />

resides. Using a portion of the cDNA fused to GFP under control of the zk370.3 promoter we observed<br />

expression in the proximal gonad, spermatheca and pharynx. Expression in the spermatheca is observed<br />

when gametogenesis is occurring, at late larval stages and early adulthood. Pharyngeal expression can<br />

first be observed in early larval stages, and is maintained through adulthood. We have raised a polyclonal<br />

antibody to zk370.3 for use in immunofluorescence microscopy experiments. The localization of zk370.3<br />

protein detected by the antibody is in agreement with our expression pattern analysis.<br />

216


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

OXIDANT STRESS RESPONSES IN C. ELEGANS<br />

Farhang Payvar, Andrew DeMatteo, Tom Hazinski<br />

Department of Pediatrics, Vanderbilt University Medical School, Nashville, TN, 37232<br />

We are intrigued by the genetic events triggered with hyperoxia and how they might differ from oxidative<br />

stress per se. Hyperoxia can influence gene expression by transcriptional (JCI 96:2083-2089, 1995)<br />

and/or post-transcriptional mechanisms leading directly or indirectly to oxidant stress (OS). In C. <strong>elegans</strong>,<br />

insulin-like signaling pathway regulates life span and sensitivity to OS. For example, animals with<br />

mutations in DAF-2 (insulin-like receptor) or AGE-1 (PI3K) are relatively resistant to OS and have<br />

extended life span; mutations in DAF-16 (fork head transcription factor) suppress the phenotypes of daf-2<br />

and age-1 mutants, suggesting that DAF-16 is target of negative regulation by upstream AGE-1 and<br />

DAF-2.<br />

Two forms of OS-induction not previously explored in C. <strong>elegans</strong> were examined. The wild type (N2) &<br />

mutant (daf-2, age-1, daf-16) animals were exposed to 3.5 days of sodium nitroprusside (NP), an OS<br />

inducer, in normoxic (21 % oxygen) or hyperoxic (95% oxygen) environments. Survival rates were<br />

measured, and the LD50 for NP treatment with and without hyperoxia was calculated.<br />

Our results indicate that survival in wild-type animals is reduced by NP but not by hyperoxia. However,<br />

hyperoxia increased the sensitivity to NP by ~ 3-fold. This increase in NP sensitivity suggests that the<br />

genetic events mediating the effects of hyperoxia are, at least in part, distinct from those for NP in C.<br />

<strong>elegans</strong>, and that there is cross-talk between hyperoxia and NP signaling pathways. Moreover, response<br />

to NP, alone or in combination with hyperoxia is altered in several mutant C. <strong>elegans</strong> strains. Consistent<br />

with the conventional DAF-16 pathway model, age-1 mutants are relatively NP-resistant, suggesting that<br />

AGE-1 is involved in NP signaling. Unexpectedly, age-1 mutants are only ~ 17 % reduced in their<br />

sensitivity to hyperoxia (+NP), pointing to alternate players/pathways for transmission of hyperoxic signal.<br />

Surprisingly, daf-2 mutants are hypersensitive to NP & have nearly lost their NP + hyperoxia-response<br />

phenotype. These findings are consistent with the notion that NP and hyperoxia signals are relayed via<br />

DAF-2 to downstream molecules that are distinct from AGE-1.<br />

217


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

PHARYNGEAL PUMPING DEFECTS IN UNC-103 MUTANTS<br />

Christina I. Petersen 1 , David J. Reiner 2 , Elizabeth M. Newton. 3 ,<br />

James H. Thomas 3 , Jeffrey R. Balser 4<br />

1 Dept. of Anesthesiology, Room 560B MRB II, Vanderbilt University, Nashville, TN 37232-6600<br />

2 Howard Hughes Medical Institute and Dept. of Molecular And Cell Biology, 401 Barker Hall #3204,<br />

University of California, Berkeley, CA 94720-3204<br />

3 Dept. of Genetics, Box 357360, University of Washington, Seattle, WA 98195<br />

4 Dept. of Anesthesiology, Room 560 MRB I, Vanderbilt University, Nashville, TN 37232-6600<br />

Human ether-a-go-go (HERG) potassium channels play a critical role in cardiac repolarization and have<br />

been genetically linked to an inherited arrhythmia, the long QT syndrome. unc-103 encodes the worm<br />

ortholog of HERG with 70 % amino acid identity in the conserved transmembrane and pore regions.<br />

unc-103 gain-of-function (gf) mutants display a variety of phenotypes reflecting reduced muscle<br />

excitation. These phenotypes include defective egg-laying, paralysis and defects in defecation. Because<br />

the worm pharynx displays many similarities to the mammalian heart, we examined unc-103 gf mutants<br />

for defects in pharyngeal pumping. We find that these mutants display lengthy pauses in pharyngeal<br />

pumping (as long as 7 seconds), reminiscent of cardiac bradydysrhythmias. Notably, in wildtype worms,<br />

HERG-specific blockers such as dofetilide and d-sotalol slow the rate of pharyngeal pumping, but do not<br />

induce pauses. Promoter-GFP fusion constructs reveal that unc-103 is expressed in I1, I2 and NSM<br />

neurons, which innervate the pharynx. The molecular nature of the unc-103 gf mutant is an A to T change<br />

at position 334 in the S6 domain, a region important for potassium channel gating. To understand the<br />

diverse effects of the unc-103 gf mutation and HERG-specific blockers, we are undertaking an<br />

electrophysiological characterization of the wild type and UNC-103 gf mutant using a mammalian<br />

heterologous expression system. These studies may clarify whether complex ion channel gating effects<br />

underlie differences between the unc-103 gf phenotype and the effects of HERG blockers in the pharynx.<br />

Furthermore, comparing the electrophysiological properties of wt UNC-103 with HERG in our expression<br />

system will also allow us to address structure/function differences using chimeric constructs of worm and<br />

human channels.<br />

218


A REQUIREMENT FOR C. ELEGANS RHO-BINDING KINASE<br />

IN EARLY CLEAVAGE<br />

Alisa J. Piekny, Paul E. Mains<br />

<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

Genes and Development Research Group, Department of Biochemistry and Molecular Biology, University<br />

of Calgary, Calgary, AB. CANADA T2N 4N1<br />

Our lab is investigating genes that regulate the actin-mediated cell shape changes that drive C.<strong>elegans</strong><br />

embryonic elongation. let-502 encodes a Rho-dependent kinase and strong antimorphic mutations display<br />

early larval arrest due to failed elongation 1 . We recently isolated hypomorphic homozygous viable let-502<br />

mutations. Some of these alleles sufficiently decrease maternal expression to reveal a new, early<br />

embryonic phenotype. The embryos either do not complete any cell cleavages or form cleavage furrows<br />

that regress and result in multinucleated embryos. Some of these embryos also show defects in<br />

asymmetry, some appear larger than wild-type embryos and this may indicate defects in oocyte<br />

formation.<br />

Consistent with a role in cleavage furrow formation, LET-502 localizes to the furrow during early cell<br />

divisions. mel-11 encodes a myosin phosphatase and suppresses let-502 elongation defects 1,2 . A<br />

hypomorphic mel-11 mutant also suppresses the cleavage defects caused by let-502 mutants suggesting<br />

that mel-11 also may play a role in cytokinesis. However, these mel-11 mutant embryos do not show<br />

cleavage defects on their own.<br />

mlc-4 encodes a nonmuscle regulatory light chain and is a downstream target for both let-502 and mel-11<br />

in embryonic elongation. mlc-4 previously has been shown to play a role in early cleavage 3 . Together,<br />

these results suggest that the pathway regulating embryonic elongation may be similar to the pathway<br />

regulating early cleavage. In higher eukaryotes nonmuscle myosin, Rho, RhoGEF and the Rho-binding<br />

kinase effector, Citron-kinase, have been shown to regulate cytokinesis and cleavage furrow formation.<br />

Since the predicted C. <strong>elegans</strong> homologue for Citron has no associated kinase domain, in C. <strong>elegans</strong><br />

LET-502 could be the kinase required for early cleavage. let-502’s role in cytokinesis is currently being<br />

investigated using various molecular and genetic tools.<br />

1. Wissmann, A., J. Ingles, J.D. McGhee and P.E. Mains, 1997. Genes Dev. 11:409-422.<br />

2. Wissmann, A., J. Ingles and P.E. Mains, 1999. Develop. Biol. 209:111-127.<br />

3. Shelton, C. A., J.C. Carter, G.C. Ellis and B. Bowerman, 1999. J. Cell. Biol. 146: 439-451.<br />

219


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

FUNCTION OF THE RECEPTOR TYROSINE KINASE<br />

CAM-1/KIN-8 IN COORDINATED MOVEMENT<br />

S. Poulson, D. Madsen, A.V. Maricq<br />

Department of Biology, University of Utah, Salt Lake City UT 84112<br />

Receptor Tyrosine Kinases (RTKs) are important for many types of cell-to-cell signaling. Many<br />

developmental processes, including the development of synapses, are mediated by signal transduction<br />

through RTKs. Recently, a Ror-like RTK, KIN-8, was identified in C. <strong>elegans</strong>. Forrester and Garriga<br />

isolated a kin-8 (renamed cam-1) mutation and described the role of CAM-1 in normal migration of the<br />

CAN neurons. Koga and Ohshima later addressed the daf-c phenotype of cam-1 mutants. Independently,<br />

we had studied the expression pattern of kin-8/cam-1, found that it was expressed in many neurons as<br />

well as muscles and muscle arms, and generated a deletion mutation (ak37) in the gene. The mutant<br />

worms were severely uncoordinated, defective in mechanosensation, and showed a kinked head and<br />

withered tail. We are interested in why these worms are so defective in locomotion.<br />

Mechanosensory defects combined with locomotory defects suggested kin-8/cam-1 may be required for<br />

the function of interneurons that subserve locomotion. Using nmr-1::GFP to visualize the command<br />

interneurons (AVA, AVB, AVD, AVE, and PVC), we found that 85% of kin-8/cam-1(ak37) mutants exhibit<br />

aberrant axon outgrowth in these interneurons. We are currently investigating whether kin-8/cam-1 is<br />

required in the commnand interneurons for axon outgrowth. Uncoordination could also result from defects<br />

in synaptic or muscle function. Using electrophysiological recording techniques, we plan to record<br />

ligand-gated currents from identified neurons and muscles.<br />

220


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

THE AUTOSOMAL SEX SIGNAL IN C. ELEGANS?<br />

Jennifer R. Powell, Barbara J. Meyer<br />

HHMI and Department of Molecular and Cell Biology, University of California at Berkeley 94720.<br />

In C. <strong>elegans</strong>, sex is determined by the ratio of X chromosomes to sets of autosomes. The low X:A ratio<br />

in XO diploid worms permits high expression of the developmental switch gene xol-1, and hence male<br />

development ensues. Conversely, a high X:A ratio in XX animals results in low xol-1 activity and<br />

hermaphrodite development. The X portion of the sex signal is comprised of several X-signal elements<br />

(XSE) that repress the activity of xol-1 in a dose-dependent manner. The nature of the autosomal<br />

component of the sex signal is unknown, but presumably increases the activity of xol-1. It is possible that<br />

discrete autosomal signal element genes (ASE) exist that are analogous to, but act in opposition to, the<br />

XSEs. To test this hypothesis, we are performing a genetic screen to isolate loss-of-function mutations in<br />

ASEs, if they exist.<br />

This screen is based on the fact that the sex signal is cumulative; therefore, we expect loss-of-function<br />

mutations in ASEs to rescue the XX-specific lethality caused by removing XSEs. Specifically, XX worms<br />

homozygous for mutations in the two XSEs fox-1, an RNA-binding protein, and sex-1, a nuclear hormone<br />

receptor, are completely inviable. We are screening for suppressor mutations that rescue this lethality;<br />

these are candidate ASE mutations. The fox-1 sex-1 starting strain can be maintained as viable<br />

hermaphrodites if these worms also overexpress sdc-2(+), a downstream gene in the pathway, from an<br />

extrachromosomal array. Mutagenized worms that contain a suppressor mutation will no longer depend<br />

on the array for viability, so we screen through F1, F2, and F3 generations to look for live<br />

non-array-bearing hermaphrodites that potentially contain dominant, recessive, or maternal effect<br />

suppressor mutations.<br />

In initial rounds of mutagenesis, we have screened approximately 2000 haploid genomes and isolated 10<br />

suppressor mutations. They include autosomal and X-linked mutations, and show a range of suppression<br />

phenotypes. There are several classes of suppressors that could be recovered from this screen, including<br />

ase (lf), xol-1 (lf), sdc-2 (gf), xse (gf), and fox-1 or sex-1 revertants. We are currently characterizing our<br />

existing mutants and continuing to screen for additional suppressors.<br />

221


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

GOT THE BLUES? TRY ANOTHER GENETIC SCREEN!<br />

Chad Rappleye 1 , Rebecca Lyczak 2 , Bruce Bowerman 2 , Raffi Aroian 1<br />

1University of California, San Diego, La Jolla, CA 92093<br />

2University of Oregon, Eugene, OR 97403<br />

To better understand the process by which cell polarity is established, we are analyzing genes required<br />

for the characteristic asymmetries of the early C. <strong>elegans</strong> embryo. Recently, we showed that the pod-1<br />

locus represents a new class of polarity genes as mutation of pod loci results in osmosensitive embryos in<br />

addition to loss of polarity (’pod’=polarity and osmotic defective). The identification of pod-2 by our lab<br />

(see abstract by A. Tagawa) further underscores the importance of the pod class of mutants. The<br />

osmosensitivity of pod mutants appears to result from permeability of the egg shell surrounding the<br />

embryo. Understanding the connection between polarity establishment and egg shell formation should<br />

begin to reveal the molecular mechanisms underlying cellular asymmetry.<br />

To further identify the set of components responsible for this aspect of polarity establishment, we have<br />

initiated a study of mutants that have osmosensitive phenotypes. Among previously identified osmotically<br />

sensitive mutants, emb-8 and emb-30 mutants also exhibit the ’Pod’ phenotype. In these, as well as a<br />

pod-1 null mutant, the polarity defect is not completely penetrant suggesting that multiple, parallel<br />

pathways might contribute to overall cell polarity. However, analysis of double mutants between pod-1<br />

and emb-8 or emb-30 indicate that these three function in a common genetic pathway.<br />

Through two different genetic screens, one of which directly identifies osmosensitive embryos, we have<br />

isolated a number of other mutations that represent new loci of the pod class. This collection includes<br />

genes which when mutated cause near 100% penetrant loss of polarity. We are currently pursuing the<br />

molecular identities of these loci which will enable us to refine our models for how cellular asymmetry is<br />

controlled.<br />

222


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

IDENTIFICATION OF COMPONENTS OF THE MEIOTIC<br />

MACHINERY IN C. ELEGANS<br />

Kirthi Reddy, Monica Colaiacovo, Gillian Stanfield, Anne Villeneuve<br />

Dept. of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305<br />

Meiosis is the specialized form of cell division that diploid cells undergo to produce gametes with a<br />

haploid chromosome number. During prophase of meiosis I, homologous chromosomes locate each other<br />

(pair), associate tightly along their lengths (synapse), and undergo recombination. Failure to complete any<br />

of these processes can result in improper chromosome segregation. We are interested in the molecular<br />

mechanisms that are involved in chromosome pairing, synapsis and crossover formation. We have<br />

initiated a functional genomics strategy to identify components of the C. <strong>elegans</strong> meiotic machinery (see<br />

abstract by Colaiacovo et al). Microarray analysis has identified a set of genes that are upregulated in the<br />

germline and are not differentially expressed in male and female germ cells (V. Reinke, S. Kim and<br />

collaborators). We are screening a subset of these genes to identify those for which RNAi elicits defects<br />

in meiosis.<br />

We initially focused on genes encoding proteins with predicted coiled-coil domains, since several proteins<br />

in this class have been implicated in chromosome structure and behavior. We have identified two genes,<br />

