24.02.2013 Views

Huge Images? - GIT Verlag

Huge Images? - GIT Verlag

Huge Images? - GIT Verlag

SHOW MORE
SHOW LESS

Create successful ePaper yourself

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

69721<br />

VOLUME 9<br />

NOVEMbER 2007<br />

4<br />

3D Orientation Microscopy<br />

FRET, FRAP and FISH<br />

Wide-field CARS-Microscopy<br />

Series: Digital Materials Analysis<br />

Imaging<br />

Microscopy<br />

&RESEARCH • DEVELOPMENT • PRODUCTION<br />

Official Partner of the EMS<br />

G.I.T. Imaging www.gitverlag.com<br />

& Microscopy 2/2007 •


A New Age of Vision:<br />

Superresolution Microscopy!<br />

New concepts in microscopy revolutionize biomedical research<br />

Our new 4Pi and STED microscopy systems provide research scientists with the tools they need<br />

to find answers to many of life’s unresolved questions. With their superb resolving power,<br />

these progressive microscopy technologies break through the physical barriers of visibility, enabling<br />

the exploration of completely new perspectives in biomedicine.<br />

www.leica-microsystems.com/Confocal_Microscopes<br />

z<br />

Confocal x<br />

4Pi<br />

x<br />

Confocal y<br />

STED


Nobel Prize for Surface Scientist Gerhard Ertl<br />

Dear Reader,<br />

Some days ago the Royal Swedish Academy<br />

of Sciences announced the 2007 Nobel<br />

Prize in Chemistry for Gerhard Ertl,<br />

professor emeritus at the Fritz-Haber Institute<br />

in Berlin.<br />

Imaging & Microscopy congratulates<br />

Gerhard Ertl for this most prestigious<br />

honour in science that is awarded due to<br />

his thorough studies of chemical reactions<br />

on solid surfaces. When having a<br />

closer look on fundamental molecular<br />

processes at the gas-solid interface, small<br />

gas molecules may either be adsorbed or<br />

bounce back at the solid surface, according<br />

to a note by the Royal Swedish Academy<br />

of Sciences (www.kva.se). The first<br />

case includes the most interesting possibilities:<br />

The gas molecule can dissociate<br />

at the interface, the chemical properties<br />

of the surface can be changed, or the absorbed<br />

molecule can chemically react<br />

with a previously absorbed one.<br />

Ertl’s giant step forward in the understanding<br />

of these scenarios led to various<br />

breakthroughs in the development of catalysts<br />

being invaluable across industry.<br />

Carbon monoxide and hydrocarbons are<br />

converted to carbon dioxide in vehicles<br />

exhaust gasses today, and even the content<br />

of nitrous gasses can significantly be<br />

reduced. Car manufactures around the<br />

globe are producing vehicles that are less<br />

harmful to the environment and more<br />

fuel efficient.<br />

Very early, in the mid seventies, Ertl<br />

unravelled the surface mechanism of ammonia<br />

synthesis, a reaction that was first<br />

discovered by Fritz Haber, reaching such<br />

a technical and economical significance.<br />

By applying new surface science methods<br />

he showed, that the active species is not<br />

molecular but dissociatively adsorbed<br />

atomic nitrogen. The nitrogen is hydrogenated<br />

in a step-process.<br />

In terms of a more general description<br />

of related applications, Ertl’s findings<br />

had impact on the microelectronics industries<br />

where thin semiconductor layers<br />

are formed by CVD, chemical vapour<br />

deposition. Corrosion protection is yet<br />

another area that crucially depends on<br />

knowledge in surface science. Ertl’s findings<br />

help solving problems caused by<br />

corrosion both in daily life and in industry,<br />

for example, related to aeronautics<br />

or nuclear power plants.<br />

Ertl’s surface studies have opened a<br />

wide span of new techniques. A citation<br />

from the Royal Swedish Academy of Sciences<br />

says that “Gerhard Ertl had been<br />

