04.03.2013 Views

PROGRAM & ABSTRACTS - Wisconsin Union - University of ...

PROGRAM & ABSTRACTS - Wisconsin Union - University of ...

PROGRAM & ABSTRACTS - Wisconsin Union - University of ...

SHOW MORE
SHOW LESS

Create successful ePaper yourself

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

Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

<strong>PROGRAM</strong> & <strong>ABSTRACTS</strong><br />

Aging, Metabolism, Stress, Pathogenesis, and<br />

Small RNAs in C. elegans<br />

Thursday, July 12 – Sunday, July 15, 2012<br />

<strong>University</strong> <strong>of</strong> <strong>Wisconsin</strong>-Madison<br />

<strong>Union</strong> South/Memorial <strong>Union</strong><br />

Madison, <strong>Wisconsin</strong> 53706<br />

Scientific Organizers<br />

Malene Hansen, Sanford-Burnham Medical Research Institute, La Jolla<br />

Emily Troemel, <strong>University</strong> <strong>of</strong> California, San Diego<br />

Organizing Committee<br />

Sean Curran, <strong>University</strong> <strong>of</strong> Southern California, Los Angeles<br />

Nils Færgeman, <strong>University</strong> <strong>of</strong> Southern Denmark, Odense, Denmark<br />

Danielle Garsin, <strong>University</strong> <strong>of</strong> Texas-Houston Medical School<br />

David Gems, <strong>University</strong> College London, United Kingdom<br />

Ao-Lin Hsu, <strong>University</strong> <strong>of</strong> Michigan, Ann Arbor<br />

Javier Irazoqui, Massachusetts General Hospital, Harvard Medical School<br />

Antony Jose, <strong>University</strong> <strong>of</strong> Maryland, College Park<br />

Ho Yi Mak, Stowers Institute for Medical Research, Kansas City, Missouri<br />

Dana Miller, <strong>University</strong> <strong>of</strong> Washington, Seattle<br />

i


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

ii<br />

www.ellisonfoundation.org<br />

www.labexpress.com<br />

www.knudra.com<br />

SPONSORS<br />

ACKNOWLEDGEMENTS<br />

www.glennfoundation.org<br />

www.eppendorfna.com<br />

www.unionbio.com<br />

All sponsoring companies and <strong>University</strong> <strong>of</strong> <strong>Wisconsin</strong> Memorial <strong>Union</strong> Conference Services.<br />

Cover Description: Front cover image and design courtesy <strong>of</strong> Malina A. Bakowski (Troemel lab).<br />

Cover represents the main themes <strong>of</strong> this C. elegans Topic Meeting.<br />

For emergencies call (608) 890-1077 or stop by conference headquarters in <strong>Union</strong> South (Varsity Lounge)


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

Conference Program<br />

Thursday, July 12 th<br />

12 Noon – 7:00 pm Registration Check-In <strong>Union</strong> South, Varsity Lounge<br />

5:00 – 7:15 pm Opening Reception <strong>Union</strong> South, Varsity Hall<br />

7:15 pm Welcome and Opening Remarks <strong>Union</strong> South, Marquee<br />

7:30 – 10:30 pm Session 1 (Abstracts 1 – 5) <strong>Union</strong> South, Marquee<br />

Chairs: Danielle Garsin and Ao-Lin Hsu<br />

7:30 pm Keynote Speaker<br />

Jonathan Ewbank<br />

Building an integrated view <strong>of</strong> anti-fungal innate immunity<br />

8:30 – 9:00 pm Refreshment Break <strong>Union</strong> South, Varsity Hall<br />

9:00 pm Filipe Cabreiro<br />

Metformin increases C. elegans lifespan by altering E. coli<br />

folate metabolism<br />

(Lab: Gems)<br />

9:15 pm Kathleen Dumas (Lab: Hu)<br />

The C. elegans Dosage Compensation Protein DPY-21<br />

Regulates Dauer Arrest<br />

9:30 pm Anat Ben-Zvi (Lab: Ben-Zvi)<br />

Germline Stem Cells Regulate Somatic Proteostasis During<br />

Caenorhabditis elegans Adulthood<br />

9:45 pm L. Ryan Baugh (Lab: Baugh)<br />

Transgenerational Effects <strong>of</strong> Starvation on Development,<br />

Reproduction and Lifespan: Bet-hedging on an Epigenetic<br />

Fitness Trade-<strong>of</strong>f<br />

Friday, July 13 th<br />

7:00 – 8:30 am Breakfast <strong>Union</strong> South, Varsity Hall<br />

8:00 am – 7:30 pm Registration/Questions <strong>Union</strong> South, Varsity Lounge<br />

8:00 am – 7:30 pm Poster Set-up Memorial <strong>Union</strong>, Great Hall/Reception Room<br />

8:30 am – 12:30 pm Session 2 (Abstracts 6 – 16) <strong>Union</strong> South, Marquee<br />

Chairs: Dana Miller and Matthew Gill<br />

8:30 am Plenary Speaker<br />

Siu Sylvia Lee<br />

Transcriptional Network Key to Longevity Determination<br />

and Stress Regulation<br />

iii


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

9:00 am Xiaoyan Guo (Lab: Garcia)<br />

SIR-2.1, an HDAC, is Required To Maintain The Male Mating<br />

Potency In C. elegans<br />

9:15 am Geert Depuydt (Lab: Braeckman)<br />

LC-MS Proteomics Analysis Reveals Both Common and<br />

Unique Changes in the Proteome <strong>of</strong> Insulin/IGF-1 Receptor<br />

Mutant and Dietary-Restricted C. elegans<br />

9:30 am Carl Franz (Lab: Wang)<br />

Characterization <strong>of</strong> Caenorhabditis Nematode Infecting<br />

Viruses<br />

9:45 am Alexandre de Lencastre (Lab: Slack)<br />

Functions <strong>of</strong> MicroRNAs During Aging<br />

10:00 am T. Keith Blackwell (Lab: Blackwell)<br />

Integration <strong>of</strong> the Unfolded Protein and Oxidative Stress<br />

Responses through SKN-1/Nrf<br />

10:15 – 10:45 am Refreshment Break <strong>Union</strong> South, Varsity Hall<br />

10:45 am Plenary Speaker <strong>Union</strong> South, Marquee<br />

Matt Kaeberlein<br />

Oxygen, Temperature, and Food: New Insights into the Role<br />

<strong>of</strong> Environment in C. elegans Aging<br />

11:15 am Louis Lapierre (Lab: Hansen)<br />

The Transcription Factor HLH-30/TFEB Regulates Autophagy<br />

and Modulates Longevity in C. elegans<br />

11:30 am Orane Visvikis (Lab: Irazoqui)<br />

HLH-30 Is a New Transcription Factor involved in C. elegans<br />

Host Response Triggered by S. aureus Infection<br />

11:45 am Nils Færgeman (Lab: Færgeman)<br />

Quantitative Proteomics Identifies SBP-1/SREBP as a General<br />

Transcriptional Regulator <strong>of</strong> Metabolic Pathways in C. elegans<br />

12:00 pm David Hall (Lab: Hall)<br />

Pathology <strong>of</strong> the Aging Gonad Viewed By TEM<br />

12:15 – 1:45 pm Luncheon Buffet <strong>Union</strong> South, Varsity Hall<br />

1:45 – 5:30 pm Session 3 (Abstracts 17 – 24) <strong>Union</strong> South, Marquee<br />

Session chairs: Ho Yi Mak and William Mair<br />

1:45 pm Plenary Speaker<br />

Kaveh Ashrafi<br />

New Insights Into Understanding the Regulation <strong>of</strong> Fat and<br />

Feeding<br />

2:15 pm Thomas Heimbucher (Lab: Dillin)<br />

Identification <strong>of</strong> DAF-16 Co-regulators with Tandem Affinity<br />

Purification and MudPIT<br />

iv


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

2:30 pm Emilie Demoinet (Lab: Roy)<br />

AMPK Regulates a Novel UPR-like Response to Mediate<br />

Survival During Nutrient Stress in C. elegans<br />

2:45 pm Panel Discussion <strong>Union</strong> South, Marquee<br />

Looking Ahead to Future Research Directions<br />

3:30 – 4:00 pm Refreshment Break <strong>Union</strong> South, Varsity Hall<br />

4:00 pm Plenary Speaker <strong>Union</strong> South, Marquee<br />

Coleen Murphy<br />

Pathways that regulate Reproductive Aging and Longevity<br />

4:30 pm Donha Park (Lab: Taubert)<br />

A Novel Role <strong>of</strong> TGF-β Pathway in Dietary Restriction<br />

Induced Longevity<br />

4:45 pm Ju-Ling Liu (Lab: Hekimi)<br />

Mitochondrial Oxidative Stress in C. elegans Alters a Pathway<br />

with Strong Similarities to that <strong>of</strong> Bile Acid Biosynthesis and<br />

Secretion in Vertebrates<br />

5:00 pm Justine Melo (Lab: Ruvkun)<br />

Surveillance <strong>of</strong> Core Cellular Processes as a Strategy for<br />

Xenobiotic and Pathogen Recognition<br />

5:30 – 7:30 pm Dinner Memorial <strong>Union</strong>, Tripp Commons<br />

7:30 – 9:30 pm Poster Session (Odd Numbered Posters) Memorial <strong>Union</strong>, 4th Floor<br />

Great Hall/Reception Room<br />

Aging 51 - 77<br />

Metabolism 79 - 93, 171<br />

Stress 95 - 131<br />

Pathogenesis 133 - 161, 169<br />

Small RNAs 61, 163 - 167<br />

Saturday, July 14 th<br />

7:00 – 8:30 am Breakfast <strong>Union</strong> South, Varsity Hall<br />

8:00 am – 7:30 pm Registration/Questions <strong>Union</strong> South, Varsity Lounge<br />

8:30 am – 12:30 pm Session 4 (Abstracts 25 – 35) <strong>Union</strong> South, Marquee<br />

Session chairs: David Gems and Stefan Taubert<br />

8:30 am Plenary Speaker <strong>Union</strong> South, Marquee<br />

Amy Pasquinelli<br />

Pinning Down MicroRNA Targets In Vivo<br />

9:00 am Nadia Storm (Lab: Antebi)<br />

Enhancing N-glycosylation Through Activation <strong>of</strong> the<br />

Hexosamine Pathway Improves ER Protein Folding Capacity<br />

and Slows Ageing<br />

v


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

9:15 am Yutao Chen (Lab: Baugh)<br />

ins-5 and ins-6 Promote Post-embryonic Development in<br />

Response to Feeding<br />

9:30 am Aimee Kao (Lab: Kenyon/Kao)<br />

A New Class <strong>of</strong> Stress-Resistance Genes<br />

9:45 am Alexander Soukas (Lab: Ruvkun/Soukas)<br />

Genetic Regulation <strong>of</strong> Age Pigment and Nile Red<br />

Accumulation in the Lysosome-Related Organelle<br />

10:00 am David Gems (Lab: Gems)<br />

A Necrotic Cascade Accelerates Stress-Induced Death and<br />

Causes a Burst <strong>of</strong> Anthranilic Acid Fluorescence in C. elegans<br />

10:15 – 10:45 am Refreshment Break <strong>Union</strong> South, Varsity Hall<br />

10:45 am Plenary Speaker <strong>Union</strong> South, Marquee<br />

Marie-Anne Felix<br />

The Real Life <strong>of</strong> Caenorhabditis and Their Natural Pathogens<br />

11:15 am Brian Onken (Lab: Driscoll)<br />

Insulin Signaling and Dietary Restriction Differentially<br />

Regulate Glucose Metabolism to Impact C. elegans<br />

Healthspan<br />

11:30 am Suzannah Szumowski (Lab: Troemel)<br />

Exit <strong>of</strong> the Intracellular Pathogen Nematocida parisii from C.<br />

elegans Intestinal Cells<br />

11:45 pm Amir Sapir (Lab: Broday)<br />

ULP-4 SUMO protease Regulates Mitochondria Homeostasis<br />

During C. elegans Development and Mitochondrial Stress<br />

12:00 pm Jeff Simske (Lab: Simske)<br />

The Diet <strong>of</strong> Worms: Expression <strong>of</strong> the agl-1 (Glycogen-<br />

Debranching Enzyme) Embryonic Arrest Phenotype Depends<br />

on Maternal Diet<br />

12:15 – 1:45 pm Luncheon Buffet <strong>Union</strong> South, Varsity Hall<br />

1:45 – 5:30 pm Session 5 (Abstracts 36 – 45) <strong>Union</strong> South, Marquee<br />

Chairs: Arjumand Ghazi and Nils Færgeman<br />

1:45 pm Plenary Speaker <strong>Union</strong> South, Marquee<br />

Jennifer Watts<br />

Fatty Acid Desaturase Activity Regulates Lipid Droplet Size<br />

2:00 pm Nicholas Stroustrup (Lab: Fontana)<br />

The Lifespan Machine: A Scalable, Automated Microscope<br />

Increases the Throughput and Statistical Quality <strong>of</strong><br />

Nematode Lifespan and Stress Resistance Assays<br />

2:15 am Parag Mahanti (Lab: Schroeder)<br />

Novel Endogenous Ligands <strong>of</strong> DAF-12 Nuclear Hormone<br />

Receptor Revealed by Comparative Metabolomics<br />

vi


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

2:30 pm Cole Haynes (Lab: Haynes)<br />

Direct Detection <strong>of</strong> Mitochondrial Dysfunction by the<br />

Transcription Factor ATFS-1<br />

2:45 pm Michael Shapira (Lab: Shapira)<br />

Dissecting Opposing Age-dependent Contributions <strong>of</strong> JNK<br />

Signaling to Stress Resistance<br />

3:15 – 3:45 pm Refreshment Break <strong>Union</strong> South, Varsity Hall<br />

3:45 pm Plenary Speaker <strong>Union</strong> South, Marquee<br />

Patrick Hu<br />

A Genetic Screen for Novel DAF-16/FoxO Regulators<br />

4:15 pm Markus Kunzler (Lab: Aebi)<br />

Interplay <strong>of</strong> Foreign and Endogenous Lectins in Innate<br />

Immunity <strong>of</strong> Caenorhabditis elegans<br />

4:30 pm Chun-Ling Sun (Lab: Crowder)<br />

Cell-Specific Hypoxic Injury in C. elegans: Modeling the<br />

Ischemic Penumbra<br />

4:45 pm Antony Jose (Lab: Jose)<br />

Movement <strong>of</strong> RNA Silencing Between C. elegans Cells<br />

5:00 pm Abigail Cabunoc (Lab: Stein)<br />

WormBase 2012: Website Redesign<br />

5:30 – 7:30 pm Dinner Memorial <strong>Union</strong>, Inn <strong>Wisconsin</strong><br />

7:30 – 9:30 pm Poster Session (Even Numbered Posters) Memorial <strong>Union</strong>, 4th Floor<br />

Great Hall/Reception Room<br />

Aging 52 - 76<br />

Metabolism 78 - 92<br />

Stress 94 - 130, 170<br />

Pathogenesis 132 - 160<br />

Small RNAs 162 - 168<br />

9:30 pm – 12 Midnight Party and Dance Memorial <strong>Union</strong>, Tripp Commons<br />

Sunday, July 15 th<br />

7:00 – 8:30 am Breakfast <strong>Union</strong> South, Varsity Hall<br />

8:00 am – 12 Noon Registration/Questions <strong>Union</strong> South, Varsity Lounge<br />

8:30 – 10:30 am Session 6 (Abstracts 46 – 50) <strong>Union</strong> South, Marquee<br />

Session Chairs: Antony Jose and Javier Irazoqui<br />

8:30 am Plenary Speaker <strong>Union</strong> South, Marquee<br />

Dennis Kim<br />

Dynamic Neuroendocrine Signaling in C. elegans Behavioral<br />

Responses to Pathogenic Bacteria<br />

vii


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

9:00 am Rebecca Taylor (Lab: Dillin)<br />

The UPRER is a Cell Non-Autonomous Regulator <strong>of</strong> Stress<br />

Resistance and Longevity<br />

9:15 am Amy Walker (Lab: Naar/Walker)<br />

Interactions Between 1-carbon Cycle and Lipid Metabolism<br />

in C. elegans<br />

9:30 am Horishi Suzuki (Lab: Suzuki)<br />

Novel Approach in Oxidative Stress Study by Targeted ROS<br />

Generation Using a Photosensitizer SuperNova in C. elegans<br />

9:45 am Plenary Speaker <strong>Union</strong> South, Marquee<br />

Gordon Lithgow<br />

Natural Products that Suppress Protein Aggregation and<br />

Slow Aging<br />

10:15 am Closing remarks<br />

10:30 am Refreshments and Goodbye <strong>Union</strong> South, Varsity Hall<br />

viii


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

TABLE OF CONTENTS<br />

Thursday, July 12, 2012 - 7:30–10:30 pm<br />

Session #1 - <strong>Union</strong> South, Marquee<br />

Abstracts 1–5<br />

Chairs: Danielle Garsin and Ao-Lin Hsu<br />

1 Keynote: Building an integrated view <strong>of</strong> anti-fungal innate immunity<br />

Jonathan Ewbank (Keynote Speaker)<br />

2 Metformin increases C. elegans lifespan by altering E. coli folate metabolism<br />

Filipe Cabreiro, Catherine Au, Kit-Yi Leung, Nick Greene, David Gems<br />

3 The C. elegans Dosage Compensation Protein DPY-21 Regulates Dauer<br />

Arrest<br />

Kathleen Dumas, Stephane Flibotte, Don Moerman, Patrick Hu<br />

4 Germline Stem Cells Regulate Somatic Proteostasis During Caenorhabditis<br />

elegans Adulthood<br />

Netta Shemesh, Nadav Shai, Anat Ben-Zvi<br />

5 Transgenerational Effects <strong>of</strong> Starvation on Development, Reproduction and<br />

Lifespan: Bet-hedging on an Epigenetic Fitness Trade-<strong>of</strong>f<br />

Meghan Jobson, L. Ryan Baugh<br />

Friday, July 13, 2012 - 8:30 am–12:30 pm<br />

Session #2 - <strong>Union</strong> South, Marquee<br />

Abstracts 6–16<br />

Chairs: Dana Miller and Matthew Gill<br />

6 Transcriptional Network Key to Longevity Determination and Stress<br />

Regulation<br />

Siu Sylvia Lee (Plenary Speaker)<br />

7 SIR-2.1, an HDAC, is Required To Maintain The Male Mating Potency In C.<br />

elegans<br />

Xiaoyan Guo, Luis rene Garcia<br />

8 LC-MS Proteomics Analysis Reveals Both Common and Unique Changes in<br />

the Proteome <strong>of</strong> Insulin/IGF-1 Receptor Mutant and Dietary Restricted C.<br />

elegans<br />

Geert Depuydt, Fang Xie, Vladislav Petyuk, Heather Brewer, Arne Smolders, Ineke Dhondt, Nilesh<br />

Shanmugam, David Camp II, Richard Smith, Bart Braeckman<br />

9 Characterization <strong>of</strong> Caenorhabditis Nematode Infecting Viruses<br />

Carl Franz, Marie-Anne Felix, Yanfang Jiang, Guoyan Zhao, Hilary Renshaw, Guang Wu, David<br />

Wang<br />

ix


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

10 Functions <strong>of</strong> MicroRNAs During Aging<br />

Alexandre de Lencastre, Frank Slack<br />

11 Integration <strong>of</strong> the Unfolded Protein and Oxidative Stress Responses through<br />

SKN-1/Nrf<br />

Kira Glover-Cutter, Stephanie Lin, T. Keith Blackwell<br />

12 Oxygen, temperature, and food: new insights into the role <strong>of</strong> environment in<br />

C. elegans aging<br />

Matt Kaeberlein (Plenary Speaker)<br />

13 The transcription factor HLH-30/TFEB regulates autophagy and modulates<br />

longevity in C. elegans<br />

Louis Lapierre, Malene Hansen<br />

14 HLH-30 is a new Transcription Factor involved in C. elegans Host Response<br />

triggered by S. aureus Infection<br />

Orane Visvikis, Nnamdi Ihuegbu, Lyly Luhachack, Amanda Wollenberg, Anna-Maria Alves, Gary<br />

Stormo, Javier Irazoqui<br />

15 Quantitative Proteomics Identifies SBP-1/SREBP as a General Transcriptional<br />

Regulator <strong>of</strong> Metabolic Pathways in C. elegans<br />

Julius Fredens, Kasper Engholm-Keller, Jakob Moeller-Jensen, Martin Roessel Larsen, Nils<br />

Færgeman<br />

16 Pathology Of The Aging Gonad Viewed By TEM<br />

David Hall, Angela Jevince, Ken Nguyen, Laura Herndon<br />

x<br />

Friday, July 13, 2012 - 1:45–5:30 pm<br />

Session #3 - <strong>Union</strong> South, Marquee<br />

Abstracts 17–24<br />

Chairs: Ho Yi Mak and William Mair<br />

17 New Insights Into Understanding The Regulation Of Fat And Feeding<br />

Kaveh Ashrafi (Plenary Speaker), Katherine Cunningham, Aude Bouagnon, Liron Noiman<br />

18 Identification <strong>of</strong> DAF-16 Co-regulators with Tandem Affinity Purification and<br />

MudPIT<br />

Thomas Heimbucher, Zheng Liu, Andrea Carrano, Carine Bossard, Bryan Fonslow, Jonathan Yates,<br />

Tony Hunter, Andrew Dillin<br />

19 AMPK regulates a novel UPR-like response to mediate survival during<br />

nutrient stress in C. elegans<br />

Emilie Demoinet, Julie Mantovani, Richard Roy<br />

20 Pathways that regulate Reproductive Aging and Longevity<br />

Coleen Murphy (Plenary Speaker)<br />

21 A Novel Role <strong>of</strong> TGF-β Pathway in Dietary Restriction Induced Longevity<br />

Donha Park, Dara Lo, Charles Yang, Donald Riddle, Stefan Taubert


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

22 Mitochondrial Oxidative Stress in C. elegans Alters a Pathway with Strong<br />

Similarities to that <strong>of</strong> Bile Acid Biosynthesis and Secretion in Vertebrates<br />

Ju-Ling Liu, David Desjardins, Robyn Branicky, Luis B. Agellon, Siegfried Hekimi<br />

23 Surveillance <strong>of</strong> Core Cellular Processes as a Strategy for Xenobiotic and<br />

Pathogen Recognition<br />

Justine Melo, Gary Ruvkun<br />

24 Abstract withdrawn<br />

Saturday, July 14, 2012 - 8:30 am–12:30 pm<br />

Session #4 - <strong>Union</strong> South, Marquee<br />

Abstracts 25–35<br />

Chairs: David Gems and Stefan Taubert<br />

25 Pinning Down MicroRNA Targets In Vivo<br />

Amy Pasquinelli (Plenary Speaker)<br />

26 Enhancing N-glycosylation Through Activation <strong>of</strong> the Hexosamine Pathway<br />

Improves ER Protein Folding Capacity and Slows Ageing.<br />

Nadia Storm, Martin Denzel, Adam Antebi<br />

27 ins-5 and ins-6 Promote Post-embryonic Development in Response to<br />

Feeding<br />

Yutao Chen, Ryan Baugh<br />

28 A New Class <strong>of</strong> Stress Resistance Genes<br />

Aimee Kao, Ayumi Nakamura, Meredith Judy, Anne Huang, Cynthia Kenyon<br />

29 Genetic Regulation <strong>of</strong> Age Pigment and Nile Red Accumulation in the<br />

Lysosome Related Organelle<br />

Alexander Soukas, Christopher Carr, Gary Ruvkun<br />

30 A Necrotic Cascade Accelerates Stress-Induced Death and Causes a Burst <strong>of</strong><br />

Anthranilic Acid Fluorescence in C. elegans<br />

Cassandra Coburn, Parag Mahanti, Abraham Mandel, Filip Matthijssens, Bart Braeckman, Frank<br />

Schroeder, David Gems<br />

31 The real life <strong>of</strong> Caenorhabditis and their natural pathogens<br />

Marie-Anne Felix, (Plenary Speaker) Fabien Duveau, Tony Belicard<br />

32 Insulin Signaling and Dietary Restriction Differentially Regulate Glucose<br />

Metabolism to Impact C. elegans Healthspan<br />

Brian Onken, Monica Driscoll<br />

33 Exit <strong>of</strong> the Intracellular Pathogen Nematocida parisii from C. elegans Intestinal<br />

Cells<br />

Suzannah Szumowski, Kathleen Estes, Margery Smelkinson, Emily Troemel<br />

34 ULP-4 SUMO protease regulates mitochondria homeostasis during C. elegans<br />

development and mitochondrial stress<br />

Amir Sapir, Assaf Tsur, Thijs Thijs Koorman, Mike Boxem, Paul Sternberg, Limor Broday<br />

xi


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

35 The Diet <strong>of</strong> Worms: Expression <strong>of</strong> the agl-1 (Glycogen Debranching Enzyme)<br />

Embryonic Arrest Phenotype Depends on Maternal Diet<br />

Jeff Simske<br />

xii<br />

Saturday, July 14, 2012 - 1:45–5:30 pm<br />

Session #5 - <strong>Union</strong> South, Marquee<br />

Abstracts 36–45<br />

Chairs: Arjumand Ghazi and Nils Færgeman<br />

36 Fatty acid desaturase activity regulates lipid droplet size<br />

Jennifer Watts (Plenary Speaker)<br />

37 The Lifespan Machine: A scalable, automated microscope increases the<br />

throughput and statistical quality <strong>of</strong> nematode lifespan and stress resistance<br />

assays<br />

Nicholas Stroustrup, Javier Apfeld, Walter Fontana<br />

38 Novel endogenous ligands <strong>of</strong> DAF-12 nuclear hormone receptor revealed by<br />

comparative metabolomics<br />

Parag Mahanti, Neelanjan Bose, Axel Bethke, Joshua Judkins, Joshua Wollam, Kathleen Dumas,<br />

Anna Zimmerman, Patrick Hu, Adam Antebi, Frank Schroeder<br />

39 Direct Detection <strong>of</strong> Mitochondrial Dysfunction by the Transcription Factor<br />

ATFS-1<br />

Cole Haynes<br />

40 Dissecting opposing age-dependent contributions <strong>of</strong> JNK signaling to stress<br />

resistance<br />

Kwame Twumasi-Boateng, Kuang-Hui Lee, Ali Salehpour, Michael Shapira<br />

41 A Genetic Screen for Novel DAF-16/FoxO Regulators<br />

Patrick Hu (Plenary Speaker), Kathleen Dumas, Albert Chen, Hung-Jen Shih, Chunfang Guo<br />

42 Interplay <strong>of</strong> Foreign and Endogenous Lectins in Innate Immunity <strong>of</strong><br />

Caenorhabditis elegans<br />

Therese Wohlschlager, Alex Butschi, Katrin Stutz, Iain Wilson, Michael Hengartner, Markus Aebi,<br />

Markus Kunzler<br />

43 Cell-Specific Hypoxic Injury in C. elegans: Modeling the Ischemic Penumbra<br />

Chun-Ling Sun, Euysoo Kim, C. Michael Crowder<br />

44 Movement <strong>of</strong> RNA Silencing Between C. elegans Cells<br />

Antony Jose, Hai Le, Snusha Ravikumar, Sindhuja Devanapally<br />

45 WormBase 2012: Website Redesign<br />

Abigail Cabunoc, Norie de la Cruz, Adrian Duong, Maher Kassim, Xiaoqi Shi, Todd Harris, Lincoln<br />

Stein


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

Sunday, July 15, 2012 - 1:45–5:30 pm<br />

Session #6 - <strong>Union</strong> South, Marquee<br />

Abstracts 46–50<br />

Chairs: Antony Jose and Javier Irazoqui<br />

46 Dynamic Neuroendocrine Signaling in C. elegans Behavioral Responses to<br />

Pathogenic Bacteria<br />

Dennis Kim (Plenary Speaker)<br />

47 The UPRER is a Cell Non-Autonomous Regulator <strong>of</strong> Stress Resistance and<br />

Longevity<br />

Rebecca Taylor, Andrew Dillin<br />

48 Interactions between 1-carbon cycle and lipid metabolism in C. elegans<br />

Amy Walker, Rene Jacobs, Jenny Watts, Veerle Rottiers, Anders Naar<br />

49 Novel approach in oxidative stress study by targeted ROS generation using a<br />

photosensitizer SuperNova in C. elegans<br />

Hiroshi SUZUKI, Donald Fu, Ippei Kotera, Po-An SU<br />

50 Natural Products that Suppress Protein Aggregation and Slow Aging<br />

Silvestre Alavez, Pedro Rodriguez, Maithili C. Vantipalli, David J. S. Zucker, Ida M. Klang Milena<br />

Price, Karla Mark, Gordon J. Lithgow (Plenary Speaker)<br />

xiii


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

xiv<br />

Friday & Saturday, July 13-14, 2012 - 7:30–9:30 pm<br />

Poster Sessions - Memorial <strong>Union</strong>, Great Hall/Reception Room (4th floor)<br />

Odd Numbered on Friday, Even Numbered on Saturday<br />

Topic: Aging<br />

Abstracts 51-77<br />

51 Expression-level Polymorphism for the Rec-8 Gene Underlies a Quantitative<br />

Trait Locus Governing Lifespan, Multiple Stress Resistances, and<br />

X-Chromosome Nondisjunction<br />

Srinivas Ayyadevara, Cagdas Tazearslan, Ramani Alla, Robert Shmookler Reis<br />

52 Effects <strong>of</strong> Dietary Restriction by Axenic Medium on Mitochondrial Function<br />

in Caenorhabditis elegans.<br />

Natascha Castelein, Berhanu Kassa, Bart Braeckman<br />

53 SGK-1 Extends Lifespan by Activating DAF-16/FoxO<br />

Albert Chen, Chunfang Guo, Kathleen Dumas, Travis Williams, Sawako Yoshina, Shohei Mitani,<br />

Kaveh Ashrafi, Patrick Hu<br />

54 Proteomic Changes Elicited by Metformin Treatment<br />

Wouter De Haes, Roel Van Assche, Steven Haenen, Bart Braeckman, Liliane Scho<strong>of</strong>s<br />

55 The p120RasGAP Family Member GAP-3 is a Novel Regulator <strong>of</strong> Dauer<br />

Arrest and Longevity<br />

Kathleen Dumas, Stephane Flibotte, Don Moerman, Patrick Hu<br />

56 Investigating the tissue-specific requirements for autophagy in C. elegans<br />

longevity mutants<br />

Sara Gelino, Malene Hansen<br />

57 Tumor suppressors and longevity in C. elegans<br />

Hakam Gharbi, Francesca Fabretti, Puneet Bharill, Bernhard Schermer, Thomas Benzing, Roman<br />

Ulrich Mueller<br />

58 Identification <strong>of</strong> a Novel Bacterial Species That Extends the Lifespan <strong>of</strong><br />

Caenorhabditis elegans<br />

Junhyeok Go, Kang-Mu Lee, Sang Sun Yoon<br />

59 The Role <strong>of</strong> Thioredoxin-1 (TRX-1) in Caenorhabditis elegans Aging<br />

Maria Gonzalez-Barrios, Juan Carlos Fierro-Gonzalez, Manuel Munoz, Peter Swoboda, Antonio<br />

Miranda-Vizuete<br />

60 Aging is a Determinant in Anoxia Stress Tolerance in Caenorhabditis elegans<br />

Jo Goy, Pamela Padilla<br />

62 Translational Effect <strong>of</strong> Hydrogen Sulfide on C. elegans<br />

Joseph Horsman, Dana Miller<br />

63 Roles <strong>of</strong> Specific daf-16/FoxO Is<strong>of</strong>orms in Dauer Regulation and Lifespan<br />

Control<br />

Chunfang Guo, Travis Williams, Kathleen Dumas, Sawako Yoshina, Shohei Mitani, Patrick Hu


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

64 Quantitative In Vivo Redox Sensors Uncover Oxidative Stress as an Early<br />

Event in Life<br />

Daniela Knoefler, Maike Thamsen, Martin Koniczek, Nicholas Niemuth, Ann-Kristin Diederich,<br />

Ursula Jakob<br />

65 Genes Acting Downstream <strong>of</strong> Sensory Neurons to Influence Longevity, Dauer<br />

Formation and Pathogen Responses in C. elegans<br />

Marta Gaglia, Dae-Eun Jeong, Eun-A Ryu, Dongyeop Lee, Seung-Jae Lee<br />

66 Age-dependent Alteration <strong>of</strong> Major Large Intestinal Granules in<br />

Caenorhabditis elegans<br />

Kenji Nishikori, Takahiro Tanji, Eisuke Kuroda, Yuki Ueda, Hirohisa Shiraishi, Ayako Ohashi-<br />

Kobayashi<br />

67 Electrolyzed-reduced water confers increased resistance to environmental<br />

stresses and longevity in C. elegans<br />

Seul-Ki Park, Soon-Young Lee, Sang-Kyu Park<br />

68 A daf-16b-specific Mutation Extends C. elegans Lifespan<br />

Andy Polzin, Chunfang Guo, Sawako Yoshina, Shohei Mitani, Patrick Hu<br />

69 Molecular Mechanisms <strong>of</strong> C. elegans Germline Stem Cell Aging<br />

Zhao Qin, E. Jane Albert Hubbard<br />

70 Nutritional Deprivation in the Late Larval Stages <strong>of</strong> C. elegans Induces<br />

Developmental Diapause at Precise Checkpoints<br />

Adam Schindler, L Baugh, David Sherwood<br />

71 Shared Targets <strong>of</strong> TGF-β and Insulin/IGF-1 Signaling Regulate Reproductive<br />

Aging through Mechanisms Distinct from Somatic Aging Regulation<br />

Shijing Luo, Cheng Shi, Jasmine Ashraf, Coleen Murphy<br />

72 The Role <strong>of</strong> FAHD1 in the Aging <strong>of</strong> Caenorhabditis elegans<br />

Andrea Taferner, Haymo Pircher, Lucia Micutkova, Nektarios Tavernarakis, Pidder Jansen-Duerr<br />

73 Physical Interaction <strong>of</strong> Half ABC Transporters HAF-4 and HAF-9, Which Are<br />

Required for the Biogenesis <strong>of</strong> the Intestinal Lysosome-related Organelles in<br />

Caenorhabditis elegans<br />

Takahiro Tanji, Kenji Nishikori, Hirohisa Shiraishi, Masatomo Maeda, Ayako Ohashi-Kobayashi<br />

74 Neurite Sprouting and Synapse Deterioration in the Aging C. elegans<br />

Nervous System<br />

Marton Toth, Ilija Melentijevic, Leena Shah, Aatish Bhatia, Kevin Lu, Amish Talwar, Haaris Naji,<br />

Carolina Ibanez-Ventoso, Piya Ghose, Angelina Jevince, Laura Herndon, Gyan Bhanot, Christopher<br />

Rongo, David Hall, Jian Xue, Monica Driscoll<br />

75 Identification <strong>of</strong> the Core Chaperone Network that Modulates Proteostasis<br />

in C. elegans<br />

Cindy Voisine, Kai Orton, Richard Morimoto<br />

76 The Conserved MicroRNA-80 Modulates C. elegans Longevity through<br />

Dietary Restriction<br />

Mehul Vora, Mitalie Shah, Jian Xue, Monica Driscoll<br />

xv


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

77 Supplemental cellular protection by a carotenoid astaxanthin extends<br />

lifespan via ins/IGF-1 signaling in C. elegans<br />

Sumino Yanase<br />

xvi<br />

Topic: Metabolism<br />

Abstracts 78-93<br />

78 The Role <strong>of</strong> Thioredoxin Reductase in Inorganic Selenium Metabolism in C.<br />

elegans<br />

Christopher Boehler, Roger Sunde<br />

79 Regulation <strong>of</strong> lipid storage and secretion: Genetics and Molecular Analysis<br />

Christopher Brey, Jun Zhang, Sanya Hashmi, Randy Gaugler, Sarwar Hashmi<br />

80 Investigation <strong>of</strong> Satiety Quiescence Signaling Using Automated Locomotion<br />

Assay and Hidden Markov Model Analysis to Identify Worm Behavioral States<br />

Thomas Gallagher, Leon Avery, Young-Jai You<br />

81 Circadian rhythms in metabolism, stress tolerance and pathogenesis: lessons<br />

from Caenorhabditis elegans.<br />

Maria Goya, Andres Romanowski, Maria Migliori, Sergio Simonetta, Diego Golombek<br />

82 Ceramide signal mediates antipsychotic drug-induced developmental delay<br />

and lethality in C. elegans.<br />

Limin Hao, Bruce Cohen, Edgar Buttner<br />

83 Functional Characterization <strong>of</strong> C. elegans N-acylethanolamine Biosynthetic<br />

Enzymes<br />

Neale Harrison, Ifedayo Victor Ogungbe, Matthew Gill<br />

84 The Mediator Subunit MED15/MDT-15 is a Conserved Transcriptional Coregulator<br />

in Lipid Homeostasis<br />

Nicole Hou, Donha Park, Stefan Taubert<br />

85 L1 longevity is determined by metabolic rate and that AMPK as a master<br />

regulator <strong>of</strong> metabolism controls<br />

Inhwan Lee, Amber Hendrix, Jennifer Yoshimoto, Jeongho Kim, Young-Jai You<br />

86 Proteomic Study and Marker Protein Identification <strong>of</strong> Caenorhabditis<br />

elegans Lipid Droplets<br />

Pingsheng Liu, Peng Zhang, Huimin Na<br />

87 WormSizer: High-Throughput Image Analysis <strong>of</strong> Nematode Size and Shape<br />

Brad Moore, James Jordan, Ryan Baugh<br />

88 Acetylcholine Dependent Starvation Signaling<br />

Robert Pollok, Leon Avery<br />

89 How to sense fasting : searching for the mechanism <strong>of</strong> energy homeostasis by<br />

IRE-1<br />

Jisun Shin, Hyungmin Moon, Jiwon Shim, Junho Lee


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

90 Pigment dispersing factor - a neuropeptidergic influence on metabolism and<br />

stress<br />

Liesbet Temmerman, Ellen Meelkop, Tom Janssen, Liliane Scho<strong>of</strong>s<br />

91 Ethanol Influence on Gonadal Development in C. elegans daf-18/PTEN<br />

Mutants<br />

Tim Wolf, Wenjing Qi, Ralf Baumeister<br />

92 Increased Levels <strong>of</strong> Hydrogen Peroxide Induce a HIF-1-dependent<br />

Remodeling <strong>of</strong> Lipid Metabolism in C. elegans<br />

Meng Xie, Richard Roy<br />

93 The Transmembrane Channel-like Protein TMC-1 Affects Adaptation to a<br />

Chemically Defined Medium in C. elegans<br />

Liusuo Zhang, L Rene Garcia<br />

Topic: Stress<br />

Abstracts 94-131, 170<br />

94 Screening for novel regulators <strong>of</strong> rnt-1 in stress response<br />

Soungyub Ahn, Junho Lee<br />

95 Screening Nuclear Receptors to Discover the Regulation <strong>of</strong> the Xenobiotic<br />

Stress Response<br />

Leah Blackwell, Amanda Marra, Andrew Davidson, Tim Lindblom<br />

96 Depletion <strong>of</strong> the Nascent Polypeptide-associated Complex in C. elegans<br />

Up-regulates ER Chaperone Expression and Engages the Unfolded Protein<br />

Response, Resulting in the Induction <strong>of</strong> Autophagy and Apoptosis<br />

Paul Arsenovic, Anthony Maldonado, Vaughn Colleluori, Tim Bloss<br />

97 Genetic analysis <strong>of</strong> the Unfolded Protein Response during pathogen infection<br />

in C. elegans<br />

Douglas Cattie, Kirthi Reddy, Claire Richardson, Dennis Kim<br />

98 BCAS2 is Essential for Drosophila Viability and Functions in Pre-mRNA<br />

Splicing<br />

Po-Han Chen, Yeou-Ping Tsao, Show-Li Chen<br />

99 Oxidative stress related PMK-1-HIF-1 signaling pathways in silver<br />

nanoparticles toxicity in C. elegans<br />

Jinhee Choi, Hyun - Jeong Eom, Jeong-Min Ahn<br />

100 Insulin-like signaling in the parasitic nematode Brugia malayi<br />

Kirsten Crossgrove, Brenda Garland, Peter Sackett<br />

101 Using Mathematical Models to Predict Gene Flow and Discover Gene<br />

Function<br />

Andrew Davidson, Marc-Andre LeBlanc, Megan Powell, Tim Lindblom<br />

102 EGL-9 Function is Required to Produce Viable Offspring from Animals<br />

Exposed to Chronic Hypoxia<br />

Jennifer Dennis, Pamela Padilla, Brent Little<br />

xvii


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

103 Analysis a Null Allele <strong>of</strong> the Heterochronic Gene lin-42, a period Homolog<br />

Theresa Edelman, Katherine McCulloch, Angela Barr, Christian Frokjaer-Jensen, Erik Jorgensen,<br />

Ann Rougvie<br />

104 SWI/SNF is Required to Maintain Memory <strong>of</strong> Adaptation to Hydrogen Sulfide<br />

Emily Fawcett, Dana Miller<br />

105 Diet & Environment Affect Oxygen-Deprivation Survival in C. elegans<br />

Anastacia Garcia, Pamela Padilla<br />

106 Transcriptional Regulation in the Oxidative Stress Response<br />

Grace Goh, Ada Kwong, Stefan Taubert<br />

107 Disruption <strong>of</strong> the ire-1/xbp-1 UPR Pathway Induces ER stress and Attenuates<br />

the Production and Processing <strong>of</strong> Secreted Proteins Such as Insulin<br />

Modi Safra, Cynthia Kenyon, Sivan Henis-Korenblit<br />

108 The Role <strong>of</strong> C. elegans BRAP-2 in Regulation <strong>of</strong> the Oxygen Radical<br />

Detoxification Response<br />

Queenie Hu, Lesley MacNeil, Marian Walhout, Terry Kubiseski<br />

109 Epigenetic regulation <strong>of</strong> stress response in C. elegans<br />

Moonjung Hyun, Young-Jai You<br />

110 The Role Of daf-2 Pathway In Primary, Secondary, And Delayed Hypoxic<br />

Injury<br />

Euysoo Kim, Chun-Ling Sun, Michael Crowder<br />

111 The Influence <strong>of</strong> Endoplasmic Reticulum Stress on the Dauer Developmental<br />

Decision in C. elegans<br />

Warakorn Kulalert, Dennis Kim<br />

112 PKC-2 in Peroxide mediated Stress and Aging<br />

Marianne Land, Charles Rubin<br />

113 High-throughput screening for small molecule modulators <strong>of</strong> SKN-1<br />

Chi Leung, Siobhan Malany, Andrew Deonarine, Ying Wang, Keith Choe<br />

114 Multisite phosphorylation fine-tunes SKN-1 activity by modulating<br />

interactions with WDR-23 and target DNA<br />

Chi Leung, Koichi Hasegawa, Keith Choe<br />

115 Functions <strong>of</strong> CLIC proteins in C. elegans<br />

Jun Liang, Cathy Savage-Dunn<br />

116 Mouse Nmnat1 protects C. elegans from hypoxic death<br />

Xianrong Mao, C. Michael Crowder<br />

117 Gene silencing based functional analysis <strong>of</strong> C. elegans cytochromes P450: roles<br />

in biotransformation, fat storage and eicosanoid formation<br />

Ralph Menzel, Christian Steinberg, Wolf-Hagen Schunck<br />

118 Deciphering the microRNA responses to High Temperature Stress<br />

Camilla Nehammer, Agnieszka Podolska, Konstantinos Kagias, Sebastian Mackowiak, Nikolaus<br />

Rajewsky, Roger Pocock<br />

xviii


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

119 Transmission <strong>of</strong> a Q/N-Rich Prion Domain Between C. elegans Tissues Causes<br />

an Extreme Form <strong>of</strong> Proteotoxicity<br />

Carmen Nussbaum-Krammer, Kyung-Won Park, Liming Li, Ronald Melki, Richard Morimoto<br />

120 The affect <strong>of</strong> carbohydrate diet, metabolism, germline function and age on<br />

oxygen deprivation response and survival in C. elegans<br />

Pamela Padilla, Anastacia Garcia, Jo Goy, Mary Ladage<br />

121 Understanding Hypoxia-dependent, HIF-independent Pathways in C. elegans<br />

Divya Padmanabha, Young-Jai You, Keith Baker<br />

122 Osmotic Stress Resistance and Cuticle Defects – Two Symptoms, One Cause<br />

Anne-Katrin Rohlfing<br />

123 Knockdown <strong>of</strong> Genes Involved in Basic Cellular Functions Impairs<br />

Mitochondrial ROS Stress Signaling to the Nucleus<br />

Eva Runkel, Shu Liu, Ralf Baumeister, Ekkehard Schulze<br />

124 The Effects <strong>of</strong> HIF-1 Over-Activation: Real-Time Assays for Toxin Response<br />

Jenifer Saldanha, Archana Parashar, Santosh Pandey, Jo Anne Powell-C<strong>of</strong>fman<br />

125 Exploring Natural Variation <strong>of</strong> Starvation Resistance and Growth Rate<br />

Moses Sandr<strong>of</strong>, Meghan Jobson, L. Ryan Baugh<br />

126 The Role <strong>of</strong> the Translation Machinery in Survival from Hypoxia<br />

Barbara Scott, Chun-Ling Sun, Xianrong Mao, Charles Yu, C. Michael Crowder<br />

127 The Role <strong>of</strong> the Ubiquitin-Proteasome System in the Formation <strong>of</strong><br />

Polyglutamine Aggregates<br />

Gregory Skibinski, Lynn Boyd<br />

128 C. elegans as a Model to Study Drug-induced Mitochondrial Dysfunction<br />

Richard de Boer, Reuben Smith, Hans van der Spek, Stanley Brul<br />

129 Identification <strong>of</strong> pathways by which a peroxiredoxin influences stress<br />

resistance reveal (i) its importance for insulin/IGF1-like-signalling and (ii) new<br />

genes important for phase 2 detoxification gene expression, stress resistance<br />

and longevity<br />

Helen Crook, Monika Olahova, Elizabeth Veal<br />

130 Energy Metabolism Abnormality Of Ultraviolet Irradiation Sensitive Mutant<br />

Rad-8 In C. elegans<br />

Kayo Yasuda, Michihiko Fujii, Hitoshi Suda, Phillip Hartman, Takamasa Ishii, Naoaki Ishii<br />

131 Graphite Nanoplatelets and Caenorhabditis elegans: Insights from an in vivo<br />

Model<br />

Elena Zanni, Giovanni De Bellis, Maria Bracciale, Alessandra Broggi, Maria Santarelli, Maria<br />

Sarto, Claudio Palleschi, Daniela Uccelletti<br />

170 Dynamics <strong>of</strong> stress response across the worm unveils cross-tissue<br />

interactions<br />

Ronen Kopito, Christian Anderson, Erel Levine<br />

xix


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

xx<br />

Topic: Pathogenesis<br />

Abstracts 132-161, 169<br />

132 Cancer-like features could facilitate rapid intracellular growth <strong>of</strong> a natural<br />

intracellular parasite in C. elegans.<br />

Malina Bakowski, Amy Ma, Christopher Desjardins, Christina Cuomo, Emily Troemel<br />

133 Identifying a genetic basis to resistance against microsporidian parasites<br />

Keir Balla, Erik Andersen, Zhi Yan, Leonid Kruglyak, Emily Troemel<br />

134 Mapping natural variation in sensitivity to the Orsay virus in C. elegans wild<br />

isolates<br />

Tony Belicard, Marie-Anne Felix<br />

135 The Human Herpesvirus Type 1 Latency Associated Transcript Produces Egg<br />

Laying and Locomotion Defects in the Nematode C. elegans<br />

Ana Bratanich, Jesica Diogo<br />

136 The Conserved 1-Cys Peroxiredoxin, PRDX-6, has Diverse Roles in Stress<br />

Resistance, Innate Immunity and Aging<br />

Emma Button, Elizabeth Veal<br />

137 Shiga-like Toxin 1 Is Required Partly for The Pathogenicity <strong>of</strong> Escherichia coli<br />

O157:H7 in Caenorhabditis elegans<br />

Chang-Shi Chen, Ting-Chen Chou, Hao-Chieh Chiu, Cheng-Ju Kuo, Ching-Ming Wu, Wan-Jr Syu<br />

138 Leucobacter Strains Are Diverse Natural Pathogens <strong>of</strong> Caenorhabditis<br />

Laura Clark, Marie-Anne Felix, Maria Joao Gravato-Nobre, Jonathan Hodgkin<br />

139 Discriminating Pathogens from Innocuous Microbes: C/EBP Surveillance<br />

Immunity in C. elegans Intestinal Defense<br />

Tiffany Dunbar, Zhi Yan, Keir Balla, Margery Smelkinson, Emily Troemel<br />

140 The invertebrate lysozyme ILYS-3 aids bacterial disruption and acts in the<br />

intestine under non-autonomous pharyngeal control to protect against<br />

bacterial infection<br />

Maria Joao Gravato-Nobre , Jonathan Hodgkin<br />

141 Dissecting ERAD Component Function in a Motor Neuron Disease Model<br />

Angela Jablonski, Robert Kalb<br />

142 A Transmembrane Leucine-Rich Repeat Protein Important for Microsporidia<br />

Infection in C. elegans<br />

Robert Luallen, Malina Bakowski, Emily Troemel<br />

143 The redox sensor TRX-1: a potential regulator <strong>of</strong> signaling in C. elegans<br />

Katie McCallum, Danielle Garsin<br />

144 Functional Characterization <strong>of</strong> Thioredoxin 3 (TRX-3), a Caenorhabditis<br />

elegans Intestine-Specific Thioredoxin<br />

Maria Jimenez-Hidalgo, Cyril Leopold Kurz, Jose Rafael Pedrajas, Francisco Jose Naranjo-Galindo,<br />

Juan Cabello, Elamparithi Jayamani, Eleftherios Mylonakis, Juan Carlos Fierro-Gonzalez, Peter<br />

Swoboda, Antonio Miranda-Vizuete


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

145 Screening Potential Anthelmintic Compounds for Novel Activity<br />

Megan Gross, Olufadakemi Awoyinka, Michael Smith, M. Shahjahan Kabir, James Cook, Aaron<br />

Monte, Jennifer Miskowski<br />

146 Characterizing C. elegans behavior in response to E. faecalis<br />

Mona Gardner, Thomas Graciano, June Middleton, Edith Myers<br />

147 Exploring Antifungal Potential <strong>of</strong> CAMPs in the C. elegans Infection Model<br />

Massimiliano Olivi, Daniela Uccelletti, Maria Mangoni, Donatella Barra, Claudio Palleschi<br />

148 Using Bacterial Infection as a Tool to Dissect the Molecular Components <strong>of</strong><br />

the Nematode Surface<br />

Delia O’Rourke, Rebecca Price, Dave Stroud, Jonathan Hodgkin<br />

149 Identification <strong>of</strong> the Transcriptional Targets <strong>of</strong> the PMK-1 p38-Dependent<br />

Transcription Factor ATF-7<br />

Daniel Pagano, Dennis Kim<br />

150 The Role <strong>of</strong> the G-protein Coupled Receptor FSHR-1 in the C. elegans Stress<br />

Response<br />

Amanda Miller, Hannah Anthony, Joseph Robinson, Shannon Hartley, Jennifer Powell<br />

151 Nematodes Living in Anoxic Conditions at a Deep-sea Methane Seep are<br />

Infected With Sexually Transmitted Parasitic Fungus-related Microsporidia<br />

Amir Sapir, Adler Dillman, Manuel Mundo-Ocampo, James Baldwin, Victoria Orphan, Paul<br />

Sternberg<br />

152 Key Residues <strong>of</strong> Cry5B Structure and Function: Mutagenesis by Alanine<br />

Scanning<br />

Jillian Sesar, Yan Hu, Sandy Chang, Arash Safavi, Raffi Aroian<br />

153 Caenorhabditis elegans Neutralizes Small Molecule Toxins Produced by<br />

Pseudomonas aeruginosa<br />

Gregory Stupp, Ramadan Ajredini, Arthur Edison<br />

154 Coprinopsis cinerea Lectins-mediated Toxicity against C. elegans<br />

Katrin Stutz, Alex Butschi, Silvia Bleuler-Martinez, Mario Schubert, Markus Aebi, Markus<br />

Kuenzler, Michael Hengartner<br />

155 Understanding the Molecular Underpinnings <strong>of</strong> Pathogen Recognition in C.<br />

elegans<br />

Kwame Twumasi-Boateng, Hae-Sung Kang, Michael Shapira<br />

156 Reactive oxygen species generated by BLI-3/Ce-Duox1 during infection<br />

triggers the activation <strong>of</strong> SKN-1 in C. elegans<br />

Ransome van der Hoeven, Katie McCallum, Melissa Cruz, Danielle Garsin<br />

157 TRPM Channels are Required for Clozapine’s Effects in C. elegans<br />

Xin Wang, Taixiang Saur, Bruce Cohen, Edgar(Ned) Buttner<br />

158 QTL Mapping <strong>of</strong> Differential Susceptibility to Bacteria in Caenorhabditis<br />

elegans<br />

Ziyi Wang, Michael Herman, Basten Snoek, Jan Kammenga<br />

xxi


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

159 C. elegans Acyl-CoA synthetase-3 And The Nuclear Receptor NHR-25<br />

Promote Epidermal Integrity And Resistance To Pathogens.<br />

Jordan Ward, Carole Couillault, Brendan Mullaney, Teresita Bernal, Marc Van Gilst, Kaveh Ashrafi,<br />

Jonathan Ewbank, Keith Yamamoto<br />

160 Genetic Mechanisms <strong>of</strong> Innate Immune Response: The interaction between<br />

Caenorhabditis elegans and the opportunistic pathogen Stenotrophomonas<br />

maltophilia<br />

Corin White, Vinod Mony, Brian Darby, Michael Herman<br />

161 Characterization <strong>of</strong> the Effects <strong>of</strong> Naturally Isolated Salmonella enterica<br />

Strains on Caenorhabditis elegans<br />

Amanda Wollenberg, Anna Maria Alves, Michael McClelland, Javier Irazoqui<br />

169 Characterizing in-host population dynamics <strong>of</strong> pathogenic bacteria during<br />

intestinal infection <strong>of</strong> C. elegans<br />

Ronen Kopito, Andrzej Nowojewski, Erel Levine<br />

xxii<br />

Topic: Small RNAs<br />

Abstracts 61, 162-168<br />

61 LEP-2/Makorin Promotes let-7 microRNA-mediated Terminal Differentiation<br />

in Male Tail Tip Morphogenesis<br />

R Antonio Herrera, Karin Kiontke, David Fitch<br />

162 Analysis Of mi-RNAS, Target Prediction Algorithms And Databases In C.<br />

elegans<br />

Hema Kasisomayajula, Frnklyn Bolander<br />

163 C. elegans rrf-1 Mutations Maintain RNAi Efficiency in the Soma in Addition<br />

to the Germline<br />

Caroline Kumsta, Malene Hansen<br />

164 Genetic requirements for viRNAs-mediated antiviral immunity in C. elegans<br />

Jeremie Le Pen, Alyson Ashe, Leonard Goldstein, Eric Miska<br />

165 Unifying Models <strong>of</strong> Biological Timers: A Molting Cycle Approach<br />

Gabriela Monsalve, Alison Frand<br />

166 C. elegans PRG-1 and piRNAs silence target transcripts through a secondary<br />

22G-siRNA pathway<br />

Alexandra Sapetschnig, Eva-Maria Weick, Marloes Bagijn, Leonard Goldstein, Amy Cording, Eric<br />

Miska<br />

167 MicroRNA Predictors <strong>of</strong> Longevity in C. elegans<br />

Zachary Pincus, Thalyana Smith-Vikos, Frank Slack<br />

168 A Genetic Suppressor Screen in Caenorhabditis elegans for Targets <strong>of</strong><br />

Antipsychotic Drugs: Role <strong>of</strong> the Nicotinic Acetylcholine Receptor Homolog<br />

acr-7<br />

Taixiang Xu, Xin Wang, Limin Hao, Bruce Cohen, Edgar Buttner


Contact: ewbank@ciml.univ-mrs.fr<br />

Lab: Ewbank<br />

Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

Building an integrated view <strong>of</strong> anti-fungal innate immunity<br />

Jonathan Ewbank<br />

Centre déImmunologie de Marseille-Luminy, INSERM, CNRS, Aix-Marseille<br />

Université , Marseille, France<br />

Our research focuses on how C. elegans responds to fungal infection, mainly using a<br />

natural pathogen, Drechmeria coniospora. Worms have a highly derived innate immune system,<br />

exemplified by the fact that they have no equivalent <strong>of</strong> NF-kappaB, central to immunity in<br />

many animal species. Nor do they have many <strong>of</strong> the receptors known from other species to be<br />

important for triggering host defenses. On the other hand, in common with most multicellular<br />

organisms, both plant and animal, C. elegans can produce antimicrobial peptides (AMPs) that<br />

contribute to pathogen resistance. We have applied forward and reverse genetics, proteomics,<br />

biochemistry, and cell biology to dissect the signaling pathways that control the expression <strong>of</strong><br />

two main AMP families, the structurally-related nlp and cnc genes.<br />

Our recent results have given insights into the different steps <strong>of</strong> host defense: how infection<br />

is recognized, what signaling networks transduce this information and how the innate immune<br />

response is integrated into cellular and organismal physiology.<br />

Session 1 Keynote Speaker<br />

1


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

Metformin increases C. elegans lifespan by altering E. coli folate<br />

metabolism<br />

Filipe Cabreiro 1 , Catherine Au 1 , Kit-Yi Leung 2 , Nick Greene 2 , David Gems 1<br />

1 Institute <strong>of</strong> Healthy Ageing, <strong>University</strong> College London, London, UK, 2 Institute <strong>of</strong><br />

Child Health, <strong>University</strong> College London, London, UK<br />

Metformin is the most widely prescribed drug for type II diabetes, and also increases<br />

lifespan in C. elegans and rodents, possibly by inducing a dietary restriction-like state. Worms<br />

are co-cultured with E. coli, which exerts complex nutritional and pathogenic effects on their<br />

host/predator affecting lifespan. In mammals, intestinal microbiota mediates effects <strong>of</strong> nutrition<br />

on host metabolic status, and risk <strong>of</strong> metabolic disease. We are investigating how metformin<br />

acts to increase worm lifespan and, perhaps, ameliorate metabolic disease. In the absence<br />

<strong>of</strong> E. coli, or on UV-irradiated or metformin-resistant E. coli, this drug only shortens lifespan.<br />

Moreover, using a range <strong>of</strong> E. coli strains, effects <strong>of</strong> metformin on worm lifespan correlate<br />

positively with its inhibitory effect on bacterial growth. This implies that E. coli mediates effects<br />

<strong>of</strong> metformin on worm lifespan. Metformin inhibits E. coli growth by impairing folate metabolism<br />

via a so-called “methyl trap” involving methionine synthase inhibition. Metformin effects on worm<br />

lifespan are phenocopied by the dihydr<strong>of</strong>olate reductase inhibitor trimethoprim (an antibiotic),<br />

but are independent <strong>of</strong> effects on bacterial proliferation. Thus, metformin has two effects on<br />

adult worms: a direct, life-shortening one (probably reflecting drug toxicity), and an indirect,<br />

life-extending effect promoted by bacterial metabolic alteration. The latter is mediated in the<br />

worm by a metabolic sensing pathway controlling the S-adenosyl-methionine/S-adenosylhomocysteine<br />

ratio. Life-extending effects <strong>of</strong> metformin involve sams-1 (S-adenosyl methionine<br />

synthetase), metr-1 (methionine synthase) and aak-2 (AMP-activated protein kinase). skn-1<br />

(homologous to mammalian Nrf2) and aak-2 also mediate the life-extending effects <strong>of</strong> metformin<br />

by protecting against drug toxicity. Consistent with this, the latter two mutants are hypersensitive<br />

to developmental retardation by metformin. Finally, elevated glucose, a risk factor for metabolic<br />

disease, abolishes life extension by metformin. In humans, side effects <strong>of</strong> metformin include<br />

gastrointestinal upset and folate deficiency. This, with our findings, suggests evolutionarily<br />

conserved bacterial mediation <strong>of</strong> metformin effects on host metabolism, and the therapeutic<br />

power <strong>of</strong> manipulating intestinal microbiota to assure metabolic health.<br />

Contact: F.cabreiro@ucl.ac.uk<br />

Lab: Gems<br />

2<br />

Session 1


Contact: kjdumas@umich.edu<br />

Lab: Hu<br />

Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

The C. elegans Dosage Compensation Protein DPY-21 Regulates<br />

Dauer Arrest<br />

Kathleen Dumas1 , Stephane Flibotte2 , Don Moerman2 , Patrick Hu1 1 2 <strong>University</strong> <strong>of</strong> Michigan, Ann Arbor, MI, USA, <strong>University</strong> <strong>of</strong> British Columbia,<br />

Vancouver, BC, Canada<br />

In C. elegans, both reducing insulin/insulin-like growth factor signaling (IIS) and ablating<br />

the germline extend life span by promoting the translocation <strong>of</strong> DAF-16/FoxO to the nucleus.<br />