F26D2.2 and F39H2.4, that appear to be involved in homologous chromosome synapsis. Both have RNAi<br />

phenotypes characteristic of animals with severe chromosome segregation defects -- affected<br />

hermaphrodites produce a high percentage of dead embryos, and among the few survivors, there is a<br />

high incidence of males. Further, affected animals show achiasmate chromosomes late in meiotic<br />

prophase, likely reflecting a failure to form crossovers. Earlier in prophase, when chromosomes should be<br />

aligned and synapsed, affected animals exhibit extensive asynapsis. The chromatin morphology seen in<br />

the affected animals is characteristic of sys-1 and sys-2 mutants, which are unable to stabilize<br />

associations between homologs (see abstract by MacQueen and Villeneuve). Indeed, the sys-1(me17)<br />

allele has a stop mutation in the F26D2.2 gene. No previously identified synapsis-defective mutants map<br />

near F39H2.4, which we have designated sys-3.<br />

Our RNAi screen for genes required for meiosis is ongoing. We will report on additional genes that we<br />

identify on the basis of meiotic defects elicited by RNAi.<br />

223


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

NOVEL AND ATYPICAL RECEPTOR TYROSINE KINASES IN<br />

MORPHOGENESIS.<br />

David J. Reiner, Lewis Leng, Barbara J. Meyer<br />

Howard Hughes Medical Institute and Department of Molecular and Cell Biology, University of California,<br />

Berkeley, CA 94720.<br />

Most receptor tyrosine kinases (RTKs) fall into one of a number well-characterized growth factor receptor<br />

families. However, in C. <strong>elegans</strong> we have used the genome sequence to identify four apparently novel<br />

receptors and two atypical receptors with mammalian homologs. Over-expression of five of these six<br />

genes results in variable, lumpy lethality, while RNAi of four of these genes results in visible phenotypes.<br />

The RNAi causes mainly morphogenetic phenotypes. For example, RNAi for one of these genes causes<br />

partially penetrant lethality due to extreme hypodermal deformities, and some escaping animals have<br />

pronounced axon guidance and fasciculation defects. The atypical receptors are apparently nematode<br />

homologs of the mammalian Discoidin Domain Receptors (DDRs), RTKs that function as collagen<br />

receptors. The in vivo function of DDRs is unknown, and we hope to address this issue in C. <strong>elegans</strong>.<br />

Candidate mutations have been identified for only one of the six receptors. T14E8.1 rescues the weakly<br />

penetrant tail phenotypes of mab-19, but we were unable to find lesions corresponding to the two mab-19<br />

alleles. A heat-shock promoter driven RNAi foldback construct of T14E8.1 causes moderate, incompletely<br />

penetrant Mab phenotypes comparable to mab-19, plus a novel truncation of certain tail sensory rays. To<br />

identify deletion mutations in these genes, we are currently constructing a large deletion library to be<br />

screened with PCR. Future directions depend on the outcome of these screens.<br />

224


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

DIFFERENTIAL EFFECTS OF HEAT SHOCK AND COLD<br />

SHOCK FOLLOWING MASSED AND DISTRIBUTED<br />

LONG-TERM HABITUATION TRAINING IN C. ELEGANS<br />

Jacqueline Rose 1 , Kenneth Eng 2 , Catharine Rankin 1<br />

1Department of Psychology, University of British Columbia, Vancouver B.C.<br />

2Neuroscience Graduate Program, University of British Columbia, Vancouver B.C.<br />

A new en masse long-term habituation (LTH) training procedure was used to examine the effects of heat<br />

and cold shock on LTH of the tap withdrawal response. In order to induce LTH a distributed training<br />

protocol was used where groups of worms received four blocks of 20 taps separated by one-hour rest<br />

periods and testing each worm individually 24 hours later. LTH following massed training was also<br />

examined by administering one block of 120 taps to worm groups. <strong>Worm</strong>s that received massed training<br />

did not show LTH when tested 24 hours later. Temperature shock was administered by submerging<br />

sealed worm-containing petri plates into either a 32° C (heat shock) or a 0° C bath for the first 40<br />

minutes following training blocks: worms were kept at room temperature for the last 20 minutes. <strong>Worm</strong>s in<br />

the control condition underwent similar submersion during rest periods at 21° C (room temperature). It<br />

was found that when heat shock is administered between distributed training blocks an attenuation of<br />

LTH results. Predictably, heat shock after massed training did not produce LTH. Interestingly, when<br />

worms receive distributed training blocks with cold shock between blocks, LTH of the tap withdrawal<br />

response is at least retained and may be enhanced. Similar to before, LTH did not result from massed<br />

training followed by cold shock. Twelve-hour retention has also been examined with distributed training<br />

and similar findings resulted where LTH was induced after room-temperature submersion and cold shock<br />

but not heat shock. Currently, studies are investigating if massed training results in LTH if measured 12<br />

hours rather than 24 hours later. As well, the effects of heat shock and cold shock on 12-hour retention<br />

following massed training are being examined. The results that have been reported thus far indicate that<br />

differential mechanisms may underlie both LTH resulting from massed versus distributed training and the<br />

effects of heat shock versus cold shock.<br />

Research funded by the Natural Sciences and Engineering Research Council of Canada<br />

225


A NEW EN MASSE TRAINING PROCEDURE TO STUDY<br />

LONG-TERM HABITUATION IN C. ELEGANS<br />

Jacqueline Rose, Catharine Rankin<br />

<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

Department of Psychology, University of British Columbia, Vancouver B.C.<br />

Past research examining long-term habituation (LTH) of the tap withdrawal response in C. <strong>elegans</strong><br />

required training individual worms with three blocks of 20 tap-trains separated by one hour rest periods<br />

with test tap-trains delivered 24 hours later. This single worm method was time and labor intensive. In an<br />

attempt to increase throughput, a between-groups design was implemented. Groups of worms were<br />

trained at a 60 s interstimulus interval by receiving either 4 blocks of 20 taps with one-hour rest periods<br />

(distributed) or 1 block of 80 taps (massed). Testing was conducted 24 hours after training with single<br />

worms individually being given one block of 10 taps. Trained groups were compared to a control group<br />

that received a single tap on training day and the same individual testing on day two. The results showed<br />

that worms that received distributed training show significantly smaller responses compared to single-tap<br />

controls. <strong>Worm</strong>s that underwent massed training showed no difference in response magnitude compared<br />

to the control group. Since LTH was only found following distributed training a follow-up experiment was<br />

conducted where worms were given either one, two, three or four training blocks separated by one hour<br />

rest periods and were tested individually 24 hours later. It was found that worms that received 4 training<br />

blocks showed the greatest LTH while worms that received a single training block showed no LTH<br />

demonstrating that an effect of accumulation of learning on LTH. Together these results effectively<br />

replicate what has previously been found with the single worm training method showing this new protocol<br />

can now be used to more effectively assess LTH in a variety of situations.<br />

Research funded by the Natural Sciences and Engineering Research Council of Canada.<br />

226


GLOBAL PATTERNS OF EXPRESSION PATTERNS IN<br />

MUSCLE USING MRNA-TAGGING<br />

Peter J. Roy, Stuart Kim<br />

<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

Department of Developmental Biology, Stanford University, 279 Campus Drive, Stanford, CA 94305<br />

Gene networks that control development must endow each unique cell with distinguishing properties.<br />

Microarray technology along with the completed C. <strong>elegans</strong> genome now makes it possible to know these<br />

gene networks in a comprehensive manner. Our microarrays contain PCR fragments representing nearly<br />

every predicted C. <strong>elegans</strong> gene. By hybridizing labeled cDNA probes from different samples to the<br />

microarray, one can simultaneously determine the expression differences between those two samples for<br />

every gene. A limitation of current microarray technology is that RNA is isolated from whole worms, in<br />

turn, making tissue specific expression patterns difficult to detect. We have developed a strategy to<br />

circumvent this problem of identifying the mRNA profiles in specific tissues or cells by using an approach<br />

we call "mRNA-tagging".<br />

To isolate cell-specific mRNA, we are using previously characterized promoters to drive the expression of<br />

epitope-tagged protein that binds mRNA in a non-biased fashion in a defined set of cells. By<br />

immunoprecipitating the tagged mRNA-binding protein from whole animal lysates, cell-specific mRNA is<br />

co-immunoprecipitated. The abundance of each mRNA species isolated from these distinct cells is then<br />

assayed using microarrays.<br />

To identify genes expressed in muscle, we are using the well-characterized promoter myo-3 (Okkema et<br />

al., Genetics 135, 385-404) to drive the expression of the epitope-tagged mRNA-binding protein in body<br />

wall muscle. To date, we have successfully and reproducibly co-immunoprecipitated muscle-specific<br />

RNA, as assayed through both northern and microarray analysis. Control germ line and neuronal RNA<br />

are not enriched. We are currently optimizing our mRNA-tagging protocol to identify gene expressed in<br />

muscle, and are also extending our studies to identify neuronal and hypodermal genes.<br />

227


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

COOPERATION BETWEEN UNC-26/SYNAPTOJANIN AND<br />

THE DYNAMIN-RELATED PROTEIN DRP-1 DURING<br />

MITOCHONDRIAL DIVISION<br />

Dan Rube 1 , Todd Harris 2 , Erik Jorgensen 2 , Alexander van der Bliek 1<br />

1Department of Biological Chemistry, UCLA School of Medicine, P.O. Box 951737, Los Angeles, CA<br />

90095-1737<br />

2Department of Biology, University of Utah, 257 South 1400 East Salt Lake City, UT 84112-0840<br />

Mitochondria often exist as a dynamic tubular network. Individual mitochondria frequently divide or fuse<br />

with neighboring mitochondria in response to a variety of metabolic and cell division cues. Our laboratory<br />

is interested in the mechanism of mitochondrial division and how this mechanism is related to<br />

endocytosis. A first indication that these processes may be related came from our studies of<br />

dynamin-related protein (DRP-1) in the nematode C. <strong>elegans</strong>. We discovered that DRP-1 is important for<br />

the final stage of division of the mitochondrial outer membrane. This was surprising, because the protein<br />

sequence is very similar to that of dynamin, which we know mediates an early stage of clathrin mediated<br />

endocytosis. We hypothesize that division of the mitochondrial outer membrane is evolutionarily and<br />

mechanistically related to endocytic vesicle formation. The very first endosymbiontic bacterium would<br />

have retained parts of the endocytic machinery to divide the incipient mitochondrial membranes.<br />

To confirm this hypothesis, we are looking for additional proteins that act both in endocytosis and in<br />

division of the mitochondrial outer membrane. We are focusing on two well-known players in endocytic<br />

vesicle formation, synaptojanin and endophilin, which might also be candidates for mitochondrial division.<br />

We tested the effect of the synaptojanin mutant unc-26(e205) on mitochondrial morphology using a GFP<br />

with a mitochondrial leader sequence under control of the myo-3 promoter. The mitochondria in C.<br />

<strong>elegans</strong> body wall muscles appear as a few large clumps rather than as numerous regularly shaped<br />

wild-type mitochondrial bodies. These clumps may arise from impaired mitochondrial division.<br />

Furthermore, this phenotype appears to be partially rescued by overexpression of DRP-1. Our data thus<br />

indicates that in C. <strong>elegans</strong>, synaptojanin itself plays a role in maintaining proper mitochondrial<br />

morphology. Currently, we are seeking to determine whether the morphology defect of unc-26(e205)<br />

mitochondria arises from impaired mitochondrial division or disruption of another mitochondrial process.<br />

228


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

CALCIUM DYNAMICS OF FERTILIZATION IN C. ELEGANS<br />

Aravinthan D.T. Samuel 1 , Venkatesh N. Murthy 1 , Michael O.<br />

Hengartner 2<br />

1Department of Molecular and Cellular Biology, Harvard University, Cambridge, Massachusetts 02138.<br />

2Cold Spring Harbor Laboratories, Cold Spring Harbor, New York 11724.<br />

In all animals, fertilization generates a pattern of intracellular calcium dynamics within the oocyte that<br />

constitutes an essential trigger for normal development. The spatiotemporal properties of the calcium<br />

dynamics differ among animals, e.g. cnidarians, nemerteans, fish and frogs have single calcium<br />

transients whereas annelids, ascidians, and mammals have multiple calcium oscillations. However, in all<br />

animals, fertilization-induced calcium dynamics are mediated by release of internal calcium stores by<br />

inositol 1,4,5-triphosphate (IP 3). Although little is known about the signaling pathway intervening<br />

fertilization and the production of IP 3, features of the pathway are likely to be widely shared among<br />

species [1]. Of the animals typically used to study fertilization-induced calcium dynamics, none is as<br />

accessible to genetics and molecular biology as the model organism <strong>Caenorhabditis</strong> <strong>elegans</strong>. Motivated<br />

by the experimental possibilities inherent in using such a well-established model organism to study<br />

fertilization-induced calcium dynamics, we have characterized these dynamics in C. <strong>elegans</strong>. Owing to<br />

the transparency of the nematode, we have been able to study the calcium signal in C. <strong>elegans</strong><br />

fertilization in vivo by monitoring the fluorescence of calcium indicator dyes that we introduce into the<br />

cytosol of oocytes. In C. <strong>elegans</strong>, fertilization induces a single calcium transient that originates at the point<br />

of sperm entry. This calcium elevation immediately spreads throughout the oocyte with an amplitude ~250<br />

nM. The duration of this solitary calcium transient is ~6 min, after which the cytosolic calcium<br />

concentration returns to that preceding fertilization. Among other effects, this calcium signal may trigger<br />

the completion of meiosis and the formation of eggshell.<br />

229


MUTANTS IN THERMOSENSORY NEURON SPECIFICATION<br />

AND FUNCTION<br />

John S. Satterlee, Piali Sengupta<br />

<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

Department of Biology, Brandeis University, 415 South St. Waltham, MA 02454<br />

The AFD neurons are required for thermosensation in C. <strong>elegans</strong>. This pair of ciliated sensory neurons<br />

has a unique morphology and presumably expresses a specific suite of genes which confer<br />

thermosensory function. For example, the promoters of both a nuclear hormone receptor homolog<br />

(nhr-38) and a receptor guanylyl cyclase homolog (gcy-8) drive GFP expression specifically in this neural<br />

pair. We are examining the mechanism by which the fate and functions of the AFD thermosensory<br />

neurons are specified, by identifying genes that work in a cascade to direct expression of these<br />

AFD-specific markers.<br />

Two mutants with reduced expression of nhr-38::GFP in the AFD neurons were found to be allelic to tax-2<br />

and tax-4. The tax-2/tax-4 genes encode subunits of a cGMP-gated channel which function in some<br />

sensory neurons, including AFD. It is possible that the TAX-2/TAX-4 channel is required for<br />

activity-dependent expression of some AFD specific genes. We have made promoter::GFP fusions to<br />

other tax-4-like channels encoded by the worm genome and found that two of these genes are also<br />

expressed in AFD, as well as a number of other sensory neurons.<br />

We have identified mutations in two genes (2 alleles each) with reduced expression of gcy-8::GFP in the<br />

AFD neurons. One of these (sns-6) has been mapped to a small region of LG IV. The other (sns-7) is<br />

allelic to the previously identified thermotaxis mutant ttx-1. Single nucleotide polymorphisms have been<br />

used to map ttx-1/sns-7 to a 130 kb region of LG V which encodes 9 predicted protein products.<br />

Both sns-6 and ttx-1/sns-7 are thermotaxis defective, but are not defective in behaviors mediated by the<br />

AWA, AWC, ASE, and ASH neurons. Mutations in both sns-6 and ttx-1/sns-7 suppress the<br />

dauer-constitutive phenotype of daf-7 mutants at 25ûC.<br />

We have isolated a mutant with a deletion of the gcy-8 guanylyl cyclase domain. This mutation has no<br />

effect on gcy-8::GFP or nhr-38::GFP expression in AFD. Further characterization of this mutant is in<br />

progress.<br />

The molecular characterization of thermotaxis mutants should reveal how AFD function is specified and<br />

may also shed light on how thermosensory signals are transduced.<br />

230


VESICULAR GABA TRANSPORT IN C. ELEGANS REQUIRES<br />

TWO PROTEINS UNC-47 AND UNC-46<br />

Kim Schuske, Erik M. Jorgensen<br />

<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

Department of Biology, University of Utah, Salt Lake City, UT 84112<br />

For neurotransmitter to be released from a neuron, it must be transported into synaptic vesicles from the<br />

cytoplasm where it is produced. Transport proteins are located in the vesicle membrane that bind and<br />

load a specific neurotransmitter into the synaptic vesicle. Previously we cloned the gene unc-47, which is<br />

defective in all GABA neurotransmission, and found that it encodes a transport protein that loads GABA<br />

into synaptic vesicles. We now report that there is a second gene that is phenotypically defective in all<br />