one of the first to see the potential of<br />

these new techniques. Step by step he<br />

had created a methodology for surface<br />

chemistry by demonstrating how different<br />

experimental procedures can be used<br />

to provide a complete picture of a surface<br />

reaction”.<br />

Ertl recognized the significance of a<br />

microscopy related methodology in surface<br />

science very early. Quite interestingly<br />

Ertl and his group have set up a<br />

scanning tunneling microscope, STM, for<br />

imaging surface reconstructions in the<br />

presence of adsorbates at a time where<br />

many others were skeptical about STM,<br />

after its invention by Binnig and Rohrer<br />

in 1982. Some years later Ertl and coworkers<br />

succeeded in directly visualizing<br />

diffusion processes using high speed STM<br />

in order to verify macroscopic laws. Another<br />

example demonstrating Ertl’s ambition<br />

to take use out of modern microscopy<br />

methods in heterogeneous catalysis<br />

research is the observation of adsoption<br />

patterns in case of CO oxidation on Pt by<br />

Photo Emission Electron Microscopy,<br />

PEEM. Some excellent illustrations of<br />

adsoption patterns are published by the<br />

Surface Imaging Group, Dept. of Physical<br />

Chemistry, Fritz-Haber Institute of the<br />

Max-Planck-Society, www.fhi-berlin.<br />

mpg.de/surfimag. Field Ion Microscopy,<br />

FIM, is another important experimental<br />

approach to study catalytic reactions at<br />

the nanometer scale. This unique microscopy<br />

method has been further developed<br />

and applied to imaging and in-situ<br />

chemical probing in heterogeneous catalysis<br />

by Norbert Kruse, former Fritz-<br />

Haber Institute staff member and scientific<br />

advisor of Imaging & Microscopy<br />

[1, 2].<br />

In this issue of Imaging & Microscopy<br />

some excellent scientific articles are published,<br />

which give a view to recent research<br />

in Scanning Probe Microscopy,<br />

Compositional Analysis, Electron- and<br />

Light Microscopy. Furthermore, we like<br />

to steer the reader’s attention to Imaging<br />

& Microscopy’s conference reports and<br />

announcements.<br />

Enjoy your reading this issue<br />

Martin Friedrich Thomas Matzelle<br />

[1] Visart de Bocarmé T., Imaging & Microscopy<br />

8 (1), 19–21 (2006).<br />

[2] News & People, Imaging & Microscopy 9 (3), 8<br />

(2007).<br />

E d i t o r i a l<br />

[3] Freund H.-J., Knözinger H., J. Phys. Chem. B,<br />

108, 38, 14183–14186 (2004).<br />

G.I.T. Imaging & Microscopy 4/2007 •


C o n t e n t s<br />

E D I TO R I A L<br />

NObEL PRIzE fOR SuRfAcE ScIENTIST<br />

GERhARD ERTL<br />

Dr. T. Matzelle, Dr. M. Friedrich,<br />

<strong>GIT</strong> VERLAG, DE 1<br />

c OV E R S TO RY<br />

ThE “f” WORDS<br />

fRET, fRAP, and fISh<br />

– Technology and<br />

Techniques<br />

K. Garsha, Photometrics,<br />

AZ, USA 46<br />

P R O D u c T S 70<br />

I & M S h OW c A S E<br />

Bruker AXS 65<br />

FEI Company 65<br />

JPK 66<br />

Leica 66<br />

Nikon 67<br />

Olympus 67<br />

c O M PA N Y P R O f I L E<br />

AGAR ScIENTIfIc 64<br />

E V E N T c A L E N DA R 4<br />

I & M N E W S T I c K E R 6<br />

c O M PA N Y N E W S 1 4<br />

Index Inside Back Cover<br />

Imprint Inside Back Cover<br />

business Partner Inside Back Cover<br />

• G.I.T. Imaging & Microscopy 4/2007<br />

N E W S f R O M E M S<br />

EMS NEWSLETTER 20, OcTObER 2007<br />

Prof. Dr. D. Schryvers, University of<br />

Antwerp, BE 10<br />

R M S I N f O c u S<br />

ThE RMS – AccESS AND PROGRESSION ...<br />

A. Winton, Royal Microscopical Society, UK 12<br />

ORGANIzING ELMI 2008 hAS ALREADY<br />

STARTED<br />

Dr. M. Friedrich, <strong>GIT</strong> VERLAG, DE 13<br />

A N N O u N c E M E N T<br />

fOcuS ON MIcROScOPY 2008<br />

Prof. Dr. G.J. Brakenhoff et al., University of<br />