Nuclear DAF-16/FoxO induces transcriptional programs that promote longevity. The IIS pathway<br />

in C. elegans consists <strong>of</strong> conserved PI3K and Akt components that antagonize DAF-16/FoxO<br />

by promoting its cytoplasmic sequestration. We have discovered the EAK pathway, a novel,<br />

conserved pathway that inhibits DAF-16/FoxO. The EAK pathway acts in parallel to PI3K/Akt<br />

signaling to inhibit nuclear DAF-16/FoxO activity without promoting its translocation to the<br />

cytoplasm. How the EAK pathway inhibits DAF-16/FoxO is not known.<br />

To illuminate mechanisms by which the EAK pathway inhibits DAF-16/FoxO, we performed<br />

a genetic screen for suppressors <strong>of</strong> the eak-7;akt-1 dauer arrest phenotype (seak mutants).<br />

Among sixteen independent seak mutants isolated, three harbored distinct missense mutations<br />

in dpy-21, which encodes a conserved component <strong>of</strong> the dosage compensation complex (DCC)<br />

that represses X-chromosome gene expression in hermaphrodite animals. All three dpy-21<br />

mutations affect conserved residues in the C-terminus <strong>of</strong> the protein. SNP mapping indicated<br />

that the seak phenotype is linked to dpy-21 in all three mutants, and dpy-21 RNAi recapitulates<br />

the seak phenotype. Therefore, dpy-21 is likely a bonafide seak gene.<br />

To determine whether the effect <strong>of</strong> dpy-21 mutations on eak-7;akt-1 dauer arrest was due<br />

to disruption <strong>of</strong> the DCC, we performed RNAi knockdown <strong>of</strong> the ten known DCC components<br />

in eak-7;akt-1 double mutant animals. Whereas RNAi <strong>of</strong> sdc-1 and dpy-30 failed to suppress<br />

eak-7;akt-1 dauer arrest, RNAi <strong>of</strong> genes encoding the other eight DCC components suppressed<br />

eak-7;akt-1 dauer arrest significantly. Importantly, RNAi <strong>of</strong> dpy-27, which encodes the only<br />

condensin complex component that is specific to the DCC, strongly suppressed eak-7;akt-1<br />

dauer arrest. These results implicate multiple components <strong>of</strong> the DCC in the control <strong>of</strong> dauer<br />

arrest. Experiments are ongoing to determine whether the effect <strong>of</strong> DCC knockdown on dauer<br />

arrest is an indirect consequence <strong>of</strong> aberrant expression <strong>of</strong> X-linked or autosomal dauer<br />

regulatory genes as opposed to a direct effect <strong>of</strong> DCC proteins on DAF-16/FoxO target gene<br />

expression.<br />

Session 1<br />

3


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

Germline Stem Cells Regulate Somatic Proteostasis During<br />

Caenorhabditis elegans Adulthood<br />

Netta Shemesh, Nadav Shai, Anat Ben-Zvi<br />

Ben-Gurion <strong>University</strong> <strong>of</strong> the Negev, Beer-Sheva, Israel<br />

All cells rely on highly conserved protein folding and clearance pathways to detect<br />

and resolve protein damage and maintain protein folding homeostasis (proteostasis). The<br />

recognition that an age-associated imbalance in proteostasis is a potent contributor to the<br />

onset <strong>of</strong> neurodegenerative diseases stresses the need to understand how protein folding is<br />

regulated in a multi-cellular organism. We have noted that the ability <strong>of</strong> Caenorhabditis elegans<br />

to maintain proteostasis declines sharply following the onset <strong>of</strong> oocyte biomass production,<br />

suggesting that a restricted folding capacity may be linked to the onset <strong>of</strong> reproduction. To<br />

test this hypothesis, we monitored the effects <strong>of</strong> different sterile mutations on proteostasis<br />

maintenance in the soma <strong>of</strong> C. elegans. We find that germline stem cell (GSC) arrest rescued<br />

protein quality control, resulting in maintenance <strong>of</strong> robust proteostasis in different somatic<br />

tissues <strong>of</strong> adult animals. We further demonstrate that cell-nonautonomous signaling via kri-1,<br />

a key player in GSC-dependent regulation <strong>of</strong> fat metabolism and longevity, is a mediator <strong>of</strong><br />

proteostasis maintenance in adulthood. We find that signaling through kri-1 and, specifically, the<br />

activity <strong>of</strong> LIPL-4, a lipase that is regulated by KRI-1, can uncouple germline proliferation from<br />

somatic proteostasis and prevent the switch between a robust and a limited state <strong>of</strong> proteostatic<br />

capacity in the soma. This decreases the age-dependent accumulation <strong>of</strong> damaged proteins<br />

and postpones the onset <strong>of</strong> age-dependent protein misfolding in a polyglutamine disease model.<br />

Contact: anatbz@bgu.ac.il<br />

Lab: Ben-Zvi<br />

4<br />

Session 1


Contact: ryan.baugh@duke.edu<br />

Lab: Baugh<br />

Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

Transgenerational Effects <strong>of</strong> Starvation on Development,<br />

Reproduction and Lifespan: Bet-hedging on an Epigenetic Fitness<br />

Trade-<strong>of</strong>f<br />

Meghan Jobson, L. Ryan Baugh<br />

Duke <strong>University</strong>, Durham, NC, USA<br />

Starvation during early human development has epigenetic effects that are thought to be<br />

adaptive if famine persists, and there may be transgenerational effects as well. Life in the<br />

wild is presumed to be feast or famine for C. elegans, suggesting that the worm has a robust<br />

epigenetic response to starvation. Our lab uses L1 arrest and recovery as a model to investigate<br />

nutritional control <strong>of</strong> development. We examined a variety <strong>of</strong> life history traits in well-fed animals<br />

following L1 arrest to characterize epigenetic effects <strong>of</strong> starvation. We show that development,<br />

reproduction and lifespan are all affected after recovery from L1 arrest and, remarkably, that<br />

these epigenetic effects persist for multiple generations. Development is delayed, producing<br />

smaller adults, and fertility is reduced, but stress resistance and lifespan increase. Egg-laying<br />

behavior is also affected, with a high frequency <strong>of</strong> internal hatching (i.e. ‘bagging’), revealing<br />

epigenetic control <strong>of</strong> reproductive behavior. Starvation causes a striking amount <strong>of</strong> phenotypic<br />

variation among isogenic individuals, and those that develop slowest are least fertile but most<br />

stress resistant. These most affected individuals live 50% longer, and they transmit the life<br />

span extension phenotype to their progeny as well as increased starvation resistance. Our<br />

work shows that environmental conditions and life history can have pr<strong>of</strong>ound transgenerational<br />

epigenetic effects on several organismal traits including lifespan. In particular, we demonstrate<br />

epigenetic control <strong>of</strong> a fitness trade-<strong>of</strong>f between reproduction and stress resistance in response<br />

to starvation, suggesting anticipation <strong>of</strong> stress. Furthermore, the resulting phenotypic variation<br />

among isogenic individuals is consistent with an evolutionary bet-hedging strategy to optimize<br />

fitness in fluctuating environmental conditions.<br />

Session 2<br />

5


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

Transcriptional Network Key to Longevity Determination and Stress<br />

Regulation<br />

Siu Sylvia Lee<br />

Cornell <strong>University</strong>, Ithaca, NY, USA<br />

The highly conserved transcriptional co-regulator HCF-1 is capable <strong>of</strong> coordinating many<br />

transcription and chromatin factor complexes and participates in a wide variety <strong>of</strong> biological<br />

processes. We previously identified the C. elegans hcf-1 null mutant to be long-lived and stress<br />

resistant. Our investigations revealed that HCF-1 modulates longevity and stress response by<br />

acting with the protein deacetylase SIR-2.1/SIRT1 to regulate the activity <strong>of</strong> DAF-16/FOXO.<br />

Interestingly, HCF-1 appears to also regulate SKN-1/NRF2 to specifically affect oxidative stress<br />

response. Our data suggest HCF-1 is an integral component <strong>of</strong> the transcriptional network key<br />

to longevity determination and stress regulation.<br />

Contact: sylvia.lee@cornell.edu<br />

Lab: Lee<br />

6<br />

Session 2 Plenary Speaker


Contact: xguo@bio.tamu.edu<br />

Lab: Garcia<br />

Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

SIR-2.1, an HDAC, is Required To Maintain The Male Mating Potency In<br />

C. elegans<br />

Xiaoyan Guo 2 , Luis rene Garcia 2,1<br />

1 HHMI, 2 Texas A&M <strong>University</strong>, College Station, TX<br />

In a previous study (1), we carefully characterized that C. elegans male mating behavior<br />

significantly deteriorates at adult age day 3. Through direct mating behavior observations,<br />

Ca2+ imaging, and pharmacological tests, we found that the muscular components used for<br />

mating become more excitable as the males age. To further uncover the molecular mechanism,<br />

we found that sir-2.1, a histone deacetylase in C. elegans might be the link between the male<br />

mating behavior decay and the increase <strong>of</strong> the sex muscle excitability. Compared to wild type,<br />

sir-2.1(ok434) males mating potency prematurely decline: a significant drop is observed at<br />

adult age day 2, correlating with an obvious increase <strong>of</strong> sex muscle excitability. Furthermore,<br />

we observed that sir-2.1(ok434) males have a series <strong>of</strong> metabolism disorders: significantly<br />

more glycogen, fat and ATP are synthesized in sir-2.1(ok434) males. Those observations lead<br />

to the hypothesis that there is more ROS stress undergoing in sir-2.1 (ok434), which might be<br />

responsible for the increase <strong>of</strong> sex muscle excitability increase and mating potency decline.<br />

Indeed, we found that sir-2.1(ok434) males are more sensitive to the oxidative stress, paraquat.<br />

Meanwhile, for wild type males, feeding small amount <strong>of</strong> paraquat decreases the male mating<br />

potency at day 2 and increase the sex muscle excitability.<br />

1. Guo X, Navetta A, Gualberto DG, Garcia LR. Behavioral decay in aging male C. elegans correlates with<br />

increased cell excitability. Neurobiol Aging. 2012.<br />

Session 2<br />

7


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

LC-MS Proteomics Analysis Reveals Both Common and Unique<br />

Changes in the Proteome <strong>of</strong> Insulin/IGF-1 Receptor Mutant and<br />

Dietary Restricted C. elegans<br />

Geert Depuydt1 , Fang Xie2 , Vladislav Petyuk2 , Heather Brewer2 , Arne Smolders1 , Ineke<br />

Dhondt1 , Nilesh Shanmugam1 , David Camp II2 , Richard Smith2 , Bart Braeckman2 1 2 Biology Department, Ghent <strong>University</strong>, B-9000 Ghent, Belgium, Biological<br />

Sciences Division and Environmental Molecular Sciences Laboratory, Pacific<br />

Northwest National Laboratory, Richland, WA 99352, USA<br />

Impaired insulin/IGF-1 signaling (IIS) and dietary restriction are two well-characterized<br />

lifespan extending interventions that operate by (partially) independent mechanisms in C.<br />

elegans. Taking advantage <strong>of</strong> recent developments in quantitative LC-MS/MS based proteomics,<br />

we sought to uncover potentially universal, as well as condition-specific, proteomic changes<br />

that could underlie daf-2(e1370)- and dietary restriction-induced longevity. A most striking<br />

pattern in the proteome fingerprints <strong>of</strong> both long-lived IIS-mutant and DR worms is the overall<br />

decrease in the abundance <strong>of</strong> a large number <strong>of</strong> ribosomal proteins <strong>of</strong> both the large (60S)<br />

and small (40S) subunits. Consistently, we found decreased rates <strong>of</strong> protein synthesis in our<br />

long-lived worms. These results indicate that a general downregulation in mRNA translation<br />

rate is part <strong>of</strong> an adaptive response that allows daf-2 and DR worms to be long-lived. A second<br />

common pattern in these worms is the increased abundance <strong>of</strong> many muscle proteins, which<br />

we could attribute to an increase in relative, but not absolute, muscle biomass. In addition,<br />

increased mRNA levels <strong>of</strong> muscle-related genes and higher concentrations <strong>of</strong> branched-chain<br />

amino acids as a result <strong>of</strong> their reduced catabolism in daf-2, suggest C. elegans attempts<br />

to preserve muscle integrity in conditions <strong>of</strong> low nutrient availability. A most striking pattern<br />

in the daf-2 proteome pr<strong>of</strong>ile is the strong upregulation <strong>of</strong> most core intermediary metabolic<br />

pathways (e.g. carbohydrate, fat, oxidative ATP generation), including several pathways that<br />

are reciprocally regulated. Despite this upregulation <strong>of</strong> the energy metabolic machinery, we<br />

found daf-2 animals fit a hypometabolic energy pr<strong>of</strong>ile, similar to dauers, but retain the ability<br />

for strong activity bursts when necessary.<br />

Contact: Geert.Depuydt@ugent.be<br />

Lab: Braeckman<br />

8<br />

Session 2


Contact: franzcj@gmail.com<br />

Lab: Wang<br />

Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

Characterization <strong>of</strong> Caenorhabditis Nematode Infecting Viruses<br />

Carl Franz 1 , Marie-Anne Felix 2 , Yanfang Jiang 1 , Guoyan Zhao 1 , Hilary Renshaw 1 , Guang<br />

Wu 1 , David Wang 1<br />

1 Washington <strong>University</strong> School <strong>of</strong> Medicine, Saint Louis, (MO), USA, 2 Institute <strong>of</strong><br />

Biology <strong>of</strong> the Ecole Normale Superieure (IBENS), Paris, France<br />

Orsay virus and Santeuil virus, the first discovered viruses capable <strong>of</strong> naturally infecting<br />

the nematodes C. elegans and C. briggsae, respectively, were recently discovered by high<br />

throughput sequencing <strong>of</strong> wild Caenorhabditis strains. By similar analysis <strong>of</strong> another wild<br />

C. briggsae isolate, we have now discovered and sequenced the complete genome <strong>of</strong><br />

another novel virus, Le Blanc virus, that shared less than 50% amino acid identity with Orsay<br />

and Santeuil viruses. All three viruses are most closely related to nodaviruses, which are<br />

positive sense RNA viruses with bipartite genomes. Comparison <strong>of</strong> their complete genomes<br />

demonstrated unique coding and noncoding features absent in known nodaviruses. Le Blanc<br />

virus, similar to Santeuil virus, was capable <strong>of</strong> infecting wild C.briggsae isolates but not the<br />

AF16 C. briggsae laboratory reference strain nor any tested C. elegans strains. In order to<br />

define the tissue tropism <strong>of</strong> nematode infection by Orsay, Santeuil and Le Blanc viruses, we<br />

developed immun<strong>of</strong>luorescence assays targeting proteins from each <strong>of</strong> the three viruses.<br />

Analysis <strong>of</strong> virally infected larval stage worms demonstrated that infection by all three viruses<br />

was localized to intestinal cells. Furthermore, the RNA dependent RNA polymerase and capsid<br />

proteins <strong>of</strong> Orsay virus exhibited distinct subcellular localization patterns. These findings identify<br />

a third novel virus and provide further characterization <strong>of</strong> the recently established viral infection<br />

system in Caenorhabditis nematodes.<br />

Session 2<br />

9


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

Functions <strong>of</strong> MicroRNAs During Aging<br />

Alexandre de Lencastre, Frank Slack<br />

Yale <strong>University</strong>, New Haven, CT, USA<br />

MicroRNAs (miRNAs) function to regulate gene expression during development in higher<br />

eukaryotes. Previous work in our lab has demonstrated that miRNAs <strong>of</strong> previously unknown<br />

function, such as miR-71, miR-238, miR-239 and miR-246 function during adulthood to promote<br />

or antagonize longevity and stress response in C. elegans (1). We found that these miRNAs are<br />

up-regulated in aging and genetically interact with components <strong>of</strong> the insulin signaling pathway<br />

and the DNA damage checkpoint response pathway. Together with our previous observation<br />

that mutations to the miRNA lin-4 and its target lin-14 significantly affect the lifespan <strong>of</strong> C.<br />

elegans (2), these results establish miRNAs as a new class <strong>of</strong> aging-associated genes, with<br />

the potential to interact with a wide range <strong>of</strong> aging pathways.<br />

In order to understand the biological function <strong>of</strong> these age-associated miRNAs, we have<br />

begun characterizing the factors downstream as well as upstream <strong>of</strong> these miRNAs that mediate<br />

their function. We have shown by genetic epistasis that the insulin-signalling and the DNA<br />

damage response pathways interact with miR-71 and miR-239. In order to characterize the<br />

detailed molecular identities <strong>of</strong> pathway genes that may transduce these miRNAs’ functions,<br />

we have surveyed the mRNA expression levels <strong>of</strong> multiple genes in these pathways in the<br />

genetic background <strong>of</strong> aging-associated miRNA mutants. We have found significantly deregulated<br />

expression <strong>of</strong> candidate targets genes in these pathways that are also predicted to<br />

be direct targets <strong>of</strong> these miRNAs. Similarly, we have also begun characterizing the identity<br />

<strong>of</strong> upstream factors that regulate the expression <strong>of</strong> our age-associated miRNAs. We have<br />

evidence that suggests that 2 well-known transcription factors regulate the expression <strong>of</strong> specific<br />

aging-associate miRNAs and suggest a possible function for these miRNAs as mediators <strong>of</strong><br />

organismal response to environmental stress. Given the high conservation <strong>of</strong> aging pathways<br />

and miRNAs across species, it is likely that insights uncovered by this research will have high<br />

relevance towards our understanding <strong>of</strong> aging in higher organisms and humans.<br />

References:<br />

1. de Lencastre A. et al., Curr Biol. 20, 1-10 (2010).<br />

2. Boehm M., Slack FJ., Science. 310, 1954-7 (2005).<br />

Contact: alexandre.delencastre@yale.edu<br />

Lab: Slack<br />

10<br />

Session 2


Contact: keith.blackwell@joslin.harvard.edu<br />

Lab: Blackwell<br />

Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

Integration <strong>of</strong> the Unfolded Protein and Oxidative Stress Responses<br />

through SKN-1/Nrf<br />

Kira Glover-Cutter, Stephanie Lin, T. Keith Blackwell<br />

Joslin Diabetes Center and Harvard Medical School, Boston, MA, USA<br />

C. elegans SKN-1 (ortholog <strong>of</strong> mammalian Nrf1/2/3) is critical for oxidative and xenobiotic<br />

stress defense, and maintenance <strong>of</strong> proteasome activity. SKN-1 also promotes longevity, and<br />

is important for lifespan extensions associated with reduced growth signaling (Insulin/IGF-<br />

1-like, TORC1, TORC2) and dietary restriction. SKN-1 regulates genes involved in various<br />

biological processes, and mobilizes distinct responses to different stresses or signals, including<br />

perturbations in protein synthesis. We now report that SKN-1 plays an essential role in the<br />

unfolded protein response (UPR) to stress in the endoplasmic reticulum (ER), where secretory<br />

and membrane proteins are synthesized, processed, and folded. SKN-1 is important for ER<br />

stress resistance, and is required for ER stresses to induce expression <strong>of</strong> core UPR signaling<br />

and transcription factors (XBP-1, ATF-5, IRE-1, HSP-4/BiP), along with downstream genes.<br />

A number <strong>of</strong> these genes are direct transcriptional targets <strong>of</strong> SKN-1. SKN-1 cooperates with<br />

UPR transcription factors at downstream regulatory targets, including its own gene, and its<br />

expression is induced by ER stress. SKN-1 also associates with the ER, suggesting a possible<br />

signaling role. Surprisingly, the UPR signaling apparatus is required for SKN-1 to accumulate<br />

in nuclei and activate downstream genes in response to oxidative stresses. Impairment <strong>of</strong><br />

UPR signaling increases oxidative stress sensitivity, consistent with its importance for SKN-1<br />

functions. The data show that SKN-1 is a key UPR factor and has a broad role in maintaining<br />

protein homeostasis, and that its functions are modulated by interactions with other stressresponse<br />

transcription factors, a paradigm that may explain how SKN-1 mounts responses<br />

that are tailored to different stress situations. They also indicate that signaling from the ER<br />

plays a licensing and possibly detection role in oxidative stress responses, a relationship that<br />

may be important because <strong>of</strong> the centrality <strong>of</strong> redox status to ER function.<br />

Session 2<br />

11


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

Oxygen, temperature, and food: new insights into the role <strong>of</strong><br />

environment in C. elegans aging<br />

Matt Kaeberlein<br />

<strong>University</strong> <strong>of</strong> Washington, Department <strong>of</strong> Pathology, Seattle, WA, United States<br />

Aging is controlled by a complex interaction between environmental and genetic factors.<br />

The most studied environmental modulator <strong>of</strong> longevity is nutrient availability, with dietary<br />

restriction known to extend lifespan in species from yeast to monkeys. In addition to food,<br />

oxygen and temperature are also known to be important environmental factors in determining<br />

adult lifespan in C. elegans. Here I will describe our recent efforts to understand how each <strong>of</strong><br />

these environmental components interacts with known genetic modifiers <strong>of</strong> aging, including<br />

insulin-like signaling, mRNA translation, and the hypoxic response. I will also describe ongoing<br />

studies to define the mechanisms <strong>of</strong> lifespan extension from stabilization <strong>of</strong> the hypoxic response<br />

transcription factor, HIF-1, under normoxic conditions.<br />

Contact: kaeber@uw.edu<br />

Lab: Kaeberlein<br />

12<br />

Session 2 Plenary Speaker


Contact: lapierre@burnham.org<br />

Lab: Hansen<br />

Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

The transcription factor HLH-30/TFEB regulates autophagy and<br />

modulates longevity in C. elegans<br />

Louis Lapierre, Malene Hansen<br />

Sanford-Burnham Medical Research Institute<br />

The cytosolic degradation process <strong>of</strong> autophagy has emerged as a key mechanism<br />

required for lifespan extension in C. elegans, yet the underlying regulatory mechanisms remain<br />

unclear. The mammalian transcription factor TFEB was recently found to coordinately upregulate<br />

several genes involved in both the formation <strong>of</strong> the autophagosome (i.e., the vesicle<br />

sequestering material for degradation) and in lysosomal processing (i.e., compartment where<br />

active degradation takes place). In this study, we asked if the nematode ortholog <strong>of</strong> TFEB,<br />

called HLH-30, is a functional homolog <strong>of</strong> mammalian TFEB.<br />

To investigate this hypothesis, we analyzed the function <strong>of</strong> HLH-30 in long-lived, germlineless<br />

glp-1 mutants. We previously reported that glp-1 mutants display increased autophagy,<br />

and require autophagy genes to live long (Lapierre et al., Current Biology, 2012). We found<br />

that glp-1 animals, along with an increased expression <strong>of</strong> hlh-30, showed significantly elevated<br />

mRNA levels <strong>of</strong> worm orthologs for multiple TFEB autophagy and lysosomal targets. Notably,<br />

the promoters <strong>of</strong> these target genes contain conserved binding sites for HLH-30. We also<br />

observed that glp-1 animals subjected to hlh-30 RNAi showed decreased induction <strong>of</strong> targets,<br />

as well as reduced numbers <strong>of</strong> GFP::LGG-1 autophagosomal puncta, indicating that HLH-30<br />

regulates autophagy in long-lived C. elegans.<br />

Finally, we asked if hlh-30 plays a role in longevity, as previously reported for multiple<br />

autophagy genes. We found that RNAi knockdown <strong>of</strong> hlh-30 in adult glp-1 animals significantly<br />

shortened their long lifespan, while not affecting the lifespan <strong>of</strong> wild-type animals. In addition,<br />

we observed a similar trend in another longevity model, the daf-2 insulin/IGF-1 receptor<br />

mutants, suggesting that hlh-30 plays a broader role in lifespan extension. Collectively, our<br />

findings indicate that HLH-30 is a novel longevity-modulating transcription factor, which displays<br />

functional similarity to its mammalian counterpart TFEB.<br />

Session 2<br />

13


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

HLH-30 is a new Transcription Factor involved in C. elegans Host<br />

Response triggered by S. aureus Infection<br />

Orane Visvikis 1 , Nnamdi Ihuegbu 2 , Lyly Luhachack 1 , Amanda Wollenberg 1 , Anna-Maria<br />

Alves 1 , Gary Stormo 2 , Javier Irazoqui 1<br />

1 Program <strong>of</strong> Developmental Immunology, Massachusetts General Hospital,<br />

Boston (MA), USA, 2 Center for Genome Sciences, Washington <strong>University</strong>, St.<br />

Louis (MO), USA<br />

The human pathogen Staphylococcus aureus can infect and kill Caenorhabditis elegans.<br />

Previous microarray analysis showed that S. aureus triggers a pathogen-specific transcriptional<br />

host response, which appears to be regulated by Toll-like receptor-independent sensing <strong>of</strong><br />

pathogen-associated molecular patterns (PAMPs). To identify components that orchestrate the<br />

C. elegans host response, we performed bioinformatic analysis and identified evolutionarily<br />

conserved DNA motifs that are over-represented in the promoters <strong>of</strong> S. aureus induced<br />

genes (SAIGs). One such motif corresponds to the HLH-30 binding E-box. hlh-30 expression<br />

analysis by RT-qPCR revealed a 2-fold up-regulation upon infection. Additionally, we generated<br />

transgenic animals over-expressing HLH-30::GFP under its own promoter. HLH-30::GFP protein<br />

was found to accumulate in the nucleus upon infection, consistent with the hypothesis that<br />

HLH-30 is a transcription factor involved in the host response to S. aureus infection.<br />

To test this hypothesis, we performed RNA-seq to determine the downstream target genes<br />

<strong>of</strong> HLH-30 in uninfected and infected animals. Of 825 SAIGs identified in wild-type animals,<br />

637 SAITs (77%) were not induced in hlh-30(-) animals, confirming that HLH-30 plays a<br />

key role in the C. elegans host response to S. aureus. GO annotation <strong>of</strong> these 637 HLH-30<br />

dependent SAIGs revealed enriched terms related to antimicrobial factors. We used RT-qPCR<br />

and in vivo reporter analysis to confirm that HLH-30 controls induction <strong>of</strong> antimicrobial factors<br />

during infection. Consistently, we found that hlh-30(-) mutants exhibit enhanced susceptibility<br />

to S.aureus-mediated killing. Therefore, we suspect that the transcriptional defect observed<br />

in hlh-30(-) animals is biologically significant for host defense. Interestingly, we found that hlh-<br />

30(-) animals also display enhanced susceptibility to other C. elegans pathogens (S.enterica,<br />

P. aeruginosa, E. faecalis) suggesting that HLH-30 might also regulate the host response to<br />

these three pathogens. We also found that HLH-30 regulates expression <strong>of</strong> stress and aging<br />

genes. Consistently, we found that hlh-30(-) mutants are short-lived on non pathogenic food,<br />

and exhibit enhanced susceptibility to heat-shock and starvation, but not to oxidative stress.<br />

Altogether, these data indicate that HLH-30 is a major transcription factor that can control<br />

a biologically significant transcriptional host response to stresses such as S. aureus infection.<br />

Contact: ovisvikis@partners.org<br />

Lab: Irazoqui<br />

14<br />

Session 2


Contact: nils.f@bmb.sdu.dk<br />

Lab: Færgeman<br />

Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

Quantitative Proteomics Identifies SBP-1/SREBP as a General<br />

Transcriptional Regulator <strong>of</strong> Metabolic Pathways in C. elegans<br />

Julius Fredens, Kasper Engholm-Keller, Jakob Moeller-Jensen, Martin Roessel Larsen,<br />

Nils Færgeman<br />

<strong>University</strong> <strong>of</strong> Southern Denmark, Odense, Denmark<br />

Stable isotope labeling by amino acids combined with mass spectrometry is a widely used<br />

methodology to quantitatively examine metabolic and signaling pathways in yeast, fruit flies,<br />

plants, cell cultures and mice. We have recently shown that C. elegans can be completely<br />

labeled with heavy-labeled lysine by feeding worms on prelabeled lysine auxotroph Escherichia<br />

coli for just one generation, and used this combined with high resolution mass spectrometry to<br />

identify novel genes regulated by nuclear hormone receptor 49 (Fredens et al., 2011, Nature<br />

Methods). We have used this methodology to examine how the multicellular organism C.<br />

elegans responds to knock down <strong>of</strong> SBP-1, a basic helix-loop-helix (bHLH) transcription factor<br />

homologous to the mammalian Sterol Regulatory Element Binding Proteins (SREBP), which is<br />

known as a master regulator <strong>of</strong> expression <strong>of</strong> lipogenic genes in eukaryotes. We have identified<br />

and quantified several thousands proteins, and among these identified several proteins that<br />

either become up-or down regulated in response to RNAi against SBP-1. As expected, several<br />

<strong>of</strong> the down-regulated proteins we identified are involved in the biosynthesis <strong>of</strong> fatty acids and<br />

glycerolipids, however, our results also indicate that SBP-1 regulate the transcription <strong>of</strong> genes<br />

involved in ceramide synthesis. Additionally, we identified a large number <strong>of</strong> proteins involved<br />

in carbohydrate- and amino acid metabolism to be down regulated upon SBP-1 knock down.<br />

Collectively, our findings suggest that SBP-1 not only regulate the expression <strong>of</strong> lipogenic genes,<br />

but also the expression <strong>of</strong> amino acid and carbohydrate metabolism. We therefore hypothesize<br />

that SBP-1 is a general transcriptional regulator <strong>of</strong> metabolic pathways in C. elegans.<br />

Session 2<br />

15


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

Pathology Of The Aging Gonad Viewed By TEM<br />

David Hall, Angela Jevince, Ken Nguyen, Laura Herndon<br />

Albert Einstein College <strong>of</strong> Medicine, Bronx, NY USA<br />

The Caenorhabditis elegans germline and somatic gonad undergo complex changes<br />

during larval development and achieve peak mature function during the first several days <strong>of</strong><br />

adult life. After about 6 days <strong>of</strong> adult life, the animal’s reproductive capabilities begin to decline<br />

sharply. We used electron microscopy to characterize these unique age-related changes in<br />

the C. elegans germline and have concentrated on two ages, 10 and 18 days. Our findings<br />

suggest that initially the distal gonad arm becomes much shorter, but otherwise seems intact<br />

and the germline may still undergo some mitotic divisions. Between days 7-10 <strong>of</strong> adulthood, the<br />

wild type animal adopts an emo phenotype characterized by an accumulation <strong>of</strong> endomitotic<br />

oocytes in the proximal gonad arm. Egg laying fails and the uterine contents become trapped<br />

by the closed vulva, sometimes including a few fertilized embryos. By 10 days, we see multiple<br />

“haploid” oocytes undergoing acrosomal activation without being fertilized, since all sperm<br />

are exhausted. After activation, these oocytes do not form an eggshell, but gradually enlarge,<br />

undergoing nuclear endoreduplication to produce large masses <strong>of</strong> chromatin, surrounded by<br />

complex cytoplasm, first filling the uterus and then moving backwards into the ovary. Aging<br />

germline cells within the ovary and uterus are thus mixed with retained embryonic tissues<br />

that together can swell into a rigid mass centered at the midbody. We are in the process <strong>of</strong><br />

determining whether the course <strong>of</strong> gonad decline is related to the decline in other tissues and<br />

if animals with longer healthspans and better body movement show less dramatic changes<br />

in their gonad.<br />

Contact: david.hall@einstein.yu.edu<br />

Lab: Hall<br />

16<br />

Session 2


Contact: kaveh.ashrafi@ucsf.edu<br />

Lab: Ashrafi<br />

Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

New Insights Into Understanding The Regulation Of Fat And Feeding<br />

Kaveh Ashrafi, Katherine Cunningham, Aude Bouagnon, Liron Noiman<br />

<strong>University</strong> <strong>of</strong> California San Francisco, San Francisco, CA, United States<br />

Serotonergic regulation <strong>of</strong> fat content and feeding behavior has been studied intensively<br />

for understanding the basic neurociruitry <strong>of</strong> energy balance in various organisms and as a<br />

therapeutic target for human obesity. Its underlying molecular mechanisms still remain poorly<br />

understood. In C. elegans, serotonin signaling regulates fat content, food intake behavior as<br />

well as a number <strong>of</strong> other behaviors. We have discovered that serotonergic regulation <strong>of</strong> feeding<br />

can be initiated by the chemosensory ADF neurons. A signaling relay comprised <strong>of</strong> the SER-5<br />

receptor, AMP-activated kinase, and glutamatergic signaling links serotonergic signaling from<br />

the ADF chemosensory neurons to the pharyngeal nervous system <strong>of</strong> the animal to modulate<br />

pumping rate. SER-5 mediated regulation <strong>of</strong> AMPK occurs in previously uncharacterized<br />

neurons that are the sole site <strong>of</strong> function <strong>of</strong> hlh-34, the C. elegans counterpart <strong>of</strong> human SIM1,<br />

a transcription factor associated with obesity. These findings highlight the deep evolutionary<br />

origins <strong>of</strong> feeding regulatory mechanisms that operate in the mammalian hypothalamus.<br />

Interestingly, the feeding relay described above does not play a role in serotonergic regulation <strong>of</strong><br />

fat content. Instead, another serotonergic relay comprised <strong>of</strong> a distinct set <strong>of</strong> neurally expressed<br />

serotonergic receptors signal to peripheral pathways to determine whether energetic resources<br />

should be directed to fat storage or fat oxidation pathways. As such, fat content is not simply<br />

a secondary consequence <strong>of</strong> food intake but regulated through neuroendocrine pathways<br />

that function independent <strong>of</strong> feeding regulatory mechanisms. We have discovered that these<br />

feeding-independent, fat regulatory pathways can be acted upon by heavily-used herbicides<br />

and other pollutants that are pervasive in the environment. This challenges the prevalent<br />

dogma that obesity is primarily a behavioral issue arising from either excessive caloric intake<br />

or reduced energy expenditure.<br />

Session 3 Plenary Speaker<br />

17


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

Identification <strong>of</strong> DAF-16 Co-regulators with Tandem Affinity<br />

Purification and MudPIT<br />

Thomas Heimbucher 1 , Zheng Liu 1 , Andrea Carrano 1 , Carine Bossard 1 , Bryan Fonslow 2 ,<br />

Jonathan Yates 2 , Tony Hunter 1 , Andrew Dillin 1<br />

1 The Salk Institute for Biological Studies, La Jolla, CA, USA, Howard Hughes<br />

Medical Institute, Glenn Center for Aging Research, 2 The Scripps Research<br />

Institute, La Jolla, CA<br />

The FoxO transcription factor DAF-16 regulates a wide range <strong>of</strong> organismal functions:<br />

it is involved in C. elegans development and reproduction, in stress response and life span<br />

regulation. Association <strong>of</strong> DAF-16 with diverse binding partners might be crucial for mediating<br />

these heterogeneous functions. To identify DAF-16 regulators we established a biochemical<br />

approach to purify DAF-16 associated proteins. DAF-16 was fused to various epitop-tags. On the<br />

basis <strong>of</strong> a reporter assay it was analyzed whether tagged DAF-16 versions were transcriptionally<br />

active. Their activity was further assessed by their ability to rescue phenotypes <strong>of</strong> a daf-16<br />

null mutant. A functional tagged DAF-16 variant was used to generate a transgenic worm line.<br />

DAF-16 protein complexes were isolated by tandem affinity purification and potential DAF-16<br />

binding partners were identified by tandem mass-spectrometry (MudPIT). In an additional<br />

reporter based screen DAF-16 co-regulators were validated. Their potential roles in dauer<br />

formation, stress resistance and longevity are currently being pursued. We will discuss their<br />

identity and potential functions in insulin/IGF-1 signaling.<br />

This work was supported in part by the Austrian Science Fund (FWF, grant J2734), by a<br />

grant from NIH, and by the Glenn Center for Aging Research.<br />

Contact: theimbucher@salk.edu<br />

Lab: Dillin<br />

18<br />

Session 3


Contact: emilie.demoinet@mcgill.ca<br />

Lab: Roy<br />

Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

AMPK regulates a novel UPR-like response to mediate survival during<br />

nutrient stress in C. elegans<br />

Emilie Demoinet, Julie Mantovani, Richard Roy<br />

McGill <strong>University</strong>, Montreal, Canada<br />

To enhance survival during periods <strong>of</strong> prolonged nutrient stress, many organisms execute<br />

developmental or reproductive diapause, which are reversible states <strong>of</strong> developmental<br />

dormancy. When growth conditions are suboptimal, Caenorhabditis elegans can arrest its<br />

development and execute a diapause like state. The best characterized <strong>of</strong> these are the first<br />

larval stage (L1) and dauer diapause. The C. elegans L1 arrest is a response to an insufficient<br />

level <strong>of</strong> nutrient to initiate postembryonic development, whereby development is suspended<br />

and environmental stress resistance is increased without obvious morphological modification.<br />

Wild type L1 hatchlings can normally survive up to 2 weeks in the absence <strong>of</strong> food under these<br />

conditions. We have shown that maximal survival in the L1 diapause requires aak-2, one <strong>of</strong><br />

the 2 homologues <strong>of</strong> the alpha subunit <strong>of</strong> AMP-activated protein kinase (AMPK). AMPK is a<br />

metabolic master switch that is activated in response to various nutritional and stress signals.<br />

Its main function is to maintain cellular energy homeostasis by enhancing energy generating<br />

pathways, while blocking energy-consuming processes. We found that larvae that lack AMPK<br />

(aak(0)), are compromised for protein folding allowing aggregates to accumulate in the L1<br />

larvae, leading to their premature death after 5 days. The addition <strong>of</strong> glucose, delays aggregate<br />

accumulation and partially rescues survival. A similar accumulation <strong>of</strong> unfolded protein is<br />

observed in starved wild type L1 larvae just prior to death, which is delayed following glucose<br />

addition, while increasing their survival tw<strong>of</strong>old. Accumulation <strong>of</strong> misfolded protein activates a<br />

highly conserved adaptive signaling cascade known as the unfolded protein response (UPR),<br />

which, when activated, can lead to adaptation or death. We have found that in the presence <strong>of</strong><br />

an energetic stress, a previously uncharacterized UPR-like response involving novel effectors<br />

becomes activated to ensure organismal survival. Based on genetic and cell biological data, we<br />

suggest that AMPK regulates this response to prolong survival in the diapause until favorable<br />

energy conditions are restored.<br />

Session 3<br />

19


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

Pathways that regulate Reproductive Aging and Longevity<br />

Coleen Murphy<br />

Princeton <strong>University</strong>, Princeton, NJ, United States<br />

The understanding <strong>of</strong> the regulation <strong>of</strong> early aging is likely to be particularly helpful in<br />

improving quality <strong>of</strong> life. To that end, we study not only life span, but also such early aging<br />

phenotypes as loss <strong>of</strong> reproductive capacity. While the daf-2 Insulin/IGF-1-like signaling<br />

pathway and daf-16/FoxO are critical regulators <strong>of</strong> both <strong>of</strong> these phenotypes, other factors are<br />

emerging as regulators <strong>of</strong> specific aspects <strong>of</strong> aging. For example, TGF-beta dauer signaling<br />

affects longevity, but not reproductive span, while TGF-beta Sma/Mab signaling regulates<br />

reproductive aging but not longevity. Both autonomous and non-autonomous activities play a<br />

role in regulating and coordinating these pathways’ regulation <strong>of</strong> their transcriptional targets<br />

as well as their final phenotypic outputs.<br />

Contact: ctmurphy@princeton.edu<br />

Lab: Murphy<br />

20<br />

Session 3 Plenary Speaker


Contact: dpark@cmmt.ubc.ca<br />

Lab: Taubert<br />

Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

A Novel Role <strong>of</strong> TGF-β Pathway in Dietary Restriction Induced<br />

Longevity<br />

Donha Park 1,2 , Dara Lo 1 , Charles Yang 1 , Donald Riddle 2 , Stefan Taubert 1<br />

1 Centre for Molecular Medicine and Therapeutics, <strong>University</strong> <strong>of</strong> British Columbia,<br />

Vancouver, BC, Canada, 2 Michael Smith laboratories, <strong>University</strong> <strong>of</strong> British<br />

Columbia, Vancouver, BC, Canada<br />

Transforming growth factor β (TGF-β) signaling plays a pivotal role to regulate cell<br />

proliferation, apoptosis, organ development and cancer metastasis. The downstream effector<br />

<strong>of</strong> TGF-β signaling, Smad proteins, interact with variety <strong>of</strong> partner proteins to achieve their<br />

function. To better understand Smad signaling, we performed immunoprecipitation (IP) <strong>of</strong> C.<br />

elegans DAF-8/R-Smad followed by mass spectrometry and identified the TGFβI receptor DAF-<br />

1 and HID-1 (High Temperature induced Dauer) as a novel DAF-8 interacting proteins. Others<br />

previously showed that HID-1 interacts physically with the E3 ubiquitin ligase WWP-1, which<br />

is required for dietary restriction (DR)-induced life span extension. To test whether HID-1 also<br />

affects longevity, we determined the life spans <strong>of</strong> hid-1 mutants and <strong>of</strong> worms over-expressing<br />

hid-1 in ad libitum (AL) and DR conditions. The hid-1 mutants showed a shortened adult life<br />

span, whereas hid-1 overexpression extended life span in AL and DR conditions. Furthermore,<br />

we found that over-expression <strong>of</strong> the TGFβI receptor daf-1 (daf-1oe) also extended worm life<br />

span in DR conditions, and this life span extension was dependent on hid-1. RNAi against skn-<br />

1/bZIP or pha-4/FoxA transcription factors only partially suppressed the daf-1oe mediated life<br />

span extension, indicating that TGF-β regulates DR-induced life span in parallel to skn-1 and<br />

pha-4. Furthermore, daf-7/TGFβ1 transcript and DAF-1 protein levels were maintained in DR<br />

conditions but decreased in AL fed worms over time, indicating that DR conditions may lead<br />

to sustained TGF-β signaling. Lastly, because mobilization <strong>of</strong> stored lipids may be affected in<br />

DR, we quantified the expression <strong>of</strong> several lipid metabolism genes. We found that several lipid<br />

β-oxidation genes, including daf-22, were induced by DR in daf-7 dependent manner, and that<br />

DR failed to extend the life span <strong>of</strong> daf-22 mutants. Overall, our results suggest that TGF-β<br />

signaling plays an important role in DR-induced life span extension, possibly be controlling<br />

lipid metabolism genes.<br />

Session 3<br />

21


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

Mitochondrial Oxidative Stress in C. elegans Alters a Pathway with<br />

Strong Similarities to that <strong>of</strong> Bile Acid Biosynthesis and Secretion in<br />

Vertebrates<br />

Ju-Ling Liu1 , David Desjardins1 , Robyn Branicky1,2 , Luis B. Agellon1 , Siegfried Hekimi1 1 2 McGill <strong>University</strong>, Montreal, Canada, MRC Laboratory <strong>of</strong> Molecular Biology,<br />

Cambridge, UK<br />

Mammalian bile acids (BAs) are oxidized breakdown products <strong>of</strong> cholesterol whose<br />

amphiphilic properties serve in lipid and cholesterol uptake. BAs also act as hormonelike<br />

substances that regulate metabolism. The C. elegans clk-1 mutants sustain elevated<br />

mitochondrial oxidative stress and display a slow defecation phenotype that is sensitive to<br />

the level <strong>of</strong> dietary cholesterol. We found that: 1) The defecation phenotype <strong>of</strong> clk-1 mutants<br />

is suppressed by mutations in tat-2, identified in a previous unbiased screen for suppressors<br />

<strong>of</strong> clk-1. TAT-2 is homologous to ATP8B1, a flippase required for normal BAs secretion in<br />

mammals. 2) The phenotype is suppressed by cholestyramine, a resin that binds BAs. 3) The<br />

phenotype is suppressed by the knock-down <strong>of</strong> C. elegans homologues <strong>of</strong> BA-biosynthetic<br />

enzymes. 4) The phenotype is enhanced by treatment with BAs but not by other detergents.<br />

5) Lipid extracts from C. elegans contain an activity that mimics the effect <strong>of</strong> BAs on clk-1 and<br />

the activity is more abundant in clk-1 extracts. 6) clk-1 and clk-1; tat-2 double mutants show<br />

altered cholesterol content. 7) The clk-1 defecation phenotype is enhanced by high dietary<br />

cholesterol and this effect requires TAT-2. 8) Suppression <strong>of</strong> clk-1 by tat-2 is rescued by BAs,<br />

and this requires dietary cholesterol. 9) The clk-1 phenotypes, including the level <strong>of</strong> activity<br />

in lipid extracts, are suppressed by antioxidants and enhanced by depletion <strong>of</strong> mitochondrial<br />

superoxide dismutases. Our observations suggest that C. elegans synthesizes and secretes<br />

molecules with properties and functions that resemble that <strong>of</strong> BAs. These molecules act in<br />

cholesterol uptake and their level <strong>of</strong> synthesis is up-regulated by mitochondrial oxidative<br />

stress. Future investigations should reveal whether these molecules are in fact BAs, which<br />

would suggest the unexplored possibility that the elevated oxidative stress that characterizes<br />

the metabolic syndrome might participate in disease processes by affecting the regulation <strong>of</strong><br />

metabolism through BAs synthesis.<br />

Contact: ju-ling.liu@mail.mcgill.ca<br />

Lab: Hekimi<br />

22<br />

Session 3


Contact: jmelo@molbio.mgh.harvard.edu<br />

Lab: Ruvkun<br />

Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

Surveillance <strong>of</strong> Core Cellular Processes as a Strategy for Xenobiotic<br />

and Pathogen Recognition<br />

Justine Melo, Gary Ruvkun<br />

Massachusetts General Hospital, Boston, MA USA<br />

The nematode C. elegans is attracted to nutritious bacteria and is repelled by pathogens<br />

and poisons. The basis for discrimination between benign and harmful food sources is poorly<br />

understood. We show that RNAi and toxin-mediated disruption <strong>of</strong> core cellular activities,<br />

including protein translation, oxidative respiration, and protein degradation, stimulate behavioral<br />

avoidance <strong>of</strong> normally attractive bacteria. RNAi <strong>of</strong> these and other essential processes<br />

induced expression <strong>of</strong> drug detoxification and innate immune reporters in the absence <strong>of</strong><br />

toxins or pathogens, suggesting that disruption <strong>of</strong> a single cellular process may be sufficient<br />

to trigger protective behavioral and physiological defenses. Disruption <strong>of</strong> core processes in<br />

individual tissues (such as the intestine or hypodermis) was sufficient to stimulate aversion<br />

behavior, revealing a neuroendocrine axis <strong>of</strong> control that additionally involved Jnk kinase and<br />

serotonin signaling circuits. We propose that surveillance pathways overseeing core cellular<br />

activities allow animals to recognize and reject poisonous food sources, including microbial<br />

pathogens that deploy toxins to sabotage vital host functions. Such a strategy should prove<br />

resilient to pathogen co-evolution and confer resistance to pathogens with which a host has<br />

no evolutionary history.<br />

Session 3<br />

23


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

Abstract Withdrawn<br />

Contact: haynesc@mskcc.org<br />

Lab: Haynes<br />

24<br />

Session 3


Contact: apasquinelli@ucsd.edu<br />

Lab: Pasquinelli<br />

Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

Pinning Down MicroRNA Targets In Vivo<br />

Amy Pasquinelli<br />

UCSD, La Jolla, CA, USA<br />

MicroRNAs (miRNAs) regulate gene expression by guiding Argonaute proteins to specific<br />

target mRNA sequences. Identification <strong>of</strong> bona fide miRNA target sites in animals is challenged<br />

by uncertainties regarding the base-pairing requirements between miRNA and target as well<br />

as the location <strong>of</strong> functional binding sites within mRNAs. To address these problems on a<br />

genome widescale, we developed a biochemical and computational strategy aimed at isolating<br />

and analyzing endogenous mRNA target sequences bound by the Argonaute protein ALG-1 in<br />

Caenorhabditis elegans. C. elegans provides several advantages for identifying miRNA target<br />

sites in vivo: a single Argonaute protein, ALG-1, is largely responsible for miRNA function, a<br />

viable alg-1 genetic mutant control allows for extensive elimination <strong>of</strong> background sequence<br />

noise, a short but well-established list <strong>of</strong> miRNA targets expected to be bound by ALG-1 at<br />

discrete positions is available, and the in vivo binding context <strong>of</strong> endogenous ALG-1:RNA<br />

interactions in live animals is preserved. Using cross-linking and ALG-1immunoprecipitation<br />

coupled with high-throughput sequencing (CLIP-seq), we identified extensive ALG-1 interactions<br />

with specific 3’UTR and coding exon sequences and discovered features that distinguish<br />

miRNA complex binding sites in 3’UTR from other genic regions. Furthermore, these studies<br />

revealed that Argonaute also binds non-codingRNAs and point to a novel role for Argonaute<br />

in the regulation <strong>of</strong> miRNA biogenesis.<br />

Session 4 Plenary Speaker<br />

25


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

Enhancing N-glycosylation Through Activation <strong>of</strong> the Hexosamine<br />

Pathway Improves ER Protein Folding Capacity and Slows Ageing.<br />

Nadia Storm, Martin Denzel, Adam Antebi<br />

Max Planck Institute, Cologne, Germany<br />

Organismal ageing is the progressive loss <strong>of</strong> cellular homeostasis - including protein<br />

homeostasis, which comprises all processes maintaining a functional proteome. The endoplasmic<br />

reticulum (ER) is the site <strong>of</strong> protein synthesis for all secreted and membrane proteins, and<br />

the ER protein folding fidelity is closely monitored. Proper ER protein folding depends on<br />

chaperones, and requires N-glycosylation <strong>of</strong> the nascent peptide chain. Insufficient folding<br />

capacity triggers a stress response pathway, termed the unfolded protein response (UPR). The<br />

UPR signaling pathway is critical for normal C. elegans lifespan and for lifespan extension via<br />

the insulin signaling pathway. We hypothesized that improving ER stress resistance might result<br />

in lifespan extension. To identify novel mutations that improve ER protein folding we carried<br />

out a developmental drug resistance screen. After chemical mutagenesis, we selected for<br />

resistance to tunicamycin, which interferes with ER protein folding by inhibiting N-glycosylation.<br />

Next, we analyzed the lifespan <strong>of</strong> the tunicamycin resistant mutants. We found that increasing<br />

metabolite flux through the hexosamine pathway (HP) by gain-<strong>of</strong>-function mutations <strong>of</strong> the<br />

pathway’s key enzyme, glucosamine-fructose 6-phosphate aminotransferase (gfat-1), results<br />

in ER stress resistance and lifespan extension. The HP provides UDP-N-acetylglucosamine<br />

(UDP-GlcNAc) that is required in the first step <strong>of</strong> N-glycan synthesis, which is inhibited by<br />

tunicamycin. Tunicamycin resistance resulting from GFAT-1 hyperacitvation could be mimicked<br />

by feeding wild type C. elegans with HP intermediates and was DAF-16/FOXO-independent.<br />

GFAT-1 hyperactivation or exposure to UDP-GlcNAc precursors reduced the expression <strong>of</strong><br />

UPR target genes during ER stress, as seen in the gain-<strong>of</strong>-function mutants, suggesting an<br />

elevated threshold for UPR activation resulting from increased folding capacity. Increased HP<br />

metabolite flux further alleviated pathology in nematode models <strong>of</strong> proteotoxic stress, such as<br />

polyglutamine expansion, a model for Huntington’s disease. These data, together with GFAT-1’s<br />

high degree <strong>of</strong> conservation makes the HP a potential target for the treatment <strong>of</strong> age-related<br />

proteotoxic diseases in humans.<br />

Contact: nstorm@age.mpg.de<br />

Lab: Antebi<br />

26<br />

Session 4


Contact: yutao.chen@duke.edu<br />

Lab: Baugh<br />

Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

ins-5 and ins-6 Promote Post-embryonic Development in Response to<br />

Feeding<br />

Yutao Chen, Ryan Baugh<br />

Department <strong>of</strong> Biology and IGSP Center for Systems Biology, Duke <strong>University</strong>,<br />

Durham, NC, USA<br />

Animals must coordinate development with fluctuating nutrient availability. Nutrient availability<br />

governs post-embryonic development in C. elegans: larvae that hatch in the absence <strong>of</strong> food<br />

do not initiate post-embryonic development but enter “L1 arrest” (or “L1 diapause”) and can<br />

survive starvation for weeks. Insulin-like signaling is a key regulator <strong>of</strong> L1 arrest and recovery.<br />

However, the C. elegans genome encodes 40 putative insulin-like peptides (ILPs), and there is<br />

evidence that they can function to promote development (“agonists”) or arrest (“antagonists”).<br />

We used the nCounter platform to measure high-resolution mRNA expression dynamics for all<br />

40 ILPs in response to feeding and fasting in recently hatched L1 larvae. Expression <strong>of</strong> 18 <strong>of</strong><br />

the ILPs is significantly affected by switching nutrient conditions. We classified several ILPs<br />

as candidate agonists or antagonists based on expression. Destabilized YFP reporter gene<br />

analysis confirms nutritional control <strong>of</strong> transcription and reveals the intestine as the primary<br />

site <strong>of</strong> transcriptional regulation. Phenotypic analysis during recovery from L1 arrest reveals<br />

that ins-5 and ins-6 deletion mutants have retarded developmental dynamics affecting M and<br />

V-lineage cell divisions. This is the first phenotype reported for ins-5, and the role <strong>of</strong> ins-6 in L1<br />

recovery is consistent with its role in dauer exit. Other ILPs that regulate dauer formation, life<br />

span or germ line proliferation do not have detectable effects on L1 development. Together these<br />

results suggest specificity <strong>of</strong> ILPs function, even among agonists. By combining expression<br />

and phenotypic analysis, we identified 2 ILPs that promote rapid post-embryonic development<br />

in response to feeding, and we report additional functional predictions based on expression<br />

analysis.<br />

Session 4<br />

27


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

A New Class <strong>of</strong> Stress Resistance Genes<br />

Aimee Kao, Ayumi Nakamura, Meredith Judy, Anne Huang, Cynthia Kenyon<br />

<strong>University</strong> <strong>of</strong> California San Francisco, San Francisco, CA<br />

Animals have many ways <strong>of</strong> protecting themselves against stress; for example, they can<br />

induce stress-protective pathways or they can kill damaged cells via apoptosis. How the<br />

choice between these different options is made is not well understood. Here we show that C.<br />

elegans mutations that block the normal course <strong>of</strong> programmed cell death confer resistance<br />

to a specific set <strong>of</strong> environmental stressors: heat, ER and osmotic stress. Surprisingly, ced-3<br />

caspase mutations that block apoptosis; ced-1 and other engulfment mutations that prevent<br />

apoptotic cell removal, and progranulin (pgrn-1) mutations that accelerate dying cell removal,<br />

all increased stress resistance. Because it is a gene that is associated with multiple diseases<br />

in humans, including neurodegeneration, auto-immunity and cancer, we characterized pgrn-1<br />

mutant-associated stress resistance in detail. We found that the ER stress resistance elicited<br />

by pgrn-1 mutations exhibits genetic dependency on the unfolded protein response (UPR)<br />

factors IRE-1 and PEK-1, the MAP kinase PMK-1 and the stress-related transcription factor<br />

DAF-16/FOXO. Mutations in pgrn-1 and the ced genes may induce a common pathway, since<br />

they do not exhibit additive effects. To explain these findings, we propose that C. elegans has a<br />

surveillance system that detects perturbations in the normal cell death pathway and responds<br />

to them by inducing an alternative, organism-wide stress-protective pathway. These genes<br />

represent a new class <strong>of</strong> stress response regulators.<br />

Contact: akao@memory.ucsf.edu<br />

Lab: Kenyon/Kao<br />

28<br />

Session 4


Contact: asoukas@chgr.mgh.harvard.edu<br />

Lab: Ruvkun/Soukas<br />

Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

Genetic Regulation <strong>of</strong> Age Pigment and Nile Red Accumulation in the<br />

Lysosome Related Organelle<br />

Alexander Soukas 1 , Christopher Carr 2 , Gary Ruvkun 1<br />

1 Massachusetts General Hospital, Boston, MA, USA, 2 Massachusetts Institute <strong>of</strong><br />

Technology, Cambridge, MA, USA<br />

When fed to C. elegans, the vital dye Nile red highlights the cellular compartment known<br />

as the lysosome related organelle. While work by us and others indicates that this cellular<br />

compartment is distinct from canonical lipid droplets, the lysosome related organelle is well<br />

implicated in aging and metabolism as it is the site <strong>of</strong> accumulation <strong>of</strong> lip<strong>of</strong>uscin, or age pigment.<br />

We have found that both aut<strong>of</strong>luorescence and feeding Nile red accumulate to a greater<br />

extent in short-lived C. elegans, and are therefore seem to be markers <strong>of</strong> progeria. To test this<br />

systematically, we have conducted large-scale genetic and genomic screens for altered feeding<br />

Nile red accumulation andaut<strong>of</strong>luorescence. ~80 genes have been confirmed to alter Nile red<br />

accumulation, and we have examined these using RNAi and quantitative, high-throughput<br />

microscopy in genetic backgrounds which have altered age pigment accumulation. The results<br />

begin to shed light on the complex genetics underlying age pigment formation and accumulation<br />

and mutants obtained from forward genetics demonstrate that it is possible to genetically<br />

separate feeding Nile red accumulation from aut<strong>of</strong>luorescent age pigment accumulation. The<br />

gene networks identified may shed light on fundamentals <strong>of</strong> the aging process as well as<br />

regulation <strong>of</strong> the lysosome related organelle and its role in aging and metabolism.<br />

Session 4<br />

29


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

A Necrotic Cascade Accelerates Stress-Induced Death and Causes a<br />

Burst <strong>of</strong> Anthranilic Acid Fluorescence in C. elegans<br />

Cassandra Coburn 1 , Parag Mahanti 2 , Abraham Mandel 1 , Filip Matthijssens 3 , Bart<br />

Braeckman 3 , Frank Schroeder 2 , David Gems 1<br />

1 <strong>University</strong> College London, London, UK, 2 Cornell <strong>University</strong>, Ithaca, NY, USA,<br />

3 Ghent <strong>University</strong>, Ghent, Belgium<br />

Under UV light, C. elegans gut granules (which are lysosome-related organelles) emit<br />

intense blue fluorescence. It has been suggested that the fluorescent material is lip<strong>of</strong>uscin, a<br />

heterogeneous aggregate <strong>of</strong> damaged proteins and lipids, that can accumulate with age within<br />

the lysosomes <strong>of</strong> mammalian cells. Aut<strong>of</strong>luorescence increases with age in worm cohorts and<br />

so has commonly been used as a biomarker <strong>of</strong> aging. Lip<strong>of</strong>uscin accumulation in aging worms<br />

would be consistent with the notion that aging is caused by the accumulation <strong>of</strong> molecular<br />

damage. However, several observations imply that the blue fluorescent substance in worms<br />

is not lip<strong>of</strong>uscin. We have observed that oxidative stress does not increase blue fluorescence<br />

levels. Moreover, when individual worms are monitored throughout life, blue fluorescence<br />

only increases at the point <strong>of</strong> death (“death fluorescence”). Death fluorescence is also seen<br />

when young worms are killed. To identify the blue fluorophore we compared wild type (N2)<br />

with glo-1(zu437) (Gut granule LOss) mutants, which also do not exhibit death fluorescence,<br />

using a 2D NMR-based comparative metabolomic approach. This identified an anthranilic acid<br />

glucosyl ester and its 3’-phosphorylated derivative as the source <strong>of</strong> the blue fluorescence.<br />

Anthranilic acid is derived from tryptophan by action <strong>of</strong> the kynurenine pathway, where the<br />

rate limiting enzyme is tryptophan 2,3-dioxygenase (TDO). RNAi knockdown <strong>of</strong> the putative<br />

TDO C28H8.11 abrogated both gut granule and death fluorescence. (This provides a useful<br />

tool to aid studies <strong>of</strong> fluorescent reporters <strong>of</strong> gut-expressed genes, which can be masked by<br />

gut granule fluorescence). Our work has shown that death fluorescence is generated by a<br />

systemic cascade <strong>of</strong> cellular necrosis. We are currently investigating the role <strong>of</strong> this cascade,<br />

and <strong>of</strong> the kynurenine pathway, in aging and death. Our findings show that blocking necrosis<br />

or anthranilate synthesis extends lifespan in stressed animals. This suggests that systemic<br />

necrosis and the associated burst <strong>of</strong> anthranilate production can hasten organismal death.<br />

Contact: david.gems@ucl.ac.uk<br />

Lab: Gems<br />

30<br />

Session 4


Contact: felix@biologie.ens.fr<br />

Lab: Felix<br />

Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

The real life <strong>of</strong> Caenorhabditis and their natural pathogens<br />

Marie-Anne Felix1 , Fabien Duveau1 , Tony Belicard1 1Institute <strong>of</strong> Biology <strong>of</strong> the Ecole Normale Supérieure, CNRS-Inserm-ENS, 46 rue<br />

d’Ulm, 75230 Paris cedex 05, France, 1Institute <strong>of</strong> Biology <strong>of</strong> the Ecole Normale<br />

Supérieure, CNRS-Inserm-ENS, 46 rue d’Ulm, 75230 Paris cedex 05, France<br />

C. elegans is a major laboratory model organism, yet its life and variation in the wild are<br />

little known. Especially, other organisms occurring in the habitats where C. elegans feeding<br />

stages are found are likely to be important players in its ecology, as potential food, pathogens<br />

or predators.<br />

We find natural populations <strong>of</strong> Caenorhabditis species in rotting vegetal matter, such<br />

as fruits, flowers and stems. Population counts may span a range <strong>of</strong> 1 to over 10,000<br />