GABA neurotransmission, unc-46, which also appears to be required for loading of GABA into synaptic<br />

vesicles. Overexpression of UNC-47 is capable of rescuing the unc-46 mutant phenotype. This suggests<br />

that an increased level of vesicular GABA transporter (UNC-47) in the neuron can compensate for the<br />

lack of UNC-46 protein. Therefore we believe that UNC-46 may be a modulator of vesicular GABA<br />

transport.<br />

To test this, we have cloned the unc-46 gene. The UNC-46 protein is novel and contains a single putative<br />

transmembrane domain. unc-46 is expressed in GABA neurons and the protein is localized to synaptic<br />

vesicles. UNC-47 protein is also localized to synaptic vesicles even in the absence of unc-46. However,<br />

UNC-46 protein is mislocalized in the absence of unc-47. This suggests that UNC-47 and UNC-46<br />

interact in the synaptic vesicle. The simplest model suggests that UNC-46 may be required for UNC-47<br />

transport activity. Current experiments are aimed at determining whether UNC-46 and UNC-47 physically<br />

interact, testing if UNC-46 can affect transport activity of GABA in cell culture, and identifiying a vertebrate<br />

homolog of UNC-46.<br />

231


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

UTILIZING TWO APPROACHES, GENETIC AND GENOMIC,<br />

TO IDENTIFY THE VESICULAR GLUTAMATE TRANSPORTER<br />

Kim Schuske, Dan Williams, Erik M. Jorgensen<br />

Department of Biology, University of Utah, Salt Lake City, UT 84112<br />

Neurotransmitter is loaded into the synaptic vesicles by transport proteins present in the synaptic vesicle<br />

membrane. Transport activity has been characterized for four distinct neurotransmitters in purified<br />

synaptic vesicles from rat brain: GABA, glutamate, acetylcholine, and catecholamines. Of these, only the<br />

vesicular glutamate transporter remains to be identified at the molecular level. We previously showed that<br />

the unc-47 gene encodes the vesicular GABA transporter. Because GABA and glutamate are structurally<br />

similar and their bioenergetics for transport into the vesicle are also similar, we proposed that the<br />

vesicular glutamate transporter is related by sequence to UNC-47. There are fourteen proteins in the C.<br />

<strong>elegans</strong> genome that have sequence similarity to UNC-47. We made GFP reporter constructs for twelve<br />

of these genes, and identified two candidates that are expressed in glutamatergic neurons. We are<br />

currently studying a gene knockout, made by the C. <strong>elegans</strong>gene knockout consortium, and GFP-tagged<br />

protein localization for one candidate cevt6. However, preliminary data suggests that cevt6 is not the<br />

vesicular glutamate transporter.<br />

It is possible that the vesicular glutamate transporter does not resemble the vesicular GABA transporter<br />

by sequence. For this reason we have undertaken a genetic screen for mutants that are likely to be<br />

defective for glutamate neurotransmission. We are therefore looking for mutants resembling eat-4 since<br />

they are defective for all known glutamate neurotransmission. These animals have an Osm phenotype.<br />

We have screened for Osm mutants using a Mos1 transposon (from Drosophila mauritiana) with the goal<br />

of identifying a mutant with a Mos1 insert in a transporter-like protein. We carried out a pilot screen and<br />

identified three Osm mutants. One of these mutants contains a single transposon inserted into exon ten<br />

of the eat-4 gene. The two other mutants are dye filling defective and are therefore not likely to encode<br />

the vesicular glutamate transporter. An increase in the efficiency of Mos1 transposition should allow us to<br />

do a saturation Osm screen in the future.<br />

232


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

ACTIN-DEPENDENT PROCESSES IN THE EARLY C.<br />

ELEGANS EMBRYO REQUIRE THE PROFILIN GENE PFN-1,<br />

THE FH GENE CYK-1, AND BEL-1<br />

Aaron F. Severson 1 , Rebecca Lyczak 1 , David L. Baillie 2 , Bruce<br />

Bowerman 1<br />

1University of Oregon, Eugene, OR 97403 USA<br />

2Simon Fraser University, Burnaby, B.C. CANADA V5A 1S6<br />

The actin cytoskeleton is essential for many processes in the early C. <strong>elegans</strong> embryo, including ruffling<br />

of the anterior cortex and pseudocleavage following fertilization, the asymmetric migration of pronuclei<br />

and position of the first mitotic spindle, and cytokinesis. A worm profilin gene, called pfn-1, is required for<br />

all of these processes. In pfn-1(RNAi) embryos, cortical ruffling and pseudocleavage are absent, the<br />

pronuclei meet in the center of the embryo, the first mitotic spindle is centrally positioned, and a cleavage<br />

furrow fails to form during cytokinesis. Profilin regulates the actin cytoskeleton by sequestering actin<br />

monomers and preventing their polymerization into microfilaments. In addition, profilin can stimulate actin<br />

assembly by converting actin into an assembly-competent GTP-bound state. In other systems, profilin can<br />

bind to FH proteins, which may recruit profilin/actin complexes to sites of actin assembly. The C. <strong>elegans</strong><br />

FH protein CYK-1 is required for embryonic cytokinesis (Swan et al, 1998), and embryos produced by<br />

worms mutant for a strong combination of cyk-1 alleles fail early in furrow ingression. We are currently<br />

characterizing anti-PFN-1 antisera to examine PFN-1 localization in wild-type and CYK-1 mutant<br />

embryos. In addition, we are examining actin, myosin, and CYK-1 localization in pfn-1(RNAi) embryos to<br />

determine the effects of profilin inactivation on the actin cytoskeleton.<br />

While PFN-1 may be required for organized contraction of the actin cytoskeleton, a second gene may<br />

function to temporally and spatially restrict contractility in the embryo. In embryos mutant for a<br />

temperature-sensitive, embryonic lethal allele of this gene, waves of contraction sweep across the early<br />

embryo, resembling mobile cleavage furrows. We have named this gene bellydancer, or bel-1. To<br />

determine if the mechanism that underlies these waves is similar to the contractile ring, we are staining<br />

bel-1 mutant embryos with antibodies to actin, myosin, and CYK-1. In addition, we are mapping bel-1,<br />

which we hope to clone by a combination of RNAi phenocopy and transgenic rescue.<br />

233


LIN-12 POST-TRANSCRIPTIONAL DOWNREGULATION<br />

DURING VPC SPECIFICATION<br />

DD Shaye 1 , I Greenwald 2<br />

<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

1Dept. of Genetics and Development, Columbia University, New York, NY 10032<br />

2HHMI and Dept. of Biochemistry, Columbia University, New York, NY 10032<br />

In wild-type hermaphrodites, each vulval precursor cell (VPC) adopts one of three distinct fates, 1°, 2° or<br />

3°, in a precise and invariant spatial pattern (reviewed in 1). Analysis of mutants defective in vulval<br />

development have suggested that at least three different signaling events are involved in the correct<br />

specification of VPC fates. An inhibitory signal originates in the hypodermal syncytium. An inductive signal<br />

originates in the gonadal anchor cell and allows VPCs to overcome the inhibitory signal from the<br />

hypodermis. Finally, a lateral signal originates in the presumptive 1° cell and is received by the<br />

presumptive 2° cells through LIN-12.<br />

Studies using a lin-12::gfp fusion have shown that one output of the inductive signal is to influence LIN-12<br />

protein accumulation (2). In wild-type hermaphrodites, all six VPCs initially express LIN-12::GFP.<br />

However, LIN-12::GFP accumulation is reduced specifically in P6.p at the time of vulval induction,<br />

although expression of a lin-12::lacZ reporter remains constant (3). The inductive signal seems to be<br />

necessary and sufficient for this downregulation to occur. These results have led to the proposal that the<br />

downregulation of LIN-12 in the presumptive 1° cell is at least one component of the mechanism by<br />

which the inductive signal influences lateral signaling.<br />

We are interested in elucidating the mechanism by which the inductive signal controls the downregulation<br />

of LIN-12 in P6.p and whether this downregulation plays a role in correctly generating a lateral signal. In<br />

order to answer these questions we are taking two approaches. First, we are examining the effects of<br />

mutations in the inductive pathway and in other genes on LIN-12::GFP protein accumulation. Second, we<br />

are determining the region of LIN-12 that targets LIN-12::GFP for downregulation in P6.p.<br />

1) I. Greenwald, in C. <strong>elegans</strong> II. CSH Press, Cold Spring Harbor, NY, (1997).<br />

2) D. Levitan and I. Greenwald, Development 125, 3101 (1998).<br />

3) H. Wilkinson and I. Greenwald, Genetics 141, 513 (1995).<br />

234


DISTINT AND REDUNDANT FUNCTIONS OF MU1 MEDIUM<br />

CHAINS OF AP-1 CLATHRIN-ASSOCIATED PROTEIN<br />

COMPLEX IN THE NEMATODE CAENORHABDITIS ELEGANS<br />

Jaegal Shim, Junho Lee<br />

<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

Department of Biology, Yonsei University, Seoul, Korea<br />

The clathrin-associated protein complex I(AP-1 complex) in C. <strong>elegans</strong> is composed of four adaptor<br />

proteins. Of the medium chains in the AP-1 complex of C. <strong>elegans</strong>, the first cloned is unc-101 and the<br />

second chain cloned is apm-1. Also, according to a search of the C. <strong>elegans</strong> genome, unlike the medium<br />

chains, only one each of the beta, gamma and sigma chains of the AP-1 complex exists in C. <strong>elegans</strong>.<br />

These facts support the possibility that the two medium chains in C. <strong>elegans</strong> compose distinct AP-1<br />

complex while sharing the other chains, similar to mammalian mu1a and mu1b. From the RNAi and<br />

expression studies of AP-1 complex in C. <strong>elegans</strong>, we concluded that this hypothesis was correct. Since<br />

the expression of unc-101 and apm-1 is ubiquitous throughout development, and the functions of unc-101<br />

and apm-1 are redundant in embryogenesis and vulval development while becoming distinct during larval<br />

development, we are interested in the distinct and similar functions of these chains as adaptor proteins.<br />

Mediums of the AP complexes are generally known to play a part in cargo selection of clathrin-coated<br />

vesicles, thus we have started experiments to elucidate their respective cargoes using the yeast<br />

two-hybrid system. We screened about 400,000 colonies with full-length apm-1 cDNA as a bait, and<br />

found several positive clones. One of them, F29G6.3A, was the same clone that Marc Vidal group had<br />

found with unc-101 as a bait(Albertha J.M Walhout et al., Science ?????). We have also found several<br />

novel proteins, and will present analyses on the candidate genes<br />

235


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

MOLECULAR MECHANISMS OF DAF-12 ACTION:<br />

IDENTIFICATIONOF RESPONSE ELEMENTS AND<br />

FUNCTIONAL ANALYSIS OF THE PROTEIN<br />

Yuriy Shostak 1 , Adam Antebi 2 , Marc R. van Gilst 3 , Kieth R.<br />

Yamamoto 1<br />

1 Program in Biochemistry and Molecular Biology and Department of Cellular and Molecular<br />

Pharmacology, UCSF, San Francisco, CA 94143-0450<br />

2 Max-Planck-Institut fuer molekulare Genetik,D-14195 Berlin, Germany<br />

3 Department of Cellular and Molecular Pharmacology, UCSF, San Francisco, CA 94143-0450<br />

Dauer larva formation in <strong>Caenorhabditis</strong> <strong>elegans</strong> is regulated by the intracellular receptor daf-12, which<br />

functions at the convergence of TGF-b, cGMP, and insulin-like signaling pathways. daf-12 has also been<br />

implicated in the regulation of C. <strong>elegans</strong> lifespan and developmental age. Alleles of daf-12 with distinct<br />

protein sequence alteration spartially uncouple its phenotypic effects.<br />

As a step toward understanding the molecular biology of Daf-12 function, we identified specific DNA sites<br />

in the C. <strong>elegans</strong> genome bound by the protein in vitro. We developed an in vitro selection and<br />

amplification method that, using immobilized recombinant daf-12 DNA binding domain, yielded a series of<br />

specific C. <strong>elegans</strong> genomic DNA fragments. We inserted some of these fragments into yeast reporter<br />

plasmids and showed that Daf-12 selectively activated transcription of the reporter genes in<br />

Saccharomyces cerevisiae. Hence, C. <strong>elegans</strong> DNA fragments bound specifically by Daf-12 in vitro<br />

display Daf-12 response element activity in vivo.<br />

Intracellular receptors are multidomain proteins with characteristic DNA binding, putative ligand binding,<br />

and potentially ligand-controlled transcriptional regulatory domains. We used the heterologous yeast<br />

expression system to investigate contributions of different Daf-12 regions to transcriptional regulation. We<br />

found thatDaf-12 derivatives with truncated ligand binding domains are potent transcriptional activators,<br />

and mapped an activation domain to the "hinge region" just downstream of the DNA binding domain.<br />

In future work, we shall determine whether the identified response elements are linked to Daf-12regulated<br />

target genes in C. <strong>elegans</strong>. Similarly, we shall examine the roles of the activation domain in Daf-12<br />

biology and molecular function in the animal.<br />

236


SEROTONIN-RESISTANT EGG-LAYING MUTANTS AND A<br />

RECEPTOR KNOCKOUT IN PROGRESS<br />

Stanley Shyn, William Schafer<br />

UCSD, La Jolla, CA 92093<br />

Serotonin (5HT) stimulates egg-laying in the nematode 1 , and we hypothesize that this occurs by<br />

modulation of vulval muscle excitability from a quiescent mode to an active egg-laying state 2 . The<br />

effectors used to accomplish this are largely unknown, so we are mapping and characterizing catalogued<br />

as well as more recently isolated mutants (5 lines from our own lab) which are 5HT-resistant for<br />

egg-laying. One such mutant is egl-24 (n572) 3 . Further characterization has confirmed its profound<br />

5HT-resistance, but perhaps more intriguingly that, in the absence of any exogenous 5HT, its temporal<br />

pattern of egg-laying is not grossly different from that of N2s. Preliminary data suggests its map interval<br />

may be further narrowed to between -0.54 and -0.79 on LGIII, an interval spanned by 26 cosmids and a<br />

portions of 2 YACs.<br />

From a reverse-genetics approach, we are also preparing to knockout 5HT-Ce, the only known vulval<br />

5HT receptor 4 . Among vertebrate classes, 5HT-Ce shows the most homology to the 5HT 1 subtype,<br />

which typically inhibits adenylate cyclase 5 . This prompted us to look at worms mutant in acy-1, an<br />

adenylate cyclase expressed in virtually all neurons and muscles, including the vulva 6 . nu329 and<br />

pk450::Tc1, both only partial loss-of-function, demonstrated 5HT-resistance despite a baseline with<br />

wildtype parameters. Whether cAMP stimulates or inhibits egg-laying is still unclear, but pharmacological<br />

manipulations are planned to clarify the issue.<br />

Finally, assuming 5HT-Ce inhibits adenylate cyclase, downstream G-protein candidates might include<br />

goa-1 or gpa-7. Yet, functional goa-1 appears to play an inhibitory role in egg-laying 7,8 while testing in our<br />

lab of gpa-7 worms did not uncover significant 5HT-resistance. Recently, the Plasterk lab surveyed the<br />

complete family of genes encoding worm G-proteins 9 . One of these genes, gpa-14, is expressed in the<br />

vulva and appears to be a novel G-protein whose downstream effectors are unknown. gpa-14 (pk347)<br />

mutants were subsequently tested and found to be 5HT-resistant, suggesting that gpa-14 might be part of<br />

a 5HT signalling pathway.<br />

1 Science 216:1012<br />

2 Neuron 21:203<br />

3 Genetics 104:619<br />

4 Tim Niacaris (Avery lab), pers comm<br />

5 J Mol Neurosci 8:53<br />

6 J Neurosci 18:2871<br />

7 Science 267:1652<br />

8 Science 267:1648<br />

9 Nat Genetics 21:414<br />

<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

237


A NOVEL GENETIC SCREEN FOR SYNAPTIC<br />

TRANSMISSION GENES ACTING IN THE DIACYGLYCEROL<br />

PATHWAY<br />

Derek S. Sieburth, Wendy Cham, Josh M. Kaplan<br />

UC Berkeley, MCB Dept , 361 LSA, Berkeley CA 94720<br />

Acetylcholine release at the neuromuscular junction is modulated by diacylglycerol (DAG) levels which<br />

are regulated by the opposing effects of two G protein pathways 1-4 . The Gqa pathway produces DAG<br />

through the activation of the egl-8 phospholipase c (PLC) 1,4 , and the Gao pathway reduces DAG levels<br />

possibly through diacylglycerol kinase, encoded by dgk-1 2 . DAG appears to modulate synaptic<br />

acetylcholine release by recruiting UNC-13 to the synapse where it facilitates vesicle release 1,2 .<br />