Caenorhabditis individuals in a single fruit or stem. Some populations with an intermediate<br />

census size (10-1000) contained no dauer larvae at all, whereas larger populations always<br />

included some larvae in the pre-dauer or dauer stages. It appears thus likely that C. elegans<br />

undergoes a population boom upon encountering a favorable rotting substrate, separated by<br />

periods <strong>of</strong> migration in the dauer stage.<br />

In our surveys in France, C. elegans and C. briggsae were found to co-occur on the same<br />

substrates. We followed their respective spatio-temporal distribution in an apple orchard over<br />

four years and found a seasonal distribution that matches their temperature preference in the<br />

laboratory in competition experiments.<br />

Wild Caenorhabditis were observed to <strong>of</strong>ten harbor a live bacterial flora in their intestinal<br />

lumen. In some instances, the flora proliferates and a large bacterial plug obstructs the whole<br />

intestinal tract.<br />

Pathogens provide strong and changing selection pressures. Natural pathogens are thus<br />

relevant to study the defense systems <strong>of</strong> C. elegans and their potentially rapid evolution, and<br />

may provide interesting models <strong>of</strong> co-evolution between host and pathogen. Several natural<br />

pathogens <strong>of</strong> C. elegans were isolated, including microsporidia, bacteria, fungi and recently<br />

the first viruses that infect C. elegans or C. briggsae (Félix, Ashe, Piffaretti et al. PLoS Biol.<br />

2011). Different C. elegans wild isolates show widely different sensitivity to the Orsay virus, as<br />

assayed by viral load upon infection. A genome-wide association study <strong>of</strong> Orsay virus sensitivity<br />

in a worldwide set <strong>of</strong> C. elegans isolates indicates one major locus segregating in the species<br />

(see abstracts by T. Belicard et al. and J. Le Pen et al.).<br />

Thanks to the many colleagues who helped this work, especially the labs <strong>of</strong> Dave Wang and Eric Miska,<br />

Emily Troemel, Jonathan Hodgkin, Nathalie Pujol, Buck Samuel and all fellow samplers!<br />

Session 4 Plenary Speaker<br />

31


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

Insulin Signaling and Dietary Restriction Differentially Regulate<br />

Glucose Metabolism to Impact C. elegans Healthspan<br />

Brian Onken, Monica Driscoll<br />

Rutgers <strong>University</strong><br />

A major goal <strong>of</strong> aging research is to understand the underlying relationship between<br />

nutritional intake, metabolism, and healthy aging. Low-glycemic index diets have been shown<br />

to reduce risk <strong>of</strong> age-related metabolic diseases such as diabetes and cardiovascular disease,<br />

and reduced caloric intake via dietary restriction increases healthspan across species. One<br />

potential approach for supporting healthy aging is via interventions that engage healthspanpromoting<br />

metabolism.<br />

In Caenorhabditis elegans, adding excess glucose to the growth medium shortens<br />

lifespan [1, 2], while inhibiting the glycolytic enzyme hexokinase with the glucose analog<br />

2-deoxyglucose increases lifespan [1]. We have shown that disrupting genes encoding two<br />

other glycolytic enzymes that catalyze unidirectional, irreversible reactions in glycolysis<br />

lengthens Caenorhabditis elegans median lifespan, induces large gains in youthful locomotory<br />

ability, and triggers a fluorescent biomarker that distinguishes a healthy metabolic state.<br />

Conversely, disrupting counterpart unidirectional gluconeogenic genes decreases nematode<br />

healthspan. In investigating potential longevity-related pathways that might impinge upon<br />

glucose metabolism, we found that disrupting glycolytic genes increases healthspan through<br />

the FOXO transcription factor DAF-16, which is also required for the increased lifespan seen<br />

with lowered levels <strong>of</strong> insulin signaling, and which is downregulated by increased glucose<br />

availability [2]. Strikingly, we also found that gluconeogenic activity is specifically required for<br />

increased healthspan under dietary restriction, and that the SKN-1 transcription factor, which<br />

is required for the beneficial effects <strong>of</strong> dietary restriction [3], is also needed for the healthspan<br />

effects seen with decreased gluconeogenesis. These results provide evidence for an intriguing<br />

new paradigm: breakdown <strong>of</strong> glucose via glycolysis negatively impacts healthy aging through<br />

insulin signaling and DAF-16, while dietary restriction engages the reciprocal gluconeogenic<br />

pathway to promote healthspan via SKN-1. Our observations support that healthspan might be<br />

optimized via dietary, pharmacological, or genetic interventions that increase gluconeogenic<br />

activity or decrease glycolysis.<br />

1. Schulz TJ, Zarse K, Voigt A, Urban N, Birringer M, et al. (2007). Cell Metab 6: 280-293.<br />

2. Lee SJ, Murhpy CT, Kenyon C (2009). Cell Metab 10: 379-391.<br />

3. Bishop NA, Guarente L (2007). Nature 447: 545-549.<br />

Contact: onken@biology.rutgers.edu<br />

Lab: Driscoll<br />

32<br />

Session 4


Contact: sszumows@ucsd.edu<br />

Lab: Troemel<br />

Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

Exit <strong>of</strong> the Intracellular Pathogen Nematocida parisii from C. elegans<br />

Intestinal Cells<br />

Suzannah Szumowski, Kathleen Estes, Margery Smelkinson, Emily Troemel<br />

<strong>University</strong> <strong>of</strong> California San Diego, San Diego, (CA), USA, <strong>University</strong> <strong>of</strong> California<br />

San Diego, San Diego, (CA), USA<br />

Microsporidia comprise a diverse phylum <strong>of</strong> fungal-related pathogens that infect a broad<br />

range <strong>of</strong> hosts, including insects and humans. As obligate intracellular parasites, microsporidia<br />

are dependent on their hosts for replication. Although they likely usurp many host processes,<br />

very little is known about the mechanisms <strong>of</strong> pathogenesis used by these ubiquitous microbes.<br />

Recently we found that the microsporidia Nematocida parisii reorganizes the host C. elegans<br />

actin cytoskeleton and terminal web prior to making a non-lytic exit from host cells. We found<br />

that N. parisii spores exit from the apical side <strong>of</strong> intestinal cells into the lumen, and that they are<br />

free <strong>of</strong> host membrane after exit. By studying how N. parisii exits from cells while minimizing<br />

damage to the host we expect to learn about key mechanisms <strong>of</strong> microsporidia transmission,<br />

as well as intestinal cell biology and intracellular trafficking.<br />

Our previous studies indicated that actin was required for N. parisii exit from intestinal<br />

cells, but we did not know the underlying mechanism. We now show that actin forms distinct<br />

“coats” on spores localized near the apical membrane. The number <strong>of</strong> actin coats in an animal<br />

is positively correlated with the number <strong>of</strong> spores that are shed. In addition, animals with<br />

actin-coated spores are more contagious than animals without actin-coated spores. Thus,<br />

we believe that actin-coated spores are exiting the host, and we are developing methods to<br />

image this process in vivo. So far, this process strongly resembles “kiss and coat” exocytosis<br />

where fusing membranes trigger actin polymerization in order to stabilize exiting cargo that<br />

is large or insoluble.<br />

Consistent with exocytic membrane topology, we observe intracellular host membrane<br />

around microsporidia prior to exit. Early during infection, microsporidia appear to be in direct<br />

contact with the cytoplasm, but later in infection EM analysis shows host membrane surrounding<br />

the pathogen. Thus, we believe that during development microsporidia steal intracellular<br />

membrane from the host. We hypothesize that this membrane comes from the mitochondria,<br />

based on imaging and analysis <strong>of</strong> mitochondrial mutants. In this way, microsporidia appear<br />

to usurp host processes in order to proceed through their life cycle and exit non-lytically from<br />

host cells.<br />

Session 4<br />

33


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

ULP-4 SUMO protease regulates mitochondria homeostasis during C.<br />

elegans development and mitochondrial stress<br />

Amir Sapir1 , Assaf Tsur2 , Thijs Thijs Koorman3 , Mike Boxem3 , Paul Sternberg1 , Limor<br />

Broday2 1Howard Hughes Medical Institute and Division <strong>of</strong> Biology, California Institute <strong>of</strong><br />

Technology, Pasadena, CA 91125, USA, 2Department <strong>of</strong> Cell and Developmental<br />

Biology, Sackler School <strong>of</strong> Medicine, Tel Aviv <strong>University</strong>, Tel Aviv 69978, Israel,<br />

3Developmental Biology, Utrecht <strong>University</strong>, Utrecht 3584, The Netherlands<br />

Mitochondria energy production and metabolism must be tightly and dynamically regulated<br />

to meet the different energetic and metabolic requirements <strong>of</strong> normal physiology, aging, and<br />

stress. Our understanding <strong>of</strong> the molecular mechanisms controlling these condition-dependent<br />

modulations <strong>of</strong> mitochondria activity, however, remains rudimentary at best. We hypothesize<br />

that post translational modification <strong>of</strong> mitochondrial proteins by organelle-specific effectors is<br />

taking place for a global regulation <strong>of</strong> mitochondria homeostasis. To address this hypothesis,<br />

we combined RNAi screening with protein localization and bioenergetics studies in C. elegans<br />

. We screened different members <strong>of</strong> the ubiquitin or SUMO effectors known to regulate protein<br />

post-translational modification. Focusing on the SUMO pathway, we identified a predicted<br />

SUMO protease, ulp-4, and found it to exhibits a complex age related and mitochondrial stressdependant<br />

expression and recruitment to the mitochondria <strong>of</strong> body wall muscles. To identify the<br />

transcription factors regulating ulp-4 expression, we conducted a small scale RNAi screen <strong>of</strong><br />

candidate transcription factors known to function during stress and aging. We Identified atsf-1,<br />

a transcription factor that is activated by mitochondrial stress, as a regulator <strong>of</strong> ulp-4 stressdependent<br />

expression. In concurrence with ULP-4 mitochondrial localization, ulp-4 mutant<br />

worms exhibit a strong age dependent attenuation <strong>of</strong> muscle activity and abnormal mitochondria<br />

morphology presumably stemming from a reduction <strong>of</strong> mitochondria activity. ULP-4 protein<br />

localization into inner organelle domains supports a role <strong>of</strong> ULP-4 in regulating matrix proteins.<br />

To identify the mitochondrial targets <strong>of</strong> ULP-4, we screened C. elegans two-hybrid libraries and<br />

found association with phi-50, an orthologous gene <strong>of</strong> human 3-hydroxy-3-methylglutaryl CoA<br />

synthase that functions in the metabolic switch <strong>of</strong> mitochondria toward stress and starvation<br />

induced ketogenesis. Our results are emerging toward a regulatory pathway in which one <strong>of</strong> the<br />

molecular readouts <strong>of</strong> various environmental and physiological stressors is atsf-1 dependent<br />

ulp-4 expression in the body wall muscles. This expression is followed by ULP-4 protein<br />

recruitment to the mitochondria matrix where it may regulate the sumoylation state <strong>of</strong> PHI-50<br />

to support the metabolic switch toward mitochondrial stress-induced ketogenesis.<br />

Contact: amirsa@caltech.edu<br />

Lab: Broday<br />

34<br />

Session 4


Contact: jsimske@metrohealth.org<br />

Lab: Simske<br />

Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

The Diet <strong>of</strong> Worms: Expression <strong>of</strong> the agl-1 (Glycogen Debranching<br />

Enzyme) Embryonic Arrest Phenotype Depends on Maternal Diet<br />

Jeff Simske<br />

Rammelkamp Center, Cleveland (OH), USA<br />

Previous analysis <strong>of</strong> agl-1 demonstrated that reduction in agl-1 activity results in reduced<br />

activity <strong>of</strong> AMPK, presumably though inhibition <strong>of</strong> AMPK via branched glycogen (limit dextrin)<br />

binding to the AMPKβ subunit AAKB-1 and/orAAKB-2. An ongoing challenge in studying agl-1<br />

mutants has been a mysterious loss <strong>of</strong> phenotype. Ultimately it was revealed that phenotypic<br />

expression depends on bacterial diet. Specifically, agl-1 mutants grown on freshly seeded,<br />

live OP50 display little embryonic arrest. Conversely, when grown on UV-killed OP50, agl-1<br />

hermaphrodites produce almost 100% arrested embryos. Similar agl-1 phenotypes are observed<br />

when grown on glucose or glycerol plates seeded with fresh OP50. A similar connection between<br />

bacterial viability and agl-1 phenotype was also observed when the drug metformin was tested<br />

for the ability to suppress agl-1 phenotypes through AMPK activation. When plated on media<br />

containing metformin, growth <strong>of</strong> OP50 itself is attenuated, and the phenotype <strong>of</strong> agl-1 mutants<br />

is enhanced. Conversely, metformin present in UV-killed bacterial plates or in glucose plates<br />

is able to suppress agl-1 phenotypes, but does not prevent bacterial growth in the presence<br />

<strong>of</strong> elevated glucose. Notably, bacteria grown on glucose or metformin+glucose are different in<br />

consistency than OP50 on normal NGM plates. These results suggest that metformin and dietary<br />

supplements influence agl-1 phenotypes primarily, but not exclusively through their effect on<br />

the bacterial food source. To test whether metformin exclusively acts via diet, metformin was<br />

injected directly into the pseudocoelom or gonad <strong>of</strong> agl-1 mutants grown on UV-killed OP50;<br />

agl-1 phenotypes were mildly suppressed. Furthermore, unlike UV-killed bacteria, live bacteria<br />

+ metformin diet resulted in animals with reduced brood size, and a pale, sickly appearance.<br />

Thus metformin affects worm physiology both through bacteria-dependent and independent<br />

pathways. Interestingly, the dietary maternal effect is reversible: when agl-1 hermaphrodites<br />

laying arrested embryos are shifted from dead or glucose-supplemented bacteria to live OP50,<br />

embryos hatch after a day or so and hermaphrodites shifted from ‘permissive’ diet to UV-killed<br />

bacteria generate dead embryos in a similar time-span. In all experimental conditions tested,<br />

severity <strong>of</strong> agl-1 phenotype correlates with reduction in AMPK activity. Ongoing research is<br />

focused on determining the metabolic factors that suppress agl-1 phenotypes.<br />

Session 4<br />

35


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

Fatty acid desaturase activity regulates lipid droplet size<br />

Jennifer Watts<br />

Washington State <strong>University</strong>, Pullman, WA, United States<br />

Fatty acid desaturation regulates membrane functions as well as fat storage in animals. C.<br />

elegans contains all <strong>of</strong> the enzymes required to synthesize long-chain polyunsaturated fatty<br />

acids, including arachidonic and eicosapentaenoic acid, from acetyl-CoA. The first double bond<br />

in a saturated fatty acyl chain is inserted by the D9 desaturases, also known as stearoyl-CoA<br />

desaturases (SCDs). C. elegans fat-6;fat-7 double mutants cannot catalyze the D9 desaturation<br />

<strong>of</strong> 18:0, leading to worms with high levels <strong>of</strong> 18:0 and a deficiency <strong>of</strong> C20 PUFAs. Similar to<br />

SCD1 knockout mice, these mutants have low fat stores and elevated rates <strong>of</strong> fat oxidation.<br />

Visualization <strong>of</strong> fat stores in the fat-6;fat-7 double mutants revealed small size droplets<br />

compared to wild type. We have constructed triple mutant strains in order to combine high fat<br />

mutants, such as daf-2, with the low fat SCD mutants. We found that droplet size resembles<br />

the fat-6;fat-7 in all triple mutant combinations, implying that SCD activity is necessary for the<br />

formation <strong>of</strong> large-sized lipid droplets. While most triple mutant strains have reduced fat stores<br />

as well as small lipid droplets, daf-2;fat-6;fat-7 triple mutants are able to store higher levels<br />

<strong>of</strong> triacyglycerols, even though the lipid droplet size is small. These studies reveal a specific<br />

role <strong>of</strong> stearoyl-CoA desaturase in regulating lipid droplet size.<br />

Contact: jwatts@wsu.edu<br />

Lab: Watts<br />

36<br />

Session 5 Plenary Speaker


Contact: stroustr@fas.harvard.edu<br />

Lab: Fontana<br />

Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

The Lifespan Machine: A scalable, automated microscope increases<br />

the throughput and statistical quality <strong>of</strong> nematode lifespan and stress<br />

resistance assays<br />

Nicholas Stroustrup, Javier Apfeld, Walter Fontana<br />

Harvard Medical School<br />

Survival assays can provide evidence <strong>of</strong> a gene or environment’s role in aging and<br />

stress resistance. The interpretability <strong>of</strong> these widely-used experiments hinges crucially on<br />

the accuracy and precision with which measurements can be acquired. To address current<br />

limitations in the throughput, statistical power, and replicability <strong>of</strong> manual survival assays, we<br />

have developed an automated system for the acquisition and analysis <strong>of</strong> C. elegans mortality<br />

data. Our goal for this technology is not only to automate and improve the accuracy <strong>of</strong> the<br />

standard methodology, but also to remain low-cost and scalable.<br />

We present our recent experiences operating a microscope consisting <strong>of</strong> fifty flatbed<br />

scanners, which together monitor 30,000 individuals across 800 agar plates and 2.5 square<br />

meters <strong>of</strong> agar lawn, imaged every fifteen minutes at 8 um resolution. Our automated s<strong>of</strong>tware<br />

analyzes this large volume <strong>of</strong> data to identify the death times <strong>of</strong> individual worms based on<br />

their movement, with a focus on subtle, late-life postural changes. Our toolset allows rapid<br />

visual validation <strong>of</strong> automated results, a critical step for the routine production <strong>of</strong> statistically<br />

rigorous mortality data.<br />

We discuss our results from a focused, high-throughput thermotolerance screen, as well as<br />

a high-resolution analysis <strong>of</strong> the lifespan <strong>of</strong> several insulin/IGF pathway mutants. We present<br />

our characterization <strong>of</strong> the strengths and limitations in sensitivity and reliability <strong>of</strong> C.elegans<br />

lifespan and stress resistance assays. We hope to spread our technology as a means for<br />

increasing the consistency and reproducibility <strong>of</strong> survival data between labs, and to support the<br />

accumulation <strong>of</strong> a self-consistent body <strong>of</strong> demographic aging data in the C. elegans community.<br />

Session 5<br />

37


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

Novel endogenous ligands <strong>of</strong> DAF-12 nuclear hormone receptor<br />

revealed by comparative metabolomics<br />

Parag Mahanti1 , Neelanjan Bose1 , Axel Bethke1 , Joshua Judkins1 , Joshua Wollam3 ,<br />

Kathleen Dumas2 , Anna Zimmerman1 , Patrick Hu2,5 , Adam Antebi3,4 , Frank Schroeder1 1Boyce Thompson Institute and Department <strong>of</strong> Chemistry and Chemical Biology,<br />

Cornell <strong>University</strong>, Ithaca, (NY), USA, 2Life Sciences Institute, <strong>University</strong> <strong>of</strong><br />

Michigan, Ann Arbor, (MI), USA, 3Max Planck Institute for Biology <strong>of</strong> Aging,<br />

Cologne, Germany, 4Huffington Center on Aging, Dept. <strong>of</strong> Molecular and Cellular<br />

Biology, Baylor College <strong>of</strong> Medicine, Houston, (TX), USA, 5Dept. <strong>of</strong> Internal<br />

Medicine and Cell and Developmental Biology, UMich Medical School, Ann Arbor,<br />

(MI), USA<br />

Small-molecule ligands <strong>of</strong> nuclear hormone receptors (NHRs) play a central role in the<br />

regulation <strong>of</strong> metazoan development, cell differentiation and metabolism. While small changes<br />

in ligand structures strongly affects NHR transcriptional activity, the endogenous ligands <strong>of</strong><br />

many metazoan NHRs still remain poorly characterized.<br />

Using comparative metabolomics, we identified the endogenous ligands <strong>of</strong> the C. elegans<br />

NHR DAF-12, a vitamin D and liver X receptor homolog that regulates larval development and<br />

adult lifespan. The identified molecules show strong bio-activity and include only one <strong>of</strong> two<br />

previously predicted DAF-12 ligands and feature unexpected structural motifs reminiscent <strong>of</strong><br />

mammalian bile acids.<br />

Comparative metabolomics <strong>of</strong> mutants <strong>of</strong> steroid-modifying enzymes provides evidence<br />

for differential regulation <strong>of</strong> DAF-12-ligand biosyntheses, indicating that different endogenous<br />

NHR ligands may serve partially divergent functions. Our results show that previous, classical<br />

genetics-based hypotheses about NHR signaling in C. elegans must be revised and demonstrate<br />

the utility <strong>of</strong> unbiased comparative metabolomics for the identification <strong>of</strong> metazoan NHR ligands.<br />

Contact: parag.mahanti@gmail.com<br />

Lab: Schroeder<br />

38<br />

Session 5


Contact: haynesc@mskcc.org<br />

Lab: Haynes<br />

Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

Direct Detection <strong>of</strong> Mitochondrial Dysfunction by the Transcription<br />

Factor ATFS-1<br />

Cole Haynes<br />

Memorial Sloan-Kettering Cancer Center<br />

Mitochondrial dysfunction contributes to the ill-effects <strong>of</strong> aging, diseases including cancer<br />

and Parkinson’s, as well as bacterial infection. The mechanisms organisms employ to protect<br />

cells from mitochondrial dysfunction are poorly understood. Our previous studies indicated<br />

that perturbations to mitochondrial function up-regulated mitochondrial chaperone genes via<br />

a signaling pathway termed the mitochondrial unfolded protein response (UPRmt), however<br />

the mechanism by which mitochondrial stress is detected and the signal communicated to the<br />

nucleus has remained elusive. An RNAi screen identified components required for signaling the<br />

stress response including the transcription factor ATFS-1. Consistently, worms lacking ATFS-<br />

1 are much more susceptible to mitochondrial dysfunction than wild-type worms. Here, we<br />

demonstrate that ATFS-1 directly monitors mitochondrial function to coordinate the induction<br />

<strong>of</strong> the UPRmt. Once activated, ATFS-1 mediates a broad transcriptional response <strong>of</strong> over 350<br />

genes to ameliorate the cellular and organismal effects <strong>of</strong> mitochondrial dysfunction. Included,<br />

are the expected mitochondrial chaperone and protease genes as well as ROS-detoxification<br />

machinery located throughout the cell. Components required for mitochondrial fission as well<br />

as autophagy were also induced by ATFS-1. Perhaps most interesting, we did not observe any<br />

induction <strong>of</strong> the electron transport chain genes. However, ATFS-1 did induce components <strong>of</strong> the<br />

glycolysis pathway suggesting that ATFS-1 mediates a shift in metabolism during mitochondrial<br />

dysfunction. The role <strong>of</strong> ATFS-1 and the UPRmt during physiological scenarios such as aging<br />

and bacterial infection will be discussed.<br />

Session 5<br />

39


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

Dissecting opposing age-dependent contributions <strong>of</strong> JNK signaling to<br />

stress resistance<br />

Kwame Twumasi-Boateng2,1 , Kuang-Hui Lee1 , Ali Salehpour1 , Michael Shapira1 1 2 Dept. <strong>of</strong> Integrative Biology, UC Berkeley (CA) USA, Graduate Group in<br />

Microbiology<br />

Physiological responses to adverse environmental conditions attempt to protect the animal<br />

from the relevant threat and maintain homeostasis. However, stress responses can also<br />

be detrimental, and this is usually thought to be the by-product <strong>of</strong> prolonged or excessive<br />

activation. We have recently identified an example in which the contribution <strong>of</strong> a stress<br />

protective mechanism changed from beneficial to detrimental without any difference in its<br />

activation, solely determined by the age <strong>of</strong> the animal (Twumasi-Boateng (2012) Aging Cell,<br />

in press). The age-dependent reversal was identified in the contribution <strong>of</strong> the conserved C.<br />

elegans JNK homolog, KGB-1. From providing protection against heavy metals and protein<br />

folding stress during development it becomes detrimental during early adulthood, sensitizing<br />

animals to environmental stress and shortening their lifespan. One mediator <strong>of</strong> kgb-1’s agedependent<br />

contributions was found to be the conserved longevity-associated transcription<br />

factor DAF-16 – DAF-16’s intestinal function was enhanced by KGB-1 in developing larvae,<br />

but attenuated in adults. Genetic analyses in daf-16 mutants corroborated the hypothesis that<br />

kgb-1-dependent lifespan shortening was mediated by DAF-16 attenuation, but showed no<br />

involvement <strong>of</strong> daf-16 in other kgb-1-dependent phenotypes, most intriguingly in its beneficial<br />

contribution to larval protection from heavy metals. Seeking to identify additional mediators <strong>of</strong><br />

kgb-1-dependent and age-dependent phenotypes, we used microarray analysis to compare<br />

gene expression patterns between kgb-1 mutants and wildtype animals. These analyses<br />

revealed a large group <strong>of</strong> genes induced following kgb-1 activation independently <strong>of</strong> daf-16,<br />

among them genes induced in an age-specific manner, and further helped recognizing the<br />

extra-intestinal involvement <strong>of</strong> an additional transcription factor, FOS-1 (a component <strong>of</strong> the AP-1<br />

stress transcription factor), in the increased heavy metal resistance following KGB-1 activation<br />

in developing worms. We are continuing in de-constructing the age-dependent contribution <strong>of</strong><br />

kgb-1 to stress responses and lifespan, and in characterizing the involvement <strong>of</strong> downstream<br />

processes and mediators. Our results to date depict KGB-1 as a hub for age-dependent and<br />

tissue-specific interactions and suggest that age is an important context determining whether<br />

KGB-1 activation would be beneficial or detrimental.<br />

Contact: mshapira@berkeley.edu<br />

Lab: Shapira<br />

40<br />

Session 5


Contact: pathu@umich.edu<br />

Lab: Hu<br />

Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

A Genetic Screen for Novel DAF-16/FoxO Regulators<br />

Patrick Hu, Kathleen Dumas, Albert Chen, Hung-Jen Shih, Chunfang Guo<br />

Life Sciences Institute, <strong>University</strong> <strong>of</strong> Michigan, Ann Arbor, MI, United States<br />

FoxO transcription factors promote longevity in invertebrates and may control aging in<br />

humans. Mouse models implicate FoxO transcription factors in the pathogenesis <strong>of</strong> cancer,<br />

Type 2 diabetes, cardiovascular disease, and osteoporosis. Thus, understanding how FoxO<br />

transcription factors are regulated is expected to illuminate fundamental biological processes<br />

that underlie aging and age-related diseases.<br />

In C. elegans, the DAF-2 insulin-like signaling pathway and the germline are the major<br />

regulators <strong>of</strong> DAF-16/FoxO activity. Both inhibit DAF-16/FoxO by promoting its cytoplasmic<br />

sequestration. However, nuclear translocation <strong>of</strong> DAF-16/FoxO is not sufficient for activation,<br />

indicating that other inputs that control nuclear DAF-16/FoxO activity exist. We recently<br />

discovered EAK-7, a conserved protein <strong>of</strong> unknown function that acts in parallel to the serine/<br />

threonine kinase AKT-1 to inhibit nuclear DAF-16/FoxO activity. How EAK-7 regulates DAF-<br />

16/FoxO is not known. Since EAK-7 is associated with the plasma membrane, it likely acts<br />

indirectly to control nuclear DAF-16/FoxO activity.<br />

To identify novel DAF-16/FoxO regulators in an unbiased manner, we have performed a<br />

genetic screen for suppressors <strong>of</strong> the dauer arrest phenotype <strong>of</strong> an eak-7;akt-1 double mutant<br />

(seak mutants). Whole genome sequencing <strong>of</strong> sixteen independent seak mutants has identified<br />

eighteen genes, each <strong>of</strong> which harbors distinct nonsynonymous mutations in more than one<br />

mutant strain. None <strong>of</strong> these genes has previously been implicated in FoxO regulation or lifespan<br />

control. We have validated two <strong>of</strong> these genes, both <strong>of</strong> which are conserved in mammals,<br />

as bonafide seak genes. The identification and characterization <strong>of</strong> seak mutants promises to<br />

yield new insights into the molecular basis <strong>of</strong> aging while engendering the development <strong>of</strong><br />

new strategies to combat aging and age-related diseases.<br />

Session 5 Plenary Speaker<br />

41


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

Interplay <strong>of</strong> Foreign and Endogenous Lectins in Innate Immunity <strong>of</strong><br />

Caenorhabditis elegans<br />

Therese Wohlschlager 1 , Alex Butschi 2 , Katrin Stutz 2 , Iain Wilson 3 , Michael Hengartner 2 ,<br />

Markus Aebi 1 , Markus Kunzler 1<br />

1 Institute <strong>of</strong> Microbiology, ETH Zurich, Switzerland, 2 Institute <strong>of</strong> Molecular Life<br />

Sciences, <strong>University</strong> <strong>of</strong> Zurich, Switzerland, 3 Department <strong>of</strong> Chemistry, <strong>University</strong><br />

<strong>of</strong> Natural Resources and Life Sciences (BOKU), Vienna, Austria<br />

Lectins are proteins that are able to recognize specific carbohydrate epitopes. As all living<br />

cells display a characteristic carbohydrate coat on their surface, lectins play an important role<br />

in intercellular and interorganismic interactions. In the context <strong>of</strong> Caenorhabditis elegans innate<br />

immunity, it has been hypothesized that endogenous lectins in the intestinal lumen shield<br />

endogenous carbohydrate epitopes from recognition by carbohydrate-binding bacterial toxins<br />

in that they display the same specificity as the toxin (1). However, only few molecular targets<br />

<strong>of</strong> endogenous and foreign lectins in C. elegans have been identified and characterized to<br />

date (2). Furthermore, it is not clear why lectins with the same specificity are toxic in one case<br />

(foreign lectins) but not in the other (endogenous lectins).<br />

During the last few years, we have isolated and characterized a variety <strong>of</strong> soluble,<br />

intracellular lectins from reproductive structures <strong>of</strong> different fungi and demonstrated that many<br />

<strong>of</strong> these lectins display toxicity towards Caenorhabditis elegans (3). We hypothesize that these<br />

lectins are part <strong>of</strong> an innate defense system <strong>of</strong> fungi against fungivorous nematodes. For some<br />

<strong>of</strong> the nematotoxic fungal lectins, we identified the carbohydrate epitopes that are recognized<br />

in C. elegans and are essential for susceptibility <strong>of</strong> the nematode to the lectin (4-7). These<br />

epitopes are nematode-specific, part <strong>of</strong> protein- or lipid-bound glycans and mostly localized<br />

to the apical surface <strong>of</strong> the intestinal epithelium. We have recently identified candidate C.<br />

elegans glycoproteins carrying glycans that are apparently targeted by different nematotoxic<br />

fungal lectins and also by endogenous C. elegans lectins. We are currently in the process <strong>of</strong><br />

purifying these proteins in sufficient amounts from C. elegans to analyze their glycans. The<br />

results may provide further insight into the toxicity mechanism <strong>of</strong> foreign lectins and a possible<br />

role <strong>of</strong> endogenous lectins in C. elegans innate immunity.<br />

(1) Ideo et al. 2009 J. Biol. Chem. 284:26493-501<br />

(2) Takeuchi et al. 2011 Biol. Pharm. Bull. 34:1139-42<br />

(3) Bleuler-Martinez et al. 2011 Mol. Ecol. 20:3056-70<br />

(4) Titz et al. 2009 J. Biol. Chem. 284:36223-33<br />

(5) Butschi et al. 2010 PLoS Pathogens 6:e1000717<br />

(6) Wohlschlager et al. 2011 J. Biol. Chem. 286:30337-43<br />

(7) Schubert et al. 2012 PLoS Pathogens, in press<br />

Contact: markus.kuenzler@micro.biol.ethz.ch<br />

Lab: Aebi<br />

42<br />

Session 5


Contact: sunc@morpheus.wustl.edu<br />

Lab: Crowder<br />

Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

Cell-Specific Hypoxic Injury in C. elegans: Modeling the Ischemic<br />

Penumbra<br />

Chun-Ling Sun 1 , Euysoo Kim 1 , C. Michael Crowder 1,2<br />

1 Department <strong>of</strong> Anesthesiology, 2 Department <strong>of</strong> Developmental Biology,<br />

Washington <strong>University</strong> School <strong>of</strong> Medicine, St. Louis, Missouri<br />

Stroke is the leading cause <strong>of</strong> disability in the US and the fourth leading cause <strong>of</strong> death yet<br />

has no approved cytoprotective therapy. In stroke, neurons in the most severely hypoxic areas<br />

die immediately while those in surrounding less hypoxic areas (termed the ischemic penumbra)<br />

are injured but may recover or have delayed cell death. The penumbral cells are the therapeutic<br />

target in stroke, but a genetically tractable model <strong>of</strong> the penumbra is lacking. Towards that end,<br />

we expressed rars-1(+) in C. elegans with cell-type-specific promoters in the background <strong>of</strong><br />

rars-1(gc47rf), a mutation that strongly protects C. elegans from organismal hypoxic death.<br />

Hypoxic incubation <strong>of</strong> strains with pharyngeal myocyte-specific rars-1(+) expression in gc47<br />

produced severe pharyngeal pathology and pumping defects without killing the animal after<br />

initial recovery. We observed hypoxic necrotic cell death in the pharynx and in other parts <strong>of</strong> the<br />

worm whereas none was observed in the gc47 background strain. Over the next four days, we<br />

observed an increasing Unc phenotype and delayed animal death. Likewise selective rescue<br />

<strong>of</strong> gc47 in GABA neurons with rars-1(+) produced initial pathology in GABA neurons and then<br />

a delayed severe Unc phenotype (distinct from that produced by GABAergic dysfunction) and<br />

delayed animal death. Thus, these strains model aspects <strong>of</strong> the ischemic penumbra, particularly<br />

delayed cell death and spread <strong>of</strong> injury from primarily injured cells to surrounding cells. We<br />

are now beginning to ask if previously identified hypoxic protective mutants/RNAis function to<br />

protect particular cell types and primarily versus secondarily injured cells.<br />

Session 5<br />

43


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

Movement <strong>of</strong> RNA Silencing Between C. elegans Cells<br />

Antony Jose, Hai Le, Snusha Ravikumar, Sindhuja Devanapally<br />

<strong>University</strong> <strong>of</strong> Maryland, College Park, MD, USA<br />

RNA silencing is triggered by double-stranded RNA (dsRNA) and transported between<br />

cells in C. elegans. The import <strong>of</strong> silencing RNA into cells requires a conserved tyrosine kinase<br />

that likely promotes endocytosis and a conserved dsRNA-selective transporter that allows<br />

entry into the cytoplasm. Genetic mosaic analyses suggest that exported RNA comprise <strong>of</strong><br />

two classes <strong>of</strong> dsRNA: long dsRNA and Dicer-processed short-interfering RNA (siRNA). Both<br />

classes <strong>of</strong> dsRNA are likely made and exported through a general mechanism that operates<br />

in all cell types because distinct tissues such as the gut, muscles, and neurons can all export<br />

silencing RNA. But, how cells decide to export silencing RNA and the molecular machinery<br />

that controls such export are unknown.<br />

Silencing RNA can either be exported from any cell within a tissue or from a specific subset<br />

that is specialized for RNA export. To distinguish between these two possibilities, we examined<br />

the silencing <strong>of</strong> a multicopy transgene that expresses green fluorescent protein (GFP) in C.<br />

elegans gut cells in the absence <strong>of</strong> RNA transport between cells. Gut cells that make and export<br />

silencing RNA will silence GFP expression, but cells that require the import <strong>of</strong> silencing RNA<br />

will fail to do so. We found that a subset <strong>of</strong> cells (~45% on average) in all animals silenced GFP<br />

expression. This subset, however, differed from one animal to another such that all gut cells<br />

could silence GFP expression in at least one animal. These results suggest that cells that export<br />

RNA silencing are determined in a stochastic manner in each animal. To identify mutants that<br />

are defective in the transport <strong>of</strong> mobile RNA between cells, we performed a forward genetic<br />

screen using dsRNA expressed from neurons (exporters) to silence GFP expression in gut<br />

cells (importers). We isolated 54 silencing exported from neurons defective (send) mutants<br />

using this screen and are currently analyzing 19 <strong>of</strong> the most strongly defective send mutants.<br />

Thus, the export <strong>of</strong> silencing RNA derived from expressed dsRNA is widespread and cells<br />

that export such RNA differ between animals <strong>of</strong> the same genotype. Our genetic screen,<br />

focused on the transport <strong>of</strong> RNA silencing between distinct tissues, is likely to uncover the<br />

proteins that enable this general RNA export mechanism.<br />

Contact: amjose@umd.edu<br />

Lab: Jose<br />

44<br />

Session 5


Contact: abigail.cabunoc@oicr.on.ca<br />

Lab: Stein<br />

Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

WormBase 2012: Website Redesign<br />

Abigail Cabunoc, Norie de la Cruz, Adrian Duong, Maher Kassim, Xiaoqi Shi, Todd<br />

Harris, Lincoln Stein<br />

Ontario Institute for Cancer Research, Toronto, Canada<br />

WormBase (www.wormbase.org) has served the ever growing needs <strong>of</strong> the nematode<br />

research community since 2000. Initially created as a resource for the C.elegans genome and<br />

its biology, WormBase now includes a wider variety <strong>of</strong> data from 15 related species. This growth<br />

in data is mirrored by increased website use. In response to the constantly growing data and<br />

user traffic, the WormBase web team has redesigned both the underlying architecture and<br />

user interface <strong>of</strong> the website. In this interactive presentation, we will review the new WormBase<br />

website, walk through basic and advanced tasks using the new interface, and examine several<br />

features which take advantage <strong>of</strong> the nematode research community.<br />

Session 5<br />

45


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

Dynamic Neuroendocrine Signaling in C. elegans Behavioral<br />

Responses to Pathogenic Bacteria<br />

Dennis Kim<br />

MIT, Cambridge, MA<br />

We have investigated host-microbe interactions in Caenorhabditis elegans, focusing on the<br />

molecular genetic analysis <strong>of</strong> immune and neural responses to microbes in this simple host<br />

organism. The TGF-ß signaling pathway functions in the neuroendocrine regulation <strong>of</strong> diverse<br />

physiological processes in C. elegans. I will present our recent studies <strong>of</strong> how bacteria induce<br />

changes in TGF-ß signaling that promote pathogen avoidance behavior.<br />

Contact: dhkim@mit.edu<br />

Lab: Kim<br />

46<br />

Session 6 Plenary Speaker


Contact: rtaylor@salk.edu<br />

Lab: Dillin<br />

Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

The UPRER is a Cell Non-Autonomous Regulator <strong>of</strong> Stress Resistance<br />

and Longevity<br />

Rebecca Taylor, Andrew Dillin<br />

Howard Hughes Medical Institute, The Salk Institute for Biological Studies, La<br />

Jolla, CA<br />

The ability to ensure proteostasis is critical for maintaining proper cell function and<br />

organismal viability, but is mitigated by aging. We analyzed the role <strong>of</strong> the endoplasmic reticulum<br />

Unfolded Protein Response (UPR ER ) in aging <strong>of</strong> C. elegans, and found that the ability to activate<br />

the UPR ER was lost with age. This age-onset loss <strong>of</strong> ER proteostasis could be reversed via the<br />

expression <strong>of</strong> a constitutively active form <strong>of</strong> XBP-1, XBP-1s. Expression <strong>of</strong> XBP-1s specifically<br />

in the nervous system was sufficient to rescue stress resistance, increase longevity and was<br />

able to specifically activate the UPR ER in distal, non-neuronal cell types through a cell nonautonomous<br />

mechanism. Loss <strong>of</strong> UPR ER signaling components in the distal cells blocked cell<br />

non-autonomous signaling from the nervous system, thereby blocking increased longevity <strong>of</strong><br />

the entire animal. Reduction <strong>of</strong> small clear vesicle (SCV) release blocked the non-autonomous<br />

signaling <strong>of</strong> the UPR ER , suggesting that the release <strong>of</strong> neurotransmitters is required for this<br />

inter-tissue signaling event. Our findings point towards a Secreted ER Stress Signal (SERSS)<br />

that promotes ER stress resistance and longevity.<br />

Session 6<br />

47


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

Interactions between 1-carbon cycle and lipid metabolism in C.<br />

elegans<br />

Amy Walker 1 , Rene Jacobs 2 , Jenny Watts 3 , Veerle Rottiers 4 , Anders NAar 4<br />

1 UMASS Medical School Worcester, MA USA, 2 <strong>University</strong> <strong>of</strong> Alberta, Edmonton,<br />

Canada, 3 <strong>University</strong> <strong>of</strong> Washington Pullman, 4 MGH Cancer Center, Boston MA<br />

USA<br />

Nutritional and metabolic pathways feed into diverse cellular processes. A growing body<br />

<strong>of</strong> evidence also suggests that some transcription factors are affected by metabolic cues to<br />

coordinate gene expression with nutrient availability. The SREBP family <strong>of</strong> transcription factors<br />

regulate genes for generating fatty acids and phospholipid in metazoans and cholesterol<br />

synthesis in vertebrates. These factors are inhibited by cholesterol in vertebrates, however the<br />

regulatory feedback mechanisms for lipogenic genes have been less clear. Using C. elegans<br />

and mammalian models, we have found that SBP-1 and mammalian SREBP-1 control the<br />

expression <strong>of</strong> genes in the 1-carbon cycle, which produces methyl groups necessary for<br />

protein and phospholipid methylation. In addition, lack <strong>of</strong> phospholipid methylation initiates a<br />

regulatory cascade that results in increased SBP-1/SREBP-1 activity. Up-regulation <strong>of</strong> SBP-1/<br />

SREBP-1 activity when methylation capacity or phospholipid synthesis is diminished results<br />

in increased lipogenesis and may be relevant to the development <strong>of</strong> fatty liver in mammals.<br />

Finally, our discovery that SREBP transcription factors are linked to metabolic pathways<br />

controlling methylation opens additional avenues in understanding how gene regulation is<br />

linked to metabolic control.<br />

Contact: amy.walker@umassmed.edu<br />

Lab: Naar/Walker<br />

48<br />

Session 6


Contact: wmadaki@gmail.com<br />

Lab: Suzuki<br />

Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

Novel approach in oxidative stress study by targeted ROS generation<br />

using a photosensitizer SuperNova in C. elegans<br />

Hiroshi Suzuki, Donald Fu, Ippei Kotera, Po-An SU<br />

<strong>University</strong> <strong>of</strong> Toronto, Toronto, Ontario, CANADA<br />

Oxidative stress has been implied innumerable diseases, including cancer, myocardial<br />

infarction, and neurodegenerative diseases such as Parkinson’s Disease (PD). Traditional<br />

approaches have mostly relied on indirect generation <strong>of</strong> reactive oxygen species(ROS) using<br />

chemical inhibitors <strong>of</strong> mitochondria respiratory chain to induce oxidative stress; however,<br />

chemicals may diffuse into unexamined cells and exert unintended side-effects. The lack <strong>of</strong><br />

specificity has hampered direct evaluation <strong>of</strong> its role in pathology and diseases.<br />

We here established a novel approach to directly generate ROS by using the photosensitizer<br />

SuperNova: a genetically-encoded, red fluorescent protein that generates ROS upon light<br />

activation, in specific cells, cellular compartments,and specific timing. SuperNova is a newly<br />

developed monomeric version <strong>of</strong> KillerRed.<br />

SuperNova was expressed specifically in mechanosensory neurons and dopaminergic<br />

(DA). Upon light activation, we induced neurodegeneration, revealed by loss <strong>of</strong> neuronal<br />

morphology, and by depletion in function as examined by behaviour assays. The behavioural<br />

response mediated by the mechanosensory neurons was rapidly abolished before the<br />

morphological change was detectable, which is analogous to various diseases that develop<br />

symptoms years before the appearance <strong>of</strong> neurodegenerations. In context <strong>of</strong> PD, we crossed<br />

our SuperNovatransgenic worms into the mutants <strong>of</strong> PD-related genes and observed a<br />

sensitization <strong>of</strong> DA neuron degeneration in lrk-1background, which is in good agreement with<br />

the proposed protection by LRRK2/lrk-1. Furthermore, our controlled activation <strong>of</strong> SuperNova<br />

killed DA neurons(PDE) while the adjacent mechanosensory neurons (PVM) remained intact<br />

in the cross <strong>of</strong> the two Tg strains, demonstrating the powerful model to address the selective<br />

death <strong>of</strong> DA neurons in PD. Moreover, we targeted SuperNova to the mitochondria <strong>of</strong> body<br />

wall muscles, which lead to deformed mitochondria, but nocell ablation was observed. We<br />

hypothesized a protective mechanism preventing the progress <strong>of</strong> damage, and addressed<br />

the protection by using deletion mutantsand feeding RNAi; cell ablation was observed when<br />

Parkin/pdr-1 mutant was combined with RNAi <strong>of</strong> sod-2 (mitochondrial superoxide dismutase)<br />

or pink-1. Thus, the novel method we established to deliver oxidative stress in specific cells,<br />

specific organelle at specific timing and duration in live animals will be a valuable tool to study<br />

oxidative stress and other physiological/pathological status where ROS plays roles.<br />

Session 6<br />

49


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

Natural Products that Suppress Protein Aggregation and Slow Aging<br />

Silvestre Alavez1 , Pedro Rodriguez1 , Maithili C. Vantipalli1 , David J. S. Zucker1,2 ,<br />

Ida M. Klang1,3 Milena Price, Karla Mark, and Gordon J. Lithgow1 1Buck Institute for Research on Aging, 8001 Redwood Blvd, Novato,<br />

California 94945, USA. 2Department <strong>of</strong> Natural Sciences and Mathematics,<br />

Dominican <strong>University</strong> <strong>of</strong> California, San Rafael, California, 94901, USA.<br />

3Karolinska Institute, Center for Biosciences at NOVUM, Department <strong>of</strong><br />

Biosciences and Nutrition, Hälsovägen 7, S-141 83 Huddinge, Sweden.<br />

We have undertaken screen <strong>of</strong> synthetic and natural compounds to find agents for aging<br />

interventions. Since aging can be considered a causal factor in a number <strong>of</strong> age-related<br />

diseases. We hope such screen could yield useful therapeutics. We focused our search on<br />

compounds that maintain protein homeostasis. Collapse <strong>of</strong> protein homeostasis results in<br />

protein misfolding cascades and the accumulation <strong>of</strong> insoluble protein fibrils and aggregates,<br />

such as amyloids. A group <strong>of</strong> small molecules, traditionally used in histopathology to stain<br />

amyloid in tissues, bind protein fibrils and slow aggregation in vitro and in cell culture. We<br />

proposed that treating animals with such compounds would promote protein homeostasis in<br />

vivo and increase longevity. Here we show that exposure <strong>of</strong> adult Caenorhabditis elegans<br />

to the amyloid-binding dye Thi<strong>of</strong>lavin T (ThT) resulted in a pr<strong>of</strong>oundly extended lifespan and<br />

slowed ageing. ThT also suppressed pathological features <strong>of</strong> mutant metastable proteins and<br />

human β-amyloid-associated toxicity. These beneficial effects <strong>of</strong> ThT depend on the protein<br />

homeostasis network. Other agents that extend lifespan also suppress protein homeostasis<br />

such as Lithium. We are testing for common mechanisms <strong>of</strong> action. This is reveling tissue<br />

non-autonomous signaling between neurons and muscle.<br />

Our results to date demonstrate that pharmacological maintenance <strong>of</strong> the protein<br />

homeostatic network by natural products has a pr<strong>of</strong>ound impact on aging rates, prompting<br />

the development <strong>of</strong> novel therapeutic interventions against ageing and age-related diseases.<br />

Contact: glithgow@buckinstitute.org<br />

Lab: Lithgow<br />

50<br />

Session 6 Plenary Speaker


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

Expression-level Polymorphism for the Rec-8 Gene Underlies<br />

a Quantitative Trait Locus Governing Lifespan, Multiple Stress<br />

Resistances, and X-Chromosome Nondisjunction<br />

Srinivas Ayyadevara 1,3 , Cagdas Tazearslan 2 , Ramani Alla 1 , Robert Shmookler Reis 1,3<br />

1 <strong>University</strong> <strong>of</strong> Arkansas for Medical Sciences, Little Rock,AR,USA, 2 Albert Einstein<br />

College <strong>of</strong> Medicine,Bronx, NY, 3 Central Arkansas Veterans Health Care System,<br />

Little Rock,AR,USA<br />

We identified a quantitative trait locus (QTL) on chromosome 4, lsq4, that affects lifespan<br />

and resistance to oxidative and thermal stresses in Caenorhabditis elegans. We have now<br />

defined lsq4 allelic effects on transcript levels <strong>of</strong> genes lying within the QTL region, and also<br />

across the entire genome. Of the twenty-eight confirmed genes in the lsq4 interval, eleven<br />

showed 2-to 15-fold allele-specific differential expression by quantitative real-time PCR. Ten<br />

differentially expressed lsq4 genes were examined for RNA-interference effects on the QTL<br />

traits. One gene, rec-8, increased lifespan when disrupted in worms bearing the shorterlived<br />

allele, but its knockdown had no effect in the longer-lived isogenic strain. RNAi to rec-8<br />

also reverts all other traits <strong>of</strong> the shorter-lived allele, including greater sensitivity to paraquat<br />

and heat shock, and reduced male frequency. Thus, allele-determined differences in rec-8<br />

expression can explain all known effects <strong>of</strong> this QTL. The rec-8 polymorphism may provide<br />

an instructive example <strong>of</strong> balancing selection (wherein each <strong>of</strong> two alleles is favored under<br />

different circumstances), and/or antagonistic pleiotropy (in which early benefits to Darwinian<br />

fitness drive selection for an allele that is deleterious in post-reproductive life).<br />

Contact: ayyadevarasrinivas@uams.edu<br />

Lab: Shmookler Reis<br />

Poster Topic: Aging<br />

51


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

Effects <strong>of</strong> Dietary Restriction by Axenic Medium on Mitochondrial<br />

Function in Caenorhabditis elegans.<br />

Natascha Castelein 1 , Berhanu Kassa 2 , Bart Braeckman 1<br />

1 Ghent <strong>University</strong>, Ghent, Belgium, 2 <strong>University</strong> <strong>of</strong> Cologne, Cologne, Germany<br />

C. elegans lifespan can be drastically extended by using a semidefined axenic culturing<br />

medium instead <strong>of</strong> the standard E. coli food source. This culturing method probably imposes<br />

dietary restriction (axenic dietary restriction, ADR) and slows down the aging process. The<br />

molecular causes <strong>of</strong> aging are not well understood, but it is widely believed that oxidative stress<br />

plays an important role in this process. Oxidative stress is a side effect <strong>of</strong> aerobic metabolism<br />

and may lead to progressive oxidative damage to macromolecules and a concomitant functional<br />

decline. Since aerobic metabolism occurs in the mitochondria, these organelles are considered<br />

to play a central role in the aging process.<br />

We hypothesize that mitochondria <strong>of</strong> the long-lived axenically cultured worms show a<br />

completely different physiology and are much more efficient compared to fully fed controls.<br />

To assess mitochondrial function, we measured oxygen consumption <strong>of</strong> isolated<br />

mitochondria, using a Clark type electrode (O2k, Oroboros instruments). Preliminary data<br />

suggest that respiration <strong>of</strong> isolated ADR mitochondria is substantially lowered compared to<br />

fully fed controls, but they appear to be better coupled. Further investigation will shed more<br />

light on how mitochondria are affected by ADR conditions. First, multiple substrate and inhibitor<br />

titration protocols will be applied to gain an extended understanding on the functions <strong>of</strong> different<br />

mitochondrial pathways under ADR conditions. Simultaneously, membrane potential will be<br />

monitored using TPP + electrodes.<br />

Second, in vivo quantification <strong>of</strong> ROS levels with transgenic worms expressing ROS<br />

biosensors will provide a better indication <strong>of</strong> real-time physiological conditions under ADR.<br />

Apart from mitochondrial physiology, we studied the role <strong>of</strong> DR-related genes in lifespan<br />

extension <strong>of</strong> ADR worms. We found that lifespan extension by ADR is partially dependent on<br />

AAK-2 and CUP-4 but independent <strong>of</strong> SKN-1 and UCP-4.<br />

Contact: Natascha.castelein@ugent.be<br />

Lab: Braeckman<br />

52<br />

Poster Topic: Aging


Contact: atchen@umich.edu<br />

Lab: Hu<br />

Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

SGK-1 Extends Lifespan by Activating DAF-16/FoxO<br />

Albert Chen1 , Chunfang Guo2 , Kathleen Dumas1 , Travis Williams2 , Sawako Yoshina3 ,<br />

Shohei Mitani3 , Kaveh Ashrafi4 , Patrick Hu2 1Cellular and Molecular Biology Program, <strong>University</strong> <strong>of</strong> Michigan, Ann Arbor, MI,<br />

USA, 2Life Sciences Institute, <strong>University</strong> <strong>of</strong> Michigan, Ann Arbor, MI, USA, 3Dept. <strong>of</strong> Physiology, Tokyo Women’s Medical <strong>University</strong> School <strong>of</strong> Medicine, Tokyo,<br />

Japan, 4Dept. <strong>of</strong> Physiology, UCSF School <strong>of</strong> Medicine, San Francisco, CA, USA<br />

The FoxO family <strong>of</strong> transcription factors is implicated in aging and age-related diseases in<br />

species as diverse as invertebrates and mammals. In C. elegans, loss-<strong>of</strong>-function mutations<br />

in the conserved insulin-like signaling pathway result in increased transcriptional activity <strong>of</strong><br />

the C. elegans FoxO ortholog DAF-16, which is sufficient to extend adult lifespan severalfold.<br />

DAF-16 and FoxO possess three highly conserved sites that lie within consensus motifs for<br />

the conserved AGC family kinases Sgk and Akt (referred to as AGC sites). Mammalian cell<br />

culture data suggest that phosphorylation <strong>of</strong> any <strong>of</strong> these sites by Sgk and Akt inhibits FoxO by<br />

promoting its cytoplasmic sequestration. Consistent with this model, C. elegans AKT-1 and AKT-<br />

2 limit lifespan by inhibiting DAF-16/FoxO. However, recent experiments using both loss- and<br />

gain-<strong>of</strong>-function mutations suggest that SGK-1 increases lifespan in C. elegans by activating<br />

DAF-16/FoxO, counter to the prevailing paradigm where SGK-1 solely inhibits DAF-16/FoxO.<br />

These results suggest that our understanding <strong>of</strong> how FoxO is regulated by Sgk is incomplete.<br />

We hypothesize that the FoxO N-AGC site acts as a conserved switch: SGK-1 phosphorylation<br />

<strong>of</strong> the C-AGC site inhibits DAF-16/FoxO when the N-AGC site is phosphorylated, but activates<br />

DAF-16/FoxO when the N-AGC site is not phosphorylated. To this end, we are building MosSCI<br />

strains harboring single-copy DAF-16/FoxO transgenes encoding various combinations<br />

<strong>of</strong> phosphomimetic and phosphodefective AGC site mutations. Because the mechanisms<br />

controlling DAF-16/FoxO activity are highly conserved, insights into the role <strong>of</strong> SGK-1 in<br />

controlling C. elegans longevity may be relevant to common human diseases associated with<br />

aging such as cancer, Type 2 diabetes, cardiovascular disease, and osteoporosis.<br />

Poster Topic: Aging<br />

53


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

Proteomic Changes Elicited by Metformin Treatment<br />

Wouter De Haes 1 , Roel Van Assche 1 , Steven Haenen 1 , Bart Braeckman 2 , Liliane<br />

Scho<strong>of</strong>s 1<br />

1 KU Leuven, Leuven, Belgium, 2 UGent, Ghent, Belgium<br />

Aging is a complex process in which molecular and cellular damage gradually accumulates,<br />

which in turn leads to increased vulnerability and ultimately death. Research has shown that<br />

aging and longevity are regulated by a combination <strong>of</strong> several pathways, although the specifics<br />

are still poorly understood. Many <strong>of</strong> these signaling pathways are involved in nutrient detection<br />

and, as such, it isn’t surprising that dietary restriction (a reduction in food uptake without<br />

malnutrition) leads to an extension <strong>of</strong> healthy lifespan. Metformin, the most widely used antidiabetic<br />

for the treatment <strong>of</strong> type-2 diabetes, also has lifespan-extending effects. Research<br />

has shown that this increase <strong>of</strong> longevity in healthy metformin-treated subjects might be the<br />

result <strong>of</strong> metformin mimicking the effect <strong>of</strong> dietary restriction.<br />

In our study, we use C. elegans and gel-based differential proteomics in order to elucidate<br />

the molecular effectors involved in the life-extending effect <strong>of</strong> metformin. Studies have already<br />

shown that the positive effect <strong>of</strong> metformin on C. elegans lifespan is dependent on AMPactivated<br />

protein kinase (AAK-2) and the skinhead (SKN)-1 transcription factor. Our proteomic<br />

study showed a significant upregulation <strong>of</strong> several proteins involved in carbohydrate catabolism,<br />

which is consistent with the predicted function <strong>of</strong> AAK-2. We also found overlap with data<br />

from several studies <strong>of</strong> dietary restriction, including downregulation <strong>of</strong> proteins involved in<br />

reproduction and the group <strong>of</strong> transthyretin-like proteins. Furthermore, we identified several<br />

upregulated proteins with unknown functions, including a protein with predicted kinase activity<br />

and a transcription factor. In the future, we will determine their effect on longevity and compare<br />

our results with planned gel-based proteomic studies <strong>of</strong> dietary restriction.<br />

Contact: wouter.dehaes@bio.kuleuven.be<br />

Lab: Scho<strong>of</strong>s<br />

54<br />

Poster Topic: Aging


Contact: kjdumas@umich.edu<br />

Lab: Hu<br />

Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

The p120RasGAP Family Member GAP-3 is a Novel Regulator <strong>of</strong> Dauer<br />

Arrest and Longevity<br />

Kathleen Dumas2 , Stephane Flibotte1 , Don Moerman1 , Patrick Hu2 1 2 <strong>University</strong> <strong>of</strong> British Columbia, Vancouver, BC, Canada, <strong>University</strong> <strong>of</strong> Michigan,<br />

Ann Arbor, MI, USA<br />

In C. elegans, both reducing insulin/insulin-like growth factor signaling (IIS) and ablating<br />

the germline extend life span by promoting the translocation <strong>of</strong> DAF-16/FoxO to the nucleus.<br />

Nuclear DAF-16/FoxO induces transcriptional programs that promote longevity. The IIS pathway<br />

in C. elegans consists <strong>of</strong> conserved PI3K and Akt components that antagonize DAF-16/FoxO<br />

by promoting its cytoplasmic sequestration. We have discovered the EAK pathway, a novel,<br />

conserved pathway that inhibits DAF-16/FoxO. The EAK pathway acts in parallel to PI3K/Akt<br />

signaling to inhibit nuclear DAF-16/FoxO activity without promoting its translocation to the<br />

cytoplasm. How the EAK pathway inhibits DAF-16/FoxO is not known.<br />

To illuminate the mechanism underlying EAK pathway inhibition <strong>of</strong> nuclear DAF-16/FoxO<br />

activity, we identified suppressors <strong>of</strong> eak-7;akt-1 dauer arrest (seak mutants). We identified<br />

gap-3, a p120 GTPase activating protein (GAP) family member, as a seak gene. GAP-3 is<br />

homologous to human RASA1, which was first identified as a RasGAP that antagonizes<br />

Ras by promoting its GTPase activity. GAP-3 is one <strong>of</strong> three putative RasGAPs encoded in<br />

the C. elegans genome. A spontaneous missense mutation in the RasGAP domain <strong>of</strong> gap-<br />

3 suppresses the 25°C dauer arrest phenotype <strong>of</strong> eak-7;akt-1 double mutant animals. Two<br />

independent loss <strong>of</strong> function alleles <strong>of</strong> gap-3, zh94 and ga139, also suppress eak-7;akt-1<br />

dauer arrest, confirming gap-3 as a seak gene. In contrast, mutations in gap-1 and gap-2 did<br />

not significantly influence eak-7;akt-1 dauer arrest. Initial experiments suggest gap-3 mutation<br />

also suppresses the life span extension phenotype <strong>of</strong> eak-7;akt-1 double mutants. Notably,<br />

the let-60/Ras gain-<strong>of</strong>-function allele n1046 suppresses eak-7;akt-1 dauer arrest to a lesser<br />

degree than gap-3 loss-<strong>of</strong>-function alleles, suggesting that GAP-3 may have Ras-independent<br />

functions in the control <strong>of</strong> dauer arrest. These results implicate GAP-3 as a novel regulator<br />

<strong>of</strong> dauer arrest and longevity. We are currently exploring the potential role <strong>of</strong> GAP-3 in the<br />

transduction <strong>of</strong> signals from both the EAK pathway and the PI3K/Akt pathway to DAF-16/FoxO.<br />

Poster Topic: Aging<br />

55


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

Investigating the tissue-specific requirements for autophagy in C.<br />

elegans longevity mutants<br />

Sara Gelino, Malene Hansen<br />

Sanford-Burnham Institute for Medical Research, La Jolla, (CA), USA<br />

Multiple conserved pathways and processes can modulate lifespan, including dietary<br />

restriction and reduced insulin/IGF-1 signaling. We and others have recently shown that<br />

autophagy – a cellular process by which cytoplasmic components are degraded and recycled<br />

– is induced in multiple long-lived C. elegans mutants, including dietary-restricted eat-2 animals<br />

and in daf-2 mutants with reduced insulin/IGF-1 signaling. Accordingly, autophagy genes are<br />

required for the long lifespan <strong>of</strong> these mutants, suggesting that autophagy promotes longevity<br />

by mechanisms still to be identified.<br />

To learn more about the role <strong>of</strong> autophagy in aging, we are investigating where autophagy<br />

may promote longevity in the organism. To address this question, we are utilizing several<br />

tissue-restricted RNA interference models to inactivate autophagy in specific tissues <strong>of</strong> eat-<br />