Mutations in genes acting in these two pathways have opposite effects on acetylcholine release as<br />

assayed by the response to the paralytic effects of the acetylcholine esterase inhibitor, aldicarb 1,4 .<br />

Mutations in the Gqa/PLC pathway result in decreased levels of DAG and resistance to Aldicarb (Ric),<br />

while mutations in Goa/DGK-1 result in increased levels of DAG and hypersensitivity to aldicarb (Hic).<br />

To identify additional components specifically acting the DAG pathway, we have screened for<br />

suppressors of the Hic phenotype of dgk-1 null mutants. Suppressor mutations are likely to define<br />

synaptic transmission genes involved in either DAG production or, like UNC-13 in the response to DAG,<br />

or alternatively in genes acting in a parallel pathway that also modulates acetylcholine release. We have<br />

identified 26 suppressors which each suppress the Hic phenotype of dgk-1 mutants to wild type or to Ric.<br />

22 of these have wild type response to the acetylcholine receptor agonist, levamisole, indicating that they<br />

define genes that are likely acting in the motor neurons. These 22 suppressors define at least eight<br />

genes, including unc-13, an expected target of this screen. Seven suppressors are resistant or partially<br />

resistant to the effects of an exogenously added DAG analogue, PMA, suggesting that they define DAG<br />

targets. Several suppressors have weak or no obvious locomotion or Ric phenotypes in a dgk-1(+)<br />

background, suggesting that they may have regulatory roles in vesicle release. At least two of the<br />

suppressors appear to define new genes not previously implicated in acetylcholine release. Progress on<br />

suppressor characterization will be reported.<br />

1 Lackner et al., Neuron 24: 335-346 1999<br />

2 Nurrish et al., Neuron 24: 231-242 1999<br />

3 Hajdu-Cronin et al., Genes Dev 13: 1780-93 1999.<br />

4 Miller et al., Neuron 24: 323-33 1999.<br />

<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

238


EVIDENCE OF A MATE-FINDING CUE IN THE FREE-LIVING<br />

SOIL NEMATODE C. ELEGANS<br />

Jasper M. Simon, Paul W. Sternberg<br />

HHMI and Caltech, Division of Biology<br />

<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

Use of mate-finding cues is well documented throughout the nematode literature, but no assays for C.<br />

<strong>elegans</strong> have come into common practice. Here we report two assays that suggest young adult males<br />

can detect young adult hermaphrodites through some unidentified cue. Accumulation of 14 males (per<br />

trial) to agar conditioned with 5 hermaphrodites (the muscle mutant unc-52 was used to construct a point<br />

source), which were sequentially removed, suggests a cue given off by hermaphrodites and detected by<br />

males [mean number of males observed in 1-cm diameter scoring circles shifted from 3.2+2.2 in control<br />

trials (n=29) to 6.5+2.5 in conditioned-agar trials (n=28); p


UNDERSTANDING C27H5.1: FROM SEQUENCE TO SENSE<br />

Jessica Smith, David Pilgrim<br />

<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

CW 405 Biological Sciences Bldg., University of Alberta, Edmonton, AB, T6G 2E9<br />

One of the goals of our lab is to determine the function of UNC-119, a protein involved in axon guidance<br />

and outgrowth. In the course of this study we have chosen to look at C27H5.1, a weak homologue of<br />

UNC-119 predicted by the C. <strong>elegans</strong> Genome Sequencing Project. Like UNC-119, C27H5.1 appears to<br />

be expressed early in the developing embryo and pan-neuronally in all subsequent stages of<br />

development, suggesting that it too plays a role in neural development. Unlike UNC-119, C27H5.1 shows<br />

additional expression in pharyngeal and vulval muscles suggesting other roles for this protein.<br />

Unfortunately there are no known mutations in C27H5.1. We are attempting to determine the phenotype<br />

of a C27H5.1 knockout through both deletion screening and in vivo RNA interference.<br />

While C27H5.1 is weakly similar to UNC-119, it is highly conserved (70% identity) with the delta subunit of<br />

mammalian rod phosphodiesterase (PDE6d). This protein was first identified in the retina as a subunit of<br />

rod phosphodiesterase (PDE). However, it has since been shown to be ubiquitously expressed and to<br />

interact with a number of other proteins including Rab13, RPGR, and Arl2. These interactions have not<br />

been fully characterized, but two general observations can be made. PDE6d solubilizes some proteins<br />

and affects the cGMP or GTP binding properties of others. These data suggest that PDE6d plays a<br />

regulatory role through interactions with other proteins. Interestingly, C27H5.1 interacts with and<br />

solubilizes PDE in the same manner as PDE6d suggesting functional conservation between these two<br />

proteins.<br />

To determine the role of the C27H5.1 protein in C. <strong>elegans</strong>, we are in the process of performing a yeast<br />

2-hybrid screen of a C. <strong>elegans</strong> cDNA library using C27H5.1 as bait. We are especially curious to see if<br />

C27H5.1 interacts with proteins similar to those which interact with PDEd in mammals. As well, we are<br />

producing antibodies against C27H5.1 which will allow us to further characterize the function and<br />

expression of this protein.<br />

240


GENETIC SCREENS FOR NOVEL COMPONENTS INVOLVED<br />

IN BLASTOMERE ASYMMETRY IN THE EARLY C. ELEGANS<br />

EMBRYO<br />

Martha Soto, Craig C. Mello<br />

<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

University of Massachusetts Medical Center, Worcester, MA 01605, USA<br />

In the early C. <strong>elegans</strong> embryo asymmetric cell divisions result from the partitioning of factors and from<br />

signaling between cells. At the four cell stage the EMS blastomere divides asymmetrically, leading to two<br />

daughters, E and MS, with distinct cell fates. The E blastomere is the only source of gut in the embryo,<br />

while MS makes pharynx and other tissues. This polarization requires a signal from the P2 blastomere to<br />

EMS. In addition, rotation of the spindle of EMS results in only the E daughter contacting P2. Mutations<br />

that disrupt this signal include homologs of WNT signaling factors. We have conducted forward genetic<br />

screens to identify new factors that control the asymmetric divisions of EMS. To date, our screens have<br />

identified many alleles of known WNT components, as well as a few novel genes.<br />

We will discuss a novel mutation involved in regulating cleavage orientation. Several components<br />

involved in spindle orientation have been identified by forward genetics in C. <strong>elegans</strong>, including the Par<br />

genes and let-99, and by reverse genetics, including the small GTPase CDC42 as well as G proteins. In<br />

our mutant, ne236, EMS divides left/right rather than anterior/posterior resulting in embryos in which both<br />

E and MS are in contact with P2. This phenotype is more severe than the slight disruptions in spindle<br />

rotation seen in other WNT mutations. ne236 embryos also have cell fate transformations, including<br />

frequent ectopic gut and pharynx. Interestingly, ne236 interacts with components of WNT signaling,<br />

enhancing partially penetrant gutless mutations. We are attempting to clone ne236, and testing models to<br />

understand how cleavage orientation is regulated in asymmetric cell divisions.<br />

241


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

PAX BE WITH YOU - PATTERNING THE PHARYNX<br />

Jeff Stevenson 1 , Andrew Chisholm 2 , Susan E. Mango 1<br />

1Dept. of Oncological Sciences, Huntsman Cancer Institute Center for Children, University of Utah, Salt<br />

Lake City, UT, 84112<br />

2Dept. of Biology, UC Santa Cruz, Santa Cruz, CA, 95064<br />

Formation of the pharynx consists of four phases: i) specification of pharyngeal precursors during early<br />

embryogenesis, ii) assembly of these precursors into a pharynx primordium, iii) terminal differentiation of<br />

the five pharyngeal cell types, and iv) morphogenesis of the pharynx into its adult form. Establishment of<br />

the pharyngeal precursors requires the pha-4 gene, which encodes a transcription factor expressed in all<br />

cells of the pharynx. In a pha-4 mutant background, no or few pharyngeal cells are generated.<br />

How are five different pharyngeal cell types produced from an apparently homogeneous population of<br />

precursor cells? And what is the role of pha-4 in this process, given its pan-pharyngeal expression<br />

pattern? To address these questions we are studying how one pharyngeal cell type, the marginal cell, is<br />

established during development. We have identified an early marker of marginal cells, PAX-9, a homolog<br />

of the paired transcription factor. C. <strong>elegans</strong> pax-9::GFP is expressed in all nine marginal cells, as well as<br />

three other pharyngeal cells. The involvement of transcription factors in most cell fate decisions suggests<br />

that control of marginal cell specification lies at the transcriptional level. Hence, an understanding of pax-9<br />

promoter elements may reveal the pathways that govern specification of distinct cell types within the<br />

pharynx.<br />

Deletion analysis of the pax-9 promoter shows that proper spatial and temporal expression of a<br />

pax-9::GFP reporter requires only 125 bp of upstream sequence. Within this region, we have identified a<br />

consensus PHA-4 binding site that alone is able to direct expression of a GFP reporter in nearly all cells<br />

of the developing pharynx. Significantly, evidence suggests that PHA-4 binds the pax-9 promoter in vivo,<br />

and that binding is not limited to marginal cells.<br />

Our working model postulates that the binding of PHA-4 to this promoter endows organ (pharyngeal)<br />

identity to pax-9 expression, while the binding of a second, as yet unidentified factor expressed only in<br />

marginal cells, imparts cell type identity. We are presently scanning this region for cis-elements required<br />

for the marginal cell specific expression of our reporter construct.<br />

242


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

THE EVOLUTION AND EXPRESSION OF FEM-2<br />

Paul Stothard 1 , Dave Hansen 2 , Tamara Checkland 1 , Dave Pilgrim 1<br />

1Department of Biological Sciences, University of Alberta, Edmonton, AB T6G 2E9<br />

2Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110<br />

FEM-2 is a protein phosphatase type 2C (PP2C) that regulates sex determination. We have recently<br />

focused on three aspects of FEM-2 biology. First, why do sex determining proteins in general have an<br />

accelerated rate of evolution? Second, how do mutations in other sex determining proteins affect the<br />

expression pattern of FEM-2? Third, what do the mutant alleles tell us about the function of the protein?<br />

To assess whether different selective pressures act on sex determining proteins we characterized the<br />

orthologs of C. <strong>elegans</strong> PP2C genes from the related nematode C. remanei. Comparison of the C.<br />

remanei sequences with their C. <strong>elegans</strong> orthologs indicates that FEM-2’s PP2C domain is much more<br />

divergent than the same domain in the other proteins. PP2C sequences with no known sex determining<br />

role were also isolated from the zebrafish, Danio rerio, and compared with their previously identified<br />

mouse orthologs. The zebrafish/mouse PP2C domains are more conserved than FEM-2’s PP2C domain,<br />

but less conserved than the other C. <strong>elegans</strong>/C. remanei PP2Cs. Experiments using transgenes suggest<br />

that C. remanei and C. briggsae fem-2 are able to promote male somatic development but not sperm<br />

production in C. <strong>elegans</strong>. To better understand how fem-2 regulates sexual development we have raised<br />

antisera against the FEM-2 protein. The antisera can specifically detect FEM-2 on <strong>West</strong>ern blots, and it<br />

stains sexually dimorphic structures when used on whole animals. We are currently looking for<br />

differences in staining between wild-type animals and animals that carry mutations in other sex<br />

determining genes. Finally, studies of two temperature sensitive missense alleles (b245 and q117) are<br />

underway. The b245 mutation is located in the PP2C domain while the q117 mutation is in the amino<br />

terminus. These two alleles are being characterized in vivo as well as through in vitro protein studies.<br />

243


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

FORMING A GUT: THE VIEW FROM AN ELT AND TWO ODDS<br />

Keith Strohmaier 1 , Morris Maduro 2 , Joel Rothman 2<br />

1Biochemistry and Molecular Biology Program, University of California, Santa Barbara, CA<br />

2MCDB Department, University of California, Santa Barbara, CA<br />

In an effort to understand the genetic events that lead to the formation of the intestine, we are<br />

investigating the network of regulatory factors expressed during gut development. Here, we describe our<br />

studies of three such factors: the GATA-type transcription factor, ELT-7, and two ODD-SKIPPED-like<br />

factors, ODD-1 (formerly EEL-1) and ODD-2.<br />

elt-7::gfp is expressed continuously in the E lineage beginning at the 2E cell stage. This expression<br />

pattern is identical to that of ELT-2, suggesting that these GATA factors may perform overlapping roles in<br />

gut development. Thus, ELT-2 and -7 might act redundantly and immediately downstream of the END-1<br />

and END-3 GATA factors, which redundantly specify E cell identity. However, elt-7(RNAi) embryos show<br />

no discernible phenotype, and the degraded gut phenotype of the elt-2(0) mutation does not appear to be<br />

enhanced by RNAi of elt-7. This contrasts with end-1/end-3 double RNAi, which results in a penetrant<br />

loss of gut not seen with each RNAi alone. Ectopic elt-7 expressed from a heat-shock promoter results in<br />

gut differentiation throughout the embryo, showing that ELT-7 is able to activate gut development.<br />

Ian Hope’s expression screen identified odd-1 as a gut-specific gene. odd-2 was discovered through its<br />

similarity to odd-1. Each encodes a 3 zinc finger protein similar to Drosophila ODD-SKIPPED family<br />

members. odd-1::gfp is expressed in two phases. The first phase corresponds to E lineage expression<br />

from the 4E cell stage through mid-elongation (about 1.5-fold). In the second phase, expression is<br />

continuous throughout development after elongation in intestinal-rectal valve cells and in a decreasing<br />

posterior to anterior gradient in the gut, with increased expression again in the four anterior (int1) cells.<br />

Expression of odd-2::lacZ::gfp is similar to the second phase of odd-1::gfp expression, albeit at lower<br />

levels and with somewhat altered kinetics. Both odd-1(0) and odd-2(RNAi) animals die as L1 larva, with<br />

no obvious effect on gut development.<br />

We are further characterizing the function of these genes to understand their action and regulatory<br />

interrelationships during development of the intestine.<br />

244


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

C. ELEGANS HOMOLOGUE OF PROTEIN PHOSPHATASE 4<br />

IS REQUIRED IN SPERMATOGENESIS<br />

Eisuke Sumiyoshi, Asako Sugimoto, Masayuki Yamamoto<br />

Dept. Biophys. Biochem., Grad. Schl. Sci., Univ. Tokyo, Japan<br />

Protein phosphatase 4 (PP4) is a ser/thr protein phosphatase, which is known to localize to centrosomes.<br />

Analysis using a fly mutant implicated that PP4 is required for the nucleation of microtubules during<br />

mitosis. However, its function in other organisms is not yet understood. In C.<strong>elegans</strong>, two possible<br />

homologues of PP4, namely Y75B8A.30 and Y49E10.3, have been identified by the genome project.<br />

Here we report characterization of Y75B8A.30.<br />

To understand the function of Y75B8A.30, we performed RNAi for it. Although the penetrance varied<br />

among injections, RNAi for Y75B8A.30 at the highest efficiency caused 25% embryonic lethality at F1 and<br />

nearly 100% embryonic lethality at F2.<br />

Y75B8A.30 (RNAi) F2 embryos showed a multi polar spindle with two male pronuclei at 1-cell stage. We<br />

suspected that an extra male pronucleus might result from a defect in spermatogenesis. To confirm this,<br />

we examined the spermatogenesis of the F1 progeny. Observation with Nomarski optics and with DAPI<br />

staining revealed that F1 spermatids often contained multiple nuclei. We also found that the nuclei of<br />

secondary spermatocytes of F1 worms were not properly separated, indicating that Y75B8A.30 is<br />

required for proper chromosome segregation in sperm meiosis. To see whether the multi polar spindle is<br />

a result of the defect in sperm, F1 males were crossed to rde-1 hermaphrodites, which is resistant to<br />

RNAi. 5/16 of cross progeny showed multi polar spindles at 1-cell stage, indicating that multi polar<br />

spindles are indeed caused by the defect of sperm at least in part. Thus, we conclude that PP4 is<br />

required for the segregation of chromosomes and centrosomes during spermatogenesis.<br />

To see whether Y75B8A.30 function is required in embryogenesis, we crossed Y75B8A.30 (RNAi) F1<br />

hermaphrodites with wild type male. Although wild type sperm were supplied in this cross, many cross<br />

progenies died during embryogenesis, showing that Y75B8A.30 is required for embryogenesis. To know<br />

the possible role of Y75B8A.30 in mitosis, we stained F2 embryos with anti-tubulin antibody and DAPI.<br />

Some fraction of 1-cell embryos had condensed chromosomes at the metaphase plate, with microtubules<br />

mislocalized around the nucleus, suggesting that Y75B8A.30 may be involved in the organization of<br />

microtubules during mitosis.<br />

245


TRANSCRIPTIONAL REGULATION OF THE TRYPTOPHAN<br />

HYDROXYLASE GENE TPH-1<br />

Ji Ying Sze<br />

<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

Dept. of Anatomy and Neurobiology, Univ. of California, Irvine CA 92697<br />

tph-1 encodes a tryptophan hydroxylase, the key enzyme for serotonin biosynthesis. When I was in Gary<br />

Ruvkun’s lab, I collaborated with Martin Victor in Yang Shi’s lab and isolated a tph-1 deletion mutation,<br />

tph-1(mg280). tph-1(mg280) shows no detectable serotonin based on anti-serotonin antibody staining<br />

and exhibits several behavioral and metabolic defects (Sze et al., 2000). It has been proposed that in<br />

mammals changes in the level of TPH mRNA correlate with long-term changes in the cellular serotonin<br />

level and that the regulation of TPH mRNA tends to be "inducer"- and region-specific (Semple-Rowland et<br />

al., 1996; Clark and Russo, 1997; Siuciak et al., 1998. Bethea, et al., 2000). Thus, the regulation of TPH<br />

expression may be a key step by which the nervous system adjusts its long-term synaptic serotonin<br />

levels.<br />

To identify genes regulating tph-1, I have started a genetic screen for mutations that alter tph-1::gfp<br />

expression in specific neurons. Thus far, mutations identified can be classified into three classes. (1) I<br />

have previously reported that mutations in the POU-homeodomain transcription factor UNC-86 abolish<br />

tph-1::gfp expression in NSM and HSN, but the expression in ADF is not affected. UNC-86 is expressed<br />

in HSN and NSM, but not in ADF. (2) I have identified 18 mutations where tph-1::gfp expression in ADF is<br />

reduced or eliminated. Laser ablation of ADF and ASI causes animals to form dauers (Bargmann and<br />

Horvitz, 1993; Schackwitz et al., 1996) and the tph-1(mg280) mutation downregulates daf-7::gfp<br />

expression and enhances daf-7(e1372) dauer formation at 15 o C. To test the role of ADF-produced<br />

serotonin is important for a non-dauer growth, I am testing if these mutations enhance daf-7(e1372) dauer<br />

formation at 15 o C. (3) I have identified one mutation that has tph-1::gfp expressed in an extra neuron.<br />

These mutants provide me as useful means to characterize the role of serotonin produced in specific<br />

neurons.<br />

246


SEARCHING FOR NEW GENES INVOLVED IN DOSAGE<br />

COMPENSATION<br />

Chun Tsai, Barbara J. Meyer<br />

<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

Howard Hughes Medical Institute and Department of Molecular and Cell Biology, University of California,<br />

Berkeley, CA 94720<br />

In C. <strong>elegans</strong> dosage compensation equalizes expression of X-linked genes between males (XO) and<br />

hermaphrodites (XX). This chromosome-wide regulatory process is essential for hermaphrodite<br />

development. A biochemically defined protein complex containing DPY-26, DPY-27, DPY-28 and MIX-1<br />

has been shown to associate with X chromosomes in a sex-specific fashion and is required for global<br />

repression of hermaphrodite X-linked genes. MIX-1 is also required for proper segregation of mitotic<br />

chromosomes. Mutations in mix-1 cause lethality in both sexes and therefore were missed from our<br />

screens for sex-specific lethals. To identify other dosage compensation components that might have been<br />

missed, we used the following observation: mutations in all known dosage compensation genes suppress<br />

the sexual transformation caused by certain sex determination mutations in a dominant fashion, allowing<br />

recessive lethal mutations to be identified by their dominant suppression effect. For example,<br />

heterozygous mix-1 mutations can suppress the masculinization of XX animals caused by homozygous<br />

sdc-3(Tra) mutations. The suppression screen is shown below.<br />

sdc-3(Tra)/+ sdc-3(Tra) 99% Tra, 1% hermaphrodites<br />

m/+; sdc-3(Tra)/+ m/+; sdc-3(Tra) 30-99% hermaphrodites isolate mutants<br />

So far we have screened 12,000 haploid genomes and identified 9 mutations that cause moderate to<br />

strong suppression: two are lethal mutations, two are maternal-effect lethal, one is a mix-1 allele, one is<br />

an sdc-3 null allele, and three show no obvious phenotypes. The two lethal and one maternal-effect lethal<br />

mutations have been mapped extensively and their map positions indicate that they are mutations in<br />

three different genes that have not previously been implicated in dosage compensation.<br />

Immunofluorescence studies showed that the two recessive lethal mutations disrupt the association of<br />

DPY-26 and DPY-27 with the X chromosome. DAPI staining of dead embryos revealed abnormal DNA<br />

morphology and multinucleated cells, suggesting an essential role in chromosome metabolism. These<br />

observations suggest that the mutations isolated in the screen identify new genes that play a role in<br />

dosage compensation as well as chromosome structure. Attempts are ongoing to clone the genes.<br />

247


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

CHARACTERIZING THE ROLE OF LET-99 IN SPINDLE<br />

ORIENTATION<br />

Meng-Fu Tsou, Adam Hayashi, Lesilee S. Rose<br />

Section of Molecular and Cellular Biology, University of California, Davis, CA 95616 USA<br />

Cell division plane, which is determined by the orientation of the mitotic spindle, is important for normal<br />

development. The dynein-dynactin complex has been shown to be part of the mechanical force for<br />

spindle orientation in C. <strong>elegans</strong> embryos. Cytoplasmic dynein and dynactin are also essential in a variety<br />

of cell division processes in different organisms, including nuclear positioning and anaphase B spindle<br />

pole separation, partially due to their role in connecting the astral microtubules and cell cortex.<br />

The let-99 gene plays an important role in spindle orientation. We have cloned the let-99 gene and found<br />

it encodes a novel protein that is found in the cytoplasm and at regions of cell-cell contacts in early<br />

embryos. let-99 mutants show three major defects in cell division: first, abnormal spindle positioning in<br />

both the AB and the P cell lineage, second, instability in position of the nuclear-centrosome complex at<br />

prophase (nuclear rocking), and third, poor separation of the spindle poles at anaphase B. Double mutant<br />

analysis indicates that the nuclear rocking activity seen in let-99 embryos requires the function of the<br />

dynein-dynactin complex , suggesting that this behavior may be due to abnormal interaction between<br />

astral microtubules and the cell cortex. However the nuclear rocking phenotype is not due to the<br />

mislocalization of dynein-dynactin complex, because both the localization of DHC-1 (dynein heavy<br />

chain-1) and DNC-1 (p150glued) appear to be normal in let-99 embryos. G protein beta subunit (GPB-1)<br />

was also shown to be required in spindle orientation. Embryos in which the GPB-1 is maternally depleted<br />

show similar phenotypes to that of let-99, suggesting that gpb-1 and let-99 may function in the same<br />

biochemical pathway. Our preliminary result from let99 gpb-1(RNAi) double mutant analysis supports this<br />

idea. The localization of LET-99 in gpb-1(RNAi) embryos appears to be normal, indicating that the<br />

localization of LET-99 does not require the gpb-1 signaling pathway. We are currently exploring the<br />

relationship among LET-99, dynein-dynactin complex and GPB-1 by a variety of techniques, in addition to<br />

searching for LET-99 interacting proteins using the two-hybrid approach.<br />

248


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

CHARACTERIZATION AND CLONING OF THE MUSCLE<br />

ACTIVATION GENE UNC-58<br />

Monika Tzoneva, James H. Thomas<br />

Dept. of Genetics, University of Washington, Seattle WA 98195<br />

unc-58 was first identified by dominant mutations that cause hypercontracted body-wall and egg-laying<br />

muscle in C. <strong>elegans</strong>. unc-58(dm) animals are rigidly paralyzed and egg-laying constitutive. Putative<br />

loss-of-function unc-58 alleles have been isolated as revertants of the unc-58(dm) phenotype. These<br />

alleles have no obvious phenotype on their own, suggesting that unc-58(dm) mutants result in an<br />

inappropriately activated gene product. unc-58(dm) animals also frequently flip around their longitudinal<br />

axis. Both the flipping and the hypercontarction phenotypes are partially rescued by the drug endosulfan,<br />

best known as an antagonist of GABA-gated chloride channels (B. Wightman and G. Garriga, personal<br />

communication; our unpublished data).<br />

I have cloned unc-58 and found that it encodes a potassium channel of the TWIK family. TWIKs are<br />

distinct from other potassium channels because they have four transmembrane domains (M1 to M4) and<br />

two pore domains. Potassium-selective currents have been recorded from TWIKs in both mammals and<br />

worms.<br />

The three unc-58 gain-of-function mutations cluster in the C-terminal (cytoplasmic) part of the M4<br />

transmembrane domain, which is equivalent to the S6 domain of the voltage-gated potassium channel<br />

family. S6 is thought to form a channel gate that opens in response to voltage changes. The location of<br />

the unc-58(dm) mutations suggests that its M4 helix may also form an activation gate, though the stimulus<br />

that opens it is unknown.<br />

The molecular identity of unc-58 raises several questions. First, how does an activated mutation in a<br />

putative potassium channel lead to muscle hypercontraction? It is possible that the hypercontraction is<br />

due to UNC-58 function in inhibitory motorneurons, while its slow movement may be due in part to its<br />

function in other tissues. We will test this by determining the unc-58 site(s) of action. Second, how is the<br />

unc-58(dm) phenotype rescued by the drug endosulfan? We will attempt to determine whether this rescue<br />

is by direct channel block. Third, how does the UNC-58 channel contribute to overall membrane<br />

excitability? It is not known whether or how UNC-58 activity is regulated and whether it acts alone or in<br />

complex with other subunits. We have isolated extragenic suppressors of unc-58(dm), which may offer<br />

insight into this question.<br />

249


THE STRUCTURE/FUNCTION RELATIONSHIP OF CLK-1 IN<br />

THE NEMATODE CAENORHABDITIS ELEGANS<br />

Antonio Ubach, Siegfried Hekimi<br />

Department of Biology, McGill University, Canada<br />

<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

The lifespan of the nematode <strong>Caenorhabditis</strong> <strong>elegans</strong> is genetically determined by several classes of<br />

genes. Our laboratory is particularly interested in the clk class of genes, which is composed of clk-1, clk-2,<br />

clk-3, and gro-1. Mutations in these genes have been shown to extend lifespan and to deregulate several<br />

developmental and behavioral processes. For example, clk-1 mutants exhibit an average slowing down of<br />

cell division, pharyngeal pumping, and defecation rates relative to wild-type animals. These phenotypes<br />

are maternally rescued, that is homozygous mutant animals coming from a heterozygous mother are<br />

phenotypically wild type. clk-1 encodes a 187 amino acid mitochondrial protein that is composed of two<br />

homologous TRC domains (TRC for Tandemly Repeated in CLK-1). Interestingly, the yeast homologue of<br />

clk-1, COQ7, has been implicated in ubiquinone biosynthesis. Moreover, coq7 mutants are respiration<br />

defective, yet the respiratory competence can be restored by the addition of exogenous ubiquinone. In<br />

contrast, cellular respiration is only slightly affected in clk-1 mutant worms. However, C. <strong>elegans</strong> clk-1 as<br />

well as the rat and the human homologues are capable of functionally complementing the Dcoq7 mutant.<br />

This observation suggests that the biochemical function of clk-1 have been conserved throughout<br />

evolution.<br />

We have shown previously that a recombinant CLK-1::GFP fusion protein is capable of rescuing the clk-1<br />

mutant phenotype when expressed from transgenic arrays. In the current study, we are taking advantage<br />

of this observation to try to understand the structural requirements for CLK-1 function. In order to address<br />

this, we are using a site-directed mutagenesis approach to generate several new CLK-1 mutant proteins.<br />

We are analysing the effect these CLK-1 mutant proteins have on the general clk-1 phenotype. We are<br />

also examining the subcellular distribution of the mutant fusion proteins in different genetic backgrounds,<br />

such as the wild type, the clk-1 partial loss of function e2519, and the two clk-1 putative nulls qm30 and<br />

qm51.<br />

250


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

CHARACTERIZATION OF TRANSCRIPTIONAL REGULATION<br />

BY A CLASS OF MONOMERIC NUCLEAR RECEPTORS<br />

FOUND IN C. ELEGANS<br />

Marc R. Van Gilst, Keith R. Yamamoto<br />

Cellular and Molecular Pharmacology, University of California-San Francisco, San Francisco, CA<br />

94143-0450<br />

Nuclear hormone receptors (NHRs) comprise a large family of metazoan transcription factors that<br />

participate in numerous developmental and metabolic processes. NHRs commonly target their<br />

transcriptional regulation to particular genes through interaction with DNA sequences called hormone<br />

response elements (HREs) and the nature of the transcriptional regulation carried out by a NHR at these<br />

genes is often determined by interaction with a small hydrophobic ligand.<br />

C. <strong>elegans</strong> provides unique advantages over other animal models for studying the physiological<br />

significance of several aspects of NHR function (i). Targeting of NHRs to promoters by HREs (ii)<br />

Characterization of important regulatory domains of NHRs (iii) Interaction of NHRs with protein cofactors<br />

and (iv) Interaction of NHRs with potential ligands.<br />

We have focused our studies on the C. <strong>elegans</strong> homologs of a class of NHRs that can activate<br />

transcription from AGGTCA DNA half-sites, a common binding motif for a mammalian sub-family of<br />

monomeric nuclear receptors that includes ROR, TLX, ERR and SF-1. To this end, we have started our<br />

investigations by utilizing the C. <strong>elegans</strong> receptor CHR3 (nhr-23). CHR3 is closely related to the<br />

mammalian receptor ROR, with nearly 100% conservation in the DNA binding domain, and would be<br />

expected to fall into the class of monomeric NHRs that we wish to study.<br />

Using yeast and worm reporter assays, we have characterized the transcriptional regulatory properties of<br />

CHR3. We found that CHR3 can activate transcription from a single AGGTCA half-site and activates<br />

synergistically from multimerized half-sites. Transcriptional activation occurs in yeast in the absence of<br />

any exogenous ligand. Similar to mammalian receptors, both the N and C terminal domains of CHR3<br />

contain activation functions. Furthermore, CHR3 requires ATP-dependent chromatin remodeling<br />

complexes for activation and can interact with a mammalian coactivator GRIP through a conserved<br />

structural mechanism. We are currently expanding these systems to find and study other C. <strong>elegans</strong><br />

NHRs and to set up screens for the identification of potential protein cofactors and small-molecule<br />

ligands.<br />

251


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

UNC-4 TARGETS ACR-5 AND DEL-1: ARE THEY<br />

DETERMINANTS OF SYNAPTIC CHOICE?<br />

Stephen E. Von Stetina, David M. Miller, III<br />

Dept. of Cell Biology, Vanderbilt University , Nashville, TN 37232-2175<br />

Mutations in the UNC-4 homeodomain transcription factor disrupt backward locomotion. EM<br />

reconstruction of an unc-4 mutant revealed that the VA motor neurons fail to receive synapses from their<br />

usual interneuron partners and instead accept inputs normally reserved for their sisters, the VB motor<br />

neurons. Work done in this laboratory has shown that UNC-4 and the Groucho homolog UNC-37 function<br />

together in the VA motor neurons to repress VB-specific genes. Two genes, del-1 (DEG/ENaC sodium<br />

channel subunit) and acr-5 (alpha-like nictonic acetylcholine receptor subunit), are ectopically expressed<br />

in the VA motor neurons in unc-4 and unc-37 mutants. As cell surface proteins and ion channel<br />

components, ACR-5 and DEL-1 are attractive candidates for mediators of synaptic specificity. We have<br />

now performed genetic experiments, however, that rule out a necessary role for either ACR-5 or DEL-1<br />

in the specification of VB-type inputs. Deletion mutants of del-1 and acr-5, as well as the double mutant<br />