2 and daf-2 mutants. Knowledge <strong>of</strong> where in the animal that autophagy appears to have a<br />

rejuvenating effect will shed light on the underlying mechanism by which autophagy may<br />

promote longevity. Interestingly, in our preliminary studies, we have observed that the longevity<br />

function <strong>of</strong> autophagy is indeed restricted to specific tissues <strong>of</strong> these long-lived animals.<br />

Based on these results, we propose that autophagy is utilized in certain tissues to remove<br />

damaged proteins and organelles that would normally accumulate during the aging process.<br />

In this way, the autophagic turnover <strong>of</strong> damaged macromolecules, the nature <strong>of</strong> which is still<br />

to be identified, may prolong the youthfulness <strong>of</strong> a cell, tissue, and organism.<br />

Contact: sgelino@sbmri.org<br />

Lab: Hansen<br />

56<br />

Poster Topic: Aging


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

Tumor suppressors and longevity in C.elegans<br />

Hakam Gharbi 1,2 , Francesca Fabretti 2 , Puneet Bharill 2 , Bernhard Schermer 1,2 , Thomas<br />

Benzing 1,2 , Roman Ulrich Mueller 1,2<br />

1 Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated<br />

Diseases, <strong>University</strong> <strong>of</strong> Cologne, Cologne, Germany, 2 Department II <strong>of</strong> Internal<br />

Medicine and Center for Molecular Medicine, <strong>University</strong> <strong>of</strong> Cologne, Cologne,<br />

Germany<br />

Caenorhabditis elegans is one <strong>of</strong> the most prominent model organisms in aging research<br />

due to its simple genetic amenability and short life-cycle. Recently several groups showed<br />

that loss <strong>of</strong> the tumor suppressor protein vhl-1 leads to longevity in the nematode. This effect<br />

is predominantly mediated by hif-1, a hypoxia-inducible factor, the degradation <strong>of</strong> which is<br />

mediated by vhl-1. This increased lifespan appears to be the consequence <strong>of</strong> an enhanced<br />

cellular stress response, since these strains do also show improved survival upon exposure<br />

to a row <strong>of</strong> stressors.<br />

In Von-Hippel-Lindau syndrome, which is a rare genetic condition that goes along with the<br />

development <strong>of</strong> various tumors, pVHL (Von-Hippel-Lindau protein) is mutated. Furthermore,<br />

pVHL was shown to be the most important renal tumor suppressor preventing the formation<br />

<strong>of</strong> Renal Cell Carcinoma.<br />

Work by both our and other groups now provides evidence that other tumor suppressor<br />

proteins are linked to longevity and regulation <strong>of</strong> cellular stress resistance in C.elegans. This<br />

finding indicates that protection from tumor formation in mammalian cells– which obviously does<br />

not play a role in a short-lived nematode – comes at the expense <strong>of</strong> reduced cellular fitness<br />

and stress resistance. The current data allow for the hypothesis that temporary inhibition <strong>of</strong><br />

certain tumor suppressor proteins in a timely fashion that does affect tumorigenesis could be<br />

a novel tool to protect organs from damaging stimuli.<br />

Contact: hakam.gharbi@uk-koeln.de<br />

Lab: Benzing<br />

Poster Topic: Aging<br />

57


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

Identification <strong>of</strong> a Novel Bacterial Species That Extends the Lifespan<br />

<strong>of</strong> Caenorhabditis elegans<br />

Junhyeok Go 1,2 , Kang-Mu Lee 1 , Sang Sun Yoon 1,2<br />

1 Department <strong>of</strong> Microbiology, Yonsei <strong>University</strong> College <strong>of</strong> Medicine, Seoul, South<br />

Korea, 2 Brain Korea 21 Project for Medical Sciences, Yonsei <strong>University</strong> College <strong>of</strong><br />

Medicine, Seoul, South Korea<br />

Caenorhabditis elegans is a nematode used in various fields <strong>of</strong> biological research. C.<br />

elegans has been a useful model organism to study innate immunity, because (i) its lifecycle<br />

is shorter than other higher order animals, (ii) it is genetically tractable, and (iii) it shares<br />

common innate immune systems with human. C. elegans has an alimentary canal, similar<br />

to the mammalian intestine. Therefore it has a potential to be used as a model organism to<br />

study microbial interaction with host intestine. At the moment, however, no bacterial species<br />

that forms commensal mutualism with C. elegans has been reported. Because C. elegans is<br />

originally isolated from soil, we tested several bacterial species <strong>of</strong> soil origin to examine whether<br />

they can formulate mutually beneficial relationship with C. elegans host. Many <strong>of</strong> tested strains<br />

exert no virulence to C. elegans. Importantly, one <strong>of</strong> them (soil isolate no. 2) let C. elegans<br />

live significantly longer than the common prey <strong>of</strong> C. elegans, Escherichia coli OP50 does. The<br />

median lethal time (LT 50) <strong>of</strong> this soil isolate no. 2 (SI-2) is over 140 % <strong>of</strong> the OP50 control. We<br />

are undertaking species identification <strong>of</strong> SI-2 at the moment. Not only extending C. elegans’<br />

lifespan, SI-2 enhances larval growth <strong>of</strong> C. elegans. The size <strong>of</strong> larvae fed with SI-2 is twice<br />

the size <strong>of</strong> larvae fed with OP50 when compared at the early developmental stage, while the<br />

grown worms are <strong>of</strong> similar size. But SI-2 cannot rescue reduced resistance to pathogen after<br />

bacterial treatment at the pre-hatch stage. The pre-hatch bacterial treatment with all bacteria<br />

that we treated induces higher susceptibility to pathogens. Our results provide clues that may<br />

broaden our understandings <strong>of</strong> C. elegans lifecycle.<br />

Contact: megasage@hotmail.com<br />

Lab: Yoon<br />

58<br />

Poster Topic: Aging


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

The Role <strong>of</strong> Thioredoxin-1 (TRX-1) in Caenorhabditis elegans Aging<br />

Maria Gonzalez-Barrios 1,2 , Juan Carlos Fierro-Gonzalez 3 , Manuel Munoz 1 , Peter<br />

Swoboda 3 , Antonio Miranda-Vizuete 1,2<br />

1 Centro Andaluz de Biologia del Desarrollo, Universidad Pablo de Olavide, Seville,<br />

Spain, 2 Instituto de Biomedicina de Sevilla (IBIS), Hospital Universitario Virgen<br />

del Rocio/CSIC, Seville, Spain, 3 Center for Biosciences at NOVUM, Dept. <strong>of</strong><br />

Biosciences and Nutrition, Karolinska Institute, Huddinge, Sweden<br />

Thioredoxins comprise a conserved family <strong>of</strong> proteins that mostly depend on their<br />

oxidoreductase attributes to reduce disulfide bonds in many target proteins. These redox<br />

regulators are involved in many biological processes, including stress resistance and aging.<br />

In C. elegans, thioredoxin-1 (TRX-1) is expressed specifically in ASJ sensory neurons, which<br />

have been shown to regulate both dauer formation and longevity. Indeed, we have previously<br />

reported that trx-1 (ok1449) null mutants show a decrease in their lifespan while worms<br />

overexpressing trx-1 have a slight increase in lifespan. Aiming to determine the pathways<br />

where trx-1 impacts longevity, we generated double mutants <strong>of</strong> trx-1 (ok1449) with eat-2<br />

(ad1116), a genetic surrogate <strong>of</strong> caloric restriction. Interestingly, trx-1 (ok1449) fully suppressed<br />

the extended longevity <strong>of</strong> eat-2 (ad1116) mutants. Insulin signaling is another major pathway<br />

regulating longevity in all organisms and in C. elegans it has been shown to act independently<br />

<strong>of</strong> caloric restriction. Surprisingly, trx-1 (ok1449) also reduces the longer lifespan <strong>of</strong> the insulin<br />

receptor mutant daf-2 (e1370) by approximately 70%. However, trx-1 (ok1449) mutants do<br />

not suppress the extended longevity <strong>of</strong> downstream insulin pathway mutants such as pdk-1<br />

(sa680) and, interestingly, double mutants <strong>of</strong> trx-1 (ok1449) with the FOXO transcription factor<br />

gene daf-16 (mu86) are even shorter lived than any <strong>of</strong> the single mutants. Furthermore, trx-1<br />

(ok1449) mutants do not suppress the nuclear translocation <strong>of</strong> DAF-16::GFP in daf-2 (m577)<br />

mutants. Together, these data suggest that TRX-1 regulates longevity through the insulin<br />

pathway, albeit independently <strong>of</strong> DAF-16. The DAF-12 transcription factor belongs to the<br />

nuclear steroid hormone receptor family and has recently been shown to regulate the longevity<br />

<strong>of</strong> some insulin pathway mutants. To determine whether TRX-1 modulates insulin pathway<br />

longevity via DAF-12, we used the reporter DAF-9::GFP, which is a direct transcriptional target<br />

<strong>of</strong> DAF-12 and is upregulated in a daf-2 (m577) mutant background. Our preliminary data<br />

indicate that trx-1 (ok1449) mutants significantly downregulate DAF-9::GFP levels in a daf-2<br />

(m577) background, suggesting that trx-1 might also act in the steroid hormone route. Current<br />

efforts in the lab are dedicated to further explore the role <strong>of</strong> trx-1 in longevity mediated by the<br />

steroid hormone pathway.<br />

Contact: mgonbar@upo.es<br />

Lab: Miranda-Vizuete<br />

Poster Topic: Aging<br />

59


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

Aging is a Determinant in Anoxia Stress Tolerance in Caenorhabditis<br />

elegans<br />

Jo Goy1,2 , Pamela Padilla1 1 2 <strong>University</strong> <strong>of</strong> North Texas, Denton, Texas, USA, Harding <strong>University</strong>, Searcy,<br />

Arkansas, USA<br />

The physiology <strong>of</strong> aging is a hot topic in biological research, driven in part by the increasing<br />

age <strong>of</strong> the human population. Oxygen deprivation is associated with health issues such as<br />

stroke, pulmonary dysfunction and heart disease that commonly affect the elderly. We previously<br />

showed that the one-day old adult hermaphrodite survives 24h <strong>of</strong> anoxia (>90% survivorship,<br />

20C) yet are sensitive to long-term anoxia (LTA, 72h, 20C); anoxia sensitivity is suppressed by<br />

mutations resulting in sterility and decreased ovulation rate. The majority <strong>of</strong> stress response<br />

studies have been conducted on one-day old adult hermaphrodites. We determined that<br />

aging has a positive effect on LTA survival with a significant increase in survivorship at day<br />

3 <strong>of</strong> adulthood and a peak in 5-day old adults. The increase in tolerance was suppressed by<br />

mating at adult day 2, reinforcing the idea that reproduction compromises anoxia tolerance. To<br />

further analyze how aging animals respond to anoxia additional strains including the wild-type<br />

Hawaiian isolate (CB4856), LTA tolerant (daf-2(e1370), glp-1(e2141), fog-2(q71)) and sensitive<br />

(aak-2(gt33), daf-16(mu86)) strains were examined. LTA tolerant strains maintained a high<br />

level <strong>of</strong> survivorship through day 5 <strong>of</strong> adulthood at which point a gradual age-related decline in<br />

survival was observed. Similar to wild-type, LTA sensitive strains showed an increase in anoxiasurvival<br />

rate at day 3 <strong>of</strong> adulthood although rates failed to reach that <strong>of</strong> age-matched wild-type<br />

and LTA tolerant strains. These observations suggest the suppressive effect <strong>of</strong> reproduction<br />

is distinct from the genetic effect. Males show an age-related decrease in anoxia-tolerance<br />

distinct from hermaphrodites. To determine if age influences the effect <strong>of</strong> genotype on anoxia<br />

tolerance we analyzed survivorship <strong>of</strong> the double mutant glp-1(e2141); daf-12(rh61rh411) at<br />

day 1, 3, and 5 <strong>of</strong> adulthood exposed to LTA. At 1 day <strong>of</strong> adulthood absence <strong>of</strong> DAF-12 did not<br />

suppress survivorship after LTA exposure compared to age-matched LTA tolerant glp-1(e2141)<br />

controls. However, at day 5 <strong>of</strong> adulthood anoxia-survival was significantly decreased (p


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

LEP-2/Makorin Promotes let-7 microRNA-mediated Terminal<br />

Differentiation in Male Tail Tip Morphogenesis<br />

R Antonio Herrera, Karin Kiontke, David Fitch<br />

New York <strong>University</strong>, New York, (NY), US<br />

We have identified a new heterochronic gene, lep-2, which is required for male tail tip<br />

morphogenesis (TTM) during L4. Heterochronic genes regulate when stage-specific events<br />

occur during larval development. They interact in a pathway to ultimately schedule terminal<br />

differentiation events at L4. In males, terminal differentiation <strong>of</strong> the tail tip occurs at L4 as<br />

the tail tip cells (hyp8-11) change from a pointed cone-like shape to a rounded dome-like<br />

shape by cell fusion, retraction, and migration. Heterochronic genes that specify L4 fates<br />

(lin-41 and let-7) schedule TTM to start at mid-L4. When lost, lin-41 causes TTM to occur<br />

earlier (in L3), resulting in adult tail tip phenotypes that are over-retracted. When let-7 activity<br />

is reduced, TTM is delayed, resulting in adults with pointed, “leptoderan” tail tips. Loss-<strong>of</strong>function<br />

mutations in lep-2 similarly produce a leptoderan phenotype, with a concomitant<br />

delay in the expression <strong>of</strong> dmd-3, the master regulator <strong>of</strong> TTM. lep-2 animals exhibit other<br />

developmental-delay phenotypes: they fail to exit the larval molting cycle or produce an “adult”<br />

cuticle. Also, in males the Lep phenotype is suppressed after passage through the dauer larvae<br />

stage. Through epistasis analysis, we have determined that lep-2 resides in the heterochronic<br />

pathway downstream <strong>of</strong> lin-14 to promote let-7. In lep-2 mutants we observe elevated levels <strong>of</strong><br />

heterochronic gene reporters that are downregulated prior to TTM (lin-28 & lin-41). Our data<br />

suggest that the function <strong>of</strong> lep-2 is to negatively regulate lin-28, the let-7 repressor, and have<br />

found that LIN-28 protein levels are elevated in lep-2 mutants. With comparative genomic<br />

hybridization on a lep-2 deletion mutant, we identified lep-2 as the sole C. elegans Makorin<br />

(Mkrn). Mkrns are ancient eukaryotic genes which have conserved motifs: a RING domain<br />

flanked by 4 C3H-zinc fingers. Although the functional role <strong>of</strong> Mkrns during development has<br />

not been well understood, a general pattern is their involvement in terminal differentiation<br />

events. Our demonstration that the C. elegans Mkrn is a heterochronic gene suggests that<br />

an anciently conserved Mkrn function in mammals and nematodes may be to promote let-7mediated<br />

differentiation by repressing LIN-28 during development.<br />

Contact: antonio.herrera@nyu.edu<br />

Lab: Fitch<br />

Poster Topic: Small RNAs<br />

61


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

Translational Effect <strong>of</strong> Hydrogen Sulfide on C. elegans<br />

Joseph Horsman, Dana Miller<br />

Deparetment <strong>of</strong> Biochemistry, <strong>University</strong> <strong>of</strong> Washington, Seattle, WA, USA<br />

Exposure to low concentrations <strong>of</strong> H 2S has been shown to increase lifespan in C. elegans<br />

by 70%. The conserved transcription factor hypoxia inducible factor-1 (HIF-1), in additional to<br />

its role in responding to low concentrations <strong>of</strong> oxygen (hypoxia), is key in the initial response<br />

to H 2S. Hypoxia induces dramatic translational effects and it has recently been shown that H 2S<br />

can effect translation in specific mammalian tissues. Our goal is to characterize the organismal<br />

affects <strong>of</strong> H 2S in C. elegans. We have shown that exposure to low levels <strong>of</strong> H 2S does not change<br />

global levels <strong>of</strong> translation in C. elegans. In contrast, we do observe a dramatic decrease in<br />

translation when animals are exposed to hypoxia, as expected. The translation repression<br />

effects <strong>of</strong> acute hypoxia are HIF-1 independent. Unexpectedly, we have found that HIF-1 is<br />

required to maintain translation in H 2S. When H 2S concentration is increased to toxic levels,<br />

translation decreases in a manner corresponding to the toxic dose. We show that decreases<br />

in translation at high concentrations <strong>of</strong> H 2S are not correlated with survival. Together, our<br />

data suggest that effects <strong>of</strong> H 2S on C. elegans, such as lifespan, are not caused by changes<br />

in translation. This work helps build a basis to continue our work in studying the molecular<br />

mechanism by which H 2S affects C. elegans.<br />

Contact: horsman@uw.edu<br />

Lab: Miller<br />

62<br />

Poster Topic: Aging


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

Roles <strong>of</strong> Specific daf-16/FoxO Is<strong>of</strong>orms in Dauer Regulation and<br />

Lifespan Control<br />

Chunfang Guo 1 , Travis Williams 1 , Kathleen Dumas 1 , Sawako Yoshina 2 , Shohei Mitani 2 ,<br />

Patrick Hu 1<br />

1 <strong>University</strong> <strong>of</strong> Michigan, Ann Arbor, MI, USA, 2 Tokyo Women’s Medical <strong>University</strong><br />

School <strong>of</strong> Medicine, Tokyo, Japan<br />

FoxO transcription factors promote longevity in invertebrates and influence the pathogenesis<br />

<strong>of</strong> mouse models <strong>of</strong> common human diseases associated with aging such as cancer, Type 2<br />

diabetes, cardiovascular disease, and osteoporosis. In C. elegans, the FoxO transcription factor<br />

DAF-16 promotes dauer arrest in the context <strong>of</strong> reduced insulin-like signaling during larval<br />

development and prolongs adult lifespan. Recent work has implicated two distinct DAF-16/FoxO<br />

is<strong>of</strong>orms that possess unique N-terminal amino acid sequences, DAF-16A and DAF-16D/F/H,<br />

in dauer regulation and lifespan control. Our experiments with RNAi constructs that target<br />

specific daf-16/FoxO is<strong>of</strong>orms are consistent with previous studies suggesting that DAF-16A is<br />

the major is<strong>of</strong>orm regulating dauer arrest, whereas both DAF-16A and DAF-16D/F/H play roles<br />

in lifespan control. To further elucidate the biological functions <strong>of</strong> DAF-16A and DAF-16D/F/H,<br />

we have isolated deletion mutations that affect specific daf-16/FoxO is<strong>of</strong>orms. Quantitative<br />

reverse transcriptase PCR confirmed the is<strong>of</strong>orm specificity <strong>of</strong> these mutations. Thus far our<br />

analysis reveals that daf-16a-specific mutations fully suppress the dauer-constitutive phenotype<br />

<strong>of</strong> akt-1(mg306) and daf-2(e1368) mutants but only weakly suppress the dauer-constitutive<br />

phenotype <strong>of</strong> daf-2(e1370) mutants. These results are commensurate with those obtained<br />

with daf-16a-specific RNAi constructs. daf-16a-specific mutations also partially suppress the<br />

longevity phenotype <strong>of</strong> daf-2(e1370) mutants, consistent with previous findings implicating both<br />

DAF-16A and DAF-16D/F/H in lifespan control. Interestingly, in animals lacking a germline, daf-<br />

16a-specific mutations suppressed lifespan extension to nearly the same extent as a daf-16/<br />

FoxO null mutation. Thus, in contrast to insulin-like signaling, which controls lifespan through<br />

both DAF-16A and DAF-16D/F/H, the germline influences longevity primarily by regulating<br />

DAF-16A activity. Experiments with daf-16d/f/h-specific mutations are ongoing.<br />

Contact: pathu@umich.edu<br />

Lab: Hu<br />

Poster Topic: Aging<br />

63


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

Quantitative In Vivo Redox Sensors Uncover Oxidative Stress as an<br />

Early Event in Life<br />

Daniela Knoefler, Maike Thamsen, Martin Koniczek, Nicholas Niemuth, Ann-Kristin<br />

Diederich, Ursula Jakob<br />

<strong>University</strong> <strong>of</strong> Michigan, Ann Arbor, MI<br />

Obstacles in elucidating the role <strong>of</strong> oxidative stress in aging include difficulties in 1) tracking<br />

in vivo oxidants, in 2) identifying proteins which are affected, and in 3) correlating changes in<br />

oxidant levels with lifespan. We used quantitative redox proteomics to determine the onset<br />

and the cellular targets <strong>of</strong> oxidative stress during the lifespan <strong>of</strong> Caenorhabditis elegans. In<br />

parallel, we used genetically encoded sensor proteins to determine peroxide levels in live<br />

animals in real time. We discovered that C. elegans encounters significant levels <strong>of</strong> oxidants<br />

as early as during larval development. Oxidant levels drop rapidly as animals mature and<br />

reducing conditions prevail throughout the reproductive age, after which age-accompanied<br />

protein oxidation sets in. Long-lived daf-2 mutants transition faster to reducing conditions,<br />

whereas short-lived daf-16 mutants retain higher oxidant levels throughout their mature life.<br />

These results suggest that animals with improved capacity to recover from early oxidative<br />

stress have significant advantages later in life.<br />

Contact: knoefler@umich.edu<br />

Lab: Jakob<br />

64<br />

Poster Topic: Aging


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

Genes Acting Downstream <strong>of</strong> Sensory Neurons to Influence<br />

Longevity, Dauer Formation and Pathogen Responses in C. elegans<br />

Marta Gaglia 1 , Dae-Eun Jeong 2 , Eun-A Ryu 2 , Dongyeop Lee 2 , Seung-Jae Lee 2<br />

1 <strong>University</strong> <strong>of</strong> California, Berkeley, Berkeley, California, USA, 2 Pohang <strong>University</strong><br />

<strong>of</strong> Science & Technology, Pohang, Gyeongbuk, South Korea<br />

Sensory neurons <strong>of</strong> C. elegans affect the decision between arrest in the dauer stage<br />

and reproductive growth during larval development, and they also modulate the lifespan <strong>of</strong><br />

the animals in adulthood. However, the molecular mechanisms underlying these effects are<br />

incompletely understood. Here we report how different effector genes act downstream <strong>of</strong><br />

sensory neurons to influence three physiological outputs – longevity, dauer formation and<br />

pathogen response. We identified transcripts whose levels are influenced by mutations in the<br />

intraflagellar transport protein daf-10, which cause defects in the morphology and function <strong>of</strong><br />

many ciliated sensory neurons in C. elegans. Among the genes with altered expression in daf-<br />

10 mutant animals, we found transcriptional targets <strong>of</strong> the DAF-12/nuclear hormone receptor.<br />

We show that DAF-12/nuclear hormone receptor influences specifically the high-temperatureinduced<br />

dauer-formation phenotype <strong>of</strong> these animals, but does not affect their extended<br />

lifespan. Moreover, we identified a solute transporter gene that is induced by daf-10 mutations<br />

and is required for their effect on longevity. Unexpectedly, we found that pathogen-responsive<br />

genes were repressed in daf-10 mutant animals and that these sensory mutants exhibited<br />

altered susceptibility to and behavioral avoidance <strong>of</strong> bacterial pathogens, independently <strong>of</strong><br />

DAF-12/NHR or the solute transporter. Thus, sensory input seems to influence a diverse set<br />

<strong>of</strong> transcriptional responses that modulate basic biological processes in C. elegans.<br />

Contact: seungjaelee1@gmail.com<br />

Lab: Lee<br />

Poster Topic: Aging<br />

65


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

Age-dependent Alteration <strong>of</strong> Major Large Intestinal Granules in<br />

Caenorhabditis elegans<br />

Kenji Nishikori, Takahiro Tanji, Eisuke Kuroda, Yuki Ueda, Hirohisa Shiraishi, Ayako<br />

Ohashi-Kobayashi<br />

Iwate Medical <strong>University</strong>, Yahaba, Iwate, Japan<br />

Dietary restriction response in C. elegans is one <strong>of</strong> the important targets <strong>of</strong> aging research.<br />

Intestine is a major organ responsible for uptake, digestion, storage and distribution <strong>of</strong> nutrients.<br />

In C. elegans intestinal cells, there are several different types <strong>of</strong> granules, which seem to be<br />

related with certain functions <strong>of</strong> the organ, however their function and regulation in response<br />

to internal and external conditions have yet to be elucidated. Therefore, we focused on the<br />

age- and nutrition-dependent changes <strong>of</strong> the intestinal granules in order to find new indexes<br />

<strong>of</strong> aging.<br />

At the late larval and early adult stages, at least three types <strong>of</strong> intestinal granules have been<br />

identified: lysosomotropic dyes (acridine orange and LysoTracker)–positive acidic granules,<br />

oil red O-positive fat storage granules, and LMP-1-localized non-acidic granules [1,2,3]. We<br />

previously reported that ABC transporters, HAF-4 and HAF-9, colocalize with LMP-1, on the<br />

surface <strong>of</strong> a major subset <strong>of</strong> intestinal granules. Interestingly, in the deletion mutants for haf-4<br />

and haf-9, loss <strong>of</strong> the non-acidic granules, but not <strong>of</strong> other granules, was observed [4].<br />

Here we report that even in the haf-4 and haf-9 mutant worms, some kinds <strong>of</strong> intestinal<br />

granules come to occupy most <strong>of</strong> the cytosolic volume at the postreproductive adult stage.<br />

We aimed to characterize those granules emerging at the later stage <strong>of</strong> adulthood by using<br />

fluorescent protein-tagged marker proteins. As a result, LMP-1, HAF-4, and HAF-9 were not<br />

localized any longer on the major intestinal granules in the aged wildtype N2 worms. In addition,<br />

we also revealed that the difference <strong>of</strong> major granules in larvae and adults in response to food<br />

deprivation. HAF-4 and HAF-9-positive larval intestinal granules selectively decrease by food<br />

deprivation, whereas HAF-4 and HAF-9-negative intestinal granules in aged worms still exist<br />

after food deprivation treatment. These results indicate that the age-associated alteration <strong>of</strong><br />

major intestinal granules happens during adulthood. Further molecular characterization <strong>of</strong><br />

these granules is on going.<br />

References<br />

1. Hermann GJ et al. Mol. Cell. Biol. 2005. 2. O’Rourke EJ et al. Cell Metab. 2009. 3. Kostich M. et al. J. Cell.<br />

Sci. 2000. 4. Kawai H. et al. Mol. Cell. Biol. 2009<br />

Contact: aohashi@iwate-med.ac.jp<br />

Lab: Ohashi-Kobayashi<br />

66<br />

Poster Topic: Aging


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

Electrolyzed-reduced water confers increased resistance to<br />

environmental stresses and longevity in C. elegans<br />

Seul-Ki Park, Soon-Young Lee, Sang-Kyu Park<br />

Soonchunhyang <strong>University</strong>, Asan, Rep. <strong>of</strong> Korea<br />

Free radical theory <strong>of</strong> aging suggests oxidative stress caused by reactive oxygen species<br />

plays a pivotal role in normal aging. To determine the effect <strong>of</strong> reduced water on stress response<br />

and aging, we used electrolyzed-reduced water. Electrolysis <strong>of</strong> water produces reduced water<br />

at the cathode and oxidized water at the anode. Previous studies show that electrolyzedreduced<br />

water could block the activity <strong>of</strong> reactive oxygen species produced as a byproduct <strong>of</strong><br />

metabolism in cells and protect cellular DNA from oxidative damage caused by free radicals in<br />

human lymphocyte. Here, we tested the effect <strong>of</strong> electrolyzed-reduced water on resistance to<br />

various environmental stresses, such as oxidative stress, heat stress, and UV in the nematode<br />

Caenorhabditis elegans. Worms grown in NGM media prepared with electrolyzed-reduced<br />

water have increased resistance to oxidative stress, thermotolerance, and UV-resistance<br />

compared to worms grown in media made with distilled water. We also observed a significant<br />

lifespan-extending effect <strong>of</strong> electrolyzed-reduced water in Caenorhabditis elegans. In addition,<br />

the fertility, one <strong>of</strong> biomarkers <strong>of</strong> healthspan in Caenorhabditis elegans, was significantly<br />

increased by electrolyzed-reduced water. These data suggest that electrolyzed-reduced water<br />

can play as a powerful radical scavenger and, as a result <strong>of</strong> that, can extend both lifespan and<br />

healthspan <strong>of</strong> Caenorhabditis elegans.<br />

Contact: skpark@sch.ac.kr<br />

Lab: Park<br />

Poster Topic: Aging<br />

67


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

A daf-16b-specific Mutation Extends C. elegans Lifespan<br />

Andy Polzin 1 , Chunfang Guo 1 , Sawako Yoshina 2 , Shohei Mitani 2 , Patrick Hu 1<br />

1 <strong>University</strong> <strong>of</strong> Michigan, Ann Arbor, MI, USA, 2 Tokyo Women’s Medical <strong>University</strong><br />

School <strong>of</strong> Medicine, Tokyo, Japan<br />

In C. elegans, the FoxO ortholog daf-16 promotes dauer arrest, stress resistance, and<br />

lifespan extension in the context <strong>of</strong> reduced insulin-like signaling. Recent evidence has<br />

suggested that individual DAF-16/FoxO is<strong>of</strong>orms may have distinct biological functions.<br />

Studies in which each is<strong>of</strong>orm was transgenically expressed in a daf-16/FoxO null background<br />

implicated DAF-16A and DAF-16D/F/H as the major is<strong>of</strong>orms governing dauer arrest, stress<br />

resistance, and lifespan control. To further explore the function <strong>of</strong> specific DAF-16/FoxO<br />

is<strong>of</strong>orms, we generated animals harboring deletion mutants that selectively eliminate the<br />

expression <strong>of</strong> individual DAF-16/FoxO is<strong>of</strong>orms. Strikingly, we found that animals with the<br />

daf-16b(tm5031) mutation live approximately 20% longer and are more thermotolerant than<br />

wild-type animals. This finding suggests that, in contrast to DAF-16A and DAF-16D/F/H, which<br />

promote longevity, DAF-16B shortens lifespan. daf-16b(tm5031) does not further extend the<br />

lifespan <strong>of</strong> daf-2 mutants or glp-1 mutants that lack a germline, suggesting that DAF-16B<br />

may control lifespan by regulating the activity <strong>of</strong> other DAF-16/FoxO is<strong>of</strong>orms. Experiments<br />

to determine the site <strong>of</strong> action <strong>of</strong> DAF-16B in lifespan control are ongoing.<br />

Contact: polzinaj@umich.edu<br />

Lab: Hu<br />

68<br />

Poster Topic: Aging


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

Molecular Mechanisms <strong>of</strong> C. elegans Germline Stem Cell Aging<br />

Zhao Qin, E. Jane Albert Hubbard<br />

Developmental Genetics Program, Skirball Institute <strong>of</strong> Biomolecular Medicine,<br />

NYU School <strong>of</strong> Medicine, New York, NY, USA<br />

Failure to maintain stem cells with age is associated with conditions such as tissue<br />

degeneration and increased susceptibility to tissue damage in many organisms, including<br />

humans. Here we use the C. elegans germ line as a general model to study stem cell aging.<br />

The C. elegans germ line combines a well-established genetic model for aging studies with an<br />

accessible stem cell system, providing a unique opportunity to dissect the effects <strong>of</strong> aging on<br />

stem cells. Our results suggest a depletion mechanism that requires insulin/IGF-like signaling<br />

via DAF-16/FOXO. Further, we found that insulin signaling acts neither in the germ line nor the<br />

intestine. These results suggest a novel mechanism for insulin/IGF-like signaling in regulating<br />

age-associated physiological changes that is anatomically separable from its previously<br />

described roles in larval germline proliferation and lifespan regulation.<br />

Contact: Zhao.Qin@med.nyu.edu<br />

Lab: Hubbard<br />

Poster Topic: Aging<br />

69


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

Nutritional Deprivation in the Late Larval Stages <strong>of</strong> C. elegans Induces<br />

Developmental Diapause at Precise Checkpoints<br />

Adam Schindler, L Baugh, David Sherwood<br />

Duke <strong>University</strong>, Durham, NC USA<br />

C. elegans has served as a model system to study the effects <strong>of</strong> nutritional conditions<br />

on development. When food is abundant, animals grow through four larval stages, L1–L4,<br />

before reaching adulthood. In the absence <strong>of</strong> sufficient nutrients, animals arrest growth either<br />

in the L1 larval stage or in dauer, an alternative developmental pathway that is initiated late<br />

in the L1 stage. The effects <strong>of</strong> nutrient deprivation on progression through later larval stages<br />

has not been thoroughly explored, and is <strong>of</strong> interest because several tissues develop during<br />

these times, including those that form the egg-laying system. To examine the effects <strong>of</strong> nutrient<br />

deprivation on later larval development, we first grew animals to approximately the L2 molt<br />

before removal from food. Under these conditions, animals arrested growth in the early-L3<br />

larval stage, as determined by molting cycle reporters and by the pattern <strong>of</strong> cell divisions in the<br />

vulval, uterine, sex muscle, and germline cell lineages. When animals were instead removed<br />

from food shortly after the L2 molt, about half the population passed the L3 checkpoint and<br />

invariantly molted to L4, where they again arrested in the early part <strong>of</strong> the larval stage. In<br />

L4-arrested animals, all <strong>of</strong> the tissue lineages examined developed beyond their L3 arrest<br />

points, suggesting that a global developmental checkpoint had been passed. The extent <strong>of</strong><br />

development upon passing the L3 checkpoint varied between tissues: vulval precursor cells<br />

completed all cell divisions and arrested at a precise developmental point, whereas uterine,<br />

sex muscle, and germ cell divisions arrested at earlier developmental times and in a variable<br />

manner. Our results demonstrate that global checkpoints regulate development through the<br />

L3 and L4 larval stages, and that tissue-specific responses determine the extent <strong>of</strong> growth<br />

upon passing the checkpoints. Our work expands the range <strong>of</strong> developmental checkpoints in<br />

C. elegans to include the later larval stages after the dauer decision.<br />

Contact: adam.schindler@duke.edu<br />

Lab: Sherwood<br />

70<br />

Poster Topic: Aging


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

Shared Targets <strong>of</strong> TGF-β and Insulin/IGF-1 Signaling Regulate<br />

Reproductive Aging through Mechanisms Distinct from Somatic<br />

Aging Regulation<br />

Shijing Luo, Cheng Shi, Jasmine Ashraf, Coleen Murphy<br />

Lewis-Sigler Institute for Integrative Genomics and Department <strong>of</strong> Molecular<br />

Biology, Princeton <strong>University</strong>, Princeton, NJ 08544, USA<br />

In humans, female reproductive capacity decline is one <strong>of</strong> the earliest aging phenotypes.<br />

We recently found that both TGF-β Sma/Mab and Insulin/IGF-1 signaling (IIS) regulate C.<br />

elegans reproductive span through maintenance <strong>of</strong> oocyte and germline quality with age.<br />

However, the molecular mechanisms downstream <strong>of</strong> IIS that regulate oocyte quality are<br />

unknown. Furthermore, it is not known whether the mechanisms that maintain oocyte quality<br />

and reproductive span are the same as those that maintain somatic tissues with age and<br />

extend the life span <strong>of</strong> IIS mutants.<br />

Here we have used transcriptional analysis to identify a set <strong>of</strong> gene targets regulated<br />

by the IIS receptor DAF-2 in oocytes. Many <strong>of</strong> the daf-2 oocyte targets are shared with the<br />

oocyte targets <strong>of</strong> the TGF-β Sma/Mab signaling mutant, sma-2, which also slows reproductive<br />

aging. Our data suggest that the IIS and TGF-β Sma/Mab pathways share important common<br />

mechanisms <strong>of</strong> oocyte quality maintenance. These mechanisms are also the processes that<br />

decline with age in mammalian oocytes. By contrast, transcriptional analyses suggest that daf-<br />

2’s somatic and oocyte maintenance targets are largely non-overlapping. Thus, IIS appears to<br />

regulate reproductive aging and somatic aging through largely different mechanisms. Our results<br />

indicate that mitotic and post-mitotic cells use distinct molecular strategies to combat aging.<br />

Contact: cshi@princeton.edu<br />

Lab: Murphy<br />

Poster Topic: Aging<br />

71


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

The Role <strong>of</strong> FAHD1 in the Aging <strong>of</strong> Caenorhabditis elegans<br />

Andrea Taferner 1 , Haymo Pircher 1 , Lucia Micutkova 1 , Nektarios Tavernarakis 2 , Pidder<br />

Jansen-Duerr 1<br />

1 Institute for Biomedical Aging Research, Innsbruck, Austria, 2 Institute <strong>of</strong><br />

Molecular Biology and Biotechnology, Foundation for Research and Technology,<br />

Heraklion, Greece<br />

The 25 kDa protein FAHD1 (FAH domain containing protein 1) is the second member <strong>of</strong> the<br />

FAH superfamily that has been identified in the human genome but it has not been researched<br />

extensively so far. In a recent study, it was shown that FAHD1 exhibits acylpyruvase activity,<br />

demonstrated by the hydrolysis <strong>of</strong> acetylpyruvate and fumarylpyruvate in vitro. The enzyme<br />

was found expressed in all tested murine tissues, with highest expression in liver and kidney<br />

and was also found in several human cell lines, where it localized to mitochondria (Pircher et<br />

al., 2011). Using a proteomic screen for mitochondrial proteins that are differentially regulated<br />

in young and senescent human cells, FAHD1 has been identified as a target for age-related<br />

post-translational modifications.<br />

Concerning organismic aging, it has been shown that the depletion <strong>of</strong> the FAHD1 gene<br />

product by RNAi led to a significant reduction in the mean lifespan <strong>of</strong> Caenorhabditis elegans.<br />

To further examine the physiological function <strong>of</strong> the FAHD1 ortholog in the worm, a FAHD1<br />

knock-out mutant C. elegans strain was generated. This mutant worm shows – in addition to<br />

the lifespan reduction - abnormalities in its phenotype compared to wild-type worms, especially<br />

concerning fecundity and locomotion. It will be used in a variety <strong>of</strong> experiments, including<br />

epistasis experiments and metabolic studies.<br />

The findings suggest an important function for the FAHD1 ortholog in C. elegans. We hope<br />

that with our work we will be able to clarify the physiological function and regulation <strong>of</strong> FAHD1,<br />

as well as its involvement in the aging process <strong>of</strong> C. elegans.<br />

Contact: andrea.taferner@oeaw.ac.at<br />

Lab: Jansen-Duerr<br />

72<br />

Poster Topic: Aging


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

Physical Interaction <strong>of</strong> Half ABC Transporters HAF-4 and HAF-9,<br />

Which Are Required for the Biogenesis <strong>of</strong> the Intestinal Lysosomerelated<br />

Organelles in Caenorhabditis elegans<br />

Takahiro Tanji, Kenji Nishikori, Hirohisa Shiraishi, Masatomo Maeda, Ayako Ohashi-<br />

Kobayashi<br />

Iwate Medical <strong>University</strong>, Yahaba, Iwate, Japan<br />

C. elegans HAF-4 and HAF-9 are ABC transporters highly homologous to human lysosomal<br />

peptide transporter TAPL (TAP-like; ABCB9). From their high identity at the amino acid sequence<br />

level, they are predicted to be paralogues. They colocalize on the membrane <strong>of</strong> non-acidic but<br />

LAMP (lysosome-associated membrane protein) homolog LMP-1-positive lysosome-related<br />

organelles in intestinal cells, and are both required for the normal formation <strong>of</strong> the organelles<br />

(Kawai et al. (2009) Mol. Biol. Cell, 20, 2979-90). Although the organelles are prominent from<br />

the late larval to young adult stage, they rapidly collapse by food deprivation, or disappear as<br />

the worm ages even under fed conditions. HAF-4 and HAF-9 might actually participate in the<br />

dynamic changes <strong>of</strong> the organelles in response to age and trophic conditions.<br />

As HAF-4 and HAF-9 are half-type transporters, they should function as homodimers and/<br />

or a heterodimer. Preceding genetic interaction analyses between haf-4 and haf-9 proposed<br />

two possible models; they function as a heterodimer, and/or they function differentially but<br />

cooperatively as homodimers (Tanji et al., 4th East Asia C. elegans Meeting). However, which<br />

mode <strong>of</strong> dimerization they employ is yet to be identified.<br />

Here, we show that HAF-4 and HAF-9 require each other for their stable expression.<br />

Western blot analysis revealed that the expression level <strong>of</strong> HAF-4 and HAF-9 decreased in<br />

both the haf-4 and haf-9 single mutants. Moreover, knock-down <strong>of</strong> either gene by means <strong>of</strong><br />

RNA interference results in the decreasing fluorescence <strong>of</strong> both HAF-4::GFP and HAF-9::GFP<br />

in the transgenic worms. These results suggest that HAF-4 and HAF-9 are not stable in the<br />

absence <strong>of</strong> each other. Furthermore, we detected the physical interaction between HAF-4::GFP<br />

and HAF-9::mCherry in the transgenic worms by co-immunoprecipitation analyses. These<br />

results strongly suggest the heterodimer formation <strong>of</strong> HAF-4 and HAF-9.<br />

Contact: ttanji@iwate-med.ac.jp<br />

Lab: Ohashi-Kobayashi<br />

Poster Topic: Aging<br />

73


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

Neurite Sprouting and Synapse Deterioration in the Aging C. elegans<br />

Nervous System<br />

Marton Toth1 , Ilija Melentijevic1 , Leena Shah1 , Aatish Bhatia1 , Kevin Lu1 , Amish Talwar1 ,<br />

Haaris Naji1 , Carolina Ibanez-Ventoso1 , Piya Ghose1 , Angelina Jevince2 , Laura<br />

Herndon2 , Gyan Bhanot1 , Christopher Rongo1 , David Hall2 , Jian Xue1 , Monica Driscoll1 1 2 Rutgers <strong>University</strong>, Piscataway, NJ, USA, Albert Einstein College <strong>of</strong> Medicine,<br />

New York, NY, USA<br />

C. elegans is a powerful model for analysis <strong>of</strong> the conserved mechanisms that modulate<br />

healthy aging. In the aging nematode nervous system, neuronal death and/or detectable loss<br />

<strong>of</strong> processes are not readily apparent, but because dendrite restructuring and loss <strong>of</strong> synaptic<br />

integrity are hypothesized to contribute to human brain decline and dysfunction, we combined<br />

fluorescence microscopy and electron microscopy (EM) to screen at high resolution for nervous<br />

system changes. We report two major components <strong>of</strong> morphological change in the aging C.<br />

elegans nervous system: 1) accumulation <strong>of</strong> novel outgrowths from specific neurons, and 2)<br />

physical decline in synaptic integrity. Novel outgrowth phenotypes, including branching from the<br />

main dendrite or new growth from somata, appear at a high frequency in some aging neurons,<br />

but not all. Mitochondria are <strong>of</strong>ten associated with age-associated branch sites. Lowered insulin<br />

signaling confers some maintenance <strong>of</strong> ALM and PLM neuron structural integrity into old age,<br />

and both DAF-16/FOXO and heat shock factor transcription factor HSF-1 exert neuroprotective<br />

functions. hsf-1 can act cell autonomously in this capacity. EM evaluation in synapse-rich<br />

regions reveals a striking decline in synaptic vesicle numbers and a dimunition <strong>of</strong> presynaptic<br />

density size. Interestingly, old animals that maintain locomotory prowess exhibit less synaptic<br />

decline than same-age decrepit animals, suggesting that synaptic integrity correlates with<br />

locomotory healthspan. Our data reveal similarities between the aging C. elegans nervous<br />

system and mammalian brain, suggesting conserved neuronal responses to age. Dissection<br />

<strong>of</strong> neuronal aging mechanisms in C. elegans may thus influence the development <strong>of</strong> brain<br />

healthspan-extending therapies.<br />

Contact: marton.l.toth@gmail.com<br />

Lab: Driscoll<br />

74<br />

Poster Topic: Aging


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

Identification <strong>of</strong> the Core Chaperone Network that Modulates<br />

Proteostasis in C. elegans<br />

Cindy Voisine, Kai Orton, Richard Morimoto<br />

Northwestern <strong>University</strong>, Evanston, (IL), USA<br />

Chaperone networks have central roles in proteostasis to prevent misfolding and the<br />

accumulation <strong>of</strong> protein aggregates that occur during aging and age-related neurodegenerative<br />

disease. While chaperones have been shown to suppress aggregation and toxicity <strong>of</strong> diverse<br />

proteins in multiple model systems, nearly all <strong>of</strong> these studies have taken candidate gene<br />

approaches, thus providing little understanding <strong>of</strong> the chaperone network. To address this,<br />

we have taken a functional domain-based, hierarchical clustering approach to identify 203<br />

chaperones belonging to sixteen families including individualized cytoplasmic and organellar<br />

folding factors that are expressed in the metazoan C. elegans. Using well-characterized<br />

folding sensors, including disease associated proteins and metastable proteins expressed in<br />

the body wall muscles, we asked whether a shared chaperone network exists that maintains<br />

functionality in vivo despite the proteotoxic stress. Reduced expression <strong>of</strong> only 16 chaperones<br />

by RNAi, corresponding to specific members <strong>of</strong> the Hsp90, Hsp70, Hsp60 (chaperonin), Hsp40,<br />

and TPR families, enhanced toxicity when Aβ or polyglutamine is expressed. Despite having<br />

distinct primary amino acid sequences and no structural commonalities other than forming<br />

aggregates suggests that these specific members function as a core chaperone network to<br />

maintain proteostasis. Knockdown <strong>of</strong> the core chaperone network also destabilized endogenous<br />

metastable proteins and accelerated the onset <strong>of</strong> sarcopenia in wild type animals. These findings<br />

implicate a small limited network <strong>of</strong> core chaperones that protects the proteome during aging<br />

and in age-related diseases.<br />

Contact: c-voisine@northwestern.edu<br />

Lab: Morimoto<br />

Poster Topic: Aging<br />

75


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

The Conserved MicroRNA-80 Modulates C. elegans Longevity through<br />

Dietary Restriction<br />

Mehul Vora, Mitalie Shah, Jian Xue, Monica Driscoll<br />

Rutgers, The State <strong>University</strong> <strong>of</strong> New Jersey, Piscataway (NJ), U.S.A<br />

Dietary or Caloric restriction (DR) is a conserved metabolic pathway that can prolong<br />

longevity and extend mid-life vigor (healthspan). In almost all model organisms, DR can delay<br />

onset <strong>of</strong> age-associated diseases. We conducted a screen to ascertain whether any <strong>of</strong> the<br />

available mir mutants are in constitutive DR, using a unique fluorimetric signature for the C.<br />

elegans DR state that our lab previously discovered. We exploited this biomarker to screen<br />

through miRNA deletion mutants to identify mir mutants that might persist in a constitutive<br />

DR-like state.<br />

We identified a single mir mutant, affecting the conserved mir-80 gene, which robustly<br />

exhibits the fluorimetric DR signature. mir-80 mutants [referred as mir-80(Δ)] also exhibit<br />

reduced fecundity and appear pale and thin. Like DR-animals, mir-80(Δ) is hypersensitive<br />

to the DR mimetic drug metformin, which is the drug response expected for animals that are<br />

already in DR (pushed over the edge into starvation). Interestingly, a transcriptional GFP<br />

reporter for mir-80 is responsive to food (upregulated in the presence <strong>of</strong> food and downregulated<br />

in absence <strong>of</strong> food) for three different DR regimens and a key molecular reporter<br />

<strong>of</strong> DR, the transcription factor skn-1, is activated by mir-80 deletion.Consistent with being in<br />

DR, mir-80(Δ) also exhibits several parameters <strong>of</strong> healthy aging, including extension <strong>of</strong> mean<br />

lifespan, reduced age pigment accumulation, and maintenance <strong>of</strong> youthful swimming and<br />

muscle physiology in old age compared to wild type animals. Gene expression analysis and<br />

a pilot screen <strong>of</strong> known DR-associated genes identified 5 conserved genes to be involved<br />

in the longevity phenotypes <strong>of</strong> mir-80(Δ). We also show genetically that mir-80(Δ) longevity<br />

effects are partially dependent on IIS pathway components. In sum, the mir-80(Δ) mutant is<br />

constitutively in DR even though it eats an ad lib diet—and it ages well as a consequence.<br />

To our knowledge, this work describes the first miRNA that modulates healthy aging by DR<br />

metabolic regulation.<br />

Contact: mehulmvora@gmail.com<br />

Lab: Driscoll<br />

76<br />

Poster Topic: Aging


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

Supplemental cellular protection by a carotenoid astaxanthin extends<br />

lifespan via ins/IGF-1 signaling in C. elegans<br />

Sumino Yanase<br />

Daito Bunka <strong>University</strong> School <strong>of</strong> Sports and Health Science, Department <strong>of</strong><br />

Health Science, Higashi-matsuyama, Saitama, Japan<br />

Astaxanthin (AX), which is produced by some marine animals, is a type <strong>of</strong> carotenoid<br />

that has antioxidative properties. In this study, we initially examined the effects <strong>of</strong> AX on the<br />

aging <strong>of</strong> a model organism C. elegans that has the conserved intracellular pathways related<br />

to mammalian longevity. The continuous treatments with AX (0.1 to 1 mM) from both the<br />

prereproductive and young adult stages extended the mean lifespans by about 16-30% in the<br />

wild-type and long-lived mutant age-1 <strong>of</strong> C. elegans. In contrast, the AX-dependent lifespan<br />

extension was not observed even in a daf-16 null mutant. Especially, the expression <strong>of</strong> genes<br />

encoding superoxide dismutases and catalases increased in two weeks after hatching, and<br />

the DAF-16 protein was translocated to the nucleus in the AX-exposed wild-type. These results<br />

suggest that AX protects the cell organelle mitochondria and nucleus <strong>of</strong> the nematode, resulting<br />

in a lifespan extension via an Ins/IGF-1 signaling pathway during normal aging, at least in part.<br />

Contact: syanase@ic.daito.ac.jp<br />

Lab: Yanase<br />

Poster Topic: Aging<br />

77


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

The Role <strong>of</strong> Thioredoxin Reductase in Inorganic Selenium Metabolism<br />

in C. elegans<br />

Christopher Boehler, Roger Sunde<br />

<strong>University</strong> <strong>of</strong> <strong>Wisconsin</strong>, Madison, WI, USA<br />

The trace element selenium (Se) is an essential mineral with a narrow range <strong>of</strong><br />

concentrations between adequacy and toxicity. Recent studies have associated higher intakes<br />

<strong>of</strong> Se with prevention <strong>of</strong> cancer and other diseases, but increased Se supplementation has<br />

also demonstrated adverse effects on human health. Therefore, further understanding <strong>of</strong> the<br />

metabolism underlying Se toxicity is needed. In this study, we used a C. elegans biological<br />

model to assess chronic inorganic Se toxicity, and to determine the essentiality <strong>of</strong> a proposed<br />

key enzyme in inorganic Se reduction and metabolism, thioredoxin reductase (trxr). Wild-type<br />

N2 Bristol (N2B) worms were grown in a defined, Se-deficient axenic liquid media (0.1 µM<br />

Se) for a period <strong>of</strong> 12 days with increasing Se concentrations ranging from 0 mM-5 mM using<br />

different inorganic Se forms (selenate, selenite, selenide). Knock-out strains for trxr-1 and<br />

trxr-2 genes were created and utilized in dose response curves to investigate the essentiality<br />

<strong>of</strong> thioredoxin reductase in handling excess Se. N2B C. elegans exposed to Se in the form <strong>of</strong><br />

selenite or selenide displayed a similar, sigmoidal growth response to increasing concentrations<br />

<strong>of</strong> Se with reduced growth at 0.1 mM Se, a 50% growth reduction (LC50) at 0.2 mM and 0.23<br />

mM respectively, and lethality at 1 mM Se. In contrast, selenate resulted in 1/5th the toxicity<br />

seen for selenite and selenide and a LC50 <strong>of</strong> 0.95 mM Se in N2B worms, with lethality not<br />

observed until 3 mM Se. trxr-1-/- and trxr-2 -/- knock-out strains were outcrossed to N2 males,<br />

with genotypes confirmed by PCR and SDS-PAGE gels. A double knock-out (dko) was created<br />

through crossing the single mutants to further determine if trxr is essential in increasing Se<br />

concentrations. The absence <strong>of</strong> the trxr-1, trxr-2, or both genes (dko) displayed no obvious<br />

growth defects, or changes in sensitivity to increasing concentrations <strong>of</strong> any form <strong>of</strong> inorganic<br />

Se. Interestingly, while the LC50 concentration ranged from 0.16-0.23 mM for selenite and<br />

selenide in wild-type, single and dko strains, the LC50 for selenate was about 4 fold higher<br />

(0.85 mM) in wild-type and knock-out strains. Results indicating a decreased toxicity <strong>of</strong> selenate<br />

compared to selenite or selenide. The absence <strong>of</strong> the thioredoxin reductase does not change<br />

the susceptibility to Se toxicity suggesting additional alternate reductase(s) can reduce selenite<br />

and selenate in C. elegans. (Funded in part by UW WIS04909 and WIS01435)<br />

Contact: cboehler@wisc.edu<br />

Lab: Sunde<br />

78<br />

Poster Topic: Metabolism


Contact: cbrey@marywood.edu<br />

Lab: Hashmi<br />

Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

Regulation <strong>of</strong> lipid storage and secretion: Genetics and Molecular<br />

Analysis<br />

Christopher Brey1 , Jun Zhang2 , Sanya Hashmi2 , Randy Gaugler3 , Sarwar Hashmi 2<br />

1 2 Marywood <strong>University</strong>, Scranton, PA, 18505, LFKI, NY Blood Center, Manhattan,<br />

NY 10065, 3Rutgers <strong>University</strong>, New Brunswick, NJ 08901<br />

To maintain lipid homeostasis, dietary lipids are absorbed from the small intestine and<br />

transported to various organs and tissues. The excess <strong>of</strong> ingested food is largely converted<br />

into intracellular TG deposits in lipid droplets that are most prominent in mammalian adipose<br />

tissues and to a lesser degree in heart, muscle and liver. The stored TG provides the primary<br />

source <strong>of</strong> energy during periods <strong>of</strong> food deficiency. However, the accumulation <strong>of</strong> high TG<br />

levels can cause hyperlipidemia, a serious medical condition with a disturbing epidemic<br />

forecast afflicting a growing number <strong>of</strong> the human population. In mammals there is a strict<br />

regulatory mechanism <strong>of</strong> lipid transport and distribution that helps maintain normal levels <strong>of</strong> TG<br />

in adipocytes. Some members <strong>of</strong> mammalian Krüppel-like factors, KLFs are the key players<br />

controlling adipogenesis. We have shown that aberrant activity <strong>of</strong> a Caenorhabditis elegans<br />

KLF-3 not only results in high TG accumulation in the intestine but also caused defects in<br />

fertility suggesting that excessive fat deposition and reproductive defects may be intimately<br />

linked. We also found reduced expression <strong>of</strong> C. elegans dsc-4 and/or vit genes, the homologs<br />

<strong>of</strong> mammalian MTP and apoB respectively that control mammalian lipoprotein assembly and<br />

transport. Similar to klf-3 (ok1975) mutation, mutation in both dsc-4 (qm182) and vit-5 (ok3239),<br />

results in elevated levels <strong>of</strong> fat accumulation in worm intestine. Our data suggests that Klf-3<br />

functions to limit fat storage and plays a role in its mobilization to other tissues. Hence, klf-3 is<br />

an essential regulator <strong>of</strong> lipid storage and transport, that mutation in klf-3 affects this regulatory<br />

function leading to fat accumulation. Our research aim is to define the mechanisms by which<br />

KLF-3 exerts its effects, and sets the groundwork for translation <strong>of</strong> these findings towards the<br />

goal <strong>of</strong> improved prevention and treatment <strong>of</strong> hyperlipidemia, and its related disorders. We are<br />

using three complementary, but interdependent, strings <strong>of</strong> research involving a) C. elegans;<br />

b) cellular and c) mouse models.<br />

Poster Topic: Metabolism<br />

79


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

Investigation <strong>of</strong> Satiety Quiescence Signaling Using Automated<br />

Locomotion Assay and Hidden Markov Model Analysis to Identify<br />

Worm Behavioral States<br />

Thomas Gallagher, Leon Avery, Young-Jai You<br />

Virginia Commonwealth <strong>University</strong>, Richmond, VA, USA<br />

Satiated animals stop eating, stop moving, and go to sleep[1]. We identified a behavior in<br />

worms that mimics this behavior, satiety quiescence[1]. We have developed an automated and<br />

quantitative system tracking worm locomotion to provide a detailed and integrated analysis <strong>of</strong><br />

worm nutritional state and signaling. Tracking worm locomotion shows long periods <strong>of</strong> inactivity<br />

under conditions that enhance satiety quiescence. This inactivity is not seen in conditions<br />

known to disrupt satiety quiescence; if worms are not fasted, if they are refed on poor quality<br />

food, or if they have a mutation in satiety signaling pathways. This validates our automated<br />

system for measuring satiety quiescence.<br />

Our tracking data show that conditions that disrupt satiety quiescence increase locomotion.<br />

Moreover, restoring pkg-1 expression in a dozen head neurons under the tax-4 promoter<br />

decreases locomotion back to the level <strong>of</strong> wildtype. Because the impaired satiety quiescence<br />

in daf-7 worms is rescued by expression <strong>of</strong> daf-7 in ASI neurons (contained in the tax-4 set)<br />

[1], we attempted to rescue pkg-1 worms by expressing pkg-1(gf) in ASI neurons. This results<br />

in a decrease in locomotion similar to the tax-4 rescue. Genetic ablation <strong>of</strong> the ASI neurons<br />

causes an increase in locomotion, further showing that satiety signals are conveyed in ASI<br />

neurons. Additionally, we used calcium imaging to find that ASI is activated by food and nutrients,<br />

suggesting a role for ASI neurons in conveying nutrient availability.<br />

To identify behavioral states from locomotion we are developing a Hidden Markov Model<br />

(HMM) analysis. This allows us to view the behavioral state <strong>of</strong> a worm over time and address<br />

more complex questions such as the effect <strong>of</strong> genes, food quality, and nutritional state on percent<br />

time spent in each behavioral state, frequency <strong>of</strong> switching between states, and whether there<br />

is a cyclic pattern <strong>of</strong> behavior.<br />

1. You, Y.J., et al., Insulin, cGMP, and TGF-beta signals regulate food intake and quiescence in C. elegans:<br />

a model for satiety. Cell Metab, 2008. 7(3): p. 249-57.<br />

Contact: gallaghertl@mymail.vcu.edu<br />

Lab: You<br />

80<br />

Poster Topic: Metabolism


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

Circadian rhythms in metabolism, stress tolerance and pathogenesis:<br />

lessons from Caenorhabditis elegans.<br />

Maria Goya 1 , Andres Romanowski 1 , Maria Migliori 1 , Sergio Simonetta 2 , Diego<br />

Golombek 1<br />

1 Universidad Nacional de Quilmes, Bernal, Buenos Aires, Argentina, 2 Fundacion<br />

Instituto Leloir, Ciudad Autonoma de Buenos Aires, Argentina<br />

Circadian rhythms in physiological patterns are ubiquitously found in nature. They are driven<br />

by endogenous biological clocks and are synchronized to environmental cues. C. elegans<br />

is a model organism widely used in diverse areas <strong>of</strong> research but not well characterized in<br />

chronobiological studies. C. elegans might provide fundamental information about the basis<br />

<strong>of</strong> circadian rhythmicity in eukaryotes, due to its ease <strong>of</strong> use and manipulations, as well as <strong>of</strong><br />

availability <strong>of</strong> genetic data and mutant strains. We have designed an automated system to track<br />

individual nematodes and demonstrated the existence <strong>of</strong> circadian activity rhythms in both LD<br />

(light:dark, 12h:12h) and DD (constant darkness) conditions; circadian periods were found to<br />

be <strong>of</strong> 24.2 ±0.44 h and 23.9 ±0.40 h respectively. These rhythms were affected by a mutation<br />

in lin-42, a homolog <strong>of</strong> the clock gene per. In addition, circadian periods were temperaturecompensated<br />

and could also be entrained by temperature cycles.<br />

C. elegans is a soil-dwelling nematode subjected to daily changes in environmental<br />

stressors; the ability to predict such variation might confer an adaptative advantage. We studied<br />

stress tolerance to abiotic and biotic stressors and found a rhythm in tolerance patterns for<br />

oxidative and osmotic stress, peaking at daytime and nightime, respectively. Expression <strong>of</strong><br />

stress-related genes was determined by qRealTime-PCR: gpdh-1 and gpx showed a significant<br />

diurnal variation. When exposed to Pseudomonas fluorescens or Pseudomonas aeruginosa<br />

(two soil-occuring bacteria that kill C. elegans), we found lower tolerance during nightime.<br />

We have also studied circadian rhythms in metabolic variables, such as food consumption<br />

and defecation. Food consumption rate (determined by decreasing OD600 <strong>of</strong> E. coli OP50)<br />

was shown to be rhythmic and a peak was found in the evening. Defecation rhythms also<br />

showed to be governed in a circadian manner. These results show that control animals have<br />

a 24 h modulation <strong>of</strong> the ultradian defecation rhythm. Finally, we have detected aaNAT activity<br />

and melatonin in this nematode. aaNAT exhibited a diurnal variation peaking at ZT12, and<br />

melatonin could serve as a possible circadian output signal.<br />

In summary, our results show that the circadian system regulates changes in behavior,<br />

metabolism, abiotic stress tolerance and host-pathogen interactions. However, the presence<br />

and function <strong>of</strong> an endogenous clock capable <strong>of</strong> orchestrating such temporal changes deserves<br />

further investigation.<br />

Contact: eugeniagoya@gmail.com<br />

Lab: Golombek<br />

Poster Topic: Metabolism<br />

81


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

Ceramide signal mediates antipsychotic drug-induced developmental<br />

delay and lethality in C. elegans.<br />

Limin Hao, Bruce Cohen, Edgar Buttner<br />

Mailman Research Center, McLean Hospital<br />

Our aim is to help define the molecular pathways and mechanisms <strong>of</strong> action <strong>of</strong> antipsychotic<br />

drugs (APDs), using C. elegans as a genetic model. Exposure to APDs early in development<br />

causes dose-dependent developmental delay and lethality in C. elegans. A genome-wide RNAi<br />

screen for suppressors <strong>of</strong> clozapine-induced developmental delay and lethality was performed<br />

and revealed 42 candidate genes, including sms-1, which encodes a sphingomyelin synthase.<br />

Specifically, this enzyme converts ceramide and phosphatidylcholine to sphingomyelin and<br />

diacylglycerol. As these lipids are important in intracellular signaling, the findings may be<br />

relevant to the molecular pathways by which APDs produce their long-term effects. By analyzing<br />

other enzymes involved in sphingolipid metabolism, we identified ceramide as a key molecule<br />

that mediates APD drug effects. One sms-1 is<strong>of</strong>orm is expressed in the C. elegans pharynx, and<br />

its transgene rescues the sms-1 mutant phenotype. We thus examined pharyngeal pumping<br />

and found that APD-induced inhibition <strong>of</strong> pharyngeal pumping requires sms-1, a finding that<br />

may explain the role <strong>of</strong> the gene in mediating APD-induced developmental delay. Total lipids<br />

extracted from wild-type worms are able to rescue APD-induced developmental delay and<br />

lethality. While limited to C. elegans, so far, similar mechanisms may be observed and can be<br />

studied in mammalian cells, to test their relevance to the clinical outcome <strong>of</strong> APD treatment.<br />

Contact: lhao@mclea.harvard.edu<br />

Lab: Buttner<br />

82<br />

Poster Topic: Metabolism


Contact: nharriso@scripps.edu<br />

Lab: Gill<br />

Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

Functional Characterization <strong>of</strong> C. elegans N-acylethanolamine<br />

Biosynthetic Enzymes<br />

Neale Harrison, Ifedayo Victor Ogungbe, Matthew Gill<br />

The Scripps Research Institute, Scripps Florida, 130 Scripps Way, Jupiter, Florida,<br />

33458<br />

N-acyl ethanolamines (NAEs) are an important class <strong>of</strong> lipid signaling molecules that<br />

includes the mammalian endocannabinoid arachidonoyl ethanolamide (also known as<br />

anandamide). We recently identified several NAEs in C. elegans and found that they are<br />

important for larval development and in adult animals they influence lifespan via nutrient sensing<br />

pathways. NAE metabolism is a tightly regulated process due to the ‘on demand’ nature <strong>of</strong> this<br />

signaling pathway. In mammals, this is achieved by balancing the activity <strong>of</strong> the biosynthetic<br />

enzyme N-acyl phosphatidylethanolamine phospholipase D (NAPE-PLD) with the activity <strong>of</strong><br />

the hydrolytic enzyme fatty acid amide hydrolase (FAAH). Our previous work focused on the<br />

effect <strong>of</strong> a specific NAE (eicosapentaenoyl ethanolamide, EPEA) and the role <strong>of</strong> FAAH in<br />

development and aging. We have now sought to characterize the role that the biosynthetic<br />

enzyme NAPE-PLD plays in NAE metabolism and life history traits in the worm.<br />

Based on sequence alignments, there are two NAPE-PLD homologs in C. elegans,<br />

nape-1 and nape-2, both <strong>of</strong> which retain the conserved-metallolactamase catalytic residues<br />

found in mammalian NAPE-PLD. We find that recombinant NAPE-1 and NAPE-2 are capable<br />

<strong>of</strong> liberating NAEs from N-acyl phosphatidylethanolamine substrates in vitro, providing<br />

biochemical evidence for a conservation <strong>of</strong> function. Although these enzymes contain strong<br />

sequence similarity, they are expressed in distinct, as well as overlapping, tissues suggesting<br />

the possibility <strong>of</strong> both divergent and redundant functions. Divergent functions are suggested<br />

by the ability <strong>of</strong> nape-1 overexpression, but not nape-2, to alter lifespan, while nape-2 overexpression,<br />

but not nape-1, leads to L1 arrest in a daf-2 mutant background. Individually,<br />

nape-1 and nape-2 over-expression have subtle effects on life history traits, but surprisingly<br />

when both are over-expressed simultaneously we observe L1 arrest. In order to understand<br />

the phenotypes conferred by the different genetic manipulations <strong>of</strong> these two genes, we are<br />

using gas chromatography - mass spectrometry to assess how nape-1 and nape-2 affect the<br />

levels <strong>of</strong> each <strong>of</strong> the different NAEs in vivo.<br />

Poster Topic: Metabolism<br />

83


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

The Mediator Subunit MED15/MDT-15 is a Conserved Transcriptional<br />

Co-regulator in Lipid Homeostasis<br />

Nicole Hou, Donha Park, Stefan Taubert<br />

<strong>University</strong> <strong>of</strong> British Columbia, Vancouver, Canada<br />

Regulation <strong>of</strong> metabolism is crucial for survival and development. Transcriptional regulation<br />

is one <strong>of</strong> the most important levels for metabolic control. The Mediator is a multi-subunit<br />

protein complex that regulates transcription by linking transcription factors to the general<br />

transcriptional machinery. MDT-15, the Caenorhabditis elegans homolog <strong>of</strong> the human<br />

Mediator subunit MED15, regulates genes involved in fatty acid metabolism and several<br />

stress responses. Specifically, MDT-15 is required for the production <strong>of</strong> polyunsaturated fatty<br />

acids, which are essential nutrients for C. elegans. Thus, worms with depleted or mutated<br />

mdt-15 display phenotypes such as developmental arrest, sterility, abnormal lipogenesis and<br />

shortened lifespan. However, defects in PUFA metabolism don’t fully explain the pleiotropic<br />

effects <strong>of</strong> mdt-15 depletion, prompting us to investigate additional metabolic pathways in<br />

these mutants. We found that MDT-15 is required to express genes involved in the uptake<br />

and metabolism <strong>of</strong> folate, a key precursor for the universal methyl-group-donor SAM. SAM<br />

is essential for various cellular reactions, including the synthesis <strong>of</strong> phospholipids (PLs),<br />

which play a key role in the construction <strong>of</strong> membranes. Therefore I hypothesize that MDT-<br />

15 is required for normal fatty acid metabolism, phospholipid biosynthesis and organelle<br />

structures. To determine whether MDT-15 affects folate, SAM, and PL levels, I am pr<strong>of</strong>iling<br />

both the expression <strong>of</strong> relevant genes and the lipid contents in the mdt-15-depleted worms. I<br />

found that several genes in the pathways responsible for phosphatidylcholine synthesis are<br />

significantly downregulated. Because lipid metabolism is closely linked to energy homeostasis<br />

as well as organelle structures and functions, I am also investigating the roles <strong>of</strong> MDT-15 in<br />

glucose metabolism and various subcellular organelles. I found that glucose level is reduced<br />

in worms with depleted or mutated mdt-15and ER stress is highly elevated in these worms. In<br />

addition, I will use an unbiased suppressor screen to identify and dissect new pathways that<br />

affect the metabolic pathways controlled by MDT-15. Lastly, I will test whether mammalian<br />

MED15 similarly regulates metabolism. Our studies will yield novel insights into a conserved<br />

regulator <strong>of</strong> metabolism that may be linked to human metabolic disorders.<br />

Contact: hjn31340@hotmail.com<br />

Lab: Taubert<br />

84<br />

Poster Topic: Metabolism


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

L1 longevity is determined by metabolic rate and that AMPK as a<br />

master regulator <strong>of</strong> metabolism controls<br />

Inhwan Lee 1 , Amber Hendrix 1 , Jennifer Yoshimoto 2 , Jeongho Kim 3 , Young-Jai You 1<br />

1 Biochemistry and Molecular Biology, Virginia Commonwealth <strong>University</strong>,<br />

VA, USA, 2 Department <strong>of</strong> Internal Medicine, <strong>University</strong> <strong>of</strong> Michigan, MI, USA,<br />

3 Department <strong>of</strong> Biological Science, Inha <strong>University</strong>, Incheon, Korea<br />

Animals have to cope with starvation. The molecular mechanisms by which animals survive<br />

long-term starvation, however, are not clearly understood. When they hatch without food, C.<br />

elegans arrests development at the first larval stage (L1) and survives more than two weeks.<br />

Here we show that the survival span <strong>of</strong> arrested L1s, which we call L1 longevity, is a starvation<br />

response regulated by metabolic rate during starvation. A high rate <strong>of</strong> metabolism shortens<br />

the L1 survival span, whereas a low rate <strong>of</strong> metabolism lengthens it. The longer worms are<br />

starved, the slower they grow once they are fed, suggesting that L1 arrest has metabolic<br />

costs. Furthermore, mutants <strong>of</strong> genes that regulate metabolism show altered L1 longevity.<br />

Among them, we found that AMP-dependent protein kinase (AMPK), as a key energy sensor,<br />

regulates L1 longevity by regulating this metabolic arrest. Our results suggest that L1 longevity<br />

is determined by metabolic rate and that AMPK as a master regulator <strong>of</strong> metabolism controls<br />

this arrest so that the animals survive long-term starvation.<br />

Contact: ilee4@vcu.edu<br />

Lab: You<br />

Poster Topic: Metabolism<br />

85


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

Proteomic Study and Marker Protein Identification <strong>of</strong> Caenorhabditis<br />

elegans Lipid Droplets<br />

Pingsheng Liu, Peng Zhang, Huimin Na<br />

Institute <strong>of</strong> Biophysics, Chinese Academy <strong>of</strong> Sciences<br />

Lipid droplets (LDs) are a neutral lipid storage organelle that is conserved across almost<br />

all species. Many metabolic syndromes are directly linked to the over-storage <strong>of</strong> neutral lipids<br />

in LDs. The study <strong>of</strong> LDs in Caenorhabditis elegans (C. elegans) has been difficult since the<br />

lack <strong>of</strong> specific LD marker proteins. Here we report the purification and proteomic analysis <strong>of</strong> C.<br />

elegans LDs for the first time. We identified 306 proteins, 63% <strong>of</strong> these proteins were previously<br />

known to be LD-proteins, suggesting a similarity between mammalian and C. elegans LDs.<br />

Using morphological and biochemical analyses, we show that a short-chain dehydrogenase<br />

is almost exclusively localized on C. elegans LDs in both in vivo and in vitro experiments,<br />

indicating that it can be used as a LD marker protein in C. elegans. We also identified other<br />

two major proteins <strong>of</strong> LDs. These results will facilitate further mechanistic studies <strong>of</strong> LDs in<br />

this powerful genetic system, C. elegans.<br />

Contact: pliu@ibp.ac.cn<br />

Lab: Liu<br />

86<br />

Poster Topic: Metabolism


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

WormSizer: High-Throughput Image Analysis <strong>of</strong> Nematode Size and<br />

Shape<br />

Brad Moore, James Jordan, Ryan Baugh<br />

Duke <strong>University</strong>, Durham, NC, USA<br />

The fundamental phenotypes <strong>of</strong> growth rate, size and morphology are the result <strong>of</strong> complex<br />

interactions between genotype and environment. We developed a high-throughput s<strong>of</strong>tware<br />

application, WormSizer, that computes size and shape <strong>of</strong> nematodes from brightfield microscopy<br />

images. Existing methods for estimating volume either coarsely model the nematode as a<br />

cylinder, or assume the worm shape or opacity is invariant. Our estimate is more robust to<br />

changes in morphology as it only assumes that the worms are radially symmetric. We use<br />

a point distribution model (PDM) to characterize the shape and variation <strong>of</strong> different classes<br />

<strong>of</strong> nematodes. This open source s<strong>of</strong>tware is written as a plugin for the well-known image<br />

processing framework Fiji/ImageJ, and it includes additional support for CellPr<strong>of</strong>iler. It may<br />

therefore be extended easily. We used this framework to analyze growth and shape <strong>of</strong> several<br />

canonical C. elegans mutants in a developmental time series. Our analysis confirmed and<br />

extended on existing phenotypic characterization, demonstrating the utility and robustness <strong>of</strong><br />

WormSizer. We show that a trained maximum likelihood classifier on the PDM can be used<br />

to discriminate strains, automatically identifying mutants that are phenotypically distinct from<br />

wild-type. We confirm quantitatively that a Dpy mutant is short and fat, that a Lon mutant is<br />

long and thin, and that a daf-2 insulin-like receptor mutant grows slow. We can also distinguish<br />

dauer larvae from normal larvae. WormSizer works with Unc and Rol mutants as well, showing<br />

wild-type morphology but delayed growth. We show for the first time that Sma mutants lay<br />

small eggs and that the larvae are actually smaller from the time <strong>of</strong> hatching onward.<br />

Contact: brad.t.moore@duke.edu<br />

Lab: Baugh<br />

Poster Topic: Metabolism<br />

87


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

Acetylcholine Dependent Starvation Signaling<br />

Robert Pollok, Leon Avery<br />

Virginia Commonwealth <strong>University</strong>, Richmond, VA, USA<br />

Starvation induced lethality in gpb-2 loss <strong>of</strong> function worms can be rescued by inhibiting<br />

acetylcholine signaling, suggesting cholinergic neurons propagate a starvation signal in C.<br />

elegans. GPB-2 is a regulator <strong>of</strong> G protein signaling, and the loss <strong>of</strong> function mutant has<br />

increased MAPK activation. You et al (Cell Metabolism 3: 237) showed that MAPK activation<br />

is increased in the presence <strong>of</strong> arecoline, an agonist to acetylcholine receptors, and decreased<br />

with atropine, an antagonist to acetylcholine receptors. When gpb-2(-) worms are treated with<br />

atropine, they are less sensitive to starvation induced lethality. Laser ablation <strong>of</strong> the cholinergic<br />

MC neurons also rescues starvation induced lethality in gpb-2(-) worms. Utilizing recombinant<br />

caspases to genetically ablate MC, I hope to further describe acetylcholine dependent starvation<br />

signaling in C. elegans.<br />

Contact: rhpollok@gmail.com<br />

Lab: Avery<br />

88<br />

Poster Topic: Metabolism


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

How to sense fasting : searching for the mechanism <strong>of</strong> energy<br />

homeostasis by IRE-1<br />

Jisun Shin 1 , Hyungmin Moon 2 , Jiwon Shim 2 , Junho Lee 1,2<br />

1 World Class <strong>University</strong> Program, Department <strong>of</strong> Biophysics and Chemical<br />

Biology, Seoul National <strong>University</strong>, Seoul, Korea, 2 Department <strong>of</strong> Biological<br />

Sciences, Institute <strong>of</strong> Molecular Biology and Genetics, Seoul National <strong>University</strong>,<br />

Seoul, Korea<br />

Most organisms store fat as an energy source and use it during fasting. In our previous<br />

study we demonstrated that fasting induced lipases fil-1 and fil-2 were up-regulated in the<br />

IRE-1/HSP-4- dependent and xbp-1-independent manner upon fasting. In this study, we are<br />

trying to establish how IRE-1 sensors the fasting condition <strong>of</strong> the animal. To address the<br />

question, we chose lipid-derived signaling molecules, nucleic acids, and energy sources<br />

from food as candidate signaling molecules <strong>of</strong> fasting. After treating these molecules without<br />

other nutrients, we are analyzing the changes in the fil-1 and fil-2 levels. We are also trying<br />

to elucidate the mechanism <strong>of</strong> IRE-1 activation upon fasting. The fact that the induction <strong>of</strong><br />

fil-1 and fil-2 was independent <strong>of</strong> xbp-1 indicates that the protein domains <strong>of</strong> IRE-1 may act<br />

differently upon fasting from those in the condition <strong>of</strong> conventional unfolded protein response<br />

(UPR). Therefore, we try to determine the roles <strong>of</strong> the domains within IRE-1 during fasting.<br />

Through these experiments, we hope to contribute to further understanding <strong>of</strong> the mechanism<br />

<strong>of</strong> energy homeostasis through IRE-1.<br />

Contact: jasminbinu@naver.com<br />

Lab: Lee<br />

Poster Topic: Metabolism<br />

89


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

Pigment dispersing factor - a neuropeptidergic influence on<br />

metabolism and stress<br />

Liesbet Temmerman, Ellen Meelkop, Tom Janssen, Liliane Scho<strong>of</strong>s<br />

KU Leuven, Leuven, Belgium<br />

On top <strong>of</strong> classical neurotransmitters, C. elegans uses neuropeptides as messengers<br />

or modulators in the nervous system, which mainly act upon G protein-coupled receptors<br />

(GPCRs). Most <strong>of</strong> the 1300 predicted GPCRs in C. elegans are uncharacterized orphans, i.e.<br />

their ligands and function(s) are unknown.<br />

Our group identified a GPCR signaling system resembling both the invertebrate PDF<br />

system as well as the widely studied vertebrate VIP signaling system. In insects, PDF function<br />

is pr<strong>of</strong>oundly studied with regard to molecular dissection <strong>of</strong> the circadian clock. The drawback<br />

<strong>of</strong> this focus is a remaining ignorance on possible other functions for PDF, which are better<br />

described for the vertebrate homolog VIP. Using the molecular toolkit available for C. elegans,<br />

we have broadened the scientific view on invertebrate PDF signaling systems and observed<br />

additional resemblances to the vertebrate VIP system.<br />

A combination <strong>of</strong> differential transcriptomic and proteomic data strongly indicates a<br />

modulatory role for the PDF signaling system in energy metabolism, especially under starvation.<br />

This aspect <strong>of</strong> PDF signaling finds its counterpart in the vertebrate VIP system, which also is<br />

involved in fatty acid metabolism and seems to be needed under starvation. Functions <strong>of</strong> the<br />

PDF signaling system in resistance to stress are also apparent from these molecular data, in<br />

addition, pdf mutant worms seem to cope better with osmotic stress than wild type worms do.<br />

Our data support the hypothesis that the PDF system in C. elegans modulates a broad<br />

range <strong>of</strong> processes, rather than being confined to the regulation <strong>of</strong> circadian rhythms.<br />

Contact: liesbet.temmerman@bio.kuleuven.be<br />

Lab: Temmerman<br />

90<br />

Poster Topic: Metabolism


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

Ethanol Influence on Gonadal Development in C. elegans daf-18/PTEN<br />

Mutants<br />

Tim Wolf 1 , Wenjing Qi 1 , Ralf Baumeister 1,2<br />

1 Bioinformatics and Molecular Genetics (Faculty <strong>of</strong> Biology), Albert-Ludwigs-<br />

<strong>University</strong> Freiburg, 79104 Freiburg, Germany, 2 Freiburg Institute for Advanced<br />

Studies,School <strong>of</strong> Life Sciences (FRIAS LIFENET),BIOSS Centre for Biological<br />

Signaling Studies, Freiburg, Germany<br />

During its development, C. elegans encounters different larval stages in which, depending<br />

on nutritional status, stress or pheromones, decisions have to be made to either proceed<br />

and become adult, or to arrest and go to dormancy. The genes encoding the components<br />

<strong>of</strong> the insulin/IIs pathway are controlling the most crucial developmental decisions during<br />

the L2 larval stage. Active insulin/IGF signaling results in developmental progression into<br />

becoming a reproducing adult, whereas reduced signaling provokes the entry into an<br />

alternative developmental stage, the dauer larva. Mutations in daf-18, the homolog <strong>of</strong> the tumor<br />

suppressor gene PTEN (phosphatase and tensin homolog mutated on chromosome 10) result<br />

in defective dauer entry and also defective L1 arrest after starvation. The abnormal L1 arrest<br />

is accompanied by a disordered cell cycle arrest in germline progenitor cells Z2 and Z3 at G2<br />

phase. Thus, focusing on the germline we discovered that daf-18(lf) animals recovering from<br />

a three days nutritional deprivation period display a phenotype composed <strong>of</strong> a disintegrated<br />

gonadal basement membrane and inordinate distribution <strong>of</strong> germline cells throughout the<br />

somatic tissue. During our studies evidence could be provided, that ethanol supplementation<br />

whilst starvation decreased the penetrance <strong>of</strong> this phenotype, not only in daf-18 single mutants,<br />

but also in distinct mutants showing a less severe phenotype. Moreover, as Ethanol was<br />

supplemented at very low concentration and N2 larvae under this treatment remained in L1<br />

diapause, it could be assumed that ethanol does not simply act as food compensation. We<br />

are investigating whether ethanol acts as a carbon source or if there is an ethanol sensitizing<br />

pathway regarding our phenotype.<br />

Contact: tim.wolf@biologie.uni-freiburg.de<br />

Lab: Baumeister<br />

Poster Topic: Metabolism<br />

91


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

Increased Levels <strong>of</strong> Hydrogen Peroxide Induce a HIF-1-dependent<br />

Remodeling <strong>of</strong> Lipid Metabolism in C. elegans<br />

Meng Xie, Richard Roy<br />

McGill <strong>University</strong>, Montreal, (Quebec), Canada<br />

Cells have evolved numerous mechanisms to circumvent stresses caused by the<br />

environment, many <strong>of</strong> which are regulated by the AMP-activated kinase (AMPK). Unlike most<br />

organisms, C. elegans AMPK null mutants are viable, but die prematurely in the “long-lived”<br />

dauer stage due to rapid exhaustion <strong>of</strong> triglyceride energy stores. Using a genome-wide RNAi<br />

approach we demonstrate that the disruption <strong>of</strong> genes that increase hydrogen peroxide levels<br />

enhance the survival <strong>of</strong> AMPK mutants by altering both the abundance, and the nature, <strong>of</strong> the<br />

fatty acid content in the animal by increasing the HIF-1-dependent expression <strong>of</strong> several key<br />

rate-liming enzymes involved in de novo fatty acid biosynthesis. Our data provide a mechanistic<br />

foundation to explain how an optimal level <strong>of</strong> an <strong>of</strong>ten vilified ROS-generating compound such<br />

as hydrogen peroxide can provide cellular benefit; a phenomenon described as hormesis, by<br />

instructing cells to readjust their lipid biosynthetic capacity through downstream HIF-1 activation,<br />

as a means to correct cellular energy deficiencies.<br />

Contact: meng.xie@mail.mcgill.ca<br />

Lab: Roy<br />

92<br />

Poster Topic: Metabolism


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

The Transmembrane Channel-like Protein TMC-1 Affects Adaptation to<br />

a Chemically Defined Medium in C. elegans<br />

Liusuo Zhang, L Rene Garcia<br />

Howard Hughes Medical Institute, Department <strong>of</strong> Biology, Texas A&M <strong>University</strong>,<br />

College Station, TX USA<br />

Animals must change their cellular metabolism and behavioral responses in order to invade<br />

and thrive in alternative nutritional environments. A chemically defined axenic medium, C.<br />

elegans maintenance medium (CeMM), has been established for culturing <strong>of</strong> C. elegans, which<br />

is a manipulable nutritional platform to dissect cell physiology. Worms grown on CeMM plates<br />

develop slower and exhibit a prolonged reproductive period with a decreased brood size. For<br />

N2 animals, about 10 percent <strong>of</strong> them develop into adults at day 8 after hatching, whereas the<br />

others reach adulthood in the following 10 days. Compared to the lab-bred wide type N2 strain,<br />

CB4856, a wild Hawaiian strain, takes greater than 16 days to reach adulthood. By screening<br />

known mutations which may affect feeding and growth, we observed that hermaphrodites<br />

containing mutations in fat-3, lev-11, ser-7, tph-1 and unc-73 developed faster than wild type<br />

in CeMM (30-80 percent reached adulthood between 5 and 7 days), while daf-2, daf-19, che-<br />

2, mdt-15, osm-9, unc-26, unc-32 and unc-42 mutants grew much slower or developmentally<br />

arrested in L1 (the majority could not develop into adults by two weeks). Through screening<br />

<strong>of</strong> EMS generated N2 mutant hermaphrodites that grow even faster in CeMM, we identified<br />

the rg1003 allele, which cause 90 percent <strong>of</strong> the worms develop into adults on the synthetic<br />

medium by day 5. Using two-point mapping, Illumina next generation sequencing, Sanger resequencing<br />

and complementation test, we mapped the rg1003 mutation to the gene tmc-1,<br />

which encodes a putative transmembrane channel-like protein, expressed on muscles and<br />

motor neurons. Other life history traits such as brood size, reproductive aging and life span,<br />

which were not directly selected for were not affected by tmc-1(rg1003). Animals must be able<br />

to sexually reproduce in order to thrive in new environments, therefore we examined the male<br />

mating potency <strong>of</strong> tmc-1(rg1003). We found that mutant males had higher mating potency than<br />

wild type on CeMM, whereas they were similar to wild type when grown on OP50 bacteria.<br />

qPCR analysis revealed that some genes associated with NADH production(fat-6, gpd-2 and<br />

gpd-3) were increased in tmc-1(rg1003) compared to N2 when synchronized starved worms<br />

were transferred to CeMM liquids for 2 hours, which suggests TMC-1 may facilitate energy<br />

homeostasis and metabolism regulation.<br />

Contact: lzhang@mail.bio.tamu.edu<br />

Lab: Garcia<br />

Poster Topic: Metabolism<br />

93


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

Screening for novel regulators <strong>of</strong> rnt-1 in stress response<br />

Soungyub Ahn 1 , Junho Lee 1,2<br />

1 Research Center for Functional Cellulomics, Institute <strong>of</strong> Molecular Biology and<br />

Genetics, School <strong>of</strong> Biological Sciences, Seoul National <strong>University</strong>, Korea, 2 World<br />

Class <strong>University</strong>, Department <strong>of</strong> Biophysics and Chemical Biology, Seoul National<br />

<strong>University</strong>, Seoul, Korea<br />

RUNX family transcription factors have various essential roles in mammalian development<br />

and carcinogenesis. Alternative splicing, post-transcriptional regulation and interaction with other<br />

nuclear components <strong>of</strong>fer various levels <strong>of</strong> regulation and essential functions <strong>of</strong> RUNX proteins.<br />

In Caenorhabditis elegans, rnt-1 is the sole homolog <strong>of</strong> the RUNX family, and expressed in the<br />

intestine and hypodermal seam cells. In addition, RNT-1 represses its own transcription with<br />

BRO-1 but DBL-1 acts to activate rnt-1 expression at the post-embryonic stages. We focused<br />

on a novel rnt-1 regulation mechanism in C. elegans. We previously identified that the stability<br />

<strong>of</strong> RNT-1 in the intestine is regulated by various stresses. The p38 MAPK pathway, which is<br />

important to stress response, mediates the increase <strong>of</strong> RNT-1 stability in the intestine. Now,<br />

we are trying to find novel regulators which involved in this rnt-1 regulation by using antibiotics<br />

selection and focusing on change <strong>of</strong> GFP localization.<br />

Contact: syahn87@snu.ac.kr<br />

Lab: Lee<br />

94<br />

Poster Topic: Stress


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

Screening Nuclear Receptors to Discover the Regulation <strong>of</strong> the<br />

Xenobiotic Stress Response<br />

Leah Blackwell, Amanda Marra, Andrew Davidson, Tim Lindblom<br />

Lyon College<br />

Worms enjoy a robust detoxification system that allows them to cope with a diverse array<br />

<strong>of</strong> chemical stressors in their environment. This system is complete with members <strong>of</strong> all the<br />

major detoxification enzyme classes in animals. We are studying the mechanisms that allow<br />

intestinal cells to upregulate the expression <strong>of</strong> specific detoxification enzymes in response<br />

to specific xenobiotics. Much <strong>of</strong> our work has centered around the nuclear receptor, NHR-8,<br />

which is required for wild type levels <strong>of</strong> toxin resistance. To probe the role <strong>of</strong> NHR-8 in the<br />

xenobiotic stress response, we have analyzed the expression <strong>of</strong> genes in animals that lack<br />

NHR-8 and are challenged with xenobiotics. Our data indicates that NHR-8 does not mediate<br />

the up-regulation <strong>of</strong> detoxification genes but is likely required for basal expression <strong>of</strong> many<br />

intestinally expressed genes. What proteins might mediate the xenobiotic stress response and<br />

do they cooperate with NHR-8 for their activity? To answer this question, we have initiated a<br />

feeding RNAi screen <strong>of</strong> the transcription factor loci expressed in the gut to find those that are<br />

required for toxin resistance. This screen also probes the RNAi targets in combination with<br />

the loss <strong>of</strong> NHR-8 to look for cooperative control. Of particular interest are the 120+ nuclear<br />

receptors with demonstrated expression in intestinal cells. These proteins are ideally suited to<br />

mediate chemically induced gene expression through their ligand and DNA-binding domains.<br />

This type <strong>of</strong> nuclear receptor action would help explain why C. elegans contains comparatively<br />

many more nuclear receptors than other animals.<br />

Contact: leah.blackwell@lyon.edu<br />

Lab: Lindblom<br />

Poster Topic: Stress<br />

95


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

Depletion <strong>of</strong> the Nascent Polypeptide-associated Complex in C.<br />

elegans Up-regulates ER Chaperone Expression and Engages the<br />

Unfolded Protein Response, Resulting in the Induction <strong>of</strong> Autophagy<br />

and Apoptosis<br />

Paul Arsenovic, Anthony Maldonado, Vaughn Colleluori, Tim Bloss<br />

James Madison <strong>University</strong>, Harrisonburg, VA, USA<br />

The nascent polypeptide-associated complex (NAC) is a highly conserved heterodimeric<br />

complex important for metazoan development, but its molecular function has not been fully<br />

defined. Recent evidence in S. cerevisiae supports the hypothesis that the NAC is a component<br />

<strong>of</strong> the cytosolic chaperone network that interacts with both ribosomal complexes and their<br />

emerging nascent peptides, such that the loss <strong>of</strong> the NAC in chaperone-depleted cells results<br />

in an increase in misfolded protein stress. We tested whether the NAC functions similarly in C.<br />

elegans and found that its homologous NAC subunits, i.e. ICD-1 and -2, have chaperone-like<br />

characteristics; loss <strong>of</strong> the NAC appears to induce misfolded protein stress in the ER, triggering<br />

the unfolded protein response (UPR). Depletion <strong>of</strong> either subunit altered the worm’s response<br />

to heat stress, and led to a dramatic up-regulation <strong>of</strong> HSP-4, the homologue <strong>of</strong> the human<br />

chaperone and ER stress sensor BiP. Worms lacking both ICD-1 and the UPR transcription<br />

factor XBP-1 generated a higher proportion <strong>of</strong> defective embryos and had a diminished survival<br />

rate relative to ICD-1-depleted animals with an intact UPR. Up-regulation <strong>of</strong> HSP-4 was celltype<br />

specific; in embryos lacking ICD-1 or -2, the gut cell region showed strong up-regulation<br />

<strong>of</strong> HSP-4 and resistance to cell death relative to the neuronal region, which showed little to no<br />

increase in HSP-4 expression and significant cell death. Furthermore, loss <strong>of</strong> ICD-1 generated<br />

the presence <strong>of</strong> lip<strong>of</strong>uscins in both embryonic and mature gut cells, indicating an induction <strong>of</strong><br />

ER-mediated autophagy. These results cast C. elegans NAC as an essential component <strong>of</strong><br />

protein folding and localization during translation, and confirm C. elegans as a valuable model<br />

for studying organismal and cell-type specific responses to misfolded protein stress in the ER.<br />

Contact: blossta@jmu.edu<br />

Lab: Bloss<br />

96<br />

Poster Topic: Stress


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

Genetic analysis <strong>of</strong> the Unfolded Protein Response during pathogen<br />

infection in C. elegans<br />

Douglas Cattie, Kirthi Reddy, Claire Richardson, Dennis Kim<br />

Department <strong>of</strong> Biology, Massachusetts Institute <strong>of</strong> Technology, Cambridge, MA.<br />

The Unfolded Protein Response (UPR) is a homeostatic mechanism that functions to oppose<br />

the accumulation <strong>of</strong> unfolded protein in the endoplasmic reticulum (ER) by carefully regulating<br />

several aspects <strong>of</strong> cellular physiology. In metazoans, there are three branches <strong>of</strong> the UPR—<br />

IRE-1/XBP-1, PERK/PEK-1, and ATF-6—each <strong>of</strong> which senses the concentration <strong>of</strong> unfolded<br />

protein in the ER lumen with a transmembrane protein and then transduces this signal to other<br />

cellular compartments. Prior work in our lab has shown that one <strong>of</strong> these branches, IRE-1/<br />

XBP-1, is critical for the development and survival <strong>of</strong> C. elegans on the pathogen Pseudomonas<br />

aeruginosa 1 . We therefore conducted a screen to identify suppressor mutations that enable<br />

xbp-1 mutant animals to develop to adulthood on P. aeruginosa. Classes <strong>of</strong> suppressor mutants<br />

that have been isolated exhibit differential responses to pathogen-induced and abiotic sources<br />

<strong>of</strong> ER stress, as well as distinct genetic interactions with pathways mediating the UPR. The<br />

identification <strong>of</strong> these mutants is ongoing and the progress will be presented.<br />

1. Richardson et al. Nature 463, 1092 (2010).<br />

Contact: dcattie@mit.edu<br />

Lab: Kim<br />

Poster Topic: Stress<br />

97


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

BCAS2 is Essential for Drosophila Viability and Functions in PremRNA<br />

Splicing<br />

Po-Han Chen 1 , Yeou-Ping Tsao 2 , Show-Li Chen 1<br />

1 National Taiwan <strong>University</strong>, Taipei, Taiwan, 2 MaKay Memorial Hospital<br />

BCAS2 (Breast carcinoma amplified sequence 2, also known as SPF27) was previously<br />

reported as a negative regulator <strong>of</strong> p53. Here, we demonstrate that BCAS2 is essential for<br />

the viability <strong>of</strong> Drosophila melanogaster in which CG4980 (dBCAS2) is an ortholog <strong>of</strong> human<br />

BCAS2 (hBCAS2). We find that whole-body silencing <strong>of</strong> dBCAS2 leads to larval lethality, and<br />

that knockdown <strong>of</strong> dBCAS2 using a tissue specific promoter results in deformed phenotypes.<br />

The deprivation <strong>of</strong> dBCAS2 in Drosophila results in the accumulation <strong>of</strong> pre-mRNA and the<br />

induction <strong>of</strong> the dp53-targeted reaper and hid genes. Most importantly, overexpression <strong>of</strong><br />

hBCAS2 can rescue lethality and wing deformities, indicating that hBCAS2 plays a similar<br />

role to dBCAS2. Furthermore, as a component <strong>of</strong> hPrp19 complex, we characterize the role <strong>of</strong><br />

hBCAS2 in mammalian cells in both constitutive and alternative splicing. The C-terminal domain<br />

<strong>of</strong> hBCAS2 contains two coiled-coil motifs that directly binds to CDC5L and recruits hPrp19/<br />

PLRG1 to form a core complex for pre-mRNA splicing. We also find that ectopic expression<br />

<strong>of</strong> the C-terminal region <strong>of</strong> human BCAS2 partially restores wing damage induced by silenced<br />

dBCAS2 in flies. In summary, Drosophila and human BCAS2 share at least two functions: one<br />

for p53 regulation and the other for RNA splicing, which affect cell growth viability.<br />

Contact: showlic@ntu.edu.tw<br />

Lab: Chen<br />

98<br />

Poster Topic: Stress


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

Oxidative stress related PMK-1-HIF-1 signaling pathways in silver<br />

nanoparticles toxicity in C.elegans<br />

Jinhee Choi, Hyun - Jeong Eom, Jeong-Min Ahn<br />

<strong>University</strong> <strong>of</strong> Seoul, Seoul, Korea<br />

Silver nanoparticles (AgNPs) have been increasingly used in both consumer and biomedical<br />

applications, due to their antimicrobial and anti-inflammatory. Despite <strong>of</strong> recent numerous<br />

research on the toxicity and safety <strong>of</strong> AgNPs, serious deficiencies in the knowledge relating<br />

to AgNPs toxicity still exist, especially on its mechanism. Reactive oxygen species (ROS)<br />

generation and oxidative stress are the best developed paradigms to explain toxic effects <strong>of</strong><br />

AgNPs. In the present study, a toxicity mechanism <strong>of</strong> AgNPs was investigated in Caenorhabditis<br />

elegans focusing on oxidative stress response. Initially, AgNPs were tested as potential<br />

oxidative stress inducers, and increased formation <strong>of</strong> ROS was observed in AgNP-exposed<br />

C. elegans. Subsequently, the potential upstream signaling pathway activated in response<br />

to AgNP exposure was investigated, paying special attention to the C. elegans PMK-1 P38<br />

mitogen-activated protein kinase (MAPK). The results indicated that AgNPs exposure led to<br />

increased ROS formation, increased expression <strong>of</strong> PMK-1 P38 MAPK and hypoxia-inducible<br />

factor (HIF-1), glutathione S-transferase (GST) enzyme activity, and decreased reproductive<br />

potential in wildtype C. elegans; whereas in the AgNP-exposed pmk-1 mutant, ROS formation<br />

and HIF-1 and GST activation were not observed, and decreased reproductive potential was<br />

rescued, suggesting the importance <strong>of</strong> PMK-1 P38 MAPK in oxidative stress response by<br />

AgNPs. In the second part, we investigated physiological consequences <strong>of</strong> HIF-1 activation<br />

by examining the response <strong>of</strong> regulators <strong>of</strong> HIF-1, such as, EGL-9 and VHL-1, and that <strong>of</strong><br />

genes known to be regulated by HIF-1, such as, FMO-2. To test whether AgNPs-induced HIF-1<br />

activation is due to hypoxic response via ROS, pharmacological rescue assay was conducted<br />

using a strong antioxidant, N-Acetyl-Cysteine (NAC), on the HIF-1 and related pathways and<br />

the results suggest oxidative stress is involved in activation <strong>of</strong> HIF-1 pathway. Overall results<br />

suggest that oxidative stress is an important mechanism <strong>of</strong> AgNP-induced toxicity in C. elegans<br />

and that PMK-1 p38 MAPK and HIF-1 play important roles in it. The results also suggest that<br />

GST, FMO-2 and HIF-1 activation by AgNP exposure are PMK-1 p38 MAPK–dependent, and<br />

that they play important roles in the PMK-1 p38 MAPK-mediated stress response to AgNP<br />

exposure in C. eleganss.<br />

Contact: jinhchoi@uos.ac.kr<br />

Lab: Choi<br />

Poster Topic: Stress<br />

99


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

Insulin-like signaling in the parasitic nematode Brugia malayi<br />

Kirsten Crossgrove, Brenda Garland, Peter Sackett<br />

UW-Whitewater, Whitewater (WI), USA<br />

Brugia malayi is a mosquito borne parasitic nematode that is a causative agent <strong>of</strong> lymphatic<br />

filariasis in humans. Transmission from the mosquito to human host triggers molting from the<br />

L3 (infective stage) to the L4 stage. We are interested in the signaling pathways that trigger<br />

this developmental transition in the new host. The free-living nematode Caenorhabditis elegans<br />

also responds to environmental cues during development. Crowding, low food and/or high<br />

temperature all contribute to sending C. elegans into an alternate third larval ‘dauer’ stage. If<br />

conditions improve, the worms recover from dauer and molt to fourth stage larvae. The dauer<br />

larva is thought to be analogous to the infective stage larva in B. malayi. In both species, an<br />

environmental cue is required to trigger the molt to the L4 stage. An insulin/insulin-like signaling<br />

(IIS) pathway is one <strong>of</strong> the known regulators <strong>of</strong> dauer formation and recovery in C. elegans.<br />

Specifically, an active DAF-2 insulin receptor signals for reproductive development (no dauer<br />

formation) and recovery from dauer. One target <strong>of</strong> the IIS pathway is the DAF-16 protein, a<br />

FOXO transcription factor. Insulin signaling results in modification <strong>of</strong> DAF-16 to keep it in the<br />

cytoplasm. In the absence <strong>of</strong> insulin signaling, DAF-16 can enter the nucleus and regulate<br />

target genes. We have identified putative orthologs in B. malayi <strong>of</strong> a number <strong>of</strong> IIS pathway<br />

genes. We used qRT-PCR to determine if these genes are expressed in a developmentally<br />

regulated manner in B. malayi. The eight genes we have analyzed to date showed different<br />

expression levels at different life cycle stages, suggesting that they may play a role in a IIS<br />

pathway in B. malayi. We are currently conducting functional characterization <strong>of</strong> the Bm-daf-16<br />

gene. We have expressed the DNA binding domain in E. coli and will present data on binding<br />

to DAF-16 response elements. Genes in the IIS pathway could be useful targets for drugs or<br />

vaccines that prevent molting <strong>of</strong> B. malayi infective stage larvae.<br />

Contact: crossgrk@uww.edu<br />

Lab: Crossgrove<br />

100<br />

Poster Topic: Stress


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

Using Mathematical Models to Predict Gene Flow and Discover Gene<br />

Function<br />

Andrew Davidson, Marc-Andre LeBlanc, Megan Powell, Tim Lindblom<br />

Lyon College<br />

Much <strong>of</strong> the work in our laboratory for several years has been to discover the functions<br />

<strong>of</strong> genes whose phenotypes might not be readily apparent in the plush environment <strong>of</strong> the<br />

laboratory. For example, we have probed the role <strong>of</strong> NHR-8 in the regulation <strong>of</strong> the detoxification<br />

response. Knockout alleles <strong>of</strong> nhr-8 have no phenotype under typical laboratory growth<br />

conditions. Similarly, many strains produced by the Gene Knockout Consortium or animals in<br />

RNAi screens do not display obvious phenotypes. We propose that worms in the environment<br />

are subject to significantly more stressors and that many <strong>of</strong> these genes encode proteins that<br />

protect worms during stress. The xenobiotic stress response is an excellent example. Until<br />

we challenge worms lacking full length NHR-8 with toxins, they appear no different from wild<br />

type worms and even then, the phenotype is weak. In order to design a more sensitive assay<br />

for gene necessity, we are developing mathematical models for predicting allele frequency in<br />

a population starting with a single heterozygote. After many generations, one would expect<br />

the typical 1:2:1 ratio <strong>of</strong> mutant and wild type alleles if there has been no selection against<br />

the mutant allele. By adding stressors to this population, we hope to statistically demonstrate<br />

selection by a reduction in the mutant allele frequency in the final gene pool. Using a panel <strong>of</strong><br />

stressors, we might begin to tag genes without obvious phenotypes to biological functions for<br />

more detailed analysis. Our pilot experiments are with nhr-8 alleles and xenobiotic challenges.<br />

At the meeting, we will report on our progress towards implementing the mathematical models<br />

and expansion <strong>of</strong> the assay.<br />

Contact: andrew.davidson@lyon.edu<br />

Lab: Lindblom<br />

Poster Topic: Stress<br />

101


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

EGL-9 Function is Required to Produce Viable Offspring from Animals<br />

Exposed to Chronic Hypoxia<br />

Jennifer Dennis, Pamela Padilla, Brent Little<br />

<strong>University</strong> <strong>of</strong> North Texas, Denton, TX, USA<br />

Conserved molecular mechanisms, such as the HIF-1/EGL-9 signaling pathway, have<br />

evolved in metazoans to respond to and survive oxygen deprivation making C. elegans a good<br />

model system to understand the mechanisms regulating responses to hypoxia. The majority <strong>of</strong><br />

studies thus far have involved acute exposures to hypoxia at specific developmental stages.<br />

In this study we investigate how C. elegans respond to chronic hypoxia exposure. We found<br />

that wild-type animals exposed to chronic hypoxia (.5% O 2 or 1% O 2) throughout development<br />

(embryo to gravid adulthood) survive. However, animals displayed various phenotypes including<br />

a decrease in developmental trajectory, a reduction in brood size, an increase in mean lifespan<br />

and an altered egg-laying behavior indicating that C. elegans can survive prolonged stress<br />

but at a cost to brood size. For wild-type animals, the embryos (F1 generation) from animals<br />

exposed to chronic hypoxia throughout development are viable. However, the F1 generation<br />

<strong>of</strong> egl-9 mutants exposed to chronic hypoxia were not viable indicating that egl-9 function is<br />

required for chronic hypoxia exposure. EGL-9 is known to regulate HIF-1 levels in that HIF-1<br />

accumulates in egl-9 mutants exposed to normoxia making it somewhat counterintuitive that<br />

the egl-9 mutants are sensitive to chronic hypoxia. Possible interpretations <strong>of</strong> these results<br />

will be presented. Further, we are conducting genetic and cellular analysis to elucidate the<br />

role EGL-9 has in chronic hypoxia exposure.<br />

Contact: jennifercdennis@gmail.com<br />

Lab: Padilla<br />

102<br />

Poster Topic: Stress


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

Analysis a Null Allele <strong>of</strong> the Heterochronic Gene lin-42, a period<br />

Homolog<br />

Theresa Edelman 1 , Katherine McCulloch 1 , Angela Barr 2 , Christian Frokjaer-Jensen 3 ,<br />

Erik Jorgensen 3 , Ann Rougvie 1<br />

1 <strong>University</strong> <strong>of</strong> Minnesota, Minneapolis, MN, USA, 2 <strong>University</strong> <strong>of</strong> Washington,<br />

Seattle, WA, USA, 3 <strong>University</strong> <strong>of</strong> Utah, Salt Lake City, Utah, USA<br />

Many events must be precisely timed and coordinated during the development <strong>of</strong> multicellular<br />

organisms. C.elegans is a premier system with which to identify genes that temporally regulate<br />

development. Disruption <strong>of</strong> these heterochronic genes typically result in either precocious<br />

or retarded defects, as a consequence <strong>of</strong> the skipping or reiteration <strong>of</strong> certain stage specific<br />

programs, respectively. One interesting player in the heterochronic gene pathway is lin-42,<br />

the C. elegans homolog <strong>of</strong> the fly and mammalian circadian clock gene period. lin-42 loss<strong>of</strong>-function<br />

alleles cause precocious defects, however the null phenotype is unknown. The<br />

lin-42 genomic locus is complex in that it produces four different is<strong>of</strong>orms, two <strong>of</strong> which do not<br />

overlap. All reported lin-42 alleles leave at least one is<strong>of</strong>orm intact, complicating genetic analysis<br />

and making it difficult to place lin-42 in the heterochronic gene pathway. We used MosDEL<br />

technology to delete the entire coding region, generating a lin-42(0) allele. lin-42(0) animals<br />

present developmental timing defects that are more severe than previously characterized<br />

alleles or lin-42(RNAi). The molting defect is also severe causing early larval lethality. Further<br />

genetic and molecular studies with the lin-42(0) allele will lead to a better understanding <strong>of</strong><br />

the role lin-42 plays in development.<br />

Contact: brixx001@umn.edu<br />

Lab: Rougvie<br />

Poster Topic: Stress<br />

103


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

SWI/SNF is Required to Maintain Memory <strong>of</strong> Adaptation to Hydrogen<br />

Sulfide<br />

Emily Fawcett1,2 , Dana Miller1 1Department <strong>of</strong> Biochemistry, <strong>University</strong> <strong>of</strong> Washington, Seattle, WA, USA,<br />

2Molecular and Cellular Biology Graduate Program, <strong>University</strong> <strong>of</strong> Washington,<br />

Seattle, WA, USA<br />

Rapid detection and response to external stress is critical for short-term survival in all<br />

organisms. In some situations, responses to environmental conditions result in long-lasting<br />

epigenetic changes, or “memories”, that allow for a more rapid or efficient response to<br />

subsequent stresses. We discovered that in the nematode C. elegans, transient exposure<br />

to the gas hydrogen sulfide (H 2S) results in a memory that protects against H 2S toxicity later<br />

in life. In a candidate screen, we identified the SWI/SNF chromatin-remodeling complex as<br />

required for persistence <strong>of</strong> H 2S memory. Our data demonstrate that the SWI/SNF complex<br />

allows for a robust transcriptional response to high H 2S in adapted animals. Therefore, we<br />

propose that formation <strong>of</strong> a memory to H 2S establishes an altered epigenetic landscape that<br />

allows for protection against subsequent stress. We propose that the SWI/SNF complex<br />

functions in response to H 2S memory to stabilize the altered landscape, maintaining phenotypic<br />

diversity. Our work establishes a great model for studying the fundamental underpinnings <strong>of</strong><br />

an environmental-induced phenotypic plasticity.<br />

Contact: efawcett@uw.edu<br />

Lab: Miller<br />

104<br />

Poster Topic: Stress


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

Diet & Environment Affect Oxygen-Deprivation Survival in C. elegans<br />

Anastacia Garcia, Pamela Padilla<br />

<strong>University</strong> <strong>of</strong> North Texas, Denton, TX, USA<br />

Oxygen deprivation plays a principal role in the pathology <strong>of</strong> a number <strong>of</strong> human diseases<br />

including stroke, cardiac dysfunction and pulmonary disease. Progression <strong>of</strong> these health<br />

issues is influenced by diet, environment and/or genetics. Consequently, there is interest in<br />

understanding the relationship(s) between diet, environment, genotype and the survival <strong>of</strong><br />

oxygen deprivation. Our previous studies have shown that diet and environment affect C.<br />

elegans anoxia survival rate. We hypothesize that media supplemented with carbohydrates<br />

(glucose or fructose) will affect response to and survival rate <strong>of</strong> anoxia exposure and that<br />

this response can be modulated genetically. We determined that wild-type animals grown<br />

with the addition <strong>of</strong> glucose had a significant decrease in anoxia survival rate. Additional<br />

methodologies are being used to determine if the glucose-induced anoxia sensitivity observed<br />

in wild-type animals is due to direct and/or indirect effects. Given that different E. coli strains<br />

display differences in carbohydrate content and ability to metabolize sugars, different strains<br />

<strong>of</strong> E. coli (OP50, ΔPTS OP50, & HT115) are being used to assay whether or not food source<br />

in conjunction with added sugar will differentially influence anoxia survival rate in C. elegans.<br />

Moreover, the glucose-induced anoxia sensitivity phenotype is suppressed by mutations in<br />

genes involved with highly conserved pathways including insulin-like signaling and O-GlcNAc<br />

post-translational modification (PTM) <strong>of</strong> proteins. Both daf-16 (mu86) and oga-1 (ok1207)<br />

mutant strains are able to partially suppress the glucose-induced anoxia sensitivity phenotype,<br />

suggesting that carbohydrate metabolism and PTMs have a role in oxygen-deprivation survival.<br />

This work will provide a greater understanding <strong>of</strong> the association between diet, environment,<br />

genetics and oxygen deprivation responses; and has relevant implications to human health<br />

related issues including cardiac dysfunction and diabetes.<br />

Contact: anastaciagarcia@my.unt.edu<br />

Lab: Padilla<br />

Poster Topic: Stress<br />

105


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

Transcriptional Regulation in the Oxidative Stress Response<br />

Grace Goh, Ada Kwong, Stefan Taubert<br />

<strong>University</strong> <strong>of</strong> British Columbia, Vancouver, Canada<br />

Proper transcription is required for organisms to respond to a multitude <strong>of</strong> external and<br />

internal signals. One player in transcriptional regulation is the Mediator, a large multi-protein<br />

complex that acts as a transcriptional coregulator. The Mediator is composed <strong>of</strong> ~25-30 subunits,<br />

some <strong>of</strong> which are required for basal transcription whereas others regulate transcription<br />

in a gene-specific manner. In the nematode Caenorhabditis elegans, the Mediator subunit<br />

MDT-15 is required for detoxification in response to certain xenobiotics and heavy metals.<br />

By comparing MDT-15-dependent genes to genes required for the response to oxidative<br />

stress, we found that several MDT-15 regulated genes, e.g. UGTs and GSTs, are induced by<br />

the stable organoperoxide tert-butylhydroperoxide(t-BOOH). Here, we present evidence that<br />

MDT-15 is required for the oxidative stress response upon t-BOOH exposure. We postulate<br />

that this response requires interactions between MDT-15 and at least two transcription factors.<br />

Interestingly, a number <strong>of</strong> our candidate transcription factors are also known to alter or increase<br />

fat content, leading us to hypothesize that the t-BOOH response, and perhaps the response<br />

to oxidative stress in general, is linked to lipid metabolism.<br />

Contact: graceg@cmmt.ubc.ca<br />

Lab: Taubert<br />

106<br />

Poster Topic: Stress


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

Disruption <strong>of</strong> the ire-1/xbp-1 UPR Pathway Induces ER stress and<br />

Attenuates the Production and Processing <strong>of</strong> Secreted Proteins Such<br />

as Insulin<br />

Modi Safra 1 , Cynthia Kenyon 2 , Sivan Henis-Korenblit 1<br />

1 Bar-Ilan <strong>University</strong>, Ramat-Gan, Israel, 2 UCSF, San Francisco, CA, USA<br />

The insulin/IGF-1 signaling pathway is a central pathway that controls many fundamental<br />

biological processes. While much attention has been focused on the pathways and effectors<br />

downstream <strong>of</strong> the insulin/IGF receptor, little is known about the molecular mechanisms that<br />

regulate the level <strong>of</strong> secreted insulin in the body cavity <strong>of</strong> the animal. As the production <strong>of</strong><br />

insulin generates a biosynthetic load on the endoplasmic reticulum (ER), it is plausible that<br />

ER stress response pathways regulate insulin production.<br />

To test this hypothesis, we investigated how inactivation <strong>of</strong> conserved ER stress response<br />

genes affects the expression <strong>of</strong> insulin in the model organism C. elegans. Using a strain<br />

expressing a GFP labeled insulin, we discovered that inactivation <strong>of</strong> the ER stress response<br />

genes ire-1 or xbp-1 altered insulin expression such that overall insulin levels significantly<br />

increased while the levels <strong>of</strong> circulating insulin were reduced compared to wild-type animals.<br />

How might inactivation <strong>of</strong> the ER stress-response gene ire-1 regulate insulin expression?<br />

One plausible model is that the ire-1/xbp-1 arm <strong>of</strong> the UPR is essential for maintaining protein<br />

folding homeostasis in the secretory pathway even under basal physiological conditions (in the<br />

absence <strong>of</strong> external ER stress). Thus, in their absence, production and processing <strong>of</strong> protein<br />

passing through the secretory pathway is severely disrupted. Consistent with this model we<br />

discovered that protein folding in the ER <strong>of</strong> ire-1/xbp-1 mutants is defective. Furthermore,<br />

we found that misfolded proteins cannot be cleared from the ER, leading to the activation <strong>of</strong><br />

alternative ER stress response pathways. Finally, we found that even when correctly folded<br />

proteins are produced in ire-1(-) cells, they remain constrained in the ER and do not reach the<br />

secretory granules, not to mention the body cavity <strong>of</strong> the animal.<br />

In summary, our findings uncover an essential role for the ire-1/xbp-1 arm <strong>of</strong> the UPR in<br />

maintaining protein folding homeostasis in the secretory pathway under basal physiological<br />

conditions in the adult C. elegans. At the organism level, this implies that in the absence <strong>of</strong> ire-<br />

1 multiple important biological processes based on long-range signaling within the organism<br />

may be severely disrupted.<br />

Contact: sivankorenblit@gmail.com<br />

Lab: Henis-Korenblit<br />

Poster Topic: Stress<br />

107


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

The Role <strong>of</strong> C. elegans BRAP-2 in Regulation <strong>of</strong> the Oxygen Radical<br />

Detoxification Response<br />

Queenie Hu 1 , Lesley MacNeil 2 , Marian Walhout 2 , Terry Kubiseski 1<br />

1 Department <strong>of</strong> Biology, York <strong>University</strong>, Toronto, ON, Canada, 2 Program in<br />

Gene Function and Expression and Program in Systems Biology, <strong>University</strong> <strong>of</strong><br />

Massachusetts Medical School, Worcester, MA<br />

The C. elegans BRCA-1-associated protein 2, BRAP-2, has been identified to have a role in<br />

preventing an inappropriate response to reactive oxygen species. A deletion mutant <strong>of</strong> brap-2<br />

is sensitive to oxidizing conditions, resulting in developmental larval arrest or lethality. We are<br />

interested in obtaining a better understanding BRAP-2 and its signaling pathway involved in the<br />

oxidative stress response. We have found that brap-2(ok1492) mutant displays an enhanced<br />

gst-4 expression in the intestine and hypodermis and that this enhancement is dependent on<br />

the transcription factor SKN-1. Although the exact mechanism <strong>of</strong> BRAP-2/SKN-1 regulation<br />

requires further investigation, our results indicate that BRAP-2 physically interacts with both<br />

LET-60/Ras and KSR-1/KSR-2, suggesting that the MAPK is the pathway involved in the<br />

regulation <strong>of</strong> SKN-1 by BRAP-2.<br />

In order to identify genes that function in the BRAP-2/SKN-1 detoxification pathway, we<br />

have used a genetic approach to help identify other novel regulatory components. An RNAi<br />

suppression screen, using a transcription factor library, was used to identify factors required<br />

for the enhanced gst-4::gfp expression in the brap-2(ok1492) strain. We have identified twenty<br />

suppressors that potentially represent either a novel transcription factor or co-activator <strong>of</strong> SKN-1<br />

to promote its biological effect. We are currently validating the candidate genes identified from<br />

this screen. Preliminary results indicate that the ELT-3 GATA transcription factor is required<br />

for enhanced gst-4 expression in brap-2(ok1492). Taken together, these results suggest a<br />

model where BRAP-2 acts as negative regulator <strong>of</strong> KSR and MAPK activity on SKN-1/ELT-3<br />

dependent gst-4 expression.<br />

Contact: qniehu@yorku.ca<br />

Lab: Kubiseski<br />

108<br />

Poster Topic: Stress


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

Epigenetic regulation <strong>of</strong> stress response in C. elegans<br />

Moonjung Hyun, Young-Jai You<br />

Department <strong>of</strong> Biochemistry and Molecular Biology, Richmond, Virginia<br />

Commonwealth <strong>University</strong><br />

Stress induces changes in physiology. In harsh environment, C. elegans undergoes major<br />

developmental changes to become a dauer, a dormant form to endure stress. Here we report<br />

that two known tumor suppressor genes, BLIMP1 and Metastasis Associated protein (MTA),<br />

interact to epigenetically regulate dauer formation downstream <strong>of</strong> TGFβ signal; knockdown <strong>of</strong><br />

blmp-1 expression in daf-7 mutants is lethal as the mutants enter dauer stage. The levels <strong>of</strong><br />

both mRNA and the protein <strong>of</strong> blmp-1 are highly upregulated in L2D stage in daf-7 mutants,<br />

supporting the specific role <strong>of</strong> blmp-1 in dauer formation. From an RNAi screen <strong>of</strong> histone<br />

modification and nuclear remodeling factors, we isolated lin-40, a homolog <strong>of</strong> mammalian<br />

MTA whose RNAi phenocopies the lethality we found in blmp-1;daf-7. From candidate gene<br />

approach, we found that BLMP-1 and MTA regulate common targets including the nuclear<br />

hormone receptor nhr-85, a homolog <strong>of</strong> human NR1 subfamily, which regulates molting in<br />

worms and insects and circadian rhythm in mammals. Taken together, we discovered a novel<br />

epigenetic mechanism to regulate stress response in worms that is conserved in tumorigenesis<br />

in mammals.<br />

Contact: mhyun@vcu.edu<br />

Lab: You<br />

Poster Topic: Stress<br />

109


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

The Role Of daf-2 Pathway In Primary, Secondary, And Delayed<br />

Hypoxic Injury<br />

Euysoo Kim, Chun-Ling Sun, Michael Crowder<br />

Washington <strong>University</strong> School <strong>of</strong> Medicine, St. Louis, (MO), USA<br />

Neurons and cardiac myocytes die secondary to hypoxic injury and the resultant strokes<br />

and myocardial infarctions are together the leading cause <strong>of</strong> US mortality. Yet, no cytoprotective<br />

therapy is approved for hypoxic/ischemic injury. Mechanisms underlying hypoxia resistance<br />

have been studied by discovery <strong>of</strong> hypoxia resistant mutants from various forward genetic<br />

screens. The first C. elegans mutant discovered to confer hypoxic resistance was a reduction<strong>of</strong><br />

function mutant <strong>of</strong> the insulin/IGF receptor homolog, daf-2. While some <strong>of</strong> the mechanisms<br />

whereby daf-2 regulates hypoxic sensitivity have been defined, basic aspects <strong>of</strong> the daf-2<br />

mechanism such as what cell types it protects and when it regulates hypoxic injury relative to<br />

the actual insult are unknown. Answering these questions has been hindered by limitations <strong>of</strong><br />

our whole animal death assay that is an all or none endpoint.<br />

To make a cell specific and delayed hypoxic injury model, we made use <strong>of</strong> rars-1 (gc47)<br />

mutation, which was previously shown to mediate hypoxic resistance. RARS-1 is an arginyltRNA<br />

synthetase expressed in all C. elegans cells. In the background <strong>of</strong> gc47, we expressed<br />

a rescuing wild-type copy <strong>of</strong> rars-1 in pharyngeal myocytes thereby making them selectively<br />

sensitive to hypoxia. During a 4 day recovery period after hypoxic incubation, we observed<br />

pathology and behavioral deficits in the pharynx and subsequently in the whole body, suggestive<br />