(acr-5;del-1), do not perturb forward locomotion as would be expected for a mutation which disrupts<br />

normal inputs to the VBs. Furthermore, assays designed to quantitate locomotion have detected no<br />

differences between wild type and these mutants. Placement of acr-5 and del-1 mutants in an unc-4<br />

background does not suppress the Unc-4 phenotype, suggesting that ACR-5 and DEL-1 are also not<br />

required to promote VB-type inputs in VA motor neurons. A future goal is to ectopically express ACR-5<br />

and DEL-1 in VA motor neurons to determine if these proteins are sufficient to cause miswiring. Also, we<br />

will define the UNC-4 mediated repression domains in the acr-5 and del-1 promoters in an effort to<br />

identify additional UNC-4 targets.<br />

252


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

NICOTINE ADAPTATION: A PROCESS INVOLVING<br />

PKC-DEPENDANT REGULATION OF NACHR PROTEIN<br />

LEVELS.<br />

Laura Waggoner, Kari Dickonson, Daniel Poole, Bill Schafer<br />

Department of Biology, UCSD, 9500 Gilman Dr., La Jolla, 92093<br />

C. <strong>elegans</strong> is capable of undergoing adaptation to prolonged exposure to nicotine, through attenuation of<br />

the activity of nicotinic acetylcholine receptors (nAChRs). We have discovered that in egg-laying behavior,<br />

acute nicotine treatment results in stimulation of egg-laying, but long-term exposure inhibits further<br />

stimulation by cholinergic agonists. Furthermore, we found that this adaptation process involves<br />

UNC-29-containing nAChRs functioning in the vulval muscles. We were interested in determining whether<br />

adaptation was due to a loss or attenuation of theseUNC-29-containing nAChRs; to address this, we<br />

analyzed fluorescence levels of an UNC-29::GFP chimera in the vulval muscles before and after nicotine<br />

treatment. Intriguingly, we found that nicotine treatment in fact decreased receptor protein levels in the<br />

vulval muscles. In addition, we found that this regulation was independent of the unc-29 promoter and 3’<br />

UTR. Lastly, we were interested in identifying other genes involved in this process, and we found that the<br />

protein kinase C homolog encoded by tpa-1 was necessary for the observed decrease in nAChR protein<br />

levels. Thus, it appears that nicotine treatment results in a PKC- dependant down-regulation of<br />

UNC-29-containing nAChRs in the vulval muscles. Further screens are being performed to identify other<br />

genes involved in this process.<br />

253


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

ANALYSIS OF GLR-7, GLR-5, IONOTROPIC GLUTAMATE<br />

RECEPTOR SUBUNITS<br />

Craig S. Walker, David M. Madsen, Penelope J. Brockie, Andres V.<br />

Maricq<br />

Department of Biology, University of Utah, Salt Lake City, UT 84112<br />

Of the ten putative ionotropic glutamate receptor subunits identified in the worm, two non-NMDA subunits,<br />

glr-5 and glr-7, show a very restricted expression pattern. When GLR-5 and GLR-7 are fused to GFP they<br />

show expression in only a single cell, the interneuron RIA. RIA receives synaptic input from the neurons<br />

AIY and AIZ. Previous studies have defined this group of cells to form part of the circuit that controls<br />

thermotaxis.<br />

To determine the possible roles of glr-5 and glr-7 in thermotaxis, we have generated deletion mutations in<br />

both genes. These deletions remove a portion of the extracellular domain and the first three<br />

transmembrane domains. In each case the mutations result in a functional null. Analysis of glr-5(ak57)<br />

deletion mutants using thermotaxis assays is underway. glr-5(ak57) worms also appear to have a<br />

diminished brood size, which is more severe at elevated temperatures.<br />

In order to examine the role of RIA in processing thermal information we have generated transgenic<br />

strains which express an activated form of the GLR-1 glutamate receptor subunit, GLR-1 (A/T), under<br />

control of the glr-5 promoter. GLR-1 (A/T) causes constant depolarization when expressed in other cells<br />

(see abstract by Zheng et al.). We are currently characterizing the thermotaxis behavior of these strains.<br />

Any thermotaxic phenotype observed in glr-5(ak57) may be a result of the inability of RIA to receive<br />

signals from the presynaptic cell AIY. Hobert et al. have demonstrated that the gene ttx-3 is expressed<br />

only in AIY. Using ttx-3::VAMP::CFP and glr-5::GLR-5::YFP constructs we are looking at the presynaptic<br />

and post-synaptic components of this synapse to determine if glr-5 could be mediating the signal from<br />

AIY.<br />

254


MICROARRAY ANALYSIS OF GENE EXPRESSION<br />

PATTERNS IN DAUER LARVAE<br />

John Wang, Stuart K. Kim<br />

<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

Dept of Developmental Biology, Stanford University, Stanford CA<br />

DNA microarrays containing about 95% of the genome can now be used to analyze gene expression<br />

differences from nearly the entire genome. We are using the full-genome microarrays to profile gene<br />

expression differences associated with the dauer larvae.<br />

Under conditions of starvation or extreme crowding, C. <strong>elegans</strong> enter an alternative arrested stage, the<br />

dauer larva. These larvae exhibit a unique morphology, an extended life span and resistance to<br />

environmental stresses. The decision to form dauers is controlled by TGF-beta and insulin signaling<br />

pathways.<br />

The dauer larvae is an ideal system for microarray analysis. The dramatic morphological and<br />

physiological changes that occur upon entrance into and exit from the dauer larvae suggest global<br />

transcriptional regulation. In principle, by examining the gene expression differences associated with the<br />

dauer larvae on the whole-genome level, we will be able to illuminate the complete set of genes that are<br />

implicated for the dauer-specific attributes. In particular, these candidate genes would provide a<br />

framework for understanding dauer-specific characteristics, such as altered energy metabolism, certain<br />

aspects of aging and longevity, stress resistance, and the coordinated regulation and execution of events<br />

necessary for a complex morphological change.<br />

To identify the genes involved with dauer exit, we are performing DNA microarray experiments with RNA<br />

isolated at different time points after addition of food to a pure dauer population. Each RNA sample is<br />

compared to a common reference RNA, and multiple RNA samples are prepared for each time point. We<br />

will identify those genes that are reproducibly altered during dauer exit and examine their kinetic profiles.<br />

This analysis should reveal the temporal sequence of events that occur during dauer recovery at the<br />

transcriptional level.<br />

255


CHARACTERIZATION OF CAN CELL AND EXCRETORY<br />

CANAL DEFECTS IN MIG-10(CT41) MUTANTS<br />

Nicole Washington, Jim Manser<br />

<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

Dept. of Biology, Harvey Mudd College, Claremont CA 91711<br />

We have continued our characterization of mig-10 mutant defects using a ceh-23-gfp reporter strain<br />

(kyIs5 IV, kindly provided by Jennifer Zallen) which allows scoring of CAN cell bodies and axons (see<br />

Manser et al. WCWM 1998 abstracts, p.167). In the current studies, we have focused on the amber<br />

(putative null) mig-10(ct41) allele.<br />

Nearly 90% of CAN cell bodies (61/68) are significantly displaced anteriorward in mig-10(ct41);kyIs5<br />

animals, compared to less than 8% (5/64) for the kyIs5 strain. Manser and Wood (1990) previously<br />

reported a penetrance of 60% for CAN cell body misplacement in mig-10(ct41). The higher penetrance<br />

observed using the gfp reporter probably reflects a synergy between mig-10 and ceh-23-gfp similar to that<br />

reported by Forrester et al. (1997) for other CAN migration mutants.<br />

A significant percentage of posteriorly directed CAN axons in mig-10(ct41);kyIs5 animals (23/67)<br />

terminate in positions significantly anterior to their normal target region near the anus. In contrast, all<br />

anteriorly directed CAN axons appear to extend to their normal target region in the head. This apparent<br />

directional bias is somewhat surprising given that mig-10(ct41) disrupts both anteriorward and<br />

posteriorward cell body migrations (Manser and Wood, 1990). One possibility is that the CAN axon<br />

defects are an indirect result of the shortening of the posterior excretory canals observed in mig-10(ct41)<br />

(Manser and Wood, 1990). Specifically, because CAN axons are closely associated anatomically with the<br />

excretory canals (CAN=canal associated neuron), perhaps the canals serve as guidance cues for axon<br />

outgrowth.<br />

To investigate this possibility, we have scored both CAN axon and posterior excretory canal defects in the<br />

same individuals. In more than 90% of mig-10(ct41);kyIs5 animals scored (28/31), the posteriorly directed<br />

CAN axon extended significantly beyond the point of termination of the posterior excretory canal. This set<br />

includes animals in which the posterior CAN axon extends to the anal region while the posterior excretory<br />

canal terminates significantly anterior to the vulval region. Due to the generally severe truncation of the<br />

posterior canals caused by mig-10(ct41), we have also observed animals in which the CAN cell body is<br />

positioned posterior to the canal terminus. In such animals, anteriorly directed CAN axons were observed<br />

to extend over regions of the anterior-posterior axis from which canals were missing. These observations<br />

do not support the view that the excretory canals serve as guidance cues for CAN axon outgrowth.<br />

256


RIC-7 ENCODES A NOVEL PRESYNAPTIC PROTEIN<br />

REQUIRED FOR NEUROTRANSMISSION<br />

Robby M. Weimer, Erik M. Jorgensen<br />

University of Utah, Salt Lake City, UT 84112<br />

<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

Regulated release of neurotransmitter from presynaptic terminals requires the coordinated function of<br />

several presynaptic proteins. Our current knowledge of this process has come primarily from biochemical<br />

analysis of synaptic proteins. We have taken a genetic approach in <strong>Caenorhabditis</strong> <strong>elegans</strong> to<br />

complement these studies. Previously, we reported the identification of a novel protein, RIC-7, identified<br />

genetically by screens for neurotransmission mutants. We now report that RIC-7 functions in neurons<br />

and is localized to presynaptic terminals.<br />

Mutations in ric-7 result in a pleiotropic neurotransmission phenotype. ric-7 animals lack enteric muscle<br />

contractions during defecation and display a weak shrinker phenotype, indicating of a loss of GABAergic<br />

function. In addition, ric-7 animals are resistant to the acetylcholinesterase inhibitor aldicarb, although<br />

they remain sensitive to the acetylcholine receptor agonist levamisole indicating a presynaptic cholinergic<br />

defect. These phenotypes are not due to defects in neuronal developmental since the overall structure of<br />

the nervous system appears normal. This suggests that mutations in ric-7 disrupt a common presynaptic<br />

step in chemical neurotransmission.<br />

The ric-7 phenotype is rescued by an 18kb PCR fragment that contains a single open reading frame<br />

encoding a predicted 694 amino acid protein with no similarities to known proteins or functional motifs.<br />

Expression of the reporter gene green fluorescent protein (GFP) from the ric-7 promoter identifies a<br />

predominantly neuronal expression pattern. RIC-7 GFP fusion protein is localized to synaptic terminals<br />

independent of UNC-104 function. Furthermore, expression of RIC-7 solely in GABAergic neurons<br />

rescues all GABA-specific phenotypes.<br />

Together, these data suggest that ric-7 encodes a neuronal protein that is localized to and functions at<br />

presynaptic terminals. In addition, ultrastructural data indicate that synaptic vesicle biogenesis is not<br />

affected in a ric-7 mutant background. Thus, RIC-7 is likely to play a role in synaptic vesicle exocytosis.<br />

Ongoing experiments are designed to identify RIC-7 interacting proteins and to determine the molecular<br />

function of RIC-7 in neurotransmission.<br />

257


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

THE REQUIREMENT OF SYNAPTIC VESICLE LOADING FOR<br />

SYNAPTIC VESICLE EXOCYTOSIS<br />

Robby M. Weimer, Janet E. Richmond, Erik M. Jorgensen<br />

Department of Biology, University of Utah, Salt Lake City, UT 84112-0840<br />

Neurotransmitters are released from presynaptic terminals by the fusion of synaptic vesicles with the<br />

plasma membrane. The amount of neurotransmitter in a vesicle causes a consistent amount of current,<br />

called a quanta, in the postsynaptic cell. Studies have shown that quanta are invariant within a synapse.<br />

This observation raises an interesting question. Does synaptic vesicle fusion require the proper loading<br />

of neurotransmitter into the vesicle? We have characterized a mutant in C. <strong>elegans</strong> that will allow us to<br />

address this question.<br />

Loading of neurotransmitter into synaptic vesicles requires a vesicular neurotransmitter-specific<br />

transporter and a driving force. Several studies have shown that the vacuolar H+ ATPase (vATPase)<br />

generates the required driving force for loading. Previously, we reported the cloning of the C. <strong>elegans</strong><br />

homolog of the vacuolar ATPase B subunit, which we now refer to as vha-12 (vacuolar-type H+ ATPase).<br />

A hypomorphic allele of vha-12 results in a pleiotropic neurotransmission phenotype which is consistent<br />

with the know function of the vATPase in neurotransmission. However, animals homozygous for the<br />

hypomorphic allele are not paralyzed. This suggests that neurotransmitter is still released from<br />

presynaptic terminals in a vha-12 animal.<br />

Two models could account for this observation (1) synaptic vesicles are fusing with the plasma membrane<br />

with incomplete loading of neurotransmitter or (2) only completely loaded vesicles are competent to fuse<br />

and the proportion of competent vesicles are decreased in a vha-12 mutant background. In order to<br />

distinguish between these two models we are characterizing neurotransmitter release events in a vha-12<br />

background by electrophysiologically recording from neuromuscular junctions.<br />

258


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

UNRAVELING THE BIOLOGICAL ROLE OF DMWD, A GENE<br />

CLOSE TO THE UNSTABLE CTG-REPEAT IN THE MYOTONIC<br />

DYSTROPHY LOCUS.<br />

J. <strong>West</strong>erlaken 1 , B. Wieringa 2 , P.E. Mains 1<br />

1Department of Biochemistry & Molecular Biology, University of Calgary, Canada<br />

2Department of Cell biology University of Nijmegen, the Netherlands<br />

Myotonic Dystrophy (DM) is a frequent autosomal dominant disorder, caused by the expansion of an<br />

unstable (CTG)n-repeat. The expanding repeat is located in the 3’-untranslated region of the DM protein<br />

kinase gene (DMPK). The DMPKgene is located in a gene dense area, therefore it is unclear if it is<br />

theonly gene involved in disease manifestation. Another possible candidate is the DMWD gene (formally<br />

known as DMR-N9 in mouse, or gene 59 in human), which is located closely upstream of the DMPK<br />

gene. The DMWD gene is mainly expressed in brain, testis and some smooth muscle tissues. In brain the<br />

protein is located in many areas, but most prominently in places where there are synaptic glomeruli. In<br />

testis the protein appears to be located in several distinct stages of spermatogenesis.<br />

The DMWD gene is conserved during evolution and has homologs in plant, yeast and nematodes. To<br />

futher understand the biological role of DMWD we started to investigate the C. <strong>elegans</strong> DMWD gene. In<br />

C. <strong>elegans</strong> DMWD RNA is expressed in embryos and gravid adults. Preliminary results from RNAi studies<br />

suggest that some progeny of the injected worms are sterile. We are further planning to do RNA in situ<br />

hybridisation, GFP tagging of the protein and antibody staining experiments.<br />

259


CALCIUM IMAGING OF THE DEFECATION RHYTHM IN C.<br />

ELEGANS<br />

Jeanna M. Wheeler, James H. Thomas<br />

Department of Genetics, University of Washington, Seattle, WA 98195<br />

Ultradian rhythms (those with a period shorter than 24 hours) have been shown to regulate a number of<br />

biological processes, such as the vertebrate heartbeat and invertebrate swimming behavior. The<br />

defecation cycle in C. <strong>elegans</strong> is an ultradian rhythm regulated by a clock-like mechanism (1). The cycle<br />

has a 45 to 50 second periodicity that is insensitive to changes in temperature between 18° and 30° C. In<br />

addition, the clock mechanism can keep time in the absence of the defecation motor program (when<br />

animals leave their food source), indicating that the clock is distinct from the motor program. It has been<br />

shown that the inositol trisphosphate (IP 3 ) receptor, a calcium channel that regulates cytoplasmic Ca 2+<br />

concentration by release from intracellular stores, is required in the intestine for the defecation cycle (2).<br />

Furthermore, calcium imaging with fura-2 revealed that periodic calcium spikes occur in the most<br />

posterior intestinal cell, just preceding the initiation of each posterior body-wall muscle contraction (pBoc).<br />

The pBoc begins at the posterior of the animal and proceeds anteriorly in a wave-like motion. There are<br />

two simple possibilities for how the calcium spike signals to the posterior body-wall muscles: 1) Ca 2+ may<br />

initiate secretion of a muscle activator only in the posterior intestine, which then propagates anteriorly<br />

through the pseudocoelomic space or by muscle-to-muscle signaling, or 2) the Ca 2+ spike may be<br />

propagated anteriorly through the intestinal cells themselves, initiating signaling to adjacent muscle cells<br />

along the way. In order to distinguish between these possibilities, I am constructing a strain that<br />

expresses fluorescent indicators for Ca 2+ ("cameleons") in the intestine. This strain will be used to<br />

characterize the defecation cycle in wild type worms, as well as in various mutant strains with cycle<br />

defects.<br />

1. D.W. Liu and J.H. Thomas, 1994. J Neurosci 14: 1953-1962.<br />

2. P. DalSanto et al, 1999. Cell 98: 757-767.<br />

<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

260


ESTABLISHING THE LEFT/RIGHT ASYMMETRY OF Q<br />

NEUROBLAST POLARISATION AND MIGRATION IN C.<br />

ELEGANS<br />

Lisa Williams, Lee Honigberg, Cynthia Kenyon<br />

University of California, San Francisco<br />

<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

The QL and QR neuroblasts are born in bilaterally symmetric positions along the A-P body axis in C.<br />

<strong>elegans</strong> (QL on the left and QR on the right). Shortly after hatching, the two cells undergo characteristic,<br />

stereotypical L/R asymmetric migrations. QL extends a long process to the posterior and its nucleus then<br />

migrates into this projection. Following this migration, the Hox gene mab-5 is expressed in QL and its<br />

descendants remain in the posterior. mab-5 expression is both necessary and sufficient for Q<br />

descendants to remain in the posterior. Conversely, QR extends a process to the anterior and<br />

subsequently migrates anteriorly. mab-5 is not expressed in QR, hence its descendants continue to<br />

migrate anteriorly. Each cell moves to a well-defined final position.<br />

In unc-40 or dpy-19 mutants, polarisation of both Q cells is randomised and the Q nuclei fail to undergo<br />

their migrations. This randomisation of Q polarisation is coupled with a randomisation of mab-5<br />

expression. Either cell, QL or QR, can express mab-5, and this expression then determines the fate of<br />

that cell’s descendants.<br />

In order to gain a more complete picture of how the initial L/R asymmetry of the Q cells is established, we<br />

are studying 11 new mutants from an extensive Q cell migration screen (Queelim Ch’ng and Mary Sym,<br />

unpublished results). These mutants fall into 4 complementation groups and have similar phenotypes to<br />

unc-40 and dpy-19, and are therefore good candidates for new genes in this pathway. Work is underway<br />

to characterise, map and clone these new genes. qid --1 (Q Is Defective, 8 alleles) maps to a 180kb<br />

region on the left arm of chromosome III, and is rescued by injection of a cosmid pool covering this<br />

region. qid-2 (1 allele) maps to the right arm of chromosome X, and further mapping is underway.<br />

Mapping and further characterisation are also planned for qid-3 (1 allele) and qid-4 (1 allele), although<br />

these mutants have less penetrant Q phenotypes and appear to be more pleiotropic.<br />

261


A SCREEN FOR CELL MIGRATION AND AXON OUTGROWTH<br />

MUTANTS<br />

Jim Withee, Gian Garriga<br />

<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

Molecular and Cellular Biology, University of California, Berkeley<br />

Proper development of the nervous system requires that individual neurons and their processes arrive at<br />

precise coordinates to form the correct pattern of connectivity. Directed migration of cells and growth<br />

cones involves recognition of environmental cues by receptor proteins and the translation of these cues<br />

into growth or movement to precise destinations. Thus, a primary goal of developmental neurobiology is<br />

the identification and characterization of directional cues, their cognate receptors and the intracellular<br />

proteins that interpret these cues.<br />

The left and right CAN cells are born in the head and migrate posterior to the gonad primordium. After<br />

migration is complete, the CAN cells each extend a single, unbranched axon anterior to the base of the<br />

nerve ring and posterior to the lumbar ganglia. <strong>Worm</strong>s expressing GFP from the ceh-23 promoter exhibit<br />

fluorescence from a subset of neurons including the CAN cell (Wang et al, 1993, Cell 74). We are utilizing<br />

a ceh-23::GFP expressing strain and fluorescence microscopy to screen directly for mutations that disrupt<br />

proper migration and axon outgrowth of the CANs. To date, we have identified at least five<br />

complementation groups that display defective cell migration and axon outgrowth. The mutants exhibit a<br />

wide variety of CAN axon defects ranging from simple truncation to extensive ectopic outgrowth and<br />

branching. We are in the process of mapping and characterizing the mutations as we screen for additional<br />

mutants. Because a large portion of cell and axon guidance mechanisms appear to be conserved from C.<br />

<strong>elegans</strong> to vertebrates, it is likely that some of the genes identified in this screen will play a conserved role<br />

in nervous system development.<br />

262


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

MAPPING AND CHARACTERIZATION OF HAD-1, AN HSN<br />

AXON GUIDANCE GENE<br />

Lianna Wong, Jim Rader, Gian Garriga<br />

Molecular and Cell Biology, University of California, Berkeley, CA 94720-3204<br />

To understand how growth cones are guided to their synaptic targets, we are studying the outgrowth of<br />

the HSN axons. The HSNs are a bilaterally symmetric pair of serotonergic motor neurons that innervate<br />

the vulval muscles and stimulate egg laying in C. <strong>elegans</strong>. The mature HSN axon morphology can be<br />

visualized using gmIs1, an integrated arrestin::GFP reporter that illuminates the HSN cell bodies and<br />

axons. In wild-type animals, each HSN axon extends ventrally to the ipsilateral ventral nerve cord (VNC)<br />

tract, turns anteriorly along it, then skirts dorsolaterally around the vulva, elaborating varicosities and<br />

small branches that innervate the vulval muscles. The left and right HSN axons then continue their<br />

anteriorly-directed extensions in their respective, parallel axon tracts until they reach the nerve ring in the<br />

head.<br />

Our lab performed a genetic screen for mutants defective in HSN axon guidance. Two of the mutants<br />

isolated, gm188 and gm204, display grossly abnormal HSN axon outgrowth patterns, consisting of circling<br />

and looping of the axons around the vulva, elaboration of excessively long branches, and multiple<br />

crossovers of the axons between the left and right VNC tracts. In some mutant animals, the HSN axons<br />

stop before reaching the nerve ring; in others, the HSN axons turn posteriorly and migrate back to or<br />

beyond the vulva. The gm188 and gm204 mutations both map to LG V and fail to complement one<br />

another. We have tentatively named the gene defined by these mutations as had-1 (HSN axon defective).<br />

Three-factor mapping places had-1 between sma-1 and him-5. There are a number of overlapping<br />

deficiencies in the region; nDf42 and yDf12 both uncover had-1, while arDf1 does not, placing had-1<br />

between the right breakpoint of arDf1 and him-5, a region of less than one map unit. We have injected<br />

had-1 mutants with cosmids in this region, but thus far have not achieved rescue of the HSN phenotype.<br />

To more precisely define the position of had-1 in this interval, we are presently mapping had-1 relative to<br />

cloned genes and Tc1 polymorphisms.<br />

In addition to mapping and cloning had-1, we are characterizing the HSN axon defects of had-1 mutants<br />

in animals lacking vulval cells and sex muscles to ascertain what roles these tissues play in the Had-1<br />

HSN axon phenotype.<br />

263


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

RECOGNITION OF X-CHROMOSOME-ENRICHED DNA<br />

ELEMENTS BY DOSAGE COMPENSATION PROTEINS<br />

Tammy F. Wu 1 , Jason D. Lieb 2 , Barbara J. Meyer 1<br />

1Howard Hughes Medical Institute and Department of Molecular and Cell Biology, University of California,<br />

Berkeley, CA, 94720<br />

2Howard Hughes Medical Institute and Department of Biochemistry, Stanford University Medical Center,<br />

Stanford, CA 94305<br />

In C. <strong>elegans</strong> X-chromosome dosage compensation is accomplished by repressing gene expression from<br />

both hermaphrodite X chromosomes to equal expression from the single male X. This essential process is<br />

carried out through the action of a large protein complex that localizes to X starting around the 50-cell<br />

stage in XX, but not XO, embryos.<br />

To study how dosage compensation proteins come to be localized to X chromosomes, we are<br />

investigating whether dosage compensation proteins recognize specific cis-acting elements. Previously,<br />

dosage compensation proteins were shown to localize to extrachromosomal arrays bearing specific<br />

regions of the promoter of the autosomal her-1 gene, whose expression is required for male development.<br />

Thus, one approach to the problem of X recognition is to test candidate elements for their ability to be<br />

recognized by dosage compensation proteins. To find such candidates, we have searched the genome<br />

for sequence elements that occur more often on X than on autosomes. Sixteen such elements were<br />

identified, and were found to occur in a wide variety of distributions throughout the X chromosome. These<br />

candidates are being tested for recognition by dosage compensation proteins on arrays in vivo, as<br />

previously described (Carmi et al., Nature 1998, and Dawes et al., Science 1999). Because the sequence<br />

context of these elements may be important for recognition, we are using PCR to amplify products<br />

containing unique surrounding sequence, as well as the candidate element itself.<br />

Two PCR products tested have yielded promising results. Interestingly, the sequences tested have a<br />

roughly 250 base pair region in common, which is itself enriched on the X chromosome. We are currently<br />

investigating the significance of these sequences.<br />

264


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

RAC-LIKE GTPASES AND CELL MIGRATION<br />

Yi-Chun Wu, Li-Chun Cheng, Nei-Yin Weng, Ting-Wen Cheng<br />

Zoology Department, National Taiwan University, No 1 Roosevelt Road Sec4, Taipei 10617, Taiwan,<br />

Republic of China<br />

Cytoskeletal rearrangement and cell polarization are important for fundamental cellular processes, such<br />

as cell migration and the phagocytosis of apoptotic cells. We are interested in how migration cues and<br />

apoptotic signals are interpreted by the receiving cells and subsequently affect their cytoskeletal<br />

rearrangement during cell migration and cell-corpse-phagocytosis in <strong>Caenorhabditis</strong> <strong>elegans</strong>.<br />

At least two C. <strong>elegans</strong> Rac-like GTPases have been shown to be important for cell migration. MIG-2 is<br />

necessary for the migration of Q cells, Q cell descendents and coelomocytes (1). CED-10 is important for<br />

the migration of distal tip cells as well as the engulfment of apoptotic cells (2). To test if these two<br />

GTPases may function redundantly in the migration of other cells, we have constructed ced-10;mig-2<br />

double mutants and are currently analyzing the mutant phenotype. It has been proposed that ced-10 may<br />

function downstream of ced-2 and ced-5 during cell-corpse-engulfment (2). We have also constructed<br />

ced-2; mig-2 and ced-5; mig-2 double mutants. Results will be presented at the meeting.<br />

1. Zipkin et al., Cell, 90, 883-894 (1997)<br />

2. Reddien and Horvitz, Nature Cell Biology, 2, 131-136 (2000)<br />

265


TWO NEW GENES REGULATING NEUROBLAST MIGRATION<br />

IN C. ELEGANS<br />

Lucie Yang, Mary Sym, Queelim Ch’ng, Cynthia Kenyon<br />

Box 0448, 513 Parnassus, Dept. of Biochemistry, San Francisco, CA 94143-0448<br />

In C. <strong>elegans</strong>, the Q cells are bilaterally symmetric neuroblasts present in the posterior body region of the<br />

worm at hatching. During the first larval stage, the Q cells divide and migrate. QR and its descendents<br />

migrate anteriorly whereas QL and its descendents migrate posteriorly. Several genes that regulate the<br />

anterior migrations of QR and its descendents have been identified. These include: 1) lin-39, a homeobox<br />

gene required in QR and its descendents for migration (Wang B. et al, 1993; Clark S. et al, 1993); 2)<br />

mig-13, a novel transmembrane protein required outside of QR and its descendents for migration (Sym M<br />

et al, 1999); and 3) egl-20, a Wnt homolog expressed in cells in the tail region (Whangbo J.. and Kenyon<br />

C, 1999). Two mutant screens (Mary Sym and Queelim Ch’ng) were conducted to identify additional<br />

genes that regulate the migration of QR and its descendents.<br />

From these screens, mutations in two new genes were identified that cause certain cells in the QR<br />

lineage to stop migrating prematurely. These two genes seem likely to be involved in guidance rather than<br />

in providing cells with the ability to migrate because, in these mutants, other cells sometimes migrate in<br />

the wrong direction. Current efforts are directed toward cloning these two genes and determining how<br />

these genes regulate the migration of QR and its descendents together with genes previously known to<br />

regulate these migrations.<br />

References:<br />

<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

Clark SG, Chisholm AD, and Horvitz HR. 1993. Control of Cell Fates in the Central Body Region of C.<br />

<strong>elegans</strong> by the Homeobox Gene lin-39. Cell 74: 43-55.<br />

Sym M, Robinson N, Kenyon C. 1999. mig-13 Positions Migrating Cells Along Anteroposterior Body Axis<br />

of C. <strong>elegans</strong>. Cell 98: 26-36.<br />

Wang BB, Muller-Immergluck MM, Austin, J, Robinson, NT, Chisholm, A, Kenyon, C. A Homeotic Gene<br />

Cluster Patterns the Anteroposterior Body Axis of C.. <strong>elegans</strong>. Cell 74: 29-42.<br />

Whangbo J, Kenyon, C. A Wnt Signaling System that Specifies Two Patterns of Cell Migration in C.<br />

<strong>elegans</strong>. Molecular Cell 4: 851-858.<br />

266


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

IDENTIFICATION AND CHARACTERIZATION OF TELOMERE<br />

BINDING PROTEINS IN THE NEMATODE C. ELEGANS<br />

Su Young Yi, Seunghyun Kim, Junho Lee<br />

Department of Biology, Yonsei University, Seoul 120-749<br />

Telomeres are multifunctional elements that shield chromosome ends from degradation and end-to-end<br />

fusions, prevent activation of DNA damage checkpoints, and modulates the maintenance of telomeric<br />

DNA by telomerase. Telomeric DNA has been found to interact with proteins in many organisms. For<br />

example, hTRF1, a human telomeric-repeat binding factor, is involved in the telomere length regulation.<br />

We wanted to identify and elucidate functions of telomere binding proteins in the nematode C. <strong>elegans</strong>.<br />

Telomeric repeat of C. <strong>elegans</strong> consists of a long stretch of (TTAGGC)n.<br />

1. We have isolated Ceh-37, a novel homeotic protein in C. <strong>elegans</strong>, as double-stranded telomere binding<br />

factor by yeast one-hybrid screening. We found that Ceh-37 specifically binds C. <strong>elegans</strong> telomere in<br />

vitro. We found that the N-terminal domain and the homeotic domain is sufficient for telomere binding. We<br />

also found that Ceh-37 binds the telomere as dimers, and that the N-terminal is required for the<br />

dimerization. Ceh-37 is expressed not in all cells, but in subsets of cells. Over-expression of Ceh-37<br />

caused death in late larval stages.<br />

2. We have identified a protein (CeST-BP) in C. <strong>elegans</strong> embryonic nuclear extract that specifically binds<br />

single-stranded telomere sequences by gel mobility shift assay. We found that the two Ts and Gs of the<br />

telomere sequence were necessary to efficiently bind the C. <strong>elegans</strong> single-stranded telomere binding<br />

protein (CeST-BP). CeST-BP did not efficiently bind with repeated RNA sequences of UUAGGC, thus we<br />

concluded that CeST-BP bound specifically with DNA only. We also found that CeST-BP needs at least<br />