<strong>of</strong> primary, secondary, and delayed hypoxic injury. Systemic RNAi <strong>of</strong> daf-2 significantly blocked<br />

the Unc and delayed organismal death phenotypes. Reduced necrotic bodies were observed in<br />

the pharynx <strong>of</strong> daf-2 RNAi-fed animals. Interestingly, daf-2 RNAi applied only after the hypoxic<br />

incubation also reduced necrosis in the pharynx and mildly blocked organismal death. Cellspecific<br />

expression <strong>of</strong> genes downstream <strong>of</strong> daf-2 in this model to restrict the action <strong>of</strong> daf-2 to<br />

either the primary or secondary hypoxic injury target will reveal which cell types are important<br />

for daf-2 mediated hypoxic sensitivity.<br />

Contact: kime@morpheus.wustl.edu<br />

Lab: Crowder<br />

110<br />

Poster Topic: Stress


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

The Influence <strong>of</strong> Endoplasmic Reticulum Stress on the Dauer<br />

Developmental Decision in C. elegans<br />

Warakorn Kulalert, Dennis Kim<br />

MIT, Cambridge, MA, USA<br />

Upon encountering unfavorable conditions such as starvation, crowding, and high<br />

temperature, C.elegans larvae arrest in a dauer developmental stage. Dauer larvae are<br />

non-aging and resistant to a broad array <strong>of</strong> physiologic insults. Several conserved pathways,<br />

such as insulin/IGF and TGF-beta, regulate dauer entry and maintenance. We propose that<br />

accumulation <strong>of</strong> unfolded proteins in the endoplasmic reticulum operates as a physiological<br />

response to adverse stimuli, influencing the dauer developmental decision. Administration <strong>of</strong><br />

agents that disrupt protein folding in the ER such as tunicamycin promoted dauer formation in<br />

a number <strong>of</strong> dauer-constitutive genetic backgrounds. Furthermore, mutants deficient in xbp-1,<br />

which encodes a bZIP transcription factor required for the Unfolded Protein Response (UPR),<br />

formed rare dauers under non-starvation condition. xbp-1 loss <strong>of</strong> function also enhanced dauer<br />

formation In the dauer-constitutive daf-1 mutants. The genetic analysis suggests that global ER<br />

stress caused by defective UPR promotes dauer formation, or that xbp-1 suppresses dauer<br />

formation. We are currently dissecting the molecular mechanisms <strong>of</strong> how the UPR contributes<br />

to regulation <strong>of</strong> the dauer decision.<br />

Contact: kulalert@mit.edu<br />

Lab: Kim<br />

Poster Topic: Stress<br />

111


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

PKC-2 in Peroxide mediated Stress and Aging<br />

Marianne Land1,2 , Charles Rubin2 1 2 New York Institute <strong>of</strong> Technology, Old Westbury, NY, US, Albert Einstein Col.<br />

Med., Bronx, NY, US<br />

Protein kinase C2 (PKC-2), which is activated by diacylglycerol (DAG) and Ca 2+ , is<br />

widely expressed in the nervous system and non-neuronal tissues. However, substrates and<br />

functions PKC-2 are incompletely understood. Candidate PKC-2 effectors were identified by<br />

differential in gel electrophoresis, using animals in which PKC-2 was elevated 40-fold (pkc-<br />

2(40x)). PRDX-2, a 2-Cys peroxiredoxin exhibited increased negative charge, consistent<br />

with increased phosphorylation, in pkc-2(40x) nematodes. 2-Cys peroxiredoxins catalyze<br />

peroxide reduction, thereby diminishing toxic oxygen species generated by metabolism,<br />

signaling pathways and various physical and biological stresses. A PRDX-2 deletion mutant,<br />

prdx-2(gk169), suppressed the cryophilic phenotype <strong>of</strong> pkc-2(40x) animals and elicited an<br />

athermotactic phenotype. pkc-2 null animals share this phenotype. Animals lacking PKC-2 or<br />

PRDX-2 have similarly reduced life spans and increased sensitivities to peroxide-mediated<br />

killing. The longevities <strong>of</strong> PKC-2 and PRDX-2 deficient C. elegans and prdx-2(gk169);pkc-<br />

2(ok328) double mutants are similar, suggesting the two proteins act in a common pathway.<br />

Animals overexpressing PKC-2 also have a reduced lifespan, indicating that PKC-2 activity<br />

must be maintained in a limited dynamic range to ensure normal ageing.<br />

Purified PRDX-2 was phosphorylated to high stoichiometry by a prototypical Ca 2+ , DAG<br />

dependent PKC in vitro. Site directed mutagenesis identified the PKC phosphorylation site<br />

in PRDX-2b as DpTIK (amino acids 183-186). Phospho-mimetic and non-phosphorylatable<br />

PRDX-2 mutants will be expressed in vivo to reveal the physiological relevance <strong>of</strong> the<br />

PKC-2 target site.<br />

Contact: mland@nyit.edu<br />

Lab: Rubin<br />

112<br />

Poster Topic: Stress


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

High-throughput screening for small molecule modulators <strong>of</strong> SKN-1<br />

Chi Leung 1 , Siobhan Malany 2 , Andrew Deonarine 1 , Ying Wang 1 , Keith Choe 1<br />

1 <strong>University</strong> <strong>of</strong> Florida, Gainesville (FL), 2 Sanford-Burnham Medical Research<br />

Institute at Lake Nona, Orlando (FL)<br />

Small molecules are extremely important research and therapeutic tools. Advances<br />

in chemistry and robotics have resulted in enormous small molecule libraries (>1 million<br />

compounds) available for high-throughput screening <strong>of</strong> specific biological processes. Although<br />

in vitro and cell-based assays can be robust and specific, hit compounds are <strong>of</strong>ten found to be<br />

inappropriate for in vivo use (e.g., toxicity or poor accumulation). Therefore, there is a need for<br />

in vivo animal screening systems. With support from the NIH Molecular Libraries Program, we<br />

have developed an in vivo fluorescent-based 1536-well plate assay for measuring activity <strong>of</strong><br />

the stress-inducible transcription factor SKN-1. Multidrug resistance is a growing problem in<br />

parasitic nematodes that is poorly understood. SKN-1 regulates multiple detoxification genes,<br />

mediates resistance to toxins, and is present in diverse parasitic nematodes. Pharmacological<br />

compounds that target SKN-1 would provide new tools for studying multidrug resistance in<br />

non-model nematodes. We completed a pilot screen <strong>of</strong> the Library <strong>of</strong> Pharmacologically Active<br />

Compounds and are currently screening the NIH small molecule library <strong>of</strong> ~360,000 compounds.<br />

We are also developing counter screens to eliminate toxic and non-specific compounds. This<br />

work is funded by NIH grant R21NS067678-01.<br />

Contact: leungchikwan@gmail.com<br />

Lab: Choe<br />

Poster Topic: Stress<br />

113


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

Multisite phosphorylation fine-tunes SKN-1 activity by modulating<br />

interactions with WDR-23 and target DNA<br />

Chi Leung 1 , Koichi Hasegawa 2 , Keith Choe 1<br />

1 <strong>University</strong> <strong>of</strong> Florida, Gainesville (FL), 2 Chubu <strong>University</strong>, Japan<br />

Cap ‘n’ collar (CNC) transcription factors regulate development, stress resistance, and<br />

longevity in animals ranging from nematodes to mammals. Molecular regulation involves<br />

ubiquitin ligases and several protein kinases, but it is unclear how these diverse signals are<br />

integrated. WDR-23 is a WD40-repeat protein that interacts with the single C. elegans CNC<br />

(SKN-1) to presumably target the transcription factor for proteasomal degradation. SKN-1 is also<br />

directly regulated by GSK-3, p38 MAPK, and SGK-1/AKT-1/2 insulin-like signaling pathways.<br />

To understand how these signals are integrated, we are defining biochemical interactions<br />

between WDR-23, SKN-1, and SKN-1 target DNA with and without phosphorylation <strong>of</strong> multiple<br />

SKN-1 residues. A forward genetic screen for disinhibition <strong>of</strong> the SKN-1 target gene gst-4<br />

found a dominant allele <strong>of</strong> SKN-1 and several missense alleles <strong>of</strong> WDR-23. Yeast 2-hybrid<br />

analysis indicates that all <strong>of</strong> these mutations disrupt interactions between SKN-1 and WDR-<br />

23. Preliminary data also suggest that WDR-23 can inhibit interactions between SKN-1 and its<br />

target DNA. Site-directed mutagenesis studies indicate that phosphorylation <strong>of</strong> SKN-1 alters its<br />

interactions with both WDR-23 and target DNA and that p38 MAPK sites are dominant to GSK-3<br />

sites, as was previously shown in vivo [An et al., 2005 PNAS 102(45):16275]. Functional amino<br />

acids map within two adjacent blades <strong>of</strong> a predicted WDR-23 β-propeller and to two regions <strong>of</strong><br />

SKN-1 providing structural insight into signaling. Our studies suggest that a complex multisite<br />

phosphorylation “code” regulates cellular detoxification by modulating interactions between<br />

SKN-1, its repressor protein, and target DNA. This mechanism may permit fine-tuning <strong>of</strong> CNC<br />

activity to match specific demands set by stress, metabolism, development, and ageing. This<br />

work is funded by NSF grant IOS-1120130 to KPC.<br />

Contact: leungchikwan@gmail.com<br />

Lab: Choe<br />

114<br />

Poster Topic: Stress


Contact: jrice@bmcc.cuny.edu<br />

Lab: Savage-Dunn<br />

Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

Functions <strong>of</strong> CLIC proteins in C. elegans<br />

Jun Liang1 , Cathy Savage-Dunn2 1 2 Borough <strong>of</strong> Manhattan Community College/CUNY, New York, Queens College/<br />

CUNY, New York<br />

Chloride intracellular channel proteins (CLIC) are multifunctional proteins that are<br />

homologous to the glutathione S-transferase family. Most <strong>of</strong> research is focused on CLIC4 who<br />

is ubiquitously expressed in most mammal cells. CLIC4 regulates cellular stress, apoptosis,<br />

carcinogenesis, angiogenesis, and macrophage innate response. Cellular stress molecules<br />

such as DNA damaging agents, transcription inhibitors, translation inhibitors, and TNF-α induce<br />

endogenous CLIC4 to translocate from cytoplasm to nucleus. Mammalian Schnurri-2 is required<br />

for CLIC4 nuclear translocation in response to TGF-β but not required for CLIC4 nuclear<br />

function. Schnurri-2 is a transcription factor in the BMP pathway and its worm homologous is<br />

SMA-9.<br />

In C. elegans, DBL-1/TGF-β signaling components includes dbl-1, sma-2, sma-3, sma-4,<br />

and sma-9 etc. Their mutants are viable compare with that <strong>of</strong> other organisms, which allow<br />

research being performed at a whole living animal level. There are two CLIC homologs in C.<br />

elegans: EXL-1 and EXC-4. exc-4 mutants develop cysts in the excretory canal, while exl-1<br />

mutants do not have any observable phenotypes. However, other functions <strong>of</strong> EXC-4 and EXL-<br />

1 are unknown, in particular whether they interact with SMA-9 in C. elegans. To address this<br />

issue, we analyzed integrated EXL-1::GFP lines in wild type background. Strong fluorescence<br />

was observed in intestine cells, which is consistent with previous study. EXL-1::GFP indeed<br />

translocated into the nucleus at 37°C. However, whether it is regulated by SMA-9, as well as<br />

other TGF-β signaling components, is unknown. In the future, we will focus on these genetic<br />

interactions among them.<br />

Poster Topic: Stress<br />

115


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

Mouse Nmnat1 protects C. elegans from hypoxic death<br />

Xianrong Mao, C. Michael Crowder<br />

Washington <strong>University</strong> in St Louis<br />

Wallerian degeneration slow (Wlds) is a naturally occurring mouse mutation created by a<br />

chimeric fusion between Ube4b gene and nicotinamide mononucleotide adenylyltransferase 1<br />

gene (Nmnat1, which is a NADsynthethase). Expression <strong>of</strong> Wlds or a cytoplasmically-localized<br />

Nmnat1(cyt-Nmnat1) strongly protects mouse and fly axons from degeneration after harmful<br />

insults. Recently, cyt-Nmnat1 was found to protect from hypoxic neuronal injury in a mouse<br />

model <strong>of</strong> stroke. Despite considerable investigation in mouse and fly, the mechanism for cyt-<br />

Nmnat1 protection from axonal degeneration or hypoxic injury is completely obscure. We<br />

expressed mouse cyt-Nmnat1(m-cyt-Nmnat1) in C. elegans under the control <strong>of</strong> a ubiquitous<br />

or pan-neuronal promoter and asked if it protected worms from hypoxic injury. Both neuronal<br />

and ubiquitous m-cyt-Nmnat1 strongly increased whole worm survival after a hypoxic insult.<br />

Ubiquitous overexpression <strong>of</strong> a C. elegans Nmnat1 homolog F26H9.4 also increased hypoxic<br />

survival in the worm; the sterile phenotype <strong>of</strong> a null allele <strong>of</strong> F26H9.4 was rescued with<br />

ubiquitous m-cyt-Nmnat1, consistent with functional homology. Besides hypoxia resistance,<br />

m-cyt-Nmnat1 expression in neurons produced a strong chemotaxis defective phenotype and<br />

ubiquitous expression gave a partial sterility phenotype. We previously found that restoring<br />

protein homeostasis is critical to recovery from hypoxia in C. elegans. In testing various<br />

protein homeostasis pathways, we found that the mitochondrial ABC transporter gene haf-1<br />

was required at least partially for all <strong>of</strong> the phenotypes produced by m-cyt-Nmnat1 expression<br />

in worm. HAF-1 is thought to be necessary for the transport <strong>of</strong> mitochondrial degradation<br />

peptides to the cytoplasm as part <strong>of</strong> the mitochondrial unfolded protein response (mito-UPR).<br />

This result suggests that Nmnat1 may function to regulate the mito-UPR as part <strong>of</strong> its hypoxia<br />

protective mechanism.<br />

Contact: maox@wustl.edu<br />

Lab: Crowder<br />

116<br />

Poster Topic: Stress


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

Gene silencing based functional analysis <strong>of</strong> C. elegans cytochromes<br />

P450: roles in biotransformation, fat storage and eicosanoid formation<br />

Ralph Menzel 1 , Christian Steinberg 1 , Wolf-Hagen Schunck 2<br />

1 Humboldt-<strong>University</strong> at Berlin, Germany, 2 MDC Berlin, Germany<br />

The genome <strong>of</strong> the nematode Caenorhabditis elegans contains 75 full length cytochrome<br />

P450 (CYP) genes whose individual enzymatic and biological functions are largely unknown<br />

yet. Using microsomes isolated from adult worms, we found that C. elegans produces spectrally<br />

active CYP proteins that display typical CO-difference spectra. The microsomes also showed<br />

NADPH-cytochrome c reductase activity, silencing <strong>of</strong> emb-8 expression resulted in a loss <strong>of</strong> this<br />

activity. Expression <strong>of</strong> emb-8 was also essential for all CYP-dependent activities measured in<br />

this study. To identify the CYP components <strong>of</strong> specific monooxygenase systems, we performed<br />

systematic gene silencing by RNAi and used cyp-mutant strains.<br />

Searching for CYP is<strong>of</strong>orms involved in the biotransformation <strong>of</strong> xenobiotics, we found<br />

that CYP-14A3 and CYP-14A5 are required for the hydroxylation <strong>of</strong> an ortho-substituted,<br />

non-coplanar tetrachlorbiphenyl (PCB52) to C3-, C4- and/or C6-hydroxy-PCB52. In contrast,<br />

is<strong>of</strong>orms <strong>of</strong> the CYP-35A subfamiliy, which are also induced by PCB52 and other xenobiotics,<br />

could be assigned to the fat storage pathway. Individual gene silencing <strong>of</strong> cyp-35A2, 3, 4, and 5<br />

resulted in a dramatic decrease <strong>of</strong> intestinal fat content <strong>of</strong> well fed young adults. Both cyp-14A<br />

and cyp-35A is<strong>of</strong>orms are expressed in the intestine, as shown by GFP reporter constructs.<br />

Searching for CYP is<strong>of</strong>orms involved in eicosanoid formation, we identified CYP-<br />

29A3 and CYP-33E2 as the major is<strong>of</strong>orms contributing to the endogenous metabolism <strong>of</strong><br />

eicosapentaenoic acid (EPA) and arachidonic acid (AA) in C. elegans. Co-expression <strong>of</strong> CYP-<br />

33E2 with the human NADPH-CYP-reductase in insect cells resulted in the reconstitution <strong>of</strong> an<br />

active microsomal monooxygenase system that metabolized EPA and, with lower activity, also<br />

AA to specific sets <strong>of</strong> regioisomeric epoxy- and hydroxy-derivatives. Using worms carrying a<br />

cyp-33E2 promoter::GFP-reporter construct, we found that CYP-33E2 is exclusively expressed<br />

in the pharynx, where it is predominantly localized in the marginal cells. RNAi-mediated cyp-<br />

33E2 expression silencing as well as treatments with inhibitors <strong>of</strong> mammalian AA-metabolizing<br />

CYP enzymes, significantly reduced the pharyngeal pumping frequency <strong>of</strong> adult worms. These<br />

findings suggest that CYP-eicosanoids known to modulate the contractility <strong>of</strong> cardiomyocytes<br />

and vascular smooth muscle in mammals serve as regulators <strong>of</strong> pharyngeal activity and feeding<br />

behavior in C. elegans.<br />

Contact: ralph.menzel@biologie.hu-berlin.de<br />

Lab: Menzel<br />

Poster Topic: Stress<br />

117


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

Deciphering the microRNA responses to High Temperature Stress<br />

Camilla Nehammer 1 , Agnieszka Podolska 1 , Konstantinos Kagias 1 , Sebastian<br />

Mackowiak 3 , Nikolaus Rajewsky 3 , Roger Pocock 1<br />

1 BRIC, Copenhagen, 3 Max-Delbruck-Center, Berlin<br />

Living organisms are constantly exposed to changing environments and the ability to<br />

maintain homeostasis whilst exposed to stress is essential for survival. Therefore, rapid<br />

responses to stress, such as high temperature, are crucial in order to avoid detrimental stress<br />

induced effects. MicroRNAs are rapidly acting regulatory elements which can fine-tune gene<br />

expression when required. Thus, microRNAs are excellent candidates to study the regulation<br />

<strong>of</strong> stress response pathways in C. elegans.<br />

We performed RNA-seq analysis to identify microRNAs that are regulated in response to<br />

high temperature. We exposed L4 animals to 35°C temperature stress for 4 hours and detected<br />

48-down and 11 up-regulated (> 2-fold) microRNAs compared to control. Of these we identified<br />

six novel microRNAs. The detection <strong>of</strong> a number <strong>of</strong> microRNAs previously associated with<br />

stress responses in C. elegans, including miR-71, miR-239 and miR-246, is encouraging and<br />

strengthens the reliability <strong>of</strong> this high throughput sequencing approach.<br />

Temperature stress regulated candidates including predicted-novel microRNAs are currently<br />

being validated by RT-qPCR. Furthermore, our phenotypic analysis <strong>of</strong> microRNA mutant strains,<br />

<strong>of</strong> those microRNAs regulated by high temperature, revealed a number <strong>of</strong> promising stress<br />

resistant and stress sensitive microRNAs. These data provide a platform for further in-depth<br />

analysis <strong>of</strong> the roles <strong>of</strong> these microRNAs in stress response pathways in C. elegans.<br />

Contact: cne@bric.ku.dk<br />

Lab: Pocock<br />

118<br />

Poster Topic: Stress


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

Transmission <strong>of</strong> a Q/N-Rich Prion Domain Between C. elegans<br />

Tissues Causes an Extreme Form <strong>of</strong> Proteotoxicity<br />

Carmen Nussbaum-Krammer 1 , Kyung-Won Park 2 , Liming Li 2 , Ronald Melki 3 , Richard<br />

Morimoto 1<br />

1 Northwestern <strong>University</strong>, Evanston, (IL), USA, 2 Northwestern <strong>University</strong>, Chicago,<br />

(IL), USA, 3 CNRS, Gif-sur-Yvette, France<br />

The yeast prion protein Sup35 harbors a glutamine/asparagine (Q/N)-rich sequence motif<br />

that can adopt alternative self-propagating conformations. Similar Q/N-rich regions have been<br />

found in a number <strong>of</strong> genetic modifiers <strong>of</strong> neurodegenerative disorders, however their properties<br />

are poorly understood in metazoans. Here, we have examined the behavior <strong>of</strong> the Q/N-rich<br />

prion domain (NM) from Sup35 expressed in Caenorhabditis elegans. NM formed multiple<br />

classes <strong>of</strong> aggregates with distinct biophysical properties associated with a severe cellular<br />

and organismal toxicity. Both aggregation and toxicity phenotypes were strictly dependent on<br />

the length <strong>of</strong> NM oligorepeats and led to pr<strong>of</strong>ound alterations in mitochondrial morphology not<br />

only in NM expressing muscle cells but also comprised non-expressing tissues. These cell<br />

non-autonomous effects correlated with the spreading <strong>of</strong> misfolded proteins. Taken together,<br />

this C. elegans model demonstrates that Q/N-rich prion domains have the potential to transmit<br />

from cell-to-cell in metazoans suggesting a possible mechanism for the prion-like propagation<br />

<strong>of</strong> pathology in diverse neurodegenerative diseases.<br />

Contact: c-krammer@northwestern.edu<br />

Lab: Morimoto<br />

Poster Topic: Stress<br />

119


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

The affect <strong>of</strong> carbohydrate diet, metabolism, germline function and<br />

age on oxygen deprivation response and survival in C. elegans<br />

Pamela Padilla 1 , Anastacia Garcia 1 , Jo Goy 2 , Mary Ladage 1<br />

1 <strong>University</strong> <strong>of</strong> North Texas, 2 Harding <strong>University</strong>, Searcy AR<br />

Oxygen deprivation, which is central to many human-health related issues (Ex: stroke,<br />

cardiac and pulmonary disorders) occurs frequently in the elderly and can have a more<br />

detrimental affect on individuals who are diabetic or obese. We are using Caenorhabditis<br />

elegans to examine how environment, diet, age and genotype influences oxygen deprivation<br />

response and survival. One-day old, wild-type adult C. elegans fed the standard OP50 diet and<br />

grown at 20C will survive severe oxygen deprivation (anoxia) for at least 24 hours however<br />

this survival rate is severely decreased if anoxia exposure is longer than three days. Factors<br />

associated with long-term anoxia tolerance include age (3-5 day old adults hermaphrodites<br />

survive long-term anoxia), a decrease in ovulation rate, development at 25C instead <strong>of</strong> 20C, a<br />

diet <strong>of</strong> HT115 E. coli or pretreatment with the diabetic drug metformin. Long-term anoxia survival<br />

rate is associated with an increase in carbohydrate stores (glycogen, treholose) in the intestine<br />

however a glucose-supplemented diet leads to sensitivity to anoxia exposure, suggesting that<br />

a homeostatic balance <strong>of</strong> carbohydrate stores is important for oxygen deprivation survival.<br />

Analysis <strong>of</strong> mutants identified the following signaling pathways as influencing anoxia survival:<br />

insulin-like signaling (daf-2/daf-16), developmental pathways that alter germline function (glp-<br />

1), carbohydrate metabolism (aak-2), hexosamine signaling (oga-1) and methyglycoxal protein<br />

modifications. Insulin-signaling, carbohydrate homeostasis, oxidative stress and hexosamine<br />

signaling impact diabetes in humans suggesting that C. elegans can be used as a cellular and<br />

genetic model to understand the relationship between diet, metabolism, oxygen deprivation<br />

stress and disease states such as type II diabetes.<br />

Contact: Pamela.Padilla@unt.edu<br />

Lab: Padilla<br />

120<br />

Poster Topic: Stress


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

Understanding Hypoxia-dependent, HIF-independent Pathways in<br />

C.elegans<br />

Divya Padmanabha, Young-Jai You, Keith Baker<br />

Virginia Commonwealth <strong>University</strong> Health Systems, Richmond (VA), USA<br />

The interface <strong>of</strong> metabolism and hypoxia in disease progression has been increasingly<br />

explored in recent years. Several transcriptional activators are markedly induced by oxygen<br />

deprivation. Cellular and systemic responses to hypoxia have been extensively studied in the<br />

context <strong>of</strong> the transcription factor hypoxia inducible factor (HIF), a critical regulator <strong>of</strong> angiogenic<br />

and glycolytic genes in hypoxia. Interestingly, gene expression studies provide clear evidence<br />

<strong>of</strong> HIF-independent pathways that are critical for adaptation to hypoxia 1 . However, relatively<br />

little is known about the oxygen sensors and regulatory pathways that mediate transcriptional<br />

responses independent <strong>of</strong> HIF. The nematode Caenorhabditis elegans has proven to be a<br />

powerful model system to study evolutionarily conserved signaling pathways that regulate<br />

hypoxia responses. Hence, we are conducting an RNAi screen to identify transcription factors<br />

regulating HIF-independent genes in hypoxia. Accordingly, we have generated GFP reporter<br />

animals with a gene that is induced in hypoxia in a HIF-independent manner. We posit that the<br />

screen will comprehensively identify key components that play a role in C.elegans adaptation<br />

to hypoxia, independent <strong>of</strong> HIF.<br />

1. ShenC, Nettleton D, Jiang M, Kim SK, Powell-C<strong>of</strong>fman JA. Roles <strong>of</strong> the HIF-1hypoxiainducible<br />

factor during hypoxia response in Caenorhabditis elegans. JBiol Chem. 2005<br />

Contact: padmanabhad@mymail.vcu.edu<br />

Lab: Baker<br />

Poster Topic: Stress<br />

121


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

Osmotic Stress Resistance and Cuticle Defects – Two Symptoms, One<br />

Cause<br />

Anne-Katrin Rohlfing<br />

<strong>University</strong> <strong>of</strong> Potsdam, Potsdam, Germany<br />

The strain osm-8(n1518) is a well defined example <strong>of</strong> osmotic stress resistant (osr) mutants,<br />

which exhibit a constitutive active osmotic stress response. A mutation in the mucin-like gene<br />

osm-8 disrupts osmoregulatory physiological processes and leads to a constant expression <strong>of</strong><br />

the enzyme gpdh-1. GDPH-1 triggers the synthesis and accumulation <strong>of</strong> high amounts <strong>of</strong> the<br />

osmolyte glycerol within the hypodermis and intestine, as we were able to demonstrate before.<br />

The OSM-8 protein is expressed in the hypodermis and is most likely secreted into the<br />

extracellular matrix (ECM). In the ECM, OSM-8 is supposed to act as part <strong>of</strong> an osmosensory<br />

structure and to activate the osmotic stress response via the transmembrane protein PTR-23,<br />

similar to the osmosensory mucins Hkr1 and Msb2 in S. cerevisiea. However, there is evidence<br />

for a second function <strong>of</strong> osm-8 in cuticle formation. osm-8 expression peaks before each molt<br />

and before the final molt in developing C. elegans. This cyclic expression pattern correlates<br />

strongly with expression pattern seen in genes typically connected to molt, like dpy-7. Unlike<br />

in dpy strains the typical surface morphology with regular, cirumferential-oriented furrows and<br />

annuli was still visible by scanning electron microscopy, as well as are alae. However, the osm-<br />

8(n1518) cuticle appeared thinner and displayed an altered fine structure as determined by<br />

transmission electron microscopy. The normal C. elegans cuticle is composed <strong>of</strong> three layers:<br />

the cortical layer, middle layer with collagen strut and the basal layer. In osm-8(n1518) the<br />

basal layer was reduced to the thickness <strong>of</strong> wildtype cuticle and the struts appeared smaller<br />

and more electron dense. This phenotype was reversible to some extent by a heat shock<br />

within in the early L4 state in osm-8(n1518) strains carrying a heat-shock inducible osm-8(+)<br />

transgene. This treatment also fully reversed the strong osr phenotype <strong>of</strong> transgenic osm-<br />

8(n1518) animals back to a normal response to hyperosmotic environment.<br />

Whether there is a connection between the physiological and the morphological phenotypes<br />

<strong>of</strong> osm-8(n1518) or whether they are caused by two distinct functions <strong>of</strong> one protein has to<br />

be further investigated.<br />

Contact: rohlfing@uni-potsdam.de<br />

Lab: Rohlfing<br />

122<br />

Poster Topic: Stress


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

Knockdown <strong>of</strong> Genes Involved in Basic Cellular Functions Impairs<br />

Mitochondrial ROS Stress Signaling to the Nucleus<br />

Eva Runkel 1,2 , Shu Liu 2 , Ralf Baumeister 3,2 , Ekkehard Schulze 2<br />

1 Spemann Graduate School <strong>of</strong> Biology and Medicine (SGBM), 2 Bioinformatics and<br />

Molecular Genetics (Faculty <strong>of</strong> Biology), Albert-Ludwigs-<strong>University</strong> <strong>of</strong> Freiburg,<br />

Germany , 3 Spemann Graduate School <strong>of</strong> Biology and Medicine (SGBM), FRIAS<br />

LIFENET, BIOSS<br />

Reactive oxygen species (ROS) are constant byproducts <strong>of</strong> mitochondrial respiration that<br />

under physiological conditions are scavenged by detoxifying enzymes. An increase in ROS<br />

production, however, requires additional specific regulatory responses preventing mitochondrial<br />

damage. These responses are mediated by retrograde signaling to the nucleus which we<br />

monitored using a hsp-6::gfp reporter.<br />

To mildly increase mitochondrial ROS production, we exposed C. elegans to 0.5 mM<br />

paraquat. Genome-scaled RNAi screening revealed the mitochondrial unfolded protein<br />

response (UPR mt ) mediating transcription factor ATFS-1 as essential for the retrograde ROS<br />

response.<br />

Knockdown <strong>of</strong> genes involved in basic cellular mechanisms such as translation and protein<br />

transport specifically impaired the retrograde ROS response. Remarkably, none <strong>of</strong> these<br />

conditions abrogated other stress signaling pathways that evoke the UPR ER , the heat shock<br />

response or the phase II oxidative stress response, however, a subset <strong>of</strong> these conditions<br />

impaired the UPR mt . We hypothesize that a stress status <strong>of</strong> the worm, which is initiated by<br />

impairment <strong>of</strong> basic cellular functions, suppresses the retrograde response towards mild ROS<br />

stress. Future research will address the hierarchic interplay between these distinguishable<br />

stress states.<br />

Contact: eva.runkel@sgbm.uni-freiburg.de<br />

Lab: Baumeister<br />

Poster Topic: Stress<br />

123


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

The Effects <strong>of</strong> HIF-1 Over-Activation: Real-Time Assays for Toxin<br />

Response<br />

Jenifer Saldanha, Archana Parashar, Santosh Pandey, Jo Anne Powell-C<strong>of</strong>fman<br />

Iowa State <strong>University</strong>, Ames, Iowa, USA<br />

In animals as diverse as nematodes, fruit flies, and humans, the hypoxia inducible factor<br />

(HIF) transcription factors affect gene expression changes that enable adaption to hypoxia<br />

(low oxygen). C. elegans HIF-1 has been shown to have roles in aging, stress resistance and<br />

neuronal development. HIF-1 activity is regulated by multiple mechanisms. When oxygen<br />

levels are sufficiently high, the EGL-9 enzyme hydroxylates HIF-1 and thus targets HIF-1 for<br />

degradation via the VHL-1-mediated proteasomal degradation pathway. EGL-9 also inhibits<br />

HIF-1 transcriptional activity. Strong loss-<strong>of</strong>-function mutations in egl-9 confer resistance to<br />

Pseudomonas aeruginosa PAO1 fast killing. More specifically, the egl-9 mutants are resistant<br />

to cyanide produced by the bacteria. Other mutations in the HIF-1 pathway also influence C.<br />

elegans resistance to cyanide. To characterize these phenotypes and to examine the roles<br />

<strong>of</strong> other genes in cyanide resistance, we employed an array <strong>of</strong> genetic tools and micr<strong>of</strong>luidic<br />

technologies. We have quantitated several behavioral parameters for wild-type and mutant<br />

strains, and we will present some <strong>of</strong> these results. Micr<strong>of</strong>luidic platforms and real-time imaging<br />

enable greater spatio-temporal resolution <strong>of</strong> the phenotypes, which, in turn, enriches genetic<br />

analyses <strong>of</strong> the HIF-1 genetic networks.<br />

Contact: jenifers@iastate.edu<br />

Lab: Powell-C<strong>of</strong>fman<br />

124<br />

Poster Topic: Stress


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

Exploring Natural Variation <strong>of</strong> Starvation Resistance and Growth Rate<br />

Moses Sandr<strong>of</strong>, Meghan Jobson, L. Ryan Baugh<br />

Department <strong>of</strong> Biology, Duke Center for Systems Biology, Duke <strong>University</strong><br />

The majority <strong>of</strong> studies in C. elegans use strains from a single genetic background. However,<br />

in the analysis <strong>of</strong> ecologically relevant traits, it is advantageous to look at strains from an<br />

array <strong>of</strong> environmental and evolutionary backgrounds. In theory, natural selection acts in an<br />

environment-specific manner on phenotypes relating to life history such that relevant traits may<br />

vary. Our laboratory uses L1 arrest and recovery as a nutritional deprivation model to examine<br />

trade-<strong>of</strong>fs between stress resistance and growth. We hypothesize that there is an inverse<br />

relationship between starvation resistance and growth rate among wild isolates, reflecting a<br />

trade-<strong>of</strong>f in resource allocation. We are investigating starvation survival and growth rate in<br />

multiple wild-type isolates to characterize phenotypic variation and address this hypothesis.<br />

Recent work in our lab reveals a variety <strong>of</strong> epigenetic effects <strong>of</strong> starvation on growth and stress<br />

resistance, and we are also investigating natural variation <strong>of</strong> these epigenetic effects.<br />

Contact: moses.sandr<strong>of</strong>@gmail.com<br />

Lab: Baugh<br />

Poster Topic: Stress<br />

125


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

The Role <strong>of</strong> the Translation Machinery in Survival from Hypoxia<br />

Barbara Scott, Chun-Ling Sun, Xianrong Mao, Charles Yu, C. Michael Crowder<br />

Washington <strong>University</strong> School <strong>of</strong> Medicine, St. Louis, Missouri, USA<br />

A reduction <strong>of</strong> protein synthesis has been associated with resistance to hypoxic cell death.<br />

Which components <strong>of</strong> the translation machinery control hypoxic sensitivity and the mechanism<br />

for this hypoxia resistance has not been systematically investigated. A reduction in oxygen<br />

consumption has been widely assumed to be the primary mechanism. Using genetic reagents<br />

in C. elegans, we examined the effect on organismal survival after hypoxia <strong>of</strong> knockdown and/<br />

or mutation <strong>of</strong> twelve translation factors functioning at various steps in translation. Reduction-<strong>of</strong>function<br />

<strong>of</strong> most translation machinery genes significantly increased hypoxic survival, however,<br />

to varying degrees. Surprisingly, translation rates as measured by FRAP did not correlate with<br />

hypoxic sensitivity except for reduction-<strong>of</strong>-function <strong>of</strong> aminoacyl-tRNA synthetases. Oxygen<br />

consumption rates correlated weakly with increased hypoxic survival (r 2 = 0.34, p=0.02).<br />

Hypoxic sensitivity had no correlation with lifespan or reactive oxygen species sensitivity, two<br />

phenotypes previously associated with reduced translation rates. Misfolded protein sensitivity<br />

was the only phenotype that strongly correlated with hypoxic sensitivity (r 2 = 0.90, p


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

The Role <strong>of</strong> the Ubiquitin-Proteasome System in the Formation <strong>of</strong><br />

Polyglutamine Aggregates<br />

Gregory Skibinski, Lynn Boyd<br />

<strong>University</strong> <strong>of</strong> Alabama in Huntsville, Huntsville, (Alabama), USA<br />

The misfolding <strong>of</strong> soluble proteins and subsequent aggregation is associated with several<br />

neurodegenerative diseases and is thought to be involved in the pathological process.<br />

Aggregates in multiple diseases, including polyglutamine (polyQ) aggregates, show positive<br />

immunostaining for ubiquitin and proteasomes. The ubiquitin-proteasome system, which<br />

degrades transient and damaged proteins, may be impaired in these diseases and is thought<br />

to be involved in aggregation. To investigate this, we are using a transgenic line <strong>of</strong> C. elegans<br />

developed by Morimoto et al. that expresses an aggregation-prone stretch <strong>of</strong> glutamine repeats<br />

(Q82) fused to GFP(green fluorescent protein). We have shown that knockdown <strong>of</strong> specific E2<br />

ubiquitin-conjugating (UBCs) enzymes alters the size and frequency <strong>of</strong> polyQ-GFPaggregates,<br />

and that ubc-1, ubc-13, and uev-1 are required for ubiquitin colocalization to aggregates<br />

(Howard et al., BMC Cell Biology2007, 8:32). To view this process in real-time, we are using<br />

time-lapse microscopy to view aggregation <strong>of</strong> soluble Q82::GFP into punctate aggregates<br />

in early-stage C. elegans larvae (Skibinski , Boyd, BMC Cell Biology 2012, 13:10).We have<br />

observed that the initial in vivo formation <strong>of</strong> polyQ aggregates in the body wall muscle cells<br />

occurs over a span <strong>of</strong> about 1 hour. RNAi <strong>of</strong> UBCs, most notably ubc-22, affects the rate <strong>of</strong> this<br />

initial aggregation, primarily by increasing the steady-state levels <strong>of</strong> Q82::GFP protein in the cell.<br />

This may indicate that the initial aggregation event is not directly dependent on ubiquitination<br />

<strong>of</strong> aggregating proteins. Beyond this initial phase, aggregation is slower, but RNAi <strong>of</strong> ubc-1,<br />

ubc-22, or uev-1 increases this rate. RNAi <strong>of</strong> ubc-13 reduces this rate. When Q82::GFP is<br />

co-expressed with an mCherry::ubiquitin fusion protein, the mCherry::ubiquitin colocalizes<br />

toQ82::GFP aggregates primarily in the late larval stages. FRAP analysis indicates that ubiquitin<br />

may be mobile within the aggregate. Currently,we are producing a strain expressing Citrine<br />

fused to proteasome subunit RPT-1, which will extend the analysis <strong>of</strong> aggregation dynamics<br />

to the proteasome.<br />

Contact: greg.skibinski@gmail.com<br />

Lab: Boyd<br />

Poster Topic: Stress<br />

127


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

C. elegans as a Model to Study Drug-induced Mitochondrial<br />

Dysfunction<br />

Richard de Boer, Reuben Smith, Hans van der Spek, Stanley Brul<br />

<strong>University</strong> <strong>of</strong> Amsterdam, Amsterdam, The Netherlands<br />

To address fundamental questions concerning drug-induced mitochondrial dysfunction,<br />

Caenorhabditis elegans was applied as a model organism. Several studies have shown the<br />

occurrence <strong>of</strong> mitochondrial dysfunction as a consequence <strong>of</strong> therapeutic drug use, especially<br />

the drugs used to treat HIV infected individuals. The cause <strong>of</strong> anti-HIV drug toxicity appears<br />

to be linked to a common mechanism: a decreased mitochondrial energy-generating capacity<br />

putatively caused by the secondary inhibition <strong>of</strong> mitochondrial DNA polymerase γ, resulting<br />

in the depletion <strong>of</strong> mitochondrial DNA (mtDNA). However, the exact mechanism behind<br />

mitochondrial toxicity remains unknown as drug effects are diverse. Most research has been<br />

done in patient- or cell culture studies, which pose limitations on the experiments that can<br />

be performed. Progress in this field is highly dependent on the development <strong>of</strong> a robust and<br />

accurate model system.<br />

Using C. elegans as a model system we show a concentration dependent decline in mtDNA<br />

copies when cultured in the presence <strong>of</strong> various anti-retroviral drugs. This decline was both<br />

absolute and relative compared to nuclear DNA. Moreover, exposure to these drugs resulted<br />

in altered aerobic respiration, increased ROS production and/or a quantifiable disruption <strong>of</strong> the<br />

mitochondrial morphological network. The severity <strong>of</strong> the observed effects is drug-specific and<br />

concentration dependent. Interestingly, the observed biochemical and morphological effects are<br />

not necessarily provoked by the same compounds and some <strong>of</strong> the effects can be alleviated<br />

by providing supplementation with compounds active on the mitochondrial respiratory chain.<br />

Since the side-effects <strong>of</strong> the drugs in patients on anti-retroviral therapy closely resemble<br />

our observed effects in C. elegans , we conclude that C. elegans is a highly suitable model<br />

organism to study drug induced mitochondrial dysfunction. Moreover, as C. elegans is a<br />

highly expedient model system, searching for compounds to alleviate the induced toxicities is<br />

straightforward. Preliminary results suggest the beneficial effect <strong>of</strong> supplementation as a way<br />

<strong>of</strong> counteracting and alleviating some <strong>of</strong> these pernicious side-effects.<br />

Contact: r.l.smith@uva.nl<br />

Lab: Brul<br />

128<br />

Poster Topic: Stress


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

Identification <strong>of</strong> pathways by which a peroxiredoxin influences stress<br />

resistance reveal (i) its importance for insulin/IGF1-like-signalling<br />

and (ii) new genes important for phase 2 detoxification gene<br />

expression, stress resistance and longevity<br />

Helen Crook, Monika Olahova, Elizabeth Veal<br />

Newcastle <strong>University</strong>, Newcastle upon Tyne<br />

Peroxiredoxins (Prx) are abundant peroxidases with an evolutionarily conserved role in<br />

longevity. As well as detoxifying peroxides, Prx are important regulators <strong>of</strong> signal transduction.<br />

For instance, Prx have a conserved role in promoting the stress-induced activation <strong>of</strong> p38-related<br />

MAPK, such as C. elegans PMK-1 [1]. PMK-1 activates the Nrf2-related transcription factor<br />

SKN-1 to increase expression <strong>of</strong> phase 2 detoxification genes and oxidative stress resistance<br />

[2]. However, despite reduced PMK-1 activation, C. elegans lacking the single ortholog <strong>of</strong> the<br />

Prx1 tumor suppressor, PRDX-2, express elevated levels <strong>of</strong> phase 2 detoxification enzymes<br />

and are hyper-resistant to some forms <strong>of</strong> oxidative stress [1]. We have undertaken a variety <strong>of</strong><br />

approaches to understand the basis for the increased stress resistance and reduced lifespan<br />

<strong>of</strong> prdx-2 mutant C. elegans. We have discovered that the increased activity <strong>of</strong> the DAF-16<br />

and SKN-1 transcription factors contribute to their increased stress resistance. Indeed our data<br />

strongly suggest that PRDX-2 is required for the insulin/IGF1-like (IIS)-dependent inhibition <strong>of</strong><br />

both DAF-16 and SKN-1, thus indicating a hitherto unknown role for Prx in IIS.<br />

In addition, we have used a genome-wide RNAi screen to identify other genes that are<br />

important for the increased expression <strong>of</strong> the SKN-1-target gene gcs-1 in prdx-2 mutant animals.<br />

Consistent with the important contribution made by SKN-1-regulated genes to longevity, a<br />

significant number <strong>of</strong> the genes identified by this screen have been previously identified as<br />

important for the lifespan-extension associated with reduced IIS [3]. Moreover, we find that<br />

many <strong>of</strong> the genes we have identified are also important for stress-induced increases in gcs-1<br />

expression in wild-type animals. Strikingly, our analysis has revealed that many <strong>of</strong> these new<br />

activators <strong>of</strong> phase 2 detoxification gene expression, including the receptor <strong>of</strong> activated protein<br />

kinase C RACK-1, act by novel mechanisms that are independent or downstream from PMK-1.<br />

We will discuss the important implications <strong>of</strong> our findings for the role <strong>of</strong> IIS, the PMK-1/SKN-1<br />

canonical pathway and Prx in stress resistance and ageing.<br />

1 Olahova et al (2008) PNAS 105; 19839-44<br />

2 Inoue et al (2005) Genes & Dev 19;2278-83<br />

3 Samuelson et al (2007) Genes & Dev 21;2976-94<br />

Contact: e.a.veal@ncl.ac.uk<br />

Lab: Veal<br />

Poster Topic: Stress<br />

129


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

Energy Metabolism Abnormality Of Ultraviolet Irradiation Sensitive<br />

Mutant Rad-8 In C. elegans<br />

Kayo Yasuda 1 , Michihiko Fujii 2 , Hitoshi Suda 3 , Phillip Hartman 4 , Takamasa Ishii 1 , Naoaki<br />

Ishii 1<br />

1 Tokai <strong>University</strong>, Isehara, Kanagawa, Japan, 2 Yokohama City <strong>University</strong>,<br />

Yokohama, Kanagawa, Japan, 3 Tokai <strong>University</strong>,Nishino, Numazu, Japan, 4 Texas<br />

Christian <strong>University</strong>, Fort Worth, Texas, USA<br />

C. elegans rad-8 has been isolated as an ultraviolet radiation hypersensitive mutant and we<br />

have also found that this mutant is oxygen hypersensitive. Reactive Oxygen Species (ROS) as<br />

well as radiation cause cellular damages and result in a variety <strong>of</strong> diseases including cancer<br />

and aging. It is well known that most endogenous ROS are produced from mitochondria.<br />

In order to know whether oxidative sensitivity <strong>of</strong> rad-8 mutant is correlated with mitochondrial<br />

oxidative stress, we examined, 1) amount <strong>of</strong> supeoxide anion (O 2 - ) one <strong>of</strong> ROS, from<br />

mitochondria and 2) carbonylated protein as a marker <strong>of</strong> oxidative stress, 3) apoptosis at<br />

embryonic stage, 4) growth rate 5) energy metabolism and 6) mitochondrial function. Our<br />

results showed that the level <strong>of</strong> O 2 - and the carbonylated protein in mitochondria produced by<br />

the O 2 - significantly increased compared to N2 wild type. The apoptosis also increased. On<br />

the other hand, the energy metabolisms and growth rate <strong>of</strong> rad-8 were lower compared to N2.<br />

Furthermore, the body size <strong>of</strong> rad-8 at the adult stage was smaller than N2.<br />

These data suggested that the function <strong>of</strong> rad-8 gene is related with mitochondria. It is<br />

assumed that the energy metabolic abnormality in rad-8 mitochondria leads to increase <strong>of</strong><br />

oxidative stress and apoptosis by the ROS over production.<br />

Contact: lilac@is.icc.u-tokai.ac.jp<br />

Lab: Ishii<br />

130<br />

Poster Topic: Stress


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

Graphite Nanoplatelets and Caenorhabditis elegans: Insights from an<br />

in vivo Model<br />

Elena Zanni, Giovanni De Bellis, Maria Bracciale, Alessandra Broggi, Maria Santarelli,<br />

Maria Sarto, Claudio Palleschi, Daniela Uccelletti<br />

Sapienza, <strong>University</strong> <strong>of</strong> Rome, Italy<br />

Nanosized particles display unique physical and chemical properties and represent an<br />

increasingly important material in the development <strong>of</strong> novel nanodevices which can be used<br />

in numerous physical, biological, biomedical and pharmaceutical applications.Intriguingly,<br />

graphene, a single sheet <strong>of</strong> sp 2 hybridized honeycombcarbon, and graphene-related materials<br />

such as graphite nanoplatelets (GNPs), have proved to be among the most promising materials<br />

due to their unique electronic properties, such as an extremely high carrier mobility and a<br />

linear dispersion relation.<br />

Here we report an in vivo toxicity assessment <strong>of</strong> graphite nanoplatelets on the animal<br />

model Caenorhabditis elegans. Our study pointed out the absence <strong>of</strong> any acute or chronic<br />

toxicity <strong>of</strong> GNPs on the nematodes. Moreover, the genotoxicity has been investigated and no<br />

effect on C. elegans reproductive potential has been found. In addition, an imaging technique<br />

based on Fourier Transform Infrared Spectroscopy (FT-IR) mapping was also developed to<br />

demonstrate the effective GNPs intake inside the nematodes. Such analysis not only indicated<br />

a GNPs distribution along the whole worm body, but gave evidence <strong>of</strong> nanoparticles transition<br />

from the intestine tract to the gonads.<br />

The ever-growing interest for graphene-based nanoparticles in potential applications such<br />

as nanomedicine prompted us to investigate the antimicrobial properties <strong>of</strong> GNPs. Given the<br />

useful properties <strong>of</strong>fered by the host-pathogen system C.elegans-Pseudomonas aeruginosa<br />

we assessed the in vivo antimicrobial potential <strong>of</strong> GNPs. Notably, the nanoplatelets treatment<br />

<strong>of</strong> Pseudomonas-infected animals led to a strikingly mortality <strong>of</strong> worm-colonizing bacteria<br />

within 30 minutes <strong>of</strong> exposure.<br />

We believe that our data obtained by using the animal model C. elegans could optimize<br />

the experimental validation procedures involved in safety issues. Infact, the combined effect<br />

<strong>of</strong> long term biocompatibility, absence <strong>of</strong> genotoxicity and antimicrobial activity demonstrated<br />

in nematodes should push to study such properties directly in mammalian models, especially<br />

those highly predictive for human applications.<br />

Contact: elena.zanni@uniroma1.it<br />

Lab: Uccelletti<br />

Poster Topic: Stress<br />

131


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

Cancer-like features could facilitate rapid intracellular growth <strong>of</strong> a<br />

natural intracellular parasite in C. elegans.<br />

Malina Bakowski2 , Amy Ma2 , Christopher Desjardins1 , Christina Cuomo1 , Emily Troemel2 1 2 The Broad Institute <strong>of</strong> MIT and Harvard, Cambridge, MA, <strong>University</strong> <strong>of</strong> California<br />

San Diego, La Jolla, CA<br />

Microsporidia are obligate intracellular parasites most closely related to fungi. They are<br />

ubiquitous in nature, but are poorly understood because <strong>of</strong> the difficulties in culturing them.<br />

We found a new species <strong>of</strong> microsporidia isolated from a wild-caught C. elegans near Paris<br />

and we named it Nematocida parisii, or nematode-killer from Paris. In addition to this N. parisii<br />

strain (ERTm1), we found Nematocida infections in wild-caught Caenorhabditis hosts from<br />

several locations around the world. This Nematocida/Caenorhabditis system provides a unique<br />

opportunity to study microsporidia infection in a tractable model organism.<br />

To elucidate the mechanisms <strong>of</strong> microsporidia pathogenesis we sequenced the genomes<br />

<strong>of</strong> three Nematocida strains. We also performed RNA-seq analysis on ERTm1 during infection<br />

within the C. elegans host to quantify infection dynamics and define the N. parisii transcriptome.<br />

Microsporidia have the smallest known eukaryotic genomes and, likewise, we find that the<br />

ERTm1 genome is only 4.1 Mb and contains just 2,661 predicted genes, 95.7% <strong>of</strong> which<br />

are expressed during infection <strong>of</strong> C. elegans based on RNA-seq. We are using this genome<br />

sequence data, together with other microsporidian genomes, to learn more about mechanisms<br />

<strong>of</strong> pathogenesis.<br />

Microsporidia have a limited metabolic capacity and cannot grow independently, instead<br />

relying on host cells as sources <strong>of</strong> energy and macromolecules. However, after they invade<br />

host cells these parasites can grow extremely rapidly in the intracellular environment. For<br />

example, N. parisii can grow 220-fold in less than 32 hours inside C. elegans intestinal cells.<br />

We utilized the microsporidia genome data to develop a model that may explain this swift<br />

growth. Early during infection N. parisii appears to deliver a key metabolic enzyme into the<br />

host cell cytoplasm where it could direct the infected cell to enter an anabolic state, generating<br />

building blocks necessary for cellular proliferation <strong>of</strong> the parasite, such as nucleosides. We also<br />

found that N. parisii and other microsporidian genomes lack a key cell cycle inhibitor, which is<br />

encoded by a tumor suppressor gene found in humans. Anabolic metabolism and disregulated<br />

cell cycle are both features shared by many types <strong>of</strong> cancer cells. Thus, microsporidia may<br />

use cancer-like strategies <strong>of</strong> metabolic reprogramming together with a fast cell cycle to rapidly<br />

grow inside <strong>of</strong> host cells.<br />

Contact: mbakowski@ucsd.edu<br />

Lab: Troemel<br />

132<br />

Poster Topic: Pathogenesis


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

Identifying a genetic basis to resistance against microsporidian<br />

parasites<br />

Keir Balla 1 , Erik Andersen 2 , Zhi Yan 1 , Leonid Kruglyak 2 , Emily Troemel 1<br />

1 UCSD, La Jolla, CA, USA, 2 Lewis-Sigler Institute for Integrative Genomics,<br />

Princeton <strong>University</strong>, New Jersey, USA<br />

Microsporidia are intracellular eukaryotic parasites that infect nearly all animal phyla. As<br />

microsporidia can cause lethal disease in both humans and agriculturally important animals,<br />

it is critical that we learn more about the basis for host defense. We have recently identified<br />

a strong genetic component to C. elegans resistance against infection by the microsporidian<br />

pathogen Nematocida parisii. Strain ERTm1 is a natural pathogen that was first isolated<br />

from a wild-caught C. elegans near Paris, and since that time has been isolated from wildcaught<br />

nematodes in many parts <strong>of</strong> the world. N. parisii kills the N2 strain more quickly than<br />

the CB4856 strain. In addition, we have found that N. parisii is present at a higher pathogen<br />

load in N2 than in CB4856, and we have exploited this difference to pursue the responsible<br />

genetic locus. We developed a liquid culture infection assay and use quantitative PCR to detect<br />

pathogen RNA levels within the host. With this assay, we are analyzing a panel <strong>of</strong> recombinant<br />

inbred advanced intercross lines (RIAILs) generated by a cross between N2 and CB4856 for<br />

quantitative trait loci (QTL) mapping. We have measured pathogen load in over 100 RIAILs<br />

to map significant QTL on chromosomes II and V. RIAILs bearing CB4856 SNPs within these<br />

intervals were significantly associated with lower pathogen loads. We are now generating nearisogenic<br />

lines containing the QTL intervals in an otherwise entirely N2 or CB4856 background<br />

to confirm the role <strong>of</strong> these QTL in resistance and to allow for fine-mapping <strong>of</strong> the prospective<br />

quantitative trait nucleotides. The CB4856 resistance phenotype appears to be recessive to the<br />

N2 phenotype and thus we hypothesize it is conferred by a loss- or reduction-<strong>of</strong>-function allele<br />

in CB4856. We will test this hypothesis by performing RNAi experiments targeting candidate<br />

genes within the identified QTL in the N2 strain and performing phenotypic rescue experiments<br />

in the CB4856 strain. These experiments will provide the first insights about the genetic basis<br />

<strong>of</strong> resistance to microsporidia in any system. In addition to investigations on the host side <strong>of</strong><br />

resistance, we have identified a new strain <strong>of</strong> N. parisii from a wild-caught C. briggsae from<br />

Kauai, Hawaii. We are comparing the relative virulence and the genome <strong>of</strong> this strain to the<br />

ERTm1 strain. Altogether, our studies will reveal insights into the co-evolutionary interactions<br />

that may be important for a variety <strong>of</strong> host-microsporidia pairs.<br />

Contact: kballa@ucsd.edu<br />

Lab: Troemel<br />

Poster Topic: Pathogenesis<br />

133


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

Mapping natural variation in sensitivity to the Orsay virus in C.<br />

elegans wild isolates<br />

Tony Belicard, Marie-Anne Felix<br />

Institut de Biologie de l?Ecole Normale Sup?rieure, CNRS-Inserm-ENS, Paris,<br />

France<br />

The first known virus naturally infecting C. elegans was recently discovered (Félix M-A,<br />

Ashe A, Piffaretti J, et al., 2011). The Orsay virus causes intestinal cell disorders to its natural<br />

host, the strain JU1580. Its genome is composed <strong>of</strong> two positive single-strand RNA segments<br />

and is close to that <strong>of</strong> known nodaviruses infecting fish and insects. Transmission is horizontal.<br />

The viral infection can be cured by submitting the embryos to bleach. Viral preparations can<br />

be obtained through 0.2 mm filtration.<br />

Here we explore inter- and intraspecific sensitivity to the virus in Caenorhabditis. We<br />

challenged strains <strong>of</strong> interest with a viral preparation and assayed replication <strong>of</strong> the virus 7<br />

days after infection.<br />

Until now there is no evidence that the Orsay virus can infect a C. briggsae culture. Two<br />

other related viruses (called Santeuil and Le Blanc viruses) have been found infecting wild<br />

isolates <strong>of</strong> C. briggsae and none <strong>of</strong> them seems able to infect C. elegans. Thus, each virus<br />

so far seems specific to C. elegans versus C. briggsae.<br />

At an intraspecific level, large variation in sensitivity to the Orsay virus is found among C.<br />

elegans wild isolates. We measured quantitatively, using RT-qPCR, the amount <strong>of</strong> RNA1 at 7<br />

days post-infection, in a set <strong>of</strong> 97 natural isolates <strong>of</strong> C. elegans. Using the RAD-sequencing<br />

data on these 97 strains (Andersen E, Gerke J, et al., 2012), we performed a Genome-Wide<br />

Association Mapping. We were able to detect a wide signal <strong>of</strong> 6 Mb in the center <strong>of</strong> Chromosome<br />

IV. This signal corresponds to a haplotype that is common to almost all sensitive strains and is<br />

found mostly in European isolates. We are now narrowing the region by screening for laboratory<br />

recombinations in this 6 Mb region and testing candidate genes in the region (see Jérémie Le<br />

Pen’s communication for complementary information).<br />

Contact: belicard@biologie.ens.fr<br />

Lab: Félix<br />

134<br />

Poster Topic: Pathogenesis


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

The Human Herpesvirus Type 1 Latency Associated Transcript<br />

Produces Egg Laying and Locomotion Defects in the Nematode C.<br />

elegans<br />

Ana Bratanich, Jesica Diogo<br />

<strong>University</strong> <strong>of</strong> Buenos Aires,Buenos Aires,Argentina<br />

C elegans is presently utilized as a model for the study <strong>of</strong> pathogen virulence factors<br />

especially in the field <strong>of</strong> bacteriology and mycology. However, its application in virology has<br />

been essentially inexistent. In this work, we have utilized C elegans to obtain transgenic strains<br />

expressing a human herpes virus type 1 gene associated with the latency process in sensory<br />

neurons. The function <strong>of</strong> this gene (Latency Associated Transcript or LAT) has been extremely<br />

difficult to study in higher eukaryotic systems. In vivo and in vitro experiments would suggest<br />

that one <strong>of</strong> its roles is to modulate apoptosis. C elegans transgenic strains expressing the LAT<br />

gene showed important phenotypic changes in the form <strong>of</strong> locomotion and egg laying defects.<br />

Treatment with different drugs suggested that these phenotypes were produced by neuronal<br />

presynaptic defects. RNAi experiments proved these defects were specifically mediated by the<br />

LAT gene. No obvious anatomic changes were observed in the egg laying neuronal machinery<br />

when transgenic hermaphrodites were crossed with strains expressing GFP. However, a smaller<br />

number <strong>of</strong> apoptotic bodies was found in the gonads <strong>of</strong> LAT transgenic strains when compared<br />

to controls. To the authors’ knowledge, this is the first time that transgenic C elegans strains<br />

are obtained expressing a gene from a mammalian virus.<br />

Contact: abratanich@fvet.uba.ar<br />

Lab: Bratanich<br />

Poster Topic: Pathogenesis<br />

135


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

The Conserved 1-Cys Peroxiredoxin, PRDX-6, has Diverse Roles in<br />

Stress Resistance, Innate Immunity and Aging<br />

Emma Button, Elizabeth Veal<br />

Newcastle <strong>University</strong>, Newcastle-upon-Tyne, United Kingdom<br />

Peroxiredoxins (Prx) are highly conserved, abundant antioxidant enzymes that are important<br />

for longevity in yeast, worms, flies and mice. For instance, C. elegans, lacking the 2-cys Prx,<br />

PRDX-2, the human ortholog <strong>of</strong> the tumour suppressor Prdx1, age more rapidly and are shortlived<br />

[1]. Prx utilise reversibly oxidized cysteines in the catalytic degradation <strong>of</strong> peroxides but<br />

have other functions in signal transduction and as chaperones. For instance, mammalian<br />

1-Cys Prx are multifunctional enzymes with separate peroxidase and phospholipase A 2 (PLA 2)<br />

activities. 1-cys Prxs have been found to be important for oxidative stress resistance in mice and<br />

yeast and increased levels <strong>of</strong> human Prdx6 have been associated with lung and breast cancer<br />

[2]. The C. elegans genome encodes a single 1-cys Prx, PRDX-6, in which the PLA 2 active<br />

site residues are not conserved. Hence, we have used prdx-6 mutant C.elegans as a model<br />

to investigate the role <strong>of</strong> the peroxidase activity in stress resistance and aging. Surprisingly,<br />

we have made the discovery that C. elegans lacking prdx-6 are oxidative stress-resistant and<br />

long-lived at 15 o C. However, at 25 o C the lifespan <strong>of</strong> prdx-6 mutant and wildtype animals are<br />

indistinguishable. Our investigations into the basis for this temperature–dependent effect on<br />

aging have led us to discover that PRDX-6 plays pathogen-specific roles in innate immunity.<br />