2.5 repeats of (GGCTTA) sequence for binding. CeST-BP was sensitive to salt concentrations, and<br />

insensitive to RNase treatment. The size of CeST-BP was found to be about 40 kD in South-<strong>West</strong>ern<br />

hybridization. We plan to identify CeST-BP by purification using DNA-streptavidin sepharose affinity<br />

chromatography and MALDI-TOF protein sequence search. Subsequently, studies of the functions of<br />

CeST-BP in vivo will follow.<br />

267


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

MOLECULAR ANALYSIS OF THE DOSAGE COMPENSATION<br />

GENE DPY-21<br />

Stephanie Yonker, Edith Cookson, Barbara J. Meyer<br />

Howard Hughes Medical Institute and Department of Molecular and Cell Biology, University of California,<br />

Berkeley, CA 94720<br />

Dosage compensation in <strong>Caenorhabditis</strong> <strong>elegans</strong> equalizes X chromosome expression between XO<br />

males and XX hermaphrodites. DPY-21 is one of several proteins required for proper dosage<br />

compensation. However, DPY-21 is unique among known dosage compensation proteins. While other<br />

dosage compensation mutations cause maternal-effect lethality and affect only XX animals, dpy-21<br />

mutations cause only modest lethality and affect both XX and XO animals. Furthermore, different assays<br />

of X-linked gene expression have shown that dpy-21 mutations cause both elevation and reduction in<br />

X-linked gene expression in XO animals. We have cloned dpy-21 by injecting single-stranded RNA from<br />

candidate ESTs that map to a YAC close to dpy-21. Two positive ESTs were found to represent the same<br />

gene and were used to assemble the dpy-21 mRNA sequence in conjunction with 5’ and 3’ RACE<br />

products. To verify that we had identified dpy-21, we sequenced six dpy-21 alleles and found DNA lesions<br />

within the predicted coding sequence. Three of the sequenced dpy-21 alleles contain premature STOP<br />

codons. The 5610 nucleotide dpy-21 transcript, which is both SL1 and SL2 spliced, encodes a novel 1651<br />

amino acid protein. While there is no sequence similarity to known proteins, putative homologues are<br />

present in Drosophila melanogaster and human genomes. Preliminary immunoflourescence experiments<br />

with DPY-21 antibodies indicate that DPY-21 localizes to the nucleus.<br />

268


A SEARCH FOR LETHAL SYNAPTIC FUNCTION MUTANTS<br />

USING A SENSITIZED BACKGROUND<br />

Karen Yook, Erik Jorgensen<br />

<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

Dept. of Biology, University of Utah, Salt Lake City, UT 84112 USA<br />

We are interested in identifying genes essential for neurotransmitter release. However mutations in<br />

genes essential for neurotransmission can result in L1 lethality, as observed with animals homozygous for<br />

null mutations in cha-1, the biosynthetic enzyme for acetylcholine. In the course of characterizing<br />

synaptic function mutants, we noticed that mutations in separate synaptic function loci do not complement<br />

one another when heterozygous, that is, they exhibit nonallelic noncomplementation. In an in-depth<br />

analysis of nonallelic noncomplementation among synaptic function alleles, we demonstrated that<br />

nonallelic noncomplementation occurs between lethal synaptic function alleles and hypomorphic alleles at<br />

other loci (manuscript in preparation). Therefore, in order to uncover synaptic function loci which have<br />

been mutated to lethality, we adopted a nonallelic noncomplementation strategy to screen for mutations<br />

that interact with a hypomorphic allele of unc-13. Specifically, we looked for double heterozygotes (mut/+;<br />

unc-13(n2813)/+) which decrease neurotransmitter release. Although the noncomplementation<br />

interaction between synaptic function loci is robust, we increased the sensitivity of our screening assay<br />

with an additional background mutation in unc-29, a subunit of the acetylcholine receptor. Therefore we<br />

carried out a screen for genes whose functions are effected by mutations in unc-13 and unc-29. Initially,<br />

we screened 2800 EMS mutagenized genomes in this sensitized background and uncovered 117<br />

potential synaptic function loci of which 37 exhibited an uncoordinated phenotype on its own and another<br />

12 exhibited L1 lethality. Characterization of the uncoordinated mutants identified mutant alleles of<br />

unc-26, unc-2, aex-3, ric-4, unc-38 all synaptic function loci known to be involved in neurotransmission.<br />

In addition, we uncovered a novel locus. These results suggest that the sensitized background is a good<br />

strategy for identifying the components of the synaptic transmission machinery. More recently, we used<br />

the ENU mutagen and screened through 3900 genomes. This screen has turned up 134 potential<br />

synaptic function mutations; 22 of these mutations are associated with a lethal phenotype, of which 13<br />

exhibit L1 lethality. We are currently pursuing the identity of the lethal loci.<br />

269


IDENTIFICATION OF DOWNSTREAM TARGET GENES IN<br />

DAF-2 PATHWAY<br />

Hui Yu, Pamela L. Larsen<br />

<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

Molecular Biology Program and Division of Biogerontology, University of Southern California, Los<br />

Angeles, CA 90089<br />

In a search for downstream genes in the daf-2 insulin-like receptor signaling pathway, nine genes were<br />

identified by differential display PCR and verified by Northern analysis to be differentially expressed in<br />

daf-2 mutants compared to N2 animals. The genes discovered have been named dao for daf-2 pathway<br />

over- and under-expressed genes. Three dao genes, dao-1, dao-8 and dao-9, are homologs of an<br />

intracellular transducer FK-506 binding protein. These three genes were down-regulated in daf-2(m41)<br />

animals and this effect in part relies upon daf-16. Among the other six genes, dao-2, dao-3 and dao-4<br />

were positively regulated by daf-2 signaling, while the opposite pattern was observed for dao-5, dao-6<br />

and dao-7. dao-2, dao-4 and dao-6 encode novel proteins. By homology, dao-3 is probably a<br />

methylenetetrahydrofolate dehydrogenase. The predicated DAO-5 protein has many phosphorylation site<br />

repeats and showed 38% identity to xNopp180, a nuclear localization sequence-binding protein shuttling<br />

between the cytoplasm and the nucleus. dao-7 contains a zinc finger region similar to mammalian ZFP36<br />

protein and thereby is a potential transcription factor. Distinct expression patterns and molecular identities<br />

of these new downstream targets in the daf-2 pathway indicates that daf-2 signaling is a complicated<br />

signaling network involved in multiple cellular processes.<br />

270


FATE SPECIFICATION IN MALE P(9-11).P EQUIVALENCE<br />

GROUP<br />

Hui Yu, Paul W. Sternberg<br />

<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

HHMI and Division of Biology, California Institute of Technology, Pasadena, CA 91125<br />

C. <strong>elegans</strong> P(9-11).p cells in male pre-anal ganglion (PAG) equivalence group have three potential fates,<br />

1° , 2° , and 3° . In wild-type males, P10.p and P11.p adopt the 2° and 1° respectively, divide to generate<br />

distinct subsets of cells. P9.p (3° fate), which is undivided, fuses with hyp7. osm-6::gfp is specifically<br />

expressed in two hook neurons HOA and HOB derived from the 2° lineage. To understand the fate<br />

specification in P(9-11).p cells, we constructed a series of strains carrying the osm-6::gfp marker with<br />

mutations in lin-12, lin-15 and mab-5 genes. Instead of normal one pair of GFP expressing cells, lin-12(gf)<br />

mutants showed 2-3 pairs of GFP expression characteristic of the HOA and HOB neurons, indicating a<br />

transformation to the 2° fate in P9.p and P11.p cells. In lin-15(lf) mutants, P9.p could divide and a second<br />

pair of GFP expression was observed anteriorly sometimes because of a P9.p Æ P10.p fate<br />

transformation. The results suggested that the lin-12 activity is sufficient to promote the 2° fate and lin-15,<br />

a negative regulator of let-23 -ras pathway, is required for the 3° fate. The roles of mab-5 and let-23/ras<br />

are under investigation. A genetic screen for other candidates involved in P(9-11).p fate determination is<br />

also in progress.<br />

271


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

LOSS OF A DYNAMIN RELATED PROTEIN MGM-1 CAUSES<br />

EXCESSIVE MITOCHONDRIAL FRAGMENTATION<br />

Mauro Zappaterra, Alexander van der Bliek<br />

Department of Biological Chemistry, UCLA School of Medicine, P.O. Box 951737, Los Angeles, CA<br />

90095-1737<br />

Our lab studies the functions of dynamin and dynamin related proteins. These proteins form a small family<br />

of large GTP-binding proteins. We have recently shown that the C. <strong>elegans</strong> dynamin related protein<br />

DRP-1, is involved in the scission of the mitochondrial outer membrane (1). Since little is known about<br />

mitochondrial division and mitochondrial morphology in animal cells, we are investigating the mechanisms<br />

that regulate these processes.<br />

MGM-1 is another member of the dynamin family present in animal cells. As of now we know that this<br />

protein is present in yeast, C. <strong>elegans</strong>, and humans. Unlike other members of the dynamin family, MGM-1<br />

has an N-terminal mitochondrial leader sequence. MGM-1 is also more closely related to bacterial<br />

dynamin-like proteins than it is to dynamin or DRP-1, suggesting that this protein has followed a different<br />

evolutionary path. We speculate that MGM-1 was introduced into eukaryotic cells by the progenitors of<br />

mitochondria. Expression studies using ß-galactosidase under the control of the mgm-1 promoter show<br />

high levels of expression in intestines, body wall muscles, and neurons. These high levels might reflect<br />

high metabolic rates in those tissues, because the MGM-1 expression pattern is similar to that of another<br />

dynamin-related protein, DRP-1, which is also important for mitochondrial maintenance. Mgm-1 RNAi<br />

drastically slows the growth rate and approximately doubles the life span of C. <strong>elegans</strong>. We show that<br />

excessive fragmentation of mitochondria is induced by RNAi and mgm-1 antisense cDNA. We present<br />

evidence that MGM-1 is localized to the mitochondrial matrix where it might regulate the morphology of<br />

the mitochondrial inner membrane.<br />

1. Labrousse, A.M., Zappaterra, M.D., Rube, D.A., and van der Bliek, A.M. Molecular Cell 4: 815-826.<br />

1999.<br />

272


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

ISOLATION AND PHENOTYPIC ANALYSIS OF SYD-7<br />

Mei Zhen 1,2 , Nikki Alvarez 1 , Yishi Jin 1<br />

1Department of Biology, 327 Sinsheimer Laboratories, University of California, Santa Cruz, CA 95064<br />

2zhen@darwin.ucsc.edu<br />

Using a synaptic vesicle-tagged GFP marker, Punc-25-SNB-1::GFP, that is localized to the presynaptic<br />

terminals of VD and DD motoneurons, we isolated two recessive loss-of-function mutations, ju43 and<br />

ju58, that define the syd-7 locus. In wild-type animals, the Punc-25-SNB-1::GFP marker is expressed as<br />

discrete fluorescent puncta spaced evenly along the ventral and dorsal sides of the animal. In syd-7<br />

mutants some of the fluorescent puncta are absent in various regions, while the remaining puncta appear<br />

normal in morphology and spacing. 90% of ju43 and ju58 animals lose SNB-1::GFP puncta in places<br />

corresponding to the presynaptic regions of VD1 and VD2 neurons. 50% of ju58 animals also lack<br />

SNB-1::GFP puncta in presynaptic regions of different DD neurons. Despite of the defects in GFP marker,<br />

the locomotion of syd-7 animals is indistinguishable from wild-type animals. Absence of SNB-1::GFP<br />

puncta can be caused by defects in either cell fate or neural differentiation. To distinguish these<br />

possibilities we are examining the axonal morphology of D neurons and the expression of cell-type<br />

specific markers in syd-7 animals.<br />

We mapped syd-7 to chromosome V, in a small region overlapped by yDf12 and arDf1. This tentatively<br />

places syd-7 between eat-6 and him-5. Currently we are further fine-mapping syd-7 and trying to rescue<br />

syd-7 with cosmids in the region. We are also in the progress of testing for the genetic interactions<br />

between syd-7 and genes known to be involved in the development of the nervous system.<br />

273


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

A SCREEN TO IDENTIFY GENES THAT REGULATE THE<br />

ACTIVITY OF THE C. ELEGANS GLUTAMATE RECEPTOR<br />

GLR-1.<br />

Yi Zheng, Heng Xie, Pene J. Brockie, Andres V. Maricq<br />

Department of Biology, University of Utah, Salt Lake City, Utah 84112<br />

Efficient synaptic transmission relies on an organized distribution of neurotransmitter receptors.<br />

Activity-dependent changes in synaptic strength are believed to be crucial for information processing and<br />

for the refinement of neuronal network during development. The presence of silent synapses and their<br />

activation indicates rapid and activity-dependent changes in the number of glutamate receptors at<br />

excitatory glutamatergic synapses. However, the molecular mechanisms underlying the regulation of the<br />

density of receptors at synapses are unclear.<br />

We have previously shown that the C. <strong>elegans</strong> glutamate receptor subunit GLR-1 is expressed in the<br />

interneuronal circuitry that is required for normal locomotion. To perturb the function of this circuit, we<br />

engineered into GLR-1 the A/T amino acid change first identified in the d2 glutamate receptor of the<br />

mutant Lurcher mouse. Transgenic worms (akIs9) that express this mutant glutamate receptor show a<br />

striking change in locomotion where they rapidly alternate between forward and backward movement,<br />

presumably secondary to the constitutive depolarization of the command interneurons by the GLR-1(A/T)<br />

channel. In contrast, worms that have a null mutation in GLR-1 (ky176) move about the same as wild-type<br />

animals.<br />

The dramatic difference between ky176 and akIs9 worms provides an opportunity to identify genes that<br />

regulate glutamate receptor density in C.<strong>elegans</strong>. Mutations that decrease the membrane density of<br />

GLR-1(A/T) should suppress the hyper-reversal phenotype of akIs9 worms and make them move more<br />

like ky176 animals. We mutagenized the akIs9 strain and screened ~25,000 haploid genomes for<br />

mutations that suppress the locomotory phenotype. We have isolated 17 candidate mutants that suppress<br />

the hyper-reversal movement and we are in the process of characterizing them. Amongst these mutants,<br />

we expect to find mutations that affect the insertion or stability of glutamate receptors at the synapse.<br />

274


<strong>West</strong> <strong>Coast</strong> <strong>Worm</strong> <strong>Meeting</strong> 2000<br />

A RESOURCE FOR C. ELEGANS MICROARRAYS<br />

Stuart K. Kim, Min Jiang, Kyle Duke<br />

Department of Developmental Biology and Genetics, Stanford University Medical Center, Stanford, CA<br />

We are entering a new age in molecular genetics in which we can use sequence data from genome<br />

projects to dissect cell, developmental and disease pathways more completely and more sensitively than<br />

ever before. One key functional genomics approach is to use DNA microarrays to define changes in gene<br />

expression patterns during development and during the onset of disease. DNA microarrays could be used<br />

to identify genes that are regulated by specific transcription factors, by specific cell signaling pathways, by<br />

expression of homologs of human disease genes in transgenic animals or by addition of various<br />

pharmaceutical drugs.<br />

We are currently producing DNA microarrays that contain every gene in the C. <strong>elegans</strong> genome. Our goal<br />

is to develop C. <strong>elegans</strong> microarray technology, and to make this technology available to all other C.<br />

<strong>elegans</strong> labs. We will provide these microarrays to any C. <strong>elegans</strong> lab by hybridizing their RNA samples<br />

to the full genome microarrays. The results are deposited in the Stanford Microarray Database, and can<br />

be accessed over the web. We have already performed about 200 microarray hybridizations using the full<br />

genome microarrays, in collaboration with 27 different academic C. <strong>elegans</strong> labs. We are also helping to<br />

provide needed tools and reagents so that other labs can establish their own microarraying facilities.<br />

275

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!