Here, we will present the results <strong>of</strong> our investigations into the underlying mechanisms by which<br />

PRDX-6 influences stress resistance, aging and responses to different bacterial pathogens.<br />

1 Olahova M. et al Proc. Natl. Acad. Sci. USA 2008, 105:19839-44<br />

2 Fisher A.B. Antioxidants & Redox Signaling. 2011, 15:831-44<br />

Contact: Emma.Button@ncl.ac.uk<br />

Lab: Veal<br />

136<br />

Poster Topic: Pathogenesis


Contact: cschen@mail.ncku.edu.tw<br />

Lab: Chen<br />

Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

Shiga-like Toxin 1 Is Required Partly for The Pathogenicity <strong>of</strong><br />

Escherichia coli O157:H7 in Caenorhabditis elegans<br />

Chang-Shi Chen1 , Ting-Chen Chou1 , Hao-Chieh Chiu2 , Cheng-Ju Kuo1 , Ching-Ming<br />

Wu3 , Wan-Jr Syu4 1Department <strong>of</strong> Biochemistry and Molecular Biology, National Cheng Kung<br />

<strong>University</strong>, Tainan, Taiwan, 2Department <strong>of</strong> Clinical Laboratory Sciences and<br />

Medical Biotechnology, National Taiwan <strong>University</strong>, Taipei, Taiwan, 3Department <strong>of</strong> Cell Biology and Anatomy, National Cheng Kung <strong>University</strong>, Tainan, Taiwan,<br />

4Institute <strong>of</strong> Microbiology and Immunology, National Yang Ming <strong>University</strong>, Taipei,<br />

Taiwan<br />

Enterohaemorrhagic Escherichia coli (EHEC) causes life-threatening infections in humans<br />

as a consequence <strong>of</strong> the production <strong>of</strong> Shiga-like toxins. Lack <strong>of</strong> a good animal model system<br />

hinders the study <strong>of</strong> EHEC virulence with systematic genetic methods in vivo. Here we applied<br />

the genetic tractable animal model, Caenorhabditis elegans, as a surrogate host to study the<br />

virulence <strong>of</strong> EHEC as well as the host innate immunity to this human pathogen. Our results<br />

show that E. coli O157:H7, a serotype <strong>of</strong> EHEC, intoxicates, induces a severe bag <strong>of</strong> worms<br />

(Bag) phenotype, and kills C. elegans. Colonization and induction <strong>of</strong> the characteristic attaching<br />

and effacing (A/E) lesions by E. coli O157:H7 are concomitantly demonstrated in the intact<br />

intestinal epithelium <strong>of</strong> C. elegans in vivo. Genetic analysis indicates that the Shiga-like toxin 1<br />

(Stx1) <strong>of</strong> E. coli O157:H7 is a virulence factor in C. elegans and is required for full toxicity. The<br />

C. elegans p38 MAP kinase and the DAF-2 insulin-like signaling pathways, two evolutionally<br />

conserved innate immune signaling pathways, are activated and mediated in the regulation <strong>of</strong><br />

host susceptibility to EHEC infection. Our results validate the EHEC-C. elegans interaction as<br />

suitable for future comprehensive genetic screens in both bacterial and host factors involved<br />

in the pathogenesis <strong>of</strong> EHEC infection.<br />

Poster Topic: Pathogenesis<br />

137


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

Leucobacter Strains Are Diverse Natural Pathogens <strong>of</strong> Caenorhabditis<br />

Laura Clark 1 , Marie-Anne Felix 2 , Maria Joao Gravato-Nobre 1 , Jonathan Hodgkin 1<br />

1 <strong>University</strong> <strong>of</strong> Oxford, Oxford, UK, 2 IBENS, Paris, France<br />

Examining wild isolates <strong>of</strong> Caenorhabditis is a productive way <strong>of</strong> discovering new nematode<br />

pathogens, some <strong>of</strong> which exhibit unusual modes <strong>of</strong> attack and reveal new aspects <strong>of</strong> host<br />

defense. Two cases, involving three bacterial strains belonging to the Gram-positive coryneform<br />

genus Leucobacter, provide illustration. First, the C. elegans isolate JU1088, obtained from<br />

an aviary in Kakegawa, Japan, was found to be chronically infected with a Leucobacter strain<br />

(CBX130/LJ) with similar effects to Microbacterium nematophilum, characterized by rectal<br />

colonization and inflammatory swelling <strong>of</strong> the tail region. The much-studied M. nematophilum<br />

has been repeatedly isolated as a lab contaminant but never found in the wild; strain LJ shows<br />

that rectal infection and host swelling response are part <strong>of</strong> the natural life <strong>of</strong> C. elegans. Infection<br />

by LJ exhibits some significant differences from infection by M. nematophilum. Second, an<br />

isolate <strong>of</strong> Caenorhabditis n. sp. 11, JU1635, found on rotting banana trunks in Cape Verde,<br />

was found to be doubly infected with two different Leucobacter strains, termed Verde1 and<br />

Verde2. The double infection could be transferred to C. elegans. Verde2 has similar but much<br />

more virulent effects to M. nematophilum and LJ: it rapidly kills most wildtype worms and elicits<br />

a strong but futile tail-swelling response. Most C. elegans Bus mutants (20+ complementation<br />

groups), which have been isolated for resistance to M. nematophilum, are also resistant<br />

to Verde2. Verde1 bacteria, in contrast, do not kill wildtype C. elegans but adhere densely<br />

over most <strong>of</strong> the nematode surface and elicit up-regulation <strong>of</strong> the defense peptide NLP-29.<br />

However, most Bus mutants (resistant to Verde2) are lethally hypersensitive to Verde1, dying<br />

from cuticle destruction, shrinkage and lysis. Verde1 therefore has latent potential as a virulent<br />

Caenorhabditis pathogen. A different kind <strong>of</strong> infection has been reported for Leucobacter strain<br />

TAN31504 (Muir & Tan, 2008, Appl Environ Microbiol 75:4185-98), which targets uterine tissue.<br />

A survey <strong>of</strong> other characterized Leucobacter species did not reveal further Caenorhabditis<br />

disease effects, but the properties <strong>of</strong> the four strains described above suggest that this genus<br />

may have a propensity to evolve anti-nematode pathogenicity. Progress on detailed description<br />

<strong>of</strong> the two Verde strains, and dissection <strong>of</strong> their virulence mechanisms, will be reported.<br />

Contact: lauracarolineclark@gmail.com<br />

Lab: Hodgkin<br />

138<br />

Poster Topic: Pathogenesis


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

Discriminating Pathogens from Innocuous Microbes: C/EBP<br />

Surveillance Immunity in C. elegans Intestinal Defense<br />

Tiffany Dunbar, Zhi Yan, Keir Balla, Margery Smelkinson, Emily Troemel<br />

<strong>University</strong> <strong>of</strong> California San Diego, La Jolla, CA, USA<br />

Intestinal epithelial cells are regularly exposed to a wide variety <strong>of</strong> microbes, and must<br />

be able to rapidly distinguish between innocuous and harmful microbes in order to mount<br />

a specific response to dangerous microbes. P. aeruginosa is an opportunistic pathogen in<br />

humans and causes a lethal intestinal infection in C. elegans that requires some <strong>of</strong> the same<br />

virulence factors required for killing mammalian hosts. To investigate the early response to<br />

infection we defined a set <strong>of</strong> genes specifically induced by pathogenic P. aeruginosa. A GFP<br />

reporter for one <strong>of</strong> these genes, called irg-1 (infection response gene 1), is robustly induced<br />

by infection with pathogenic P. aeruginosa, but is not induced by other pathogens tested so<br />

far. Furthermore, irg-1 is not induced by non-pathogenic P. aeruginosa strains. Thus, the irg-<br />

1::GFP reporter provides a convenient and specific read-out for the early events in intestinal<br />

cell response to pathogenic P. aeruginosa. Induction <strong>of</strong> irg-1 is controlled by a basic leucine<br />

zipper (bZIP) transcription factor called zip-2 that is required specifically for irg-1 induction and<br />

provides defense against P. aeruginosa infection.<br />

We performed a full-genome RNAi screen to identify other factors in the zip-2/irg-1 pathway.<br />

From this screen we identified 57 RNAi clones that cause irg-1::GFP induction in the absence <strong>of</strong><br />

infection. Many <strong>of</strong> these hits block mRNA translation, and we further confirmed that attenuation<br />

<strong>of</strong> translation triggers largely zip-2 dependent irg-1 induction. Surprisingly, we found that this<br />

attenuation <strong>of</strong> translation leads to activation <strong>of</strong> ZIP-2::GFP expression in the nuclei <strong>of</strong> intestinal<br />

cells. Our data indicates that regulation <strong>of</strong> ZIP-2 translation is conferred by a region in the<br />

zip-2 coding transcript that contains a conserved upstream open reading frame (uORF). We<br />

are currently investigating the mechanism by which this uORF mediates the response to P.<br />

aeruginosa infection. We also identified a candidate binding partner for ZIP-2, based on in<br />

vitro binding studies (Aaron Reinke and Amy Keating, personal communication) and RNAi<br />

knock-down in C. elegans. This binding partner is encoded by the C48E7.11 gene, which we<br />

have renamed cebp-2, since it is a likely ortholog <strong>of</strong> the C/EBP-gamma bZIP transcription<br />

factor in mammals. We are examining C/EBP signaling in mammals to determine whether the<br />

surveillance pathway used in C. elegans is also important in mammalian cells to discriminate<br />

pathogens from innocuous microbes.<br />

Contact: tldunbar@ucsd.edu<br />

Lab: Troemel<br />

Poster Topic: Pathogenesis<br />

139


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

The invertebrate lysozyme ILYS-3 aids bacterial disruption and acts<br />

in the intestine under non-autonomous pharyngeal control to protect<br />

against bacterial infection<br />

Maria Joao Gravato-Nobre , Jonathan Hodgkin<br />

<strong>University</strong> <strong>of</strong> Oxford, Oxford, UK<br />

Multiple immune effectors that fight pathogen infections have been identified in C. elegans:<br />

many act promptly taking just a few hours to reach peak levels and attain rapid microbial<br />

clearance. We hypothesize that there is a second group <strong>of</strong> effectors that “mop-up” pathogens<br />

that escape the initial response. We show that the C. elegans invertebrate lysozyme, ILYS-3<br />

functions in this way and is required for proper defense against pathogen infection. A full-genome<br />

microarray analysis previously identified a rapid up-regulation <strong>of</strong> ILYS-3 in response to M.<br />

nematophilum. Subsequent quantitative RT-PCR experiments revealed that ILY-3 levels peak<br />

at 72 h after challenge with different Gram-positive bacteria, including M. nem. and M. luteus.<br />

We find that ILYS-3 is critical for the survival <strong>of</strong> C. elegans exposed to Leucobacter Verde2,<br />

which evokes a swelling response in the rectum followed by almost 100% larval mortality 96 h<br />

after infection. Besides a hypersensitive response to Verde2, ilys-3 mutations permit increased<br />

intestinal proliferation <strong>of</strong> non-pathogenic E. coli strains, reflecting an impaired ability <strong>of</strong> larvae<br />

and young adults to reduce bacterial burden in the gut lumen. Conversely, over-expression<br />

<strong>of</strong> ILYS-3 confers striking protection against pathogen-mediated lethality by Verde2. ILYS-3 is<br />

most highly expressed in the pharynx and in the intestine <strong>of</strong> C. elegans. Intestinal induction<br />

<strong>of</strong> ilys-3p::GFP in response to infection by M. nematophilum requires activation <strong>of</strong> the ERK<br />

MAP kinase pathway. In the intestine <strong>of</strong> ERK mutants, basal expression <strong>of</strong> ilys-3p::GFP is<br />

reduced and its induction is abolished. Tissue specific experiments demonstrate that induction<br />

<strong>of</strong> ilys-3 is controlled in a cell-non-autonomous fashion. Expressing MPK-1 in the intestine <strong>of</strong><br />

mpk-1 mutants did not restore ilys-3 expression or its induction upon exposure to M. nem. In<br />

contrast, the expression <strong>of</strong> MPK-1 in the pharyngeal muscle resulted in an increased constitutive<br />

expression <strong>of</strong> ilys-3p::GFP in the intestine and rescued reporter gene induction in the mpk-1<br />

(ku1) mutant upon infection. These findings indicate that activation <strong>of</strong> MPK-1 in the pharynx<br />

regulates intestinal expression <strong>of</strong> ILYS-3 and suggest a mechanism whereby pharyngeal cells<br />

sense danger signals and respond by activating an antimicrobial response in distal tissues,<br />

where pathogen clearance will be key to organismal survival.<br />

Contact: maria.gravato-nobre@bioch.ox.ac.uk<br />

Lab: Hodgkin<br />

140<br />

Poster Topic: Pathogenesis


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

Dissecting ERAD Component Function in a Motor Neuron Disease<br />

Model<br />

Angela Jablonski, Robert Kalb<br />

<strong>University</strong> <strong>of</strong> Pennsylvania, Philadelphia, PA, USA<br />

Agingis a major risk factor for the development <strong>of</strong> neurodegenerative disease. Accumulation<br />

<strong>of</strong> aberrantly folded proteins is seen in neurodegenerative disease and these insoluble<br />

proteins may be toxic to neurons. Cellular processes that ensure correct protein folding may<br />

suppress toxicity and other age-related defects in neurodegeneration. One protein quality<br />

control mechanism is ER-associated degradation (ERAD). Improperly folded cell-surface or<br />

secreted proteins are retrotranslocated from the ER to the proteasome for ubiquitin-dependent<br />

degradation. ERAD dysfunction can lead to ER stress and, if unchecked, apoptosis.<br />

Several studies have linked dysfunction <strong>of</strong> ERAD to neurodegenerative diseases, including<br />

amyotrophiclateral sclerosis (ALS). ERAD has been studied extensively in yeast, but our<br />

understanding <strong>of</strong> this process is still limited. We had two goals: First, we wanted to determine<br />

if loss-<strong>of</strong>-function (LOF) alleles <strong>of</strong> individual ERAD components led to a phenotype (such as<br />

uncoordinated movement, sterility, or larval arrest). This would allow us to identify essential<br />

ERAD components versus components that were dispensable. Second, we wanted to determine<br />

if LOF alleles <strong>of</strong> individual ERAD components suppressed or enhanced proteotoxic insults<br />

incurred by expression <strong>of</strong> mutant TDP43 in the nervous system - C. elegans engineered to<br />

express a mutant version <strong>of</strong> TDP43 (M337V – implicated in familial ALS) display locomotor<br />

deficits and motor neuron death. To do this, we made double animals <strong>of</strong> mutant TDP43 and LOF<br />

ERAD component alleles, including ubxn-4 (tm3247), ufd-3 (tm2915), and cdc-48.1 (tm544).<br />

We found that ufd-3 (tm2915) improved average speed <strong>of</strong> animals by two-fold at L4+1d and<br />

L4+5d when compared to mutant TDP43 animals. Interestingly ufd-3 (tm2915) alone showed<br />

no difference to N2 on average locomotor speed at L4, L4+1d, L4+3d,L4+5d, or L4+7d. Ubxn-4<br />

(tm3247) enhanced toxicity and worsened average locomotor speed at stage L4. Meanwhile,<br />

the cdc-48.1 (tm544) double showed no change. These data support our hypothesis that some<br />

ERAD components are necessary, some may be dispensable, and others may be modifiers<br />

with the capability <strong>of</strong> suppressing or enhancing toxicity in models <strong>of</strong> motor neuron disease.<br />

Contact: ajablo@mail.med.upenn.edu<br />

Lab: Kalb<br />

Poster Topic: Pathogenesis<br />

141


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

A Transmembrane Leucine-Rich Repeat Protein Important for<br />

Microsporidia Infection in C. elegans<br />

Robert Luallen, Malina Bakowski, Emily Troemel<br />

<strong>University</strong> <strong>of</strong> California-San Diego, La Jolla, (CA), USA<br />

C. elegans has proven to be an excellent organism for the study <strong>of</strong> host/pathogen<br />

interactions and innate immunity. This is exemplified by the discovery <strong>of</strong> several signal<br />

transduction pathways used in host defense and recent discoveries showing that C. elegans<br />

surveils for disruption <strong>of</strong> core host cellular processes to detect infection. However, little is<br />

known about the earliest steps <strong>of</strong> C. elegans host/pathogen interactions. In particular, we do<br />

not know which host receptors are used to detect pathogens, nor which receptors may be<br />

exploited by pathogens to facilitate infection. To identify such receptors, we are studying a<br />

natural eukaryotic pathogen <strong>of</strong> C. elegans, a species <strong>of</strong> microsporidia called Nematocida parisii.<br />

N. parisii is an obligate intracellular pathogen that infects the intestine. Using larval arrest as<br />

an early readout for pathogen detection and infection, we conducted a targeted RNAi screen<br />

for genes that inhibited C. elegans arrest after N. parisii infection. One hit from this screen was<br />

a transmembrane (tm) protein containing an extracellular leucine-rich repeat (LRR) domain,<br />

which is found on numerous pathogen receptors from other systems, such as Toll-like receptors.<br />

Worms in which this LRR-tm gene was knocked down exhibited less arrest upon infection.<br />

We find that this defect in arrest is specific to N. parisii infection, because knockdown <strong>of</strong> this<br />

protein has no effect on arrest <strong>of</strong> worms infected with the bacterial pathogen Pseudomonas<br />

aeruginosa. We generated a tagged version <strong>of</strong> this protein and showed that it was expressed<br />

in the intestine, pharynx, and neurons <strong>of</strong> transgenic worms. Furthermore, it localized to the<br />

apical and vesicular membranes <strong>of</strong> the intestine, which poises this LRR protein for pathogen<br />

interaction in the intestinal lumen. Intestinal specific RNAi in the VP303 strain recapitulated<br />

inhibition <strong>of</strong> larval arrest after infection, suggesting this LRR-tm protein functions in the intestine<br />

to control a response to N. parisii. Knockdown <strong>of</strong> the LRR-tm protein resulted in slower N. parisii<br />

infection progression and lower pathogen load, as measured by FISH and qPCR. Together,<br />

the data suggests that this LRR protein may be involved early in C. elegans infection, either<br />

as a receptor used by N. parisii for efficient infection or as a suppressor <strong>of</strong> immunity. As this<br />

LRR-tm protein is conserved in mammals, dissecting its mechanism <strong>of</strong> action in early infection<br />

may inform us about intestinal microsporidia infection in humans.<br />

Contact: rluallen@ucsd.edu<br />

Lab: Troemel<br />

142<br />

Poster Topic: Pathogenesis


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

The redox sensor TRX-1: a potential regulator <strong>of</strong> signaling in C.<br />

elegans<br />

Katie McCallum, Danielle Garsin<br />

The <strong>University</strong> <strong>of</strong> Texas Health Science Center at Houston, Houston, Texas, USA<br />

Our lab has previously demonstrated that in C. elegans intestinal infections stimulate<br />

the production <strong>of</strong> reactive oxygen species (ROS) by Ce-Duox1/BLI-3. Though overall this<br />

response has protective effects, it challenges intracellular redox homeostasis, resulting in<br />

the simultaneous up-regulation <strong>of</strong> intracellular detoxification enzymes. This response occurs<br />

through the activation <strong>of</strong> SKN-1, an oxidative stress response transcription factor, in a p38<br />

MAPK pathway dependent manner. It is currently unknown how BLI-3-generated ROS activates<br />

the p38 MAPK pathway. This question is <strong>of</strong> broad interest because ROS can serve as an<br />

important signaling molecule in not only the immune response, but in the regulation <strong>of</strong> other<br />

vital processes such as cell growth and differentiation. We propose that activation <strong>of</strong> the p38<br />

MAPK pathway is regulated by a thioredoxin sensor, which is sensitive to the ROS generated<br />

by BLI-3. In C. elegans, the role <strong>of</strong> thioredoxins is understudied. It has been demonstrated<br />

that a trx-1 null mutant exhibits decreased longevity on E. coli OP50. Classically, thioredoxin’s<br />

major role is thought to be as a cytosolic anti-oxidant, and therefore important for detoxifying<br />

reactive molecules. However, recent studies suggest that thioredoxins can also play a role in<br />

regulation by affecting the activity <strong>of</strong> signaling components. Our current hypothesis is that in the<br />

C. elegans intestine, thioredoxin, in conjunction with other factors, influences SKN-1 activation<br />

during infection. Specifically, we postulate that intracellular BLI-3 generated ROS is sensed<br />

by TRX-1 and relives repression <strong>of</strong> the p38 MAPKKK, NSY-1, ultimately allowing activation<br />

<strong>of</strong> SKN-1. In support <strong>of</strong> this hypothesis, preliminary data suggests that TRX-1 influences<br />

expression <strong>of</strong> SKN-1-regulated genes and susceptibility during infection.<br />

Contact: katie.c.mccallum@uth.tmc.edu<br />

Lab: Garsin<br />

Poster Topic: Pathogenesis<br />

143


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

Functional Characterization <strong>of</strong> Thioredoxin 3 (TRX-3), a<br />

Caenorhabditis elegans Intestine-Specific Thioredoxin<br />

Maria Jimenez-Hidalgo 1 , Cyril Leopold Kurz 2 , Jose Rafael Pedrajas 3 , Francisco Jose<br />

Naranjo-Galindo 1 , Juan Cabello 4 , Elamparithi Jayamani 5 , Eleftherios Mylonakis 5 , Juan<br />

Carlos Fierro-Gonzalez 6 , Peter Swoboda 6 , Antonio Miranda-Vizuete 1,7<br />

1 Centro Andaluz de Biologia del Desarrollo, Universidad Pablo de Olavide, Seville,<br />

Spain, 2 Centre d’Immunologie de Marseille-Luminy, Universite de la Mediterranee,<br />

Marseille, France, 3 Departamento de Biologia Experimental, Universidad de Jaen,<br />

Jaen, Spain, 4 Center for Biomedical Research <strong>of</strong> La Rioja (CIBIR), Logrono, Spain,<br />

5 Infectious Disease Division, Massachusetts General Hospital, Harvard Medical<br />

School, Boston, USA, 6 Center for Biosciences at NOVUM, Dept. <strong>of</strong> Biosciences<br />

and Nutrition, Karolinska Institute, Huddinge, Sweden, 7 Instituto de Biomedicina<br />

de Sevilla (IBIS), Hospital Universitario Virgen del Rocio/CSIC, Seville, Spain<br />

Thioredoxins are a class <strong>of</strong> evolutionarily conserved proteins that have been demonstrated to<br />

play a key role in many cellular processes involving redox reactions. We report here the genetic and<br />

biochemical characterization <strong>of</strong> C. elegans TRX-3, the first metazoan thioredoxin with an intestinespecific<br />

expression pattern. By using GFP reporters we have found that trx-3 is expressed in the nucleus<br />

but also in the cytoplasm <strong>of</strong> intestinal cells where it shows a more prominent apical localization. trx-3<br />

(tm2820) loss <strong>of</strong> function mutants are viable, have normal longevity and progeny size and have no<br />

obvious phenotype other than a slightly smaller size. Despite the restricted expression in intestinal cells,<br />

trx-3 mutants do not show any defects in the gut during embryogenesis, accumulate normal levels <strong>of</strong><br />

lipids, maintain a normal structural and functional integrity <strong>of</strong> the gut apical membrane and only exhibit<br />

a minor reduction in defecation cycle timing. The C. elegans intestine is constantly exposed to chemical<br />

stressors and toxins as well as pathogens and thioredoxins are well known antioxidant proteins, which<br />

have been shown to function as protective systems against different types <strong>of</strong> stresses. However, when<br />

subjected to different treatments that induce oxidative stress such as heat shock, acrylamide, juglone<br />

or paraquat exposure, trx-3 (tm2820) mutants did not display enhanced sensitivity. Furthermore, trx-3<br />

(tm2820) mutants did not modify the levels or subcellular localization <strong>of</strong> DAF-16, HSP-16.2 or GST-<br />

4 stress reporters. Interestingly, trx-3 is induced upon infection with Photorhabdus luminescens and<br />

Candida albicans but not other pathogens such as Serratia marcescens. Preliminary data show that<br />

trx-3 (tm2820) mutants are as resistant as wild type controls to P. luminescens killing but, in contrast,<br />

trx-3 overexpression protects against killing with this pathogen. We are currently evaluating the role<br />

<strong>of</strong> TRX-3 in Candida albicans infection. In this context, it has been shown that trx-3 is equally induced<br />

upon exposure to live or heat-killed Candida, thus suggesting that TRX-3 could function at early steps<br />

<strong>of</strong> infection, such as pathogen recognition or colonization. Together, our data indicate that TRX-3<br />

function in the intestine is dispensable for C. elegans development but might play an important role in<br />

the immune response elicited against some bacterial and fungal infections.<br />

Contact: amiranda-ibis@us.es<br />

Lab: Miranda-Vizuete<br />

144<br />

Poster Topic: Pathogenesis


Contact: jmiskowski@uwlax.edu<br />

Lab: Miskowski<br />

Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

Screening Potential Anthelmintic Compounds for Novel Activity<br />

Megan Gross1 , Olufadakemi Awoyinka1 , Michael Smith1 , M. Shahjahan Kabir2 , James<br />

Cook2 , Aaron Monte1 , Jennifer Miskowski1 1 2 <strong>University</strong> <strong>of</strong> <strong>Wisconsin</strong>-La Crosse; La Crosse, WI 54601, <strong>University</strong> <strong>of</strong><br />

<strong>Wisconsin</strong>-Milwaukee; Milwaukee, WI USA<br />

Helminths are parasitic worms that infect plants, animals, and humans worldwide leading to a<br />

decreased food supply, economic hardship, and significant morbidity and mortality. Anthelmintics<br />

that combat these infections are represented by only six major classes <strong>of</strong> compounds. Misuse<br />

<strong>of</strong> these drugs has contributed to widespread anthelmintic resistance in livestock helminths<br />

and emerging drug resistance in human-infecting helminths. The identification <strong>of</strong> new lead<br />

compounds that target helminths is imperative. The non-parasitic nematode Caenorhabditis<br />

elegans has long been a model system for helminths - available anthelmintics kill C. elegans<br />

at doses similar to that <strong>of</strong> helminths, the molecular mechanism <strong>of</strong> action for all anthelmintics<br />

was elucidated using C. elegans, and the development <strong>of</strong> drug resistance has also been<br />

studied in C. elegans.<br />

Previously, we developed and performed two microassays to screen a unique collection <strong>of</strong><br />

synthetic compounds for anthelmintic activity. These analogs were derived from an antimicrobial<br />

stilbene product originally isolated from Comptonia peregrina (“sweet fern”). The motility<br />

assay screened C. elegans for paralysis and the developmental assay screened worms<br />

for developmental arrest prior to sexual maturity, a decrease in fecundity, or lethality. Sixtyseven<br />

stilbenoid compounds were tested in one or both assays, and six test compounds were<br />

prioritized for further study.<br />

These test compounds possess a novel structure compared to existing anthelmintics, yet<br />

the potencies <strong>of</strong> the compounds do not warrant their development as bona fide anthelmintics<br />

at this time. Thus, we are utilizing these agents as probes to explore mechanisms by which<br />

worms might be targeted. To this end, the stilbenoids are being tested against mutant C. elegans<br />

strains that are resistant to existing anthelmintics using our two assays. This will reveal if any<br />

<strong>of</strong> the new compounds act via a novel molecular mechanism. We have initially focused on<br />

CL-5, which showed the strongest activity in both assays. To date, CL-5 has been tested on<br />

ivermectin-resistant strains, DA1302 and DA1306. Ivermectin is one <strong>of</strong> the most widely used<br />

anthelmintics on the market today, and ivermectin resistance has been documented in helminth<br />

strains. Interestingly, CL-5 affects ivermectin-resistant C. elegans in a dose-dependent manner,<br />

similar to wild-type worms. We are currently testing CL-5 against other anthelmintic-resistant<br />

C. elegans strains and will report our progress.<br />

Poster Topic: Pathogenesis<br />

145


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

Characterizing C. elegans behavior in response to E. faecalis<br />

Mona Gardner, Thomas Graciano, June Middleton, Edith Myers<br />

Fairleigh Dickinson <strong>University</strong>, College at Florham, Madison, NJ<br />

Caenorhabditis elegans feed on soil bacteria that may sometimes be pathogenic. We<br />

decided to develop a screen to determine the pathogenicity <strong>of</strong> environmentally-isolated strains<br />

<strong>of</strong> Enterococci. We assayed survival <strong>of</strong> C. elegans fed on various strains <strong>of</strong> E. faecalis. None<br />

<strong>of</strong> these strains had a significant effect on C. elegans survival. However, C.elegans behavior<br />

was altered in the presence <strong>of</strong> several strains. The behaviors observed included egg retention<br />

and avoidance <strong>of</strong> the E.faecalis lawns. While we know that in the total absence <strong>of</strong> food<br />

C.elegans leave their environment in search <strong>of</strong> food, and cease egg laying, their behavioral<br />

responses to Enterococcus (a potentially pathogenic food source) are not well characterized.<br />

Egg retention assays involved counting the number <strong>of</strong> eggs retained by adult worms cultured<br />

for several days on strains <strong>of</strong> E. faecalis. Worms exposed to some but not all strains showed<br />

an increase in egg retention. In avoidance assays, the number <strong>of</strong> worms occupying a lawn<br />

<strong>of</strong> E faecalis was counted periodically over 24 hours. Worms showed significant avoidance<br />

<strong>of</strong> all bacterial strains used. We have identified several strains <strong>of</strong> environmental Enterococci<br />

that altered C. elegans behavior and are now assaying those strains for antibiotic resistance,<br />

hemolysis, bacteriocin, and gelatinase production. We hope to correlate C. elegans behavioral<br />

responses to the expression <strong>of</strong> known virulence factors in E.faecalis strains. We also plan<br />

to investigate the molecular mechanisms by which E. faecalis affects behavior by assaying<br />

behavioral responses to bacteria in different genetic backgrounds.<br />

Contact: emyers@fdu.edu<br />

Lab: Myers<br />

146<br />

Poster Topic: Pathogenesis


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

Exploring Antifungal Potential <strong>of</strong> CAMPs in the C.elegans Infection<br />

Model<br />

Massimiliano Olivi, Daniela Uccelletti, Maria Mangoni, Donatella Barra, Claudio<br />

Palleschi<br />

Sapienza, <strong>University</strong> <strong>of</strong> Rome, Italy<br />

The continuous development <strong>of</strong> multi drug resistant microorganisms, both bacterial and<br />

fungal, increased the interest in discovering new antimicrobial compounds. Using a wholeanimal<br />

model such as the nematode Caenorhabditis elegans for the screening <strong>of</strong> putative<br />

antimicrobial compounds will help studying not only the molecular mechanisms <strong>of</strong> microbial<br />

pathogenesis but also the toxicity <strong>of</strong> this molecules.<br />

In this study we focus our attention on the opportunistic human pathogen Candida<br />

albicans, whose infections have emerged as an important cause <strong>of</strong> morbidity and mortality<br />

in immunocompromised patients. It is well known that C. albicans, as well as other Candida<br />

species, are ingested by the nematode C. elegans and can establish a persistent lethal infection<br />

in the worm intestinal tract. Moreover, this model <strong>of</strong> pathogenesis replays key aspects <strong>of</strong> the<br />

fungus infections, such as iphae development, occurring in mammalian hosts.<br />

Naturally occurring membrane-active cationic antimicrobial peptides (CAMPs) serve as<br />

attractive candidates for the development <strong>of</strong> new therapeutic agents. Amphibian skin is one <strong>of</strong><br />

the richest sources for such peptides whose in vivo activity against the opportunistic pathogen<br />

Pseudomonas aeruginosa has been demonstrated. We investigated the antifungal action in<br />

vivo <strong>of</strong> CAMPs from frog skin (e.g., temporins and esculentin fragments) on the ATCC10231<br />

strain <strong>of</strong> C. albicans utilizing Caenorhabditis elegans as minihost model.<br />

Contact: massimiliano.olivi@uniroma1.it<br />

Lab: Palleschi<br />

Poster Topic: Pathogenesis<br />

147


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

Using Bacterial Infection as a Tool to Dissect the Molecular<br />

Components <strong>of</strong> the Nematode Surface<br />

Delia O’Rourke, Rebecca Price, Dave Stroud, Jonathan Hodgkin<br />

<strong>University</strong> <strong>of</strong> Oxford, Oxford, UK.<br />

Previous forward genetic screens for worms resistant to the non-lethal bacterial infection by<br />

M. nematophilum identified 25+ bus genes that prevent infection, many <strong>of</strong> which are involved<br />

in the synthesis <strong>of</strong> the nematode surface. These mutants also show altered resistance and<br />

sensitivity to infections with two Gram-positive Leucobacter strains, Verde1 and Verde2,<br />

isolated from an infected worm found on the Cape Verde islands (J. Hodgkin and M. A. Felix<br />

unpublished). Verde1 infection, which is non-lethal to wild type worms, kills many <strong>of</strong> the bus<br />

mutants. This allowed us to conduct powerful suppressor screens to identify 4 subs (suppressors<br />

<strong>of</strong> bus sensitivity) <strong>of</strong> bus-2, bus-10, srf-2 and srf-5. The subs mutants show phenotypic features<br />

<strong>of</strong> altered surface coat including fragility and bleach sensitivity.<br />

Strikingly, subs-1 suppresses bus-10, srf-2 and srf-5 sensitivity to Verde1. Using whole<br />

genome sequencing we identified subs-1 (e3016) as an opal stop in exon3 <strong>of</strong> agmo-1<br />

(alkylglycerol monooxygenase) an enzyme that cleaves the O-alkyl bonds <strong>of</strong> ether lipids and is<br />

dependent on the co-factor tetrahydrobiopterin. Multiple alleles <strong>of</strong> subs-1 have been identified;<br />

three alleles include changes in the conserved residues <strong>of</strong> the fatty acid hydroxylase motif. I<br />

will describe the bus-10 suppressor screen and the identification <strong>of</strong> subs-1 and discuss how<br />

the molecular identification <strong>of</strong> these suppressors furthers our understanding <strong>of</strong> the surface<br />

coat <strong>of</strong> the worm.<br />

Contact: delia.orourke@bioch.ox.ac.uk<br />

Lab: Hodgkin<br />

148<br />

Poster Topic: Pathogenesis


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

Identification <strong>of</strong> the Transcriptional Targets <strong>of</strong> the PMK-1 p38-<br />

Dependent Transcription Factor ATF-7<br />

Daniel Pagano, Dennis Kim<br />

MIT, Cambridge, MA, USA<br />

Innate immunity in C. elegans requires a conserved PMK-1 p38 mitogen-activated protein<br />

kinase (MAPK) pathway that regulates the basal and pathogen-induced expression <strong>of</strong> immune<br />

effectors. We identified ATF-7, a conserved basic-region leucine zipper (bZIP) transcription<br />

factor, as a downstream target <strong>of</strong> PMK-1 p38 MAPK and a regulator <strong>of</strong> immune effector gene<br />

expression. Unlike PMK-1 p38 MAPK, which is required for resistance against multiple stresses<br />

in addition to infection, ATF-7 appears to function specifically in the response to infection.<br />

Prior gene expression analyses <strong>of</strong> PMK-1 pathway mutants have identified putative immune<br />

effector genes; however, validating that these genes function in pathogen defense has been<br />

challenging due to the likely functional redundancy <strong>of</strong> immune effectors. The identification <strong>of</strong><br />

ATF-7 and its prominent and specific role in PMK-1 p38-dependent immunity suggests that<br />

analysis <strong>of</strong> ATF-7 transcriptional targets will identify the genes that comprise the immune<br />

response. We performed RNA-Seq on RNA extracted from wild-type and atf-7 mutant animals<br />

fed non-pathogenic E. coli OP50 and pathogenic P. aeruginosa PA14 to determine the genes<br />

regulated by ATF-7 both basally and upon infection. We anticipate that the results <strong>of</strong> our wholegenome<br />

expression analysis will better define the immune response in C. elegans.<br />

Contact: dpagano@mit.edu<br />

Lab: Kim<br />

Poster Topic: Pathogenesis<br />

149


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

The Role <strong>of</strong> the G-protein Coupled Receptor FSHR-1 in the C. elegans<br />

Stress Response<br />

Amanda Miller, Hannah Anthony, Joseph Robinson, Shannon Hartley, Jennifer Powell<br />

Gettysburg College, Gettysburg, PA, USA<br />

Innate immunity is a critical defense mechanism against infection by pathogenic<br />

microorganisms that is present in virtually all metazoans. One <strong>of</strong> the major mediators <strong>of</strong> innate<br />

immunity in C. elegans is the conserved p38 MAPK pathway. This signaling pathway not only<br />

controls the expression <strong>of</strong> a set <strong>of</strong> putative antimicrobial effectors via the transcription factor<br />

ATF-7, but also coordinately regulates the response to oxidative stress via the transcription<br />

factor SKN-1 (Shivers, et al. PLoS Genetics 2010). FSHR-1 is a G-protein coupled receptor that<br />

is required in C. elegans for the defense against infection by diverse pathogens, including Gram<br />

negative bacteria, Gram positive bacteria, and fungi. This receptor is necessary and sufficient<br />

in the intestine for its immune function and has been shown genetically to act in parallel to the<br />

p38 MAPK pathway while converging on many <strong>of</strong> the same transcriptional targets. We are<br />

investigating whether FSHR-1 also plays a role in the response to various cellular stresses,<br />

including oxidative stress, thermal stress, and heavy metal stress. Preliminary results suggest<br />

that, like p38 MAPK, FSHR-1 is important for the response to oxidative stress.<br />

Contact: jpowell@gettysburg.edu<br />

Lab: Powell<br />

150<br />

Poster Topic: Pathogenesis


Contact: amirsa@caltech.edu<br />

Lab: Sternberg<br />

Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

Nematodes Living in Anoxic Conditions at a Deep-sea Methane Seep<br />

are Infected With Sexually Transmitted Parasitic Fungus-related<br />

Microsporidia<br />

Amir Sapir1 , Adler Dillman1 , Manuel Mundo-Ocampo2,3 , James Baldwin2 , Victoria<br />

Orphan4 , Paul Sternberg1 1Howard Hughes Medical Institute and Division <strong>of</strong> Biology, California Institute <strong>of</strong><br />

Technology, Pasadena, CA 91125, USA, 2Department <strong>of</strong> Nematology, <strong>University</strong><br />

<strong>of</strong> California, Riverside, CA 92521, USA, 3CIIDIR-IPN, Unidad Sinaloa, Guasave,<br />

Sin. Mex C.P. 81000, 4Division <strong>of</strong> Geological and Planetary Sciences, California<br />

Institute <strong>of</strong> Technology, Pasadena, CA 91125, USA<br />

Intracellular parasites <strong>of</strong> the phylum Microsporidia infect and thrive on large range <strong>of</strong> hosts<br />

including nematodes from the elegans group such as C. elegans. Microsporidia biodiversity,<br />

host specificity, and infection modes in the context <strong>of</strong> their natural environment are poorly<br />

understood but are <strong>of</strong> growing medical and agricultural interests due to the proposed roles<br />

<strong>of</strong> microsporidia infection in AIDS patients mortality and colony collapse disorder <strong>of</strong> infected<br />

honey bees. In a survey aimed to identify and characterize deep sea nematodes along with<br />

their symbiotic microbes in methane cold seeps, we surprisingly discovered a new marine<br />

nematode species that is infected with microsporidia. The infected worms were found primarily<br />

as male and female adults that consume methane and sulfide-oxidizing microorganisms in<br />

an anoxic, chemosynthetic niche. Microsporidia infection was strictly species-specific but<br />

found in samples taken from the same site in the Pacific (Hydrate Ridge) in two successive<br />

years demonstrating an ecologically stable interaction between microsporidia and deep sea<br />

nematodes. Microsporidia spores and intracellular meronts localization in the reproductive<br />

systems <strong>of</strong> both infected males and females nematodes suggests that the fungus is transmitted<br />

sexually in contrast to most microsporidia species, which are known to be transmitted by oralfaucal<br />

cycles. Detailed morphological analysis <strong>of</strong> infected tissues by transmission electron<br />

microscopy revealed that the fungus targets cells <strong>of</strong> the body wall muscles causing muscle<br />

malformation and fiber rearrangements. An 18S rRNA-based phylogeny suggests there<br />

were two independent events <strong>of</strong> microsporidia-nematode parasitism. Additionally, infection <strong>of</strong><br />

deep sea worms may represent the first host-switch outside the basal bryozoan hosts during<br />

microsporidia evolution. The discovery <strong>of</strong> a parasitic fungus that thrives on a marine nematode<br />

host is the first reported case <strong>of</strong> deep sea microsporidia highlighting the potential for additional<br />

as yet uncharacterized species <strong>of</strong> Microsporidia and other parasitic fungi living in anoxic, deep<br />

sea environments.<br />

Poster Topic: Pathogenesis<br />

151


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

Key Residues <strong>of</strong> Cry5B Structure and Function: Mutagenesis by<br />

Alanine Scanning<br />

Jillian Sesar, Yan Hu, Sandy Chang, Arash Safavi, Raffi Aroian<br />

<strong>University</strong> <strong>of</strong> California, San Diego, San Diego, CA, USA<br />

Soil-transmitted helminthes (hookworms, whipworms, and Ascaris) infect more than 2<br />

billion people worldwide. Only one drug (albendazole) is able to show a high enough efficacy<br />

against parasite worms under conditions for mass drug administration. However, recent studies<br />

have shown an increase in resistance to this drug, stressing the importance <strong>of</strong> finding a new<br />

treatment option. Crystal (Cry) proteins produced from the soil bacterium Bacillus thuringiensis<br />

have been used for decades as a means to control insects that destroy crops and transmit<br />

human diseases, and studies have shown these proteins to be safe to humans. Our lab has<br />

shown that crystal proteins, specifically Cry5B, are able to kill both the free-living nematode<br />

Caenorhabditis elegans, as well as parasitic roundworms (eg. Ancylostoma ceylanicum,<br />

hookworm). We are currently investigating several <strong>of</strong> these crystal proteins to be safe and<br />

effective anthelmintics.<br />

Cry proteins intoxicate invertebrates by acting as pore-forming toxins. Several defined<br />

steps in their mechanism <strong>of</strong> action have been suggested from insect studies, but there is still<br />

great uncertainty as to the importance <strong>of</strong> these various steps. We believe that the C. elegans<br />

– Cry5B system has great potential to unlock mysteries surrounding Cry proteins.<br />

Here, I have mutated all <strong>of</strong> the 698 amino acids in the toxin domain <strong>of</strong> Cry5B, and<br />

subsequently tested these mutants on C. elegans to assess for changes in toxicity levels.<br />

These results display either an increase or decrease in toxicity as compared to the wild type<br />

Cry5B. This information has two main applications. First, it can tell us which residues are<br />

important in Cry5B protein function, with the eventual goal being to correlate these changes<br />

in activity with specific changes in protein functionality (eg. receptor binding, pore formation).<br />

Second, we can use the information gathered from the screen against C. elegans to identify<br />

Cry protein mutants that could have potential increases in toxicity against the parasites. Our<br />

goal here would be to ultimately identify improved Cry protein variant candidates for treating<br />

one <strong>of</strong> the most neglected diseases <strong>of</strong> our time, parasitic worms.<br />

Contact: jsesar@ucsd.edu<br />

Lab: Aroian<br />

152<br />

Poster Topic: Pathogenesis


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

Caenorhabditis elegans Neutralizes Small Molecule Toxins Produced<br />

by Pseudomonas aeruginosa<br />

Gregory Stupp, Ramadan Ajredini, Arthur Edison<br />

<strong>University</strong> <strong>of</strong> Florida, Gainesville, Fl, USA<br />

Caenorhabditis elegans, which is found in soil and decaying fruit, is exposed throughout its<br />

life to a variety <strong>of</strong> pathogenic microbes, which makes it an ideal candidate to study bacterial<br />

resistance. Although C. elegans has a well-developed innate immune system, it still remains<br />

susceptible to many pathogens, including many that affect humans, such as Pseudomonas<br />

aeruginosa, a common pathogen <strong>of</strong> both plants and animals. P. aeruginosa possesses a<br />

variety <strong>of</strong> virulence factors, including toxic phenazine compounds such as pyocyanin and<br />

1-hydroxyphenazine (1-HP) which are <strong>of</strong>ten found in lungs <strong>of</strong> P. aeruginosa-infected cystic<br />

fibrosis patients. These redox-active compounds are thought to cause damage to cells by<br />

producing reactive oxygen species and disrupting normal redox reactions.<br />

P. aeruginosa grown on high-osmolarity media is able to kill L4 stage C. elegans in as<br />

little as 6 hours through the production <strong>of</strong> phenazines such as phenazine-1-carboxylic acid<br />

(PCA), 1-HP, and pyocyanin. We have found that upon exposure to 1-HP, the worms modify<br />

the compound into at least 5 metabolites. These metabolites, all glycosides <strong>of</strong> 1-HP, are found<br />

in differing concentrations and forms in the worm media and in worm bodies. All <strong>of</strong> these<br />

glycosides <strong>of</strong> 1-HP are less toxic to the worm than 1-HP itself, suggesting that C. elegans is<br />

actively detoxifying its environment. We have identified several <strong>of</strong> the major components <strong>of</strong><br />

the worm media and homogenized worm pellet <strong>of</strong> young adult C. elegans exposed to 1-HP.<br />

The worm media contains 1-(β-glucopyranose)-phenazine (mono), 1-(6-β-glucopyranoseβ-glucopyranose)-phenazine<br />

(di), and at least two phenazine-trisaccharides (tri), while the<br />

homogenized worm pellet contains 1-(3-phospho-glucopyranose)-phenazine (G3P) along with<br />

the mono, di, and tri. We have shown that at concentrations <strong>of</strong> 200 μM, 1-HP kills ~80% <strong>of</strong> L4s<br />

after 6 hours, while mono, di, tri, and G3P all kill less than 5%. We are currently investigating<br />

the concentration and time-dependence <strong>of</strong> 1-HP exposure on glucoside production and are<br />

screening other species for this detoxification activity.<br />

Contact: stuppie@ufl.edu<br />

Lab: Edison<br />

Poster Topic: Pathogenesis<br />

153


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

Coprinopsis cinerea Lectins-mediated Toxicity against C. elegans<br />

Katrin Stutz 3 , Alex Butschi 3 , Silvia Bleuler-Martinez 1 , Mario Schubert 2 , Markus Aebi 1 ,<br />

Markus Kuenzler 1 , Michael Hengartner 3<br />

1 Institute <strong>of</strong> Microbiology, ETH Zurich, Switzerland, 2 Institute <strong>of</strong> Molecular Biology<br />

and Biophysics, ETH Zurich, Switzerland, 3 Institute <strong>of</strong> Molecular Life Sciences,<br />

<strong>University</strong> <strong>of</strong> Zurich, Switzerland<br />

Lectins are non-immunoglobulin, carbohydrate-binding proteins without catalytic activity<br />

towards the recognized carbohydrate. They are widely distributed among eukaryotes such<br />

as plants, fungi and mammals as well as among prokaryotes. Several lectins <strong>of</strong> Coprinopsis<br />

cinerea and <strong>of</strong> other fungi display toxicity towards Caenorhabditis elegans and other organisms<br />

such as Aedes aegypti, Acanthamoeba castellanii and HeLa cells. Thus, they may be part <strong>of</strong><br />

a lectin-mediated defense system <strong>of</strong> higher fungi against predators and parasites.<br />

Our goal is to unravel the glycotargets that are bound by the C. cinerea lectins CGL2 and<br />

CCL2 in C. elegans as well as the underlying toxicity mechanisms. The results may reveal an<br />

Achilles heel in this organism and might pave the way to new approaches in fighting animal<br />

parasitic nematodes.<br />

Toxicity <strong>of</strong> lectins against C. elegans is assessed by feeding E. coli that overexpress lectins.<br />

This results in inhibition <strong>of</strong> development and reproduction and eventually leads to premature<br />

death. As a pro<strong>of</strong> <strong>of</strong> principle, we could show for the galectin CGL2 that its toxicity depends<br />

on its ability to bind to a galactose-containing glycoconjugate present on the apical surface<br />

<strong>of</strong> the intestinal lumen. We used Mos1-transposon mutagenesis to isolate mutants resistant<br />

to CGL2. The molecular analysis <strong>of</strong> the resistance genes allowed us to predict the structure<br />

<strong>of</strong> the glycoepitope bound by CGL2, and also revealed a novel, evolutionary conserved<br />

glycosyltransferase (GALT-1).<br />

Interestingly, toxicity assays <strong>of</strong> a second C. cinerea lectin, CCL2, showed toxicity exclusively<br />

towards C. elegans and Drosophila melanogaster; unlike CGL2 that is toxic to a broad range<br />

<strong>of</strong> species. Glycan array analysis <strong>of</strong> recombinant CCL2 revealed a pronounced carbohydratespecificity<br />

for Fucα1,3GlcNAc-containing glycans. Resistance <strong>of</strong> C. elegans mutants (bre-1,<br />

ger-1, fut-1) defective in the biosynthesis <strong>of</strong> the α1,3-core fucoside confirmed this glycoepitope.<br />

Feeding C. elegans with a dTomato::CCL2 fusion protein showed that CCL2 binds, like CGL2,<br />

to the surface <strong>of</strong> the intestinal epithelium.<br />

By EMS mutagenesis and RNAi screens as well as biochemical purification <strong>of</strong> the<br />

glycotarget, we want to learn more about the structure <strong>of</strong> the CCL2 target. In addition, we<br />

will perform transmission electron microscopy and metabolomics to further elucidate toxicity<br />

mechanisms.<br />

Contact: bluekeye@gmx.net<br />

Lab: Hengartner<br />

154<br />

Poster Topic: Pathogenesis


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

Understanding the Molecular Underpinnings <strong>of</strong> Pathogen Recognition<br />

in C. elegans<br />

Kwame Twumasi-Boateng, Hae-Sung Kang, Michael Shapira<br />

<strong>University</strong> <strong>of</strong> California, Berkeley, Berkeley,CA,USA<br />

Caenorhabditis elegans has been used for over a decade to characterize signal transduction<br />

pathways involved in innate immunity. However, the identity <strong>of</strong> the signals responsible for<br />

initiating immune responses are not clear. While the distinct C.elegans immune response to<br />

different pathogens suggests specific recognition, the molecular basis for this remains unknown.<br />

To better understand what initiates the immune response, we took advantage <strong>of</strong> natural variation<br />

in colonization <strong>of</strong> the C.elegans intestine following exposure to Pseudomonas aeruginosa.<br />

Genome-wide analysis <strong>of</strong> gene expression in worms that were non-colonized versus those that<br />

were fully-colonized revealed that non-colonized worms displayed an immune response which<br />

was at least as robust as their colonized counterparts, indicating that the immune response<br />

preceded substantial colonization-associated damage. Further analyses using quantitative<br />

RT-PCRshowed that pathogen-secreted factors were not sufficient to induce immune gene<br />

expression, but an intact non-pathogenic Pseudomonas species was. This suggests recognition<br />

<strong>of</strong> Pseudomonas-specific structural components. Further analysis is underway, using transgenic<br />

worms expressing GFP from the promoter <strong>of</strong> an early infection-response gene (induced within<br />

one hour <strong>of</strong> exposure to Pseudomonas),to study the molecular underpinnings <strong>of</strong> this response.<br />

Finally, results will be presented from the preliminary characterization <strong>of</strong> a gene family encoding<br />

a protein domain that in plants is involved in microbial envelope recognition, and its roles in<br />

worm immune responses and protection.<br />

Contact: kwametb@berkeley.edu<br />

Lab: Shapira<br />

Poster Topic: Pathogenesis<br />

155


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

Reactive oxygen species generated by BLI-3/Ce-Duox1 during<br />

infection triggers the activation <strong>of</strong> SKN-1 in C. elegans<br />

Ransome van der Hoeven, Katie McCallum, Melissa Cruz, Danielle Garsin<br />

<strong>University</strong> <strong>of</strong> Texas, Health Science Center - Houston, Texas, USA<br />

Coordinated regulation <strong>of</strong> immune reactions is not only crucial to fight invading pathogens,<br />

but also vital in preventing injury to host tissue. Uncontrolled immune responses can lead to<br />

damage, sepsis and ultimately death <strong>of</strong> the host. An example <strong>of</strong> an immune response that can<br />

potentially harm host tissue is the purposeful generation <strong>of</strong> reactive oxygen species (ROS)<br />

by NADPH oxidase family <strong>of</strong> proteins. Recent studies using C. elegans and mammalian cell<br />

lines have shown that intestinal mucosal cells are involved in innate immunity through several<br />

mechanisms, one being the production <strong>of</strong> ROS by dual oxidases (Duox) as an antimicrobial.<br />

However the mechanisms by which the mucosal cells maintain homeostasis during infection are<br />

relatively unknown. Using C. elegans as a model system, we tested the hypothesis that ROS<br />

produced by intestinal cells in response to infection activates the oxidative stress transcription<br />

factor SKN-1, which is an ortholog <strong>of</strong> the mammalian Nrf transcription factor. Following oxidative<br />

stress, SKN-1 promotes survival by upregulating genes involved in the detoxification <strong>of</strong> ROS<br />

and other xenobiotic compounds. We demonstrate that SKN-1 is activated in the intestine <strong>of</strong><br />

the worm when infected with the human pathogens Enterococcus faecalis and Pseudomonas<br />

aeruginosa. Furthermore, we show several SKN-1 dependent genes gcs-1, gst-4, gst-5, gst-7<br />

and gst-10 are upregulated during infection. More importantly we establish that ROS produced<br />

by Ce-Duox1/BLI-3 activates SKN-1 through the p38 MAPK signaling pathway. Systematic<br />

analysis <strong>of</strong> the p38 MAPK pathway revealed that components NSY-1, SEK-1 and PMK-1 are<br />

required for activation <strong>of</strong> SKN-1. Loss <strong>of</strong> skn-1 decreased resistance to the pathogens, whereas<br />

overexpression <strong>of</strong> SKN-1 resulted in enhanced survival, suggesting a protective role for SKN-<br />

1 during infection. Overall, SKN-1 is activated by the p38 MAPK signaling axis in response<br />

to ROS generated by Ce-Duox1 during infection in the intestinal lining <strong>of</strong> C. elegans. The<br />

aim <strong>of</strong> the current study is to elucidate the regulation <strong>of</strong> Ce-Duox1 in response to pathogenic<br />

bacteria. Using RNA-mediated interference (RNAi) we have identified several components <strong>of</strong><br />

signal transduction cascades that are involved in the regulation <strong>of</strong> BLI-3/Ce-Duox1 activity in<br />

response to E. faecalis.<br />

Contact: ransome.vanderhoeven@uth.tmc.edu<br />

Lab: Garsin<br />

156<br />

Poster Topic: Pathogenesis


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

TRPM Channels are Required for Clozapine’s Effects in C. elegans<br />

Xin Wang, Taixiang Saur, Bruce Cohen, Edgar(Ned) Buttner<br />

McLean Hospital & Harvard Medical School, Belmont MA<br />

Clozapine is the most effective drug for refractory schizophrenia, but its widespread use<br />

has been limited due to severe adverse effects, like agranulocytosis, seizures, tachycardia,<br />

and weight gain. The mechanisms <strong>of</strong> clozapine’s therapeutic and side effects remain unknown.<br />

Clozapine can cause larval arrest and lethality in C. elegans. We took advantage <strong>of</strong> these<br />

phenotypes by performing a genome-wide feeding RNAi screen for suppressors <strong>of</strong> clozapineinduced<br />

larval arrest (Scla). The screen yielded 42 suppressors, one <strong>of</strong> which is gtl-2. GTL-2<br />

is orthologous to the human transient receptor potential cation channel subfamily M member 3<br />

with 38% sequence identity. We validated the RNAi result by confirming the Scla phenotype in<br />

a partial loss-<strong>of</strong>-function strain, gtl-2(n2618). The Scla phenotype was not observed in two other<br />

TRPM loss-<strong>of</strong>-function mutants, namely gtl-1(ok375) and gon-2(q362). However, gtl-1(ok375),<br />

gon-2(q362), gtl-1(dx171);gtl-2(tm1463) and gon-2(q388);gtl-2(tm1463) suppress clozapineinduced<br />

lethality, while gtl-2 mutants do not. GTL-2 is expressed in the pharynx and excretory<br />

cell and regulates magnesium levels in C. elegans. GTL-1 and GON-2 also regulate Mg 2+ levels<br />

by taking up Mg 2+ in the intestinal cells. Inactivation <strong>of</strong> GTL-2 causes hypermagnesemia[1],<br />

and gtl-1;gon-2 and gtl-1;gtl-2 double mutants show hypomagnesemia[2]. Suppression <strong>of</strong><br />

clozapine-induced lethality in gtl-1 and gon-2 animals could be due to hypomagnesemia.<br />

1. Teramoto T, et al,. PLoS One, 2010. 5(3): p. e9589.<br />

2. Teramoto T, et al,. Cell Metab, 2005 1(5): p. 343-54.<br />

Contact: xwang@mclean.harvard.edu<br />

Lab: Buttner<br />

Poster Topic: Pathogenesis<br />

157


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

QTL Mapping <strong>of</strong> Differential Susceptibility to Bacteria in<br />

Caenorhabditis elegans<br />

Ziyi Wang 1 , Michael Herman 1 , Basten Snoek 2 , Jan Kammenga 2<br />

1 Kansas State <strong>University</strong>, Manhattan (KS), USA, 2 Wageningen <strong>University</strong>,<br />

Wageningen, The Netherlands<br />

C. elegans is a free-living nematode that naturally consume bacteria as food source.<br />

However, they face a challenge in that some dietary bacteria could also be pathogenic. The<br />

bacterial environments C. elegans naturally encounter are unlikely to be homogeneous; in<br />

addition, no evidence suggests the animals can selectively ingest certain bacteria from the<br />

environment. Therefore, as the animals feed on heterogeneous food sources, they have to<br />

simultaneously defend against the potential pathogens. Escherichia coli OP50, for example, the<br />

standard laboratory food for C. elegans, is generally considered as non-pathogenic. However,<br />

a natural isolate <strong>of</strong> the bacterium Stenotrophomonas maltophilia JCMS was surprisingly<br />

pathogenic to C. elegans. S. maltophilia is a ubiquitous bacterium that can cause nosocomial<br />

infections, especially in immune-compromised individuals, and has been suggested to be<br />

an emerging human pathogen. We scored adult survivorship <strong>of</strong> two C. elegans isolates, N2<br />

(Bristol, England) and CB4856 (Hawaii), exposed to E. coli OP50 or S. maltophilia JCMS,<br />

as an indirect measurement <strong>of</strong> pathogenicity <strong>of</strong> the bacteria. The results demonstrated adult<br />

survivorship in different bacterial environment is a complex trait that is determined by both<br />

genetic and environmental factors, as well as genotype by environment (GxE) interactions.<br />

In order to identify the underlying genetic basis, we mapped quantitative trait loci (QTL) in a<br />

N2xCB4856 recombinant inbred panel for differential susceptibility between E. coli OP50 and S.<br />

maltophilia JCMS. We have identified two QTL, one on LGI and the other on LGX. Focusing on<br />

the LGX QTL first, we used introgression lines (ILs) that contain distinct lengths <strong>of</strong> chromosome<br />

X from CB4856 in an otherwise N2 genomic background to confirm the position <strong>of</strong> this QTL.<br />

We have detected a robust GxE effect in one IL that carries an introgression overlapped with<br />

LGX QTL. Our results clearly show a genetic basis for variation in C. elegans susceptibility to<br />

bacteria. However, further investigation is needed to dissect both QTL and pinpoint the causal<br />

loci or the genes.<br />

Contact: ziyiwang@ksu.edu<br />

Lab: Herman<br />

158<br />

Poster Topic: Pathogenesis


Contact: jordan.ward@ucsf.edu<br />

Lab: Yamamoto<br />

Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

C. elegans Acyl-CoA synthetase-3 And The Nuclear Receptor NHR-25<br />

Promote Epidermal Integrity And Resistance To Pathogens.<br />

Jordan Ward2 , Carole Couillault1 , Brendan Mullaney2 , Teresita Bernal2 , Marc Van Gilst3 ,<br />

Kaveh Ashrafi2 , Jonathan Ewbank1 , Keith Yamamoto2 1 2 3 CIML, Marseilles, France, <strong>University</strong> <strong>of</strong> California, San Francisco, USA, Fred<br />

Hutchinson Cancer Research Center, Seattle, USA<br />

We have previously demonstrated that the acyl-CoA synthetase, acs-3, regulates the<br />

nuclear hormone receptor, nhr-25, to promote an endocrine program <strong>of</strong> lipid uptake and<br />

synthesis(1). Given that NHR-25 is a transcription factor, we performed microarray analysis to<br />

identify regulated genes promoting this metabolic program. Much to our surprise, despite the<br />

antagonistic genetic relationship between acs-3 and nhr-25, there were virtually no differentially<br />

regulated genes discovered by the microarrays. Rather, loss <strong>of</strong> either acs-3 or nhr-25 activity<br />

leads to very similar transcriptional pr<strong>of</strong>iles following EASE and DAVID analysis, with significant<br />

upregulation <strong>of</strong> genes involved in cuticle formation and pathogen response. Elevated pathogen<br />

response gene expression can be caused by osmotic stress resistance phenotypes and<br />

cuticular damage. acs-3 mutants have: i) a subtle resistance to acute hyperosmotic stress;<br />

ii) a partially penetrant sensitivity to hypo-osmotic stress; and iii) a severe cuticular barrier<br />

defect, as exhibited by elevated staining with the cuticle impermeable Hoechst 33358 dye in<br />

tail nuclei. nhr-25 mutants showed essentially wild-type responses in all <strong>of</strong> these assays. We<br />

then performed sensitivity assays to the pathogens Drechmeria coniospora and Pseudomonas<br />

aeruginosa to test whether the compromised epidermal barrier affects resistance to pathogens.<br />

Loss <strong>of</strong> acs-3 activity causes hypersensitivity to Drechmeria, while nhr-25 mutants are sensitive<br />

to Pseudomonas. Intriguingly, acs-3 mutation suppresses the sensitivity <strong>of</strong> nhr-25 mutants to<br />

Pseudomonas. Together, these results highlight a link between the ACS-3-NHR-25 endocrine<br />

program, the cuticle barrier and resistance to pathogens.<br />

1. B. C. Mullaney et al., Regulation <strong>of</strong> C. elegans fat uptake and storage by acyl-CoA synthase-3 is dependent<br />

on NR5A family nuclear hormone receptor nhr-25, Cell Metab 12, 398–410 (2010).<br />

Poster Topic: Pathogenesis<br />

159


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

Genetic Mechanisms <strong>of</strong> Innate Immune Response: The interaction<br />

between Caenorhabditis elegans and the opportunistic pathogen<br />

Stenotrophomonas maltophilia<br />

Corin White1 , Vinod Mony1 , Brian Darby2 , Michael Herman 1<br />

1 2 Kansas State <strong>University</strong>, Manhattan, KS, USA, <strong>University</strong> <strong>of</strong> North Dakota, Grand<br />

Forks, ND, USA<br />

Stenotrophomonas maltophilia is a ubiquitous aerobic gram-negative bacterium that can<br />

cause nosocomial and community-acquired infections. S. maltophilia isolates, including clinical<br />

K279a, have been shown to have nematocidal activities. In the course <strong>of</strong> our studies, we<br />

discovered a pathogenic interaction between Caenorhabditis elegans and a local S. maltophilia<br />

environmental isolate,JCMS. S. maltophilia strains reduce C. elegans lifespan, which is used<br />

as an indicator <strong>of</strong> bacterial pathogenesis. Based on several analyses, we find JCMS to be<br />

more virulent than the reference environmental isolate R551-3 and clinical isolate K279a. In<br />

addition, liveJCMS accumulates in the intestine significantly more than the standard laboratory<br />

C. elegans food source E. coli OP50. We have also found that both heat and UV-killed JCMS<br />

kills C. elegans less effectively than live bacteria, suggesting that the accumulation <strong>of</strong> living<br />

bacteria contributes significantly to JCMS virulence. Several innate immune pathways that<br />

serve to protect C. elegans from various pathogenic bacteria have been discovered, including<br />

the p38 MAP kinase and Sma/MabTGFβ-related pathways. Mutants that disrupt numerous<br />

components <strong>of</strong> these pathways are hypersensitive to both JCMS and OP50, suggesting that<br />

the functions <strong>of</strong> these genes are conserved in non-specific bacterial response. Surprisingly,<br />

ins-7/insulin-like ligand or daf-2/insulin receptor mutants displayed shortened lifespans on S.<br />

maltophilia JCMS. This observation is striking as daf-2 mutants are long lived on most other<br />

bacterial pathogens. We are currently using a forward genetic approach to identify candidate<br />

genes that may explain this S. maltophilia JCMS specific evasion <strong>of</strong> the DAF-2/16 pathway.<br />

Over 1350 ethyl methane sulfonate (EMS) induced mutant lines have been screened for hypersusceptibility<br />

or resistance to JCMS and we have identified both types <strong>of</strong> mutants. Future work<br />

includes the confirmation <strong>of</strong> these phenotypes and subsequent fine gene mapping. In addition<br />

to further understanding the interaction <strong>of</strong> C. eleganswith a novel pathogen, S. maltophilia,<br />

our studies might also provide insight on the mechanisms <strong>of</strong> adaptation C. elegans employs<br />

for diverse bacterial environments.<br />

Contact: cwhite21@k-state.edu<br />

Lab: Herman<br />

160<br />

Poster Topic: Pathogenesis


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

Characterization <strong>of</strong> the Effects <strong>of</strong> Naturally Isolated Salmonella<br />

enterica Strains on Caenorhabditis elegans<br />

Amanda Wollenberg 1 , Anna Maria Alves 1 , Michael McClelland 2 , Javier Irazoqui 1<br />

1 Massachusetts General Hospital, Boston, (MA), USA, 2 San Diego Institute <strong>of</strong><br />

Biological Research, San Diego, (CA), USA<br />

Salmonella enterica is a gastrointestinal pathogen that is responsible for both many cases<br />

<strong>of</strong> food-borne illness and enteric (typhoid) fever. We are studying the immune response <strong>of</strong> C.<br />

elegans to a range <strong>of</strong> Salmonella subspecies and serovars. In particular, we are studying a<br />

set <strong>of</strong> 66 isolates whose genomes have been fully sequenced. These isolates include many<br />

serovars within the well-studied S. enterica subspecies I, whose members are <strong>of</strong>ten specialized<br />

for vertebrate hosts (e.g. paratyphi), but the set also includes strains from the other five S.<br />

enterica subspecies. Here, we present our preliminary analysis <strong>of</strong> pathogenicity and persistence<br />

in the C. elegans model and discuss directions for future research.<br />

Contact: acwollenberg@gmail.com<br />

Lab: Irazoqui<br />

Poster Topic: Pathogenesis<br />

161


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

Analysis Of mi-RNAS, Target Prediction Algorithms And Databases In<br />

C.elegans<br />

Hema Kasisomayajula, Frnklyn Bolander<br />

<strong>University</strong> <strong>of</strong> South Carolina<br />

Current methods for target prediction for mi-RNA in C.elegans include complementarity,<br />

thermodynamic approaches and support vector based methods.<br />

The most valid evidence comes from wetlab procedures, but the information from the<br />

conservation <strong>of</strong> target sequence is not used in current method.<br />

This study hopes to address the need for a new database that takes all the evidence into<br />

account and pairs information from in-silico predictions with literature mining matches that use<br />

experimental evidence into account, to rate confidence levels in prediction.<br />

Contact: HEMA090A@gmail.COM<br />

Lab: Bolander<br />

162<br />

Poster Topic: Small RNAs


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

C. elegans rrf-1 Mutations Maintain RNAi Efficiency in the Soma in<br />

Addition to the Germline<br />

Caroline Kumsta, Malene Hansen<br />

Sanford-Burnham Medical Research Institute, La Jolla, CA, USA<br />

Gene inactivation through RNA interference (RNAi) has proven to be a valuable tool for<br />

studying gene function in C. elegans. When combined with tissue-specific gene inactivation<br />

methods RNAi has the potential to shed light on the function <strong>of</strong> a gene in distinct tissues. In this<br />

study we characterized C. elegans rrf-1 mutants to determine their ability to process RNAi in<br />

various tissues. These mutants have been widely used in RNAi studies to assess the function<br />

<strong>of</strong> genes specifically in the C. elegans germline.<br />

Upon closer analysis we found that two rrf-1 mutants carrying different loss-<strong>of</strong>-function alleles<br />

were capable <strong>of</strong> processing RNAi targeting several somatically expressed genes. Specifically,<br />

we observed that the intestine was able to process RNAi triggers efficiently, whereas cells in<br />

the hypodermis showed partial susceptibility to RNAi in rrf-1 mutants. Other somatic tissues<br />

in rrf-1 mutants, such as the muscles and the somatic gonad, appeared resistant to RNAi.<br />

In addition to these observations, we found that the rrf-1 mutation induced the expression <strong>of</strong><br />

several transgenic arrays, including the FOXO transcription factor DAF-16. Unexpectedly, rrf-1<br />

mutants showed increased endogenous expression <strong>of</strong> the DAF-16 target gene sod-3; however,<br />

the lifespan and thermo-tolerance <strong>of</strong> rrf-1 mutants were similar to those <strong>of</strong> wild-type animals.<br />

In sum, our data show that rrf-1 mutants display several phenotypes not previously<br />

appreciated, including broader tissue-specific RNAi-processing capabilities, and our results<br />

underscore the need for careful characterization <strong>of</strong> tissue-specific RNAi tools.<br />

Contact: ckumsta@sbmri.org<br />

Lab: Hansen<br />

Poster Topic: Small RNAs<br />

163


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

Genetic requirements for viRNAs-mediated antiviral immunity in C.<br />

elegans<br />

Jeremie Le Pen, Alyson Ashe, Leonard Goldstein, Eric Miska<br />

Gurdon Institute, <strong>University</strong> <strong>of</strong> Cambridge, Cambridge, United Kingdom<br />

The Orsay virus is a natural pathogen <strong>of</strong> C. elegans (Felix M-A, Ashe A, Piffaretti J, et<br />

al., 2011). It can enter and replicate in the nematode’s intestinal cells in normal solid culture<br />

conditions, and it exhibits horizontal transmission only. This virus, which contains two positive<br />

single-strand RNA molecules, is distantly related to viruses in the family Nodaviridae. It provides<br />

a unique material to study antiviral immunity in C. elegans.<br />

The virus-derived small interfering RNAs (viRNAs) play a key role in the resistance to the<br />

Orsay virus in the N2 C. elegans strain. However, the strain JU1580, which is a natural host <strong>of</strong><br />

the Orsay virus, fails to produce a robust viRNA response. This correlates with an increased<br />

sensitivity to the virus <strong>of</strong> the strain JU1580, when compared to N2. We are currently using<br />

the Orsay virus to investigate the genetic requirements for viRNA-mediated immunity using<br />

a quantitative trait locus (QTL) approach as well as a candidate gene approach (also see the<br />

abstract from our collaborators, Tony Belicard & Marie-Anne Felix, who measured the viral<br />

sensitivity <strong>of</strong> 97 natural isolates <strong>of</strong> C. elegans). We aim to enhance the anti-viral immunity <strong>of</strong><br />

JU1580 animals by rescuing key components <strong>of</strong> the viRNAs pathway.<br />

Moreover, recent findings using a transgene <strong>of</strong> the Flock House virus, a fly pathogen,<br />

integrated in the C. elegans genome suggested that viRNAs can be transmitted across<br />

generations in a template-independent manner, leading to a form <strong>of</strong> transgenerational<br />

vaccination (Rechavi O, Minevich G, Hobert O, 2011). We are using the Orsay virus as a model<br />

to question the possibility <strong>of</strong> an inheritance <strong>of</strong> viRNAs in nature.<br />

Contact: jl572@cam.ac.uk<br />

Lab: Miska<br />

164<br />

Poster Topic: Small RNAs


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

Unifying Models <strong>of</strong> Biological Timers: A Molting Cycle Approach<br />

Gabriela Monsalve, Alison Frand<br />

<strong>University</strong> <strong>of</strong> California-Los Angeles, Los Angeles, CA, USA<br />

The temporal coordination <strong>of</strong> reiterative and sequential processes is critical for animal<br />

development. The heterochronic gene network programs the successive temporal fates <strong>of</strong><br />

epithelial stem cells (seam cells). These cells undergo asymmetric divisions early in every<br />

larval stage, and yet contribute to the synthesis <strong>of</strong> new cuticles late in every larval stage,<br />

as part <strong>of</strong> the molting cycle. Relatively little is known about the mechanisms that link the respecification<br />

<strong>of</strong> seam cell fates with the four molts characteristic <strong>of</strong> C. elegans development.<br />

We recently reported that cyclical and stage-specific actions <strong>of</strong> LIN-42/PERIOD coordinate the<br />

temporal re-specification <strong>of</strong> epidermal stem cells and related cell divisions with rapid molts.<br />

Using bioinformatic approaches, we have uncovered additional evidence that interconnected<br />

regulatory interactions among LIN-42, the conserved nuclear hormone receptors NHR-23<br />

and NHR-25, and the let-7 family <strong>of</strong> microRNAs together compose a novel developmental<br />

oscillator that drives larval molting cycles. This timer likely couples the periodic molts with<br />

progressive development <strong>of</strong> the epidermis, as both lin-42 and let-7 also function in the<br />

heterochronic network to program the quality <strong>of</strong> sequential life stages and the expression <strong>of</strong><br />

additional stage-specific heterochronic genes. The discovery that LIN-42 regulates both the<br />

sleep-like behavior <strong>of</strong> lethargus and epidermal stem cell dynamics further supports the model<br />

<strong>of</strong> functional conservation between LIN-42 and mammalian PERIOD proteins. The molting<br />

timer may therefore represent an ancient paradigm for integrating rhythmic and developmental<br />

processes. Further studies <strong>of</strong> the molting timer may uncover novel but conserved mechanisms<br />

by which PERIOD-based oscillators control rhythmic cellular processes in both developing<br />

and mature animals.<br />

Contact: gmonsalve@ucla.edu<br />

Lab: Frand<br />

Poster Topic: Small RNAs<br />

165


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

C. elegans PRG-1 and piRNAs silence target transcripts through a<br />

secondary 22G-siRNA pathway<br />

Alexandra Sapetschnig, Eva-Maria Weick, Marloes Bagijn, Leonard Goldstein, Amy<br />

Cording, Eric Miska<br />

Wellcome Trust/Cancer Research UK Gurdon Institute, Cambridge, United<br />

Kingdom<br />

The Piwi proteins <strong>of</strong> the Argonaute superfamily and their associated Piwi-interacting RNAs<br />

(piRNAs) are required for normal germline development in animals. In C. elegans, the Piwi<br />

protein PRG-1 associates with 21U-RNAs (the piRNAs <strong>of</strong> C. elegans) and is required for Tc3<br />

transposon silencing. prg-1 mutant animals display germline defects and a reduction in brood<br />

size. The molecular mechanism <strong>of</strong> PRG-1/21U-RNA-mediated silencing <strong>of</strong> target genes is<br />

currently not understood.<br />

To analyse piRNA-dependent gene silencing, we generated a transgenic C. elegans strain<br />

carrying a germline-expressed GFP reporter with a sequence antisense to an endogenous<br />

21U-RNA. The GFP transgene is silenced in a wild-type background and becomes de-silenced<br />

in prg-1 mutant animals. High-throughput sequencing revealed prg-1 dependent 22G-siRNAs<br />

(secondary endo-siRNAs) that are synthesised from the GFP transgene in close proximity<br />

to the target site. In a candidate gene approach, we identified additional factors required for<br />

GFP silencing and prg-1 dependent 22G-RNA synthesis including the dicer-related helicase<br />

drh-3 and genes <strong>of</strong> the mutator class like mut-16, mut-7 and mut-2/rde-3. We postulate that<br />

22G-siRNAs are the effector molecules generated downstream <strong>of</strong> a primary piRNA trigger.<br />

The gene silencing appears to be achieved by both transcriptional and post-transcriptional<br />

mechanisms. Since PRG-1 does not localise to the nucleus, transcriptional gene silencing<br />

might be achieved through a nuclear 22G-siRNA pathway. We have identified a nuclear<br />

“worm-specific” Argonaute family member HRDE-1/WAGO-9 that could act to silence piRNA<br />

targets through heterochromatin formation. We currently investigate a potential role <strong>of</strong><br />

heterochromatin-associated factors in the piRNA pathway by a candidate genetic approach<br />

and ChIP experiments.<br />

PRG-1 has a conserved catalytic centre (amino acid triad DDH) that is required for “slicing”<br />

(cleavage) <strong>of</strong> target mRNAs in other Argonaute proteins. To elucidate the role <strong>of</strong> the putative<br />

slicer activity in PRG-1 dependent gene silencing, we generated transgenic strains expressing<br />

wild-type or a slicer-dead mutant <strong>of</strong> PRG-1 in the germline. Both transgenes rescue the<br />

abnormal phenotype observed in prg-1 mutant animals indicating that slicer activity is not<br />

required for PRG-1 function.<br />

Contact: as878@cam.ac.uk<br />

Lab: Miska<br />

166<br />

Poster Topic: Small RNAs


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

MicroRNA Predictors <strong>of</strong> Longevity in C. elegans<br />

Zachary Pincus, Thalyana Smith-Vikos, Frank Slack<br />

Yale <strong>University</strong>, New Haven, CT, USA<br />

Neither genetic nor environmental factors fully account for variability in individual longevity:<br />

genetically identical invertebrates in homogenous environments <strong>of</strong>ten experience no less<br />

variability in lifespan than outbred human populations. The identification <strong>of</strong> early-life gene<br />

expression states that predict future longevity would suggest that lifespan is at least in part<br />

epigenetically determined. Such ‘’biomarkers <strong>of</strong> aging,’’ genetic or otherwise, nevertheless<br />

remain rare. In this work, we sought early-life differences in organismal robustness in<br />

unperturbed individuals and examined the utility <strong>of</strong> microRNAs, known regulators <strong>of</strong> lifespan,<br />

development, and robustness, as aging biomarkers. We analyzed biometrics <strong>of</strong> C. elegans<br />

observed throughout their lives using novel single-animal solid-media culture techniques.<br />

Additionally, we confirmed these results by rearing worms on individual plates, and mounting<br />

and recovering worms from slides for single-timepoint imaging at early-adulthood. Early-tomid–adulthood<br />

measures <strong>of</strong> homeostatic ability jointly predict 62% <strong>of</strong> longevity variability. We<br />

further identified three microRNAs in which early-adulthood expression patterns individually<br />

predict up to 47% <strong>of</strong> lifespan differences. Though expression <strong>of</strong> each increases throughout<br />

this time, miR-71 and miR-246 correlate with lifespan, while miR-239 anti-correlates. We also<br />

examined expression <strong>of</strong> these microRNAs simultaneously in the same worm to identify any<br />

potential synergistic effects. Two <strong>of</strong> these three microRNA ‘’biomarkers <strong>of</strong> aging’’ act upstream<br />

in insulin/IGF-1–like signaling (IIS) and other known longevity pathways; thus, we infer that<br />

these microRNAs not only report on but also likely determine longevity. Therefore, fluctuations<br />

in early-life IIS, due to variation in these microRNAs and from other causes, may determine<br />

individual lifespan.<br />

Contact: thalyana.smith-vikos@yale.edu<br />

Lab: Slack<br />

Poster Topic: Small RNAs<br />

167


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

A Genetic Suppressor Screen in Caenorhabditis elegans for Targets<br />

<strong>of</strong> Antipsychotic Drugs: Role <strong>of</strong> the Nicotinic Acetylcholine Receptor<br />

Homolog acr-7<br />

Taixiang Saur, Xin Wang, Limin Hao, Bruce Cohen, Edgar Buttner<br />

McLean Hospital, Belmont, MA<br />

We performed the first genetic screen for antipsychotic drug (APD) targets in an animal, a<br />

genome-wide RNAi screen for Suppressors <strong>of</strong> Clozapine-induced Larval Arrest (scla genes).<br />

Using this approach, we identified acr-7, which encodes a homolog <strong>of</strong> the human a-like<br />

nicotinic acetylcholine receptors (nAChRs), as a potential APD target. We validated our RNAi<br />

result by showing that the acr-7(tm863) deletion suppresses clozapine-induced developmental<br />

delay. A full-length translational Pacr-7::acr-7::gfp fusion construct rescues this phenotype. We<br />

investigated the specificity <strong>of</strong> acr-7(lf) suppression by testing 21 other C.e. nAChR subunits and<br />

found that only acr-7(lf) suppresses clozapine-induced developmental delay. Mutations in cha-<br />

1 and unc-17 are not suppressors, suggesting that clozapine does not cause developmental<br />

delay by stimulating Ach release.<br />

Both translational and transcriptional acr-7::gfp fusion constructs are strongly expressed<br />

in the pharynx. Clozapine inhibits pharyngeal pumping, and acr-7(lf) suppresses this effect.<br />

Thus, one mechanism by which clozapine delays development is likely by inhibiting pharyngeal<br />

pumping. The effects <strong>of</strong> clozapine on pharyngeal pumping and development are mimicked<br />

by the nAChR agonists nicotine and levamisole and blocked by the nAChR antagonists DHβ.<br />

Our results are consistent with the notion that clozapine inhibits pharyngeal pumping, in part,<br />

by activating ACR-7 receptors in the pharynx.<br />

Effects <strong>of</strong> APDs on the spectrum <strong>of</strong> nAChR subunit combinations have not been tested in<br />

mammals. Therefore, whether APDs can activate one or more these receptors is an important<br />

question. α7-nAChR agonists are currently in development for the treatment <strong>of</strong> psychosis, but<br />

no known APDs have been demonstrated to act by this mechanism. α-like nAChR signaling is a<br />

mechanism through which clozapine may produce its therapeutic and/or toxic effects in human.<br />

Contact: txu@mclean.harvard.edu<br />

Lab: Buttner<br />

168<br />

Poster Topic: Small RNAs


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

Characterizing in-host population dynamics <strong>of</strong> pathogenic bacteria<br />

during intestinal infection <strong>of</strong> C. elegans<br />

Ronen Kopito, Andrzej Nowojewski, Erel Levine<br />

Department <strong>of</strong> Physics and FAS Center for Systems Biology, Harvard <strong>University</strong><br />

Host-pathogen interactions drive the progression and outcome <strong>of</strong> bacterial infection, and are<br />

believed to be key determinants in evolution <strong>of</strong> virulence and resistance to infection. Intestinal<br />

infection <strong>of</strong> worms by pathogenic bacteria provide and excellent opportunity to characterize<br />

these interactions and link them to the development and outcome <strong>of</strong> infection. To this end,<br />

we developed tools that allow long-time high-resolution imaging <strong>of</strong> the infection process in<br />

individual worms. We track the spatiotemporal proliferation <strong>of</strong> bacteria in a worm intestine from<br />

the onset <strong>of</strong> infection until the death <strong>of</strong> the worm. At the same time, we follow the expression<br />

<strong>of</strong> relevant genes in both bacteria and worms. Physiological cues are monitored to evaluate<br />

the progression <strong>of</strong> the infection process. Thus, we aim to compose a complete timeline <strong>of</strong> the<br />

disease <strong>of</strong> each individual worm, and to use computational tools to deduce interactions and<br />

causality. We demonstrate the approach and our preliminary results focusing on “slow killing”<br />

<strong>of</strong> C. elegans by P. aeruginosa PA14. We find that bacterial population dynamics in the worm<br />

intestine is highly unstable even late in the infection process, identify cumulative bacterial<br />

sub-populations that correlate with infection outcome and immune response, and show how<br />

individual behavior contributes to variable outcome.<br />

Contact: rkopito@gmail.com<br />

Lab: Levine<br />

Poster Topic: Pathogenesis<br />

169


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

Dynamics <strong>of</strong> stress response across the worm unveils cross-tissue<br />

interactions<br />

Ronen Kopito, Christian Anderson, Erel Levine<br />

Department <strong>of</strong> Physics and FAS Center for Systems Biology, Harvard <strong>University</strong><br />

Small heat shock proteins are mediators <strong>of</strong> stress response in animals. Accumulation <strong>of</strong><br />

these proteins is linked with resilience to stress as well as with aging and longevity. In addition,<br />

heat-shock promoters are popular experimental tools, used to induce gene expression. Even<br />

under optimized conditions worms demonstrate a highly inconsistent accumulation <strong>of</strong> heat<br />

shock proteins in response to stress. We carried out a detailed quantitative study <strong>of</strong> the<br />

accumulation <strong>of</strong> Hsp-16.2 in response to stress signals, using micr<strong>of</strong>luidic tools developed<br />

for imaging gene expression at high spatiotemporal resolution over long periods <strong>of</strong> time, and<br />

analysis approaches borrowed from statistical physics. We find that stress response occurs at<br />

two time scales – short (about 2-3 hours after the onset <strong>of</strong> stress) and long (around 12 hours),<br />

followed by slow decay. We show how the total level <strong>of</strong> Hsp-16.2 across the worm, used as<br />

a proxy for gene expression in many studies, is in fact unrepresentative <strong>of</strong> the real complex<br />

dynamics. Our data suggests that a major contribution to the worm-to-worm variability stems<br />

from the decay <strong>of</strong> the stress response signal, implying that heat shock promoters should not<br />

be used to induce gene expression for long periods <strong>of</strong> time. Correlations and temporal ordering<br />

in the response dynamics <strong>of</strong> different tissues allow us to formulate hypotheses about crosstissue<br />

interactions. We find similar kinetic features in other stress response pathways, and<br />

trace them to molecular mediators <strong>of</strong> tissue-tissue interactions.<br />

Contact: rkopito@gmail.com<br />

Lab: Levine<br />

170<br />

Poster Topic: Stress


Contact: mmondoux@holycross.edu<br />

Lab: Mondoux<br />

Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

Glucose Stress Regulates Insulin Signaling, Fertility, and Mating in C.<br />

elegans<br />

Amanda K. Engstrom1 , Marjorie R. Liggett1 , John A. Hanover2 , Michael W. Krause2 ,<br />

Michelle A. Mondoux1 1 2 College <strong>of</strong> the Holy Cross, Worcester, MA, National Institutes <strong>of</strong> Health,<br />

Bethesda, MD<br />

Energy homeostasis occupies a central position in biology, as it regulates gene expression,<br />

signal transduction, cellular survival, fertility, and lifespan. However, the molecular and genetic<br />

responses to excess nutrients are poorly understood. Using C. elegans as a genetic model<br />

to study the pathways that respond to nutrient stress, we have identified a number <strong>of</strong> insulindependent<br />

processes that are sensitive to glucose stress, including fertility, reproductive timing,<br />

and dauer formation. Each <strong>of</strong> these has a different threshold for sensitivity to glucose excess.<br />

We have further characterized the C. elegans response to excess glucose in both<br />

hermaphrodites and males. We find that the reduction in fertility and delayed reproductive<br />

timing are separable phenotypes dependent on glucose exposure during adulthood and<br />

development, respectively. Glucose stress during development results in a 1-2 day delay in<br />

reproductive timing but has no effect on adult fertility. Likewise, glucose stress during adulthood<br />

reduces fertility with no change in the reproductive pr<strong>of</strong>ile. Glucose stress also reduces the<br />

ability <strong>of</strong> hermaphrodites and males to mate, as evidenced by reduced brood sizes and fewer<br />

male cross-progeny.<br />

In order to identify the pathways that respond to nutrient stress, we are screening a C.<br />

elegans RNAi library to identify factors that suppress insulin signaling specifically in the<br />

presence <strong>of</strong> glucose stress. This screen will provide insight into how cells respond to nutrient<br />

stress when insulin signaling is compromised.<br />

Poster Topic: Metabolism<br />

171


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

172<br />

NOTES


A<br />

Aebi, Markus................................ 42, 154<br />

Agellon, Luis B..................................... 22<br />

Ahn, Jeong-Min ................................... 99<br />

Ahn, Soungyub .................................... 94<br />

Ajredini, Ramadan ............................. 153<br />

Alla, Ramani ........................................ 51<br />

Alves, Anna Maria........................ 14, 161<br />

Andersen, Erik C ............................... 133<br />

Anderson, Christian ........................... 170<br />

Antebi, Adam ................................. 26, 38<br />

Anthony, Hannah L ............................ 150<br />

Apfeld, Javier ....................................... 37<br />

Aroian, Raffi ....................................... 152<br />

Arsenovic, Paul T ................................ 96<br />

Ashe, Alyson ...................................... 164<br />

Ashraf, Jasmine M ............................... 71<br />

Ashrafi, Kaveh ....................... 17, 53, 159<br />

Au, Catherine......................................... 2<br />

Avery, Leon .................................... 80, 88<br />

Awoyinka, Olufadakemi ..................... 145<br />

Ayyadevara, Srinivas ........................... 51<br />

B<br />

Bagijn, Marloes P .............................. 166<br />

Baker, Keith ....................................... 121<br />

Bakowski, Malina A .................... 132, 142<br />

Baldwin, James G.............................. 151<br />

Balla, Keir M .............................. 133, 139<br />

Barr, Angela ....................................... 103<br />

Barra, Donatella................................. 147<br />

Baugh, L Ryan ......................... 5, 70, 125<br />

Baugh, Ryan L ..................................... 27<br />

Baugh, Ryan ........................................ 87<br />

Baumeister, Ralf .......................... 91, 123<br />

Belicard, Tony .............................. 31, 134<br />

Benzing, Thomas ................................. 57<br />

Ben-Zvi, Anat ......................................... 4<br />

Bernal, Teresita .................................. 159<br />

Bethke, Axel......................................... 38<br />

Bhanot, Gyan....................................... 74<br />

Author Index<br />

Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

AUTHOR INDEX<br />

Bharill, Puneet ..................................... 57<br />

Bhatia, Aatish....................................... 74<br />

Blackwell, Leah.................................... 95<br />

Blackwell, T. Keith ................................ 11<br />

Bleuler-Martinez, Silvia ...................... 154<br />

Bloss, Tim A ......................................... 96<br />

Boehler, Christopher J ......................... 78<br />

Bolander, Frnklyn F ........................... 162<br />

Bose, Neelanjan .................................. 38<br />

Bossard, Carine .................................. 18<br />

Bouagnon, Aude .................................. 17<br />

Boxem, Mike ....................................... 34<br />

Boyd, Lynn ......................................... 127<br />

Bracciale, Maria P ............................. 131<br />

Braeckman, Bart P .............. 8, 30, 52, 54<br />

Branicky, Robyn ................................... 22<br />

Bratanich, Ana ................................... 135<br />

Brewer, Heather M................................. 8<br />

Brey, Christopher W ............................ 79<br />

Broday, Limor ..................................... 34<br />

Broggi, Alessandra ............................ 131<br />

Brul, Stanley ...................................... 128<br />

Butschi, Alex ................................ 42, 154<br />

Buttner, Edgar...................... 82, 157, 168<br />

Button, Emma L ................................. 136<br />

C<br />

Cabello, Juan..................................... 144<br />

Cabreiro, Filipe ...................................... 2<br />

Cabunoc, Abigail.................................. 45<br />

Camp II, David G ................................... 8<br />

Carr, Christopher E .............................. 29<br />

Carrano, Andrea ................................. 18<br />

Castelein, Natascha ............................ 52<br />

Cattie, Douglas J ................................. 97<br />

Chang, Sandy .................................... 152<br />

Chen, Albert T ................................ 41, 53<br />

Chen, Chang-Shi .............................. 137<br />

Chen, Po-Han ..................................... 98<br />

Chen, Show-Li ..................................... 98<br />

Chen, Yutao ......................................... 27<br />

Chiu, Hao-Chieh ............................... 137<br />

173


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

Choe, Keith P .............................113, 114<br />

Choi, Jinhee......................................... 99<br />

Chou, Ting-Chen .............................. 137<br />

Clark, Laura C ................................... 138<br />

Coburn, Cassandra ............................. 30<br />

Cohen, Bruce....................... 82, 157, 168<br />

Colleluori, Vaughn D ............................ 96<br />

Cook, James M.................................. 145<br />

Cording, Amy ..................................... 166<br />

Couillault, Carole ............................... 159<br />

Crook, Helen M.................................. 129<br />

Crossgrove, Kirsten ........................... 100<br />

Crowder, C. Michael ............ 43, 116, 126<br />

Crowder, Michael ............................... 110<br />

Cruz, Melissa R ................................. 156<br />

Cunningham, Katherine ....................... 17<br />

Cuomo, Christina A ............................ 132<br />

D<br />

Darby, Brian ...................................... 160<br />

Davidson, Andrew........................ 95, 101<br />

De Bellis, Giovanni ............................ 131<br />

de Boer, Richard ................................ 128<br />

De Haes, Wouter ................................. 54<br />

de la Cruz, Norie.................................. 45<br />

de Lencastre, Alexandre ...................... 10<br />

Demoinet, Emilie ................................. 19<br />

Dennis, Jennifer C ............................. 102<br />

Denzel, Martin S .................................. 26<br />

Deonarine, Andrew ............................ 113<br />

Depuydt, Geert G .................................. 8<br />

Desjardins, Christopher A .................. 132<br />

Desjardins, David ............................... 22<br />

Devanapally, Sindhuja ......................... 44<br />

Dhondt, Ineke ........................................ 8<br />

Diederich, Ann-Kristin ......................... 64<br />

Dillin, Andrew ................................. 18, 47<br />

Dillman, Adler R ................................. 151<br />

Diogo, Jesica ..................................... 135<br />

Driscoll, Monica ....................... 32, 74, 76<br />

Dumas, Kathleen J .. 3, 38, 41, 53, 55, 63<br />

Dunbar, Tiffany L................................ 139<br />

Duong, Adrian ...................................... 45<br />

Duveau, Fabien ................................... 31<br />

174<br />

E<br />

Edelman, Theresa LB ........................ 103<br />

Edison, Arthur S................................. 153<br />

Engholm-Keller, Kasper ....................... 15<br />

Engstrom, Amanda K ........................ 171<br />

Eom, Hyun - Jeong .............................. 99<br />

Estes, Kathleen ................................... 33<br />

Ewbank, Jonathan J ...................... 1, 159<br />

F<br />

Fabretti, Francesca.............................. 57<br />

Færgeman, Nils J ................................ 15<br />

Fawcett, Emily M ............................... 104<br />

Felix, Marie-Anne ............ 9, 31, 134, 138<br />

Fierro-Gonzalez, Juan Carlos...... 59, 144<br />

Fitch, David.......................................... 61<br />

Flibotte, Stephane ........................... 3, 55<br />

Fonslow, Bryan ................................... 18<br />

Fontana, Walter ................................... 37<br />

Frand, Alison R .................................. 165<br />

Franz, Carl J .......................................... 9<br />

Fredens, Julius .................................... 15<br />

Frokjaer-Jensen, Christian ................ 103<br />

Fu, Donald ........................................... 49<br />

Fujii, Michihiko ................................... 130<br />

G<br />

Gaglia, Marta M ................................... 65<br />

Gallagher, Thomas L ........................... 80<br />

Garcia, Anastacia M .................. 105, 120<br />

Garcia, L Rene .................................... 93<br />

Garcia, Luis rene ................................... 7<br />

Gardner, Mona................................... 146<br />

Garland, Brenda ................................ 100<br />

Garsin, Danielle A ...................... 143, 156<br />

Gaugler, Randy.................................... 79<br />

Gelino, Sara......................................... 56<br />

Gems, David .................................... 2, 30<br />

Gharbi, Hakam .................................... 57<br />

Ghose, Piya ......................................... 74<br />

Gill, Matthew S .................................... 83<br />

Glover-Cutter, Kira M ........................... 11<br />

Go, Junhyeok ...................................... 58<br />

Goh, Grace YS .................................. 106<br />

Author Index


Goldstein, Leonard D................. 164, 166<br />

Golombek, Diego A .............................. 81<br />

Gonzalez-Barrios, Maria ...................... 59<br />

Goy, Jo M .................................... 60, 120<br />

Goya, Maria E...................................... 81<br />

Graciano, Thomas ............................. 146<br />

Gravato-Nobre, Maria Joao ....... 138, 140<br />

Greene, Nick D ...................................... 2<br />

Gross, Megan .................................... 145<br />

Guo, Chunfang .................. 41, 53, 63, 68<br />

Guo, Xiaoyan ......................................... 7<br />

H<br />

Haenen, Steven ................................... 54<br />

Hall, David ..................................... 16, 74<br />

Hanover, John A ................................ 171<br />

Hansen, Malene .................... 13, 56, 163<br />

Hao, Limin ................................... 82, 168<br />

Harris, Todd W ..................................... 45<br />

Harrison, Neale.................................... 83<br />

Hartley, Shannon ............................... 150<br />

Hartman, Phillip ................................. 130<br />

Hasegawa, Koichi ............................. 114<br />

Hashmi, Sanya .................................... 79<br />

Hashmi, Sarwar ................................... 79<br />

Haynes, Cole M ................................... 39<br />

Heimbucher, Thomas ......................... 18<br />

Hekimi, Siegfried ................................. 22<br />

Hendrix, Amber .................................... 85<br />

Hengartner, Michael O................. 42, 154<br />

Henis-Korenblit, Sivan ....................... 107<br />

Herman, Michael ....................... 158, 160<br />

Herrera, R Antonio ............................... 61<br />

Herndon, Laura A........................... 16, 74<br />

Hodgkin, Jonathan............. 138, 140, 148<br />

Horsman, Joseph ................................ 62<br />

Hou, Nicole S....................................... 84<br />

Hu, Patrick J ...... 3, 38, 41, 53, 55, 63, 68<br />

Hu, Queenie ...................................... 108<br />

Hu, Yan .............................................. 152<br />

Huang, Anne ........................................ 28<br />

Hubbard, E. Jane Albert ...................... 69<br />

Hunter, Tony ....................................... 18<br />

Hyun, Moonjung ................................ 109<br />

Author Index<br />

Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

I<br />

Ibanez-Ventoso, Carolina .................... 74<br />

Ihuegbu, Nnamdi ................................ 14<br />

Irazoqui, Javier E ......................... 14, 161<br />

Ishii, Naoaki ....................................... 130<br />

Ishii, Takamasa .................................. 130<br />

J<br />

Jablonski, Angela M........................... 141<br />

Jacobs, Rene....................................... 48<br />

Jakob, Ursula....................................... 64<br />

Jansen-Duerr, Pidder........................... 72<br />

Janssen, Tom ...................................... 90<br />

Jayamani, Elamparithi ....................... 144<br />

Jeong, Dae-Eun................................... 65<br />

Jevince, Angela ................................... 16<br />

Jevince, Angelina................................. 74<br />

Jiang, Yanfang ....................................... 9<br />

Jimenez-Hidalgo, Maria ..................... 144<br />

Jobson, Meghan ............................ 5, 125<br />

Jordan, James ..................................... 87<br />

Jorgensen, Erik M.............................. 103<br />

Jose, Antony M .................................... 44<br />

Judkins, Joshua C ............................... 38<br />

Judy, Meredith ..................................... 28<br />

K<br />

Kabir, M. Shahjahan .......................... 145<br />

Kaeberlein, Matt .................................. 12<br />

Kagias, Konstantinos ......................... 118<br />

Kalb, Robert G ................................... 141<br />

Kammenga, Jan ................................ 158<br />

Kang, Hae-Sung ................................ 155<br />

Kao, Aimee W ...................................... 28<br />

Kasisomayajula, Hema ..................... 162<br />

Kassa, Berhanu ................................... 52<br />

Kassim, Maher..................................... 45<br />

Kenyon, Cynthia J ....................... 28, 107<br />

Kim, Dennis H.................46, 97, 111, 149<br />

Kim, Euysoo ................................ 43, 110<br />

Kim, Jeongho....................................... 85<br />

Kiontke, Karin ...................................... 61<br />

Klang, Ida M ........................................ 50<br />

Knoefler, Daniela ................................. 64<br />

175


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

Koniczek, Martin .................................. 64<br />

Kopito, Ronen ............................ 169, 170<br />

Kotera, Ippei ........................................ 49<br />

Krause, Michael W ............................ 171<br />

Kruglyak, Leonid ................................ 133<br />

Kubiseski, Terry ................................. 108<br />

Kuenzler, Markus ............................... 154<br />

Kulalert, Warakorn ..............................111<br />

Kumsta, Caroline ............................... 163<br />

Kunzler, Markus ................................... 42<br />

Kuo, Cheng-Ju .................................. 137<br />

Kuroda, Eisuke .................................... 66<br />

Kurz, Cyril Leopold ............................ 144<br />

Kwong, Ada ....................................... 106<br />

L<br />

Ladage, Mary..................................... 120<br />

Land, Marianne.................................. 112<br />

Lapierre, Louis R ................................. 13<br />

Le Pen, Jeremie ................................ 164<br />

Le, Hai ................................................. 44<br />

LeBlanc, Marc-Andre ......................... 101<br />

Lee, Dongyeop .................................... 65<br />

Lee, Inhwan ......................................... 85<br />

Lee, Junho ..................................... 89, 94<br />

Lee, Kang-Mu ...................................... 58<br />

Lee, Kuang-Hui.................................... 40<br />

Lee, Seung-Jae ................................... 65<br />

Lee, Siu Sylvia ....................................... 6<br />

Lee, Soon-Young ................................. 67<br />

Leung, Chi K ...............................113, 114<br />

Leung, Kit-Yi .......................................... 2<br />

Levine, Erel................................ 169, 170<br />

Li, Liming ........................................... 119<br />

Liang, Jun .......................................... 115<br />

Liggett, Marjorie R ............................. 171<br />

Lin, Stephanie...................................... 11<br />

Lindblom, Tim H........................... 95, 101<br />

Lithgow, Gordon J................................ 50<br />

Little, Brent ........................................ 102<br />

Liu, Ju-Ling .......................................... 22<br />

Liu, Pingsheng ..................................... 86<br />

Liu, Shu ............................................. 123<br />

Liu, Zheng ........................................... 18<br />

Lo, Dara ............................................... 21<br />

176<br />

Lu, Kevin.............................................. 74<br />

Luallen, Robert J ............................... 142<br />

Luhachack, Lyly G ............................... 14<br />

Luo, Shijing .......................................... 71<br />

M<br />

Ma, Amy T ......................................... 132<br />

Mackowiak, Sebastian ....................... 118<br />

MacNeil, Lesley ................................. 108<br />

Maeda, Masatomo ............................... 73<br />

Mahanti, Parag .............................. 30, 38<br />

Malany, Siobhan ................................ 113<br />

Maldonado, Anthony T ......................... 96<br />

Mandel, Abraham ................................ 30<br />

Mangoni, Maria L ............................... 147<br />

Mantovani, Julie................................... 19<br />

Mao, Xianrong R........................ 116, 126<br />

Mark, Karla .......................................... 50<br />

Marra, Amanda ................................... 95<br />

Matthijssens, Filip ................................ 30<br />

McCallum, Katie C ..................... 143, 156<br />

McClelland, Michael .......................... 161<br />

McCulloch, Katherine A ..................... 103<br />

Meelkop, Ellen ..................................... 90<br />

Melentijevic, Ilija .................................. 74<br />

Melki, Ronald ..................................... 119<br />

Melo, Justine A .................................... 23<br />

Menzel, Ralph.................................... 117<br />

Micutkova, Lucia .................................. 72<br />

Middleton, June H.............................. 146<br />

Migliori, Maria L ................................... 81<br />

Miller, Amanda J ................................ 150<br />

Miller, Dana L ............................... 62, 104<br />

Miranda-Vizuete, Antonio ............ 59, 144<br />

Miska, Eric A .............................. 164, 166<br />

Miskowski, Jennifer A ........................ 145<br />

Mitani, Shohei .......................... 53, 63, 68<br />

Mondoux, Michelle A.......................... 171<br />

Moeller-Jensen, Jakob ........................ 15<br />

Moerman, Don ................................. 3, 55<br />

Monsalve, Gabriela C ........................ 165<br />

Monte, Aaron ..................................... 145<br />

Mony, Vinod ....................................... 160<br />

Moon, Hyungmin ................................. 89<br />

Moore, Brad T ...................................... 87<br />

Author Index


Morimoto, Richard I ..................... 75, 119<br />

Mueller, Roman Ulrich ......................... 57<br />

Mullaney, Brendan ............................. 159<br />

Mundo-Ocampo, Manuel ................... 151<br />

Munoz, Manuel .................................... 59<br />

Murphy, Coleen T .......................... 20, 71<br />

Myers, Edith M................................... 146<br />

Mylonakis, Eleftherios........................ 144<br />

N<br />

Na, Huimin ........................................... 86<br />

NAar, Anders ....................................... 48<br />

Naji, Haaris .......................................... 74<br />

Nakamura, Ayumi ................................ 28<br />

Naranjo-Galindo, Francisco Jose ...... 144<br />

Nehammer, Camilla .......................... 118<br />

Nguyen, Ken CQ ................................. 16<br />

Niemuth, Nicholas J............................. 64<br />

Nishikori, Kenji ............................... 66, 73<br />

Noiman, Liron ...................................... 17<br />

Nowojewski, Andrzej.......................... 169<br />

Nussbaum-Krammer, Carmen I ......... 119<br />

O<br />

Ogungbe, Ifedayo Victor ..................... 83<br />

Ohashi-Kobayashi, Ayako ............. 66, 73<br />

Olahova, Monika................................ 129<br />

Olivi, Massimiliano ............................. 147<br />

Onken, Brian........................................ 32<br />

O’Rourke, Delia ................................. 148<br />

Orphan, Victoria J .............................. 151<br />

Orton, Kai ........................................... 75<br />

P<br />

Padilla, Pamela A......... 60, 102, 105, 120<br />

Padmanabha, Divya .......................... 121<br />

Pagano, Daniel J ............................... 149<br />

Palleschi, Claudio ...................... 131, 147<br />

Pandey, Santosh ............................... 124<br />

Parashar, Archana ............................ 124<br />

Park, Donha................................... 21, 84<br />

Park, Kyung-Won ............................. 119<br />

Park, Sang-Kyu ................................... 67<br />

Park, Seul-Ki........................................ 67<br />

Author Index<br />

Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

Pasquinelli, Amy E ............................... 25<br />

Pedrajas, Jose Rafael ....................... 144<br />

Petyuk, Vladislav A ................................ 8<br />

Pincus, Zachary ................................. 167<br />

Pircher, Haymo .................................... 72<br />

Pocock, Roger .................................. 118<br />

Podolska, Agnieszka ........................ 118<br />

Pollok, Robert H .................................. 88<br />

Polzin, Andy ......................................... 68<br />

Powell, Jennifer R.............................. 150<br />

Powell, Megan ................................... 101<br />

Powell-C<strong>of</strong>fman, Jo Anne .................. 124<br />

Price, Milena ........................................ 50<br />

Price, Rebecca ................................. 148<br />

Q<br />

Qi, Wenjing .......................................... 91<br />

Qin, Zhao ............................................. 69<br />

R<br />

Rajewsky, Nikolaus ............................ 118<br />

Ravikumar, Snusha ............................. 44<br />

Reddy, Kirthi C ..................................... 97<br />

Renshaw, Hilary..................................... 9<br />

Richardson, Claire E............................ 97<br />

Riddle, Donald L .................................. 21<br />

Robinson, Joseph D .......................... 150<br />

Rodriguez, Pedro................................. 50<br />

Roessel Larsen, Martin........................ 15<br />

Rohlfing, Anne-Katrin......................... 122<br />

Romanowski, Andres ........................... 81<br />

Rongo, Christopher ............................. 74<br />

Rottiers, Veerle ................................... 48<br />

Rougvie, Ann E .................................. 103<br />

Roy, Richard .................................. 19, 92<br />

Rubin, Charles ................................... 112<br />

Runkel, Eva D.................................... 123<br />

Ruvkun, Gary................................. 23, 29<br />

Ryu, Eun-A .......................................... 65<br />

S<br />

Sackett, Peter .................................... 100<br />

Safavi, Arash ..................................... 152<br />

Safra, Modi ........................................ 107<br />

177


Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

Saldanha, Jenifer............................... 124<br />

Salehpour, Ali....................................... 40<br />

Sandr<strong>of</strong>, Moses ................................. 125<br />

Santarelli, Maria L .............................. 131<br />

Sapetschnig, Alexandra ..................... 166<br />

Sapir, Amir ................................... 34, 151<br />

Sarto, Maria S.................................... 131<br />

Saur, Taixiang ............................ 157, 168<br />

Savage-Dunn, Cathy ......................... 115<br />

Schermer, Bernhard ............................ 57<br />

Schindler, Adam J ................................ 70<br />

Scho<strong>of</strong>s, Liliane ............................. 54, 90<br />

Schroeder, Frank C ....................... 30, 38<br />

Schubert, Mario ................................. 154<br />

Schulze, Ekkehard............................. 123<br />

Schunck, Wolf-Hagen ........................ 117<br />

Scott, Barbara A................................. 126<br />

Sesar, Jillian L ................................... 152<br />

Shah, Leena ........................................ 74<br />

Shah, Mitalie ........................................ 76<br />

Shai, Nadav ........................................... 4<br />

Shanmugam, Nilesh .............................. 8<br />

Shapira, Michael .......................... 40, 155<br />

Shemesh, Netta ..................................... 4<br />

Sherwood, David R.............................. 70<br />

Shi, Cheng ........................................... 71<br />

Shi, Xiaoqi ........................................... 45<br />

Shih, Hung-Jen .................................... 41<br />

Shim, Jiwon ......................................... 89<br />

Shin, Jisun ........................................... 89<br />

Shiraishi, Hirohisa.......................... 66, 73<br />

Shmookler Reis, Robert J.................... 51<br />

Simonetta, Sergio H ............................ 81<br />

Simske, Jeff ......................................... 35<br />

Skibinski, Gregory A .......................... 127<br />

Slack, Frank J .............................. 10, 167<br />

Smelkinson, Margery G ............... 33, 139<br />

Smith, Michael ................................... 145<br />

Smith, Reuben ................................... 128<br />

Smith, Richard D ................................... 8<br />

Smith-Vikos, Thalyana ....................... 167<br />

Smolders, Arne ...................................... 8<br />

Snoek, Basten ................................... 158<br />

Soukas, Alexander A............................ 29<br />

Stein, Lincoln D ................................... 45<br />

178<br />

Steinberg, Christian EW .................... 117<br />

Sternberg, Paul W ....................... 34, 151<br />

Storm, Nadia J ..................................... 26<br />

Stormo, Gary D.................................... 14<br />

Stroud, Dave...................................... 148<br />

Stroustrup, Nicholas ............................ 37<br />

Stupp, Gregory S ............................... 153<br />

Stutz, Katrin ................................. 42, 154<br />

Su, Po-An ............................................ 49<br />

Suda, Hitoshi ..................................... 130<br />

Sun, Chun-Ling.................... 43, 110, 126<br />

Sunde, Roger A ................................... 78<br />

Suzuki, Hiroshi..................................... 49<br />

Swoboda, Peter ........................... 59, 144<br />

Syu, Wan-Jr ...................................... 137<br />

Szumowski, Suzannah ........................ 33<br />

T<br />

Taferner, Andrea .................................. 72<br />

Talwar, Amish....................................... 74<br />

Tanji, Takahiro ................................ 66, 73<br />

Taubert, Stefan ...................... 21, 84, 106<br />

Tavernarakis, Nektarios ....................... 72<br />

Taylor, Rebecca C ............................... 47<br />

Tazearslan, Cagdas ............................ 51<br />

Temmerman, Liesbet ........................... 90<br />

Thamsen, Maike .................................. 64<br />

Thijs Koorman, Thijs ........................... 34<br />

Toth, Marton......................................... 74<br />

Troemel, Emily R . 33, 132, 133, 139, 142<br />

Tsao, Yeou-Ping................................... 98<br />

Tsur, Assaf .......................................... 34<br />

Twumasi-Boateng, Kwame .......... 40, 155<br />

U<br />

Uccelletti, Daniela ...................... 131, 147<br />

Ueda, Yuki ........................................... 66<br />

V<br />

Van Assche, Roel ................................ 54<br />

van der Hoeven, Ransome ................ 156<br />

van der Spek, Hans ........................... 128<br />

Van Gilst, Marc .................................. 159<br />

Vantipalli1, Maithili C ........................... 50<br />

Author Index


Veal, Elizabeth A ........................ 129, 136<br />

Visvikis, Orane ..................................... 14<br />

Voisine, Cindy ...................................... 75<br />

Vora, Mehul ......................................... 76<br />

W<br />

Walhout, Marian ................................ 108<br />

Walker, Amy K ..................................... 48<br />

Wang, David .......................................... 9<br />

Wang, Xin .................................. 157, 168<br />

Wang, Ying ........................................ 113<br />

Wang, Ziyi .......................................... 158<br />

Ward, Jordan D ................................. 159<br />

Watts, Jennifer .............................. 36, 48<br />

Weick, Eva-Maria .............................. 166<br />

White, Corin V ................................... 160<br />

Williams, Travis W ......................... 53, 63<br />

Wilson, Iain BH .................................... 42<br />

Wohlschlager, Therese ........................ 42<br />

Wolf, Tim .............................................. 91<br />

Wollam, Joshua ................................... 38<br />

Wollenberg, Amanda C ................ 14, 161<br />

Wu, Ching-Ming ................................ 137<br />

Wu, Guang ............................................ 9<br />

X<br />

Xie, Fang ............................................... 8<br />

Author Index<br />

Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans Topic Meeting 2012<br />

Xie, Meng ............................................ 92<br />

Xue, Jian........................................ 74, 76<br />

Y<br />

Yamamoto, Keith R ............................ 159<br />

Yan, Zhi ..................................... 133, 139<br />

Yanase, Sumino .................................. 77<br />

Yang, Charles ...................................... 21<br />

Yasuda, Kayo .................................... 130<br />

Yates, Jonathan .................................. 18<br />

Yoon, Sang Sun ................................... 58<br />

Yoshimoto, Jennifer ............................. 85<br />

Yoshina, Sawako ..................... 53, 63, 68<br />

You, Young-Jai ............... 80, 85, 109, 121<br />

Yu, Charles ........................................ 126<br />

Z<br />

Zanni, Elena ...................................... 131<br />

Zhang, Jun........................................... 79<br />

Zhang, Liusuo ...................................... 93<br />

Zhang, Peng ........................................ 86<br />

Zhao, Guoyan ........................................ 9<br />

Zimmerman, Anna M ........................... 38<br />

Zucker, David....................................... 50<br />

179


VE.<br />

DR.<br />

NIVERSITY<br />

ELM<br />

D R.<br />

UNIVERSITY BUILDINGS, ACCOMMODATIONS, PARKING and POINTS OF INTEREST<br />

Conference Site Location Accommodation Location Parking Lot Location Point <strong>of</strong> Interest Location<br />

1. Memorial <strong>Union</strong> C1 A. Lowell Hall F2 Lot 6 C1 <strong>University</strong> Bookstore D2<br />

2 <strong>Union</strong> South & Hotel B2 B. Chadbourne Hall C2 Lot 17 A2 Walgreens D2<br />

C. <strong>University</strong> Inn D2 Lot 29 C3 Walgreens Pharmacy D2<br />

D. Doubletree Hotel E3 Lot 46 D2 Monona Terrace G2<br />

E Dahlmann Campus Inn D2 Lot 83 D2 Overture Center F2<br />

1<br />

2<br />

3<br />

2012<br />

CAMPUS<br />

AVE.<br />

BREESE<br />

TER.<br />

DR.<br />

Aging, Metabolism, Stress, Pathogenesis, and Small RNAs in C. elegans<br />

P17<br />

Camp Randall<br />

Stadium<br />

Thursday, July 12–Sunday, July 15, 2012<br />

A B C D E<br />

F<br />

ENGINEERING<br />

<strong>Union</strong> South<br />

and Hotel<br />

2<br />

Chadbourne<br />

Hall<br />

B<br />

Memorial <strong>Union</strong><br />

P6<br />

1<br />

Fountain<br />

Walgreens<br />

Pharmacy<br />

P29<br />

<strong>University</strong><br />

Bookstore<br />

Lowell Hall<br />

Walgreens<br />

Lake St.<br />

Parking Ramp<br />

P83<br />

P46<br />

A<br />

E<br />

Dahlmann<br />

Campus Inn<br />

C<br />

<strong>University</strong><br />

Inn<br />

D<br />

Doubletree<br />

Hotel<br />

A B C D E<br />

F<br />

Overture<br />

Center<br />

State<br />

Capitol<br />

G<br />

Walkway<br />

Monona<br />

Terrace<br />

LAKE<br />

MONONA<br />

G<br />

1<br />

2<br />

3


Phone<br />

•<br />

N<br />

W<br />

Studio B<br />

E<br />

Studio A<br />

Entrance<br />

<br />

Play Circle<br />

Theater<br />

<strong>Wisconsin</strong> <strong>Union</strong><br />

Theater<br />

Park Street<br />

Entrance<br />

Stairs<br />

Stairs to studiosW<br />

Rosewood<br />

Room<br />

Stairs to<br />

2nd floor<br />

CE Aging Locations<br />

Poster Sessions<br />

u Great Hall (4th floor)<br />

v Reception Room (4th floor)<br />

Party & Dance<br />

w Tripp Commons (2nd floor East)<br />

Dinner<br />

x Inn <strong>Wisconsin</strong> (2nd floor East)<br />

Plenary Sessions<br />

<strong>Union</strong> South-See Campus Map<br />

S<br />

Great Hall<br />

Phone<br />

•<br />

M<br />

Langdon<br />

Room<br />

3rd Floor West<br />

NOTE: Studios are not handicapped<br />

accessible. Accessible from West stairs only.<br />

W<br />

Stairs<br />

To <strong>Union</strong> Theater<br />

Box<br />

Office<br />

Stairs<br />

Hotel Rooms<br />

<br />

Class <strong>of</strong>'24<br />

Recept. Room<br />

Elevator<br />

Browsing<br />

Library<br />

Stairs<br />

Stairs<br />

Stairs<br />

To Park Street<br />

4th Floor<br />

Elevator<br />

Capitol View<br />

Main Lounge<br />

Outside Terrace Area<br />

Stairs<br />

Rathskeller<br />

MEMORIAL UNION<br />

Board<br />

Room<br />

Beefeaters<br />

Room<br />

To Terrace<br />

W/M<br />

Stairs<br />

Stairs<br />

Tripp Deck<br />

To MU Games Room (downstairs)<br />

Paul Bunyan Room<br />

Art Gallery<br />

Elevator<br />

Main Entrance<br />

W<br />

Round<br />

Table<br />

Room<br />

Elevator<br />

Od l Madison Room<br />

rs<br />

M<br />

<br />

Tripp Commons<br />

Elevator<br />

Stairs<br />

M W<br />

Stairs<br />

Phone•<br />

Stairs<br />

Lakefront onLangdon<br />

Elevator<br />

<br />

Central Reservations<br />

and Conference Services<br />

MU Floor Legend<br />

Floor Room in Use<br />

Fourth<br />

Third<br />

Second<br />

First<br />

3rd Floor East<br />

NOTE: Accessible from East elevator/<br />

East end <strong>of</strong> building only.<br />

2nd Floor<br />

Pr<strong>of</strong>ile Room<br />

Inn <strong>Wisconsin</strong> Room<br />

M<br />

W<br />

Inn WI Deck<br />

1st Floor<br />

Stairs Elevator Information Desk<br />

Tyme<br />

Phones<br />

Stairs<br />

Daily Scoop<br />

Deli<br />

Front<br />

Entrance


Level 1<br />

R A N D A L L A V E N U E<br />

S T A I R S<br />

E N T R A N C E<br />

E N T R A N C E<br />

Badger Market<br />

S T A I R S<br />

Ginger Root<br />

Harvest Grains<br />

<strong>Union</strong> South Building Map<br />

Level 2<br />

R A N D A L L A V E N U E<br />

S T A I R S<br />

E L E V A T O R<br />

E L E V A T O R<br />

S T A I R S<br />

S T A I R S<br />

S T A I R S<br />

CE Aging Locations<br />

Registration/Conference HQ<br />

Varsity Lounge (2nd floor)<br />

Plenary Sessions<br />

Marquee (2nd floor)<br />

Breakfast<br />

Varsity Hall (2nd floor)<br />

Luncheon Buffet<br />

Varsity Hall (2nd floor)<br />

E N T R A N C E<br />

S T A I R S<br />

S T A I R S<br />

The Roost<br />

Varsity Hall<br />

Prairie Fire<br />

Urban Slice<br />

T H E P L A Z A<br />

S E R V I C E<br />

Varsity Lounge<br />

C<strong>of</strong>feehouse Lounge<br />

S T A I R S<br />

S T A I R S<br />

E N T R A N C E<br />

W . J O H N S O N<br />

E N T R A N C E<br />

SUN GARDEN<br />

ELEVATOR<br />

M E N ’ S R E S T R O O M<br />

W O M E N ’ S R E S T R O O M<br />

Daily Scoop<br />

D A Y T O N S T R E E T<br />

S T O R A G E<br />

T H E P L A Z A<br />

E N T R A N C E<br />

S T A I R S<br />

Sun Garden<br />

Gallery 1308<br />

Wiscard<br />

VIP Info Desk<br />

E N T R A N C E<br />

E N T R A N C E<br />

E N T R A N C E<br />

W . J O H N S O N<br />

Sift & Winnow<br />

M E N ’ S<br />

R E S T R O M<br />

S T A I R S<br />

W O M E N ’ S<br />

R E S T R O O M<br />

SUN GARDEN<br />

ELEVATOR<br />

Traditions<br />

D A Y T O N S T R E E T<br />

S T A I R S<br />

<strong>Wisconsin</strong> Idea<br />

The Marquee<br />

S T O R A G E<br />

S T A I R S<br />

Lounge<br />

Alumni<br />

Governance<br />

Scholars<br />

UW Credit <strong>Union</strong><br />

S T A I R S<br />

M A I N<br />

E L E V A T O R<br />

M E N ’ S<br />

R E S T R O O M<br />

The Sett<br />

M A I N<br />

E L E V A T O R<br />

Hotel Desk<br />

Hotel Lobby<br />

WOMEN’S<br />

RESTROOM<br />

Upper<br />

Climbing Wall<br />

WUD Office<br />

The Sett Balcony<br />

The Sett<br />

<strong>Union</strong> South<br />

1st Floor<br />

Office<br />

Space<br />

S T A I R S<br />

E N T R A N C E<br />

E N T R A N C E<br />

Fifth Quarter Studio<br />

Info Lounge<br />

S T A I R S<br />

T H E O R C H A R D<br />

D R I V E<br />

T U R N A R O U N D<br />

<strong>Union</strong> South<br />

2nd Floor<br />

T H E O R C H A R D

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!