Amsterdam, NL 16<br />

E V E N T R E P O RT<br />

SEE YOu LATER ALLIGATOR<br />

Dr. M. Friedrich, <strong>GIT</strong> VERLAG, DE 18<br />

AT ThE REGION Of fORMER<br />

IRONWORKS<br />

Prof. Dr. P. Mestres-Ventura, Prof. Dr. U.<br />

Hartmann, University of Saarland, DE 22<br />

E L E c T R O N M I c R O S c O P Y<br />

ENAbLING 3D TEM/STEM Of<br />

NANOPARTIcLES<br />

Dr. K. F. Jarausch, Hitachi High<br />

Technologies America, Inc., CA, USA<br />

Dr. D.N. Leonard, Appalachian State<br />

University, NC, USA 24<br />

VERIfYING ENGINEERING AT ThE<br />

NANOScALE<br />

Dr. I. F. Uchegbu, University of London, UK 28<br />

cRYO ELEcTRON TOMOGRAPhY<br />

M. Harris, FEI Company, NL 31<br />

S c A N N I N G<br />

S E c T I O N<br />

uLTRAfAST cONfOcAL<br />

RAMAN IMAGING<br />

O. Hollricher et al., Witec, DE 34<br />

uLTRASONIc MAchINING AT ThE<br />

NANOMETER ScALE<br />

Dr. M. T. Cuberes, University of<br />

Castilla – La Mancha, ES 36<br />

3D ORIENTATION MIcROScOPY<br />

S.I. Wright, EDAX-TSL, Draper, Utah, USA,<br />

S. Zaefferer, Max-Planck-Institute for Iron<br />

Research, DE 40<br />

cOMbINING OPTIcAL uPRIGhT<br />

MIcROScOPY AND AfM<br />

J. Barner, JPK Instruments, DE 42<br />

M I c R O S c O P Y fAc I L I T I E S<br />

cANcER RESEARch IN GLASGOW<br />

Dr. K. I. Anderson, Beatson Research<br />

Center, UK 44<br />

L I G h T M I c R O S c O P Y<br />

SYSTEMATIc ANALYSIS Of fRAP<br />

ExPERIMENTS<br />

Dr. S. Seiffert et al., Clausthal University of<br />

Technology, DE 48<br />

WIDE-fIELD cARS MIcROScOPY<br />

Prof. Dr. M.A.M. Ritsch-Marte et al., Innsbruck<br />

Medical University, AT 52<br />

NExT GENERATION LIGhT SOuRcES<br />

fOR IMAGING<br />

Dr. J. Clowes, Fianium, UK 55<br />

MIcROScOPY SERIES ON DI<strong>GIT</strong>AL<br />

MATERIALS ANALYSIS – PART 1<br />

Esther Ahrent, Olympus, DE 58<br />

I M AG E P R O c E S S I N G<br />

AIR QuALITY cONTROL fOR hAzARDOuS<br />

bIO-MATERIAL<br />

Dr. P. Perner, Institute of Computer Vision<br />

and Applied Computer Sciences, DE 62<br />

N OT E S f R O M N I KO N<br />

cONTROLLED LIGhT ExPOSuRE<br />

MIcROScOPY<br />

Dr. M. Balzar, Nikon Instruments<br />

Europe BV, NL 68


� ��������������������������<br />

����������������������<br />

� ���������������������������������<br />

���������������������������������������������������������������<br />

������������������������������������������������������������������������<br />

����������������������������������������������������������������������<br />

�����������������������������������������������������������������<br />

��������������������������������������������������������������������������<br />

���������������������������������������������������������������������<br />

������������������������������������������������������������������<br />

��������������������������������������������������������������������<br />

�����������������������������������������������������������������<br />

������������������������������������������������������������������<br />

����������������������������������������������������������������������<br />

������������������������������������������������������������������������<br />

������������������������������������������������������������������<br />

�������������������������������������<br />

�������������������������������<br />

��������������������������������<br />

�����������������������������<br />

�������������������������������������<br />

����������������������<br />

� � � � � � � � � � � � � � � � � �


E v E n t C a l E n d a r<br />

• G.I.T. Imaging & Microscopy 4/2007<br />

EVENT CALENDAR<br />

EVENT WhEN WhERE SouRCE of INfoRmATIoN<br />

Celebrating 50 Years of Multislice – Symposium November 6 Monash University, Brighton, Australia www.conferences.monash.org/multislice/index.<br />

cfm?p=455<br />

Scottish Microscopy Symposium, The Scottish<br />

Microscopy Group<br />

November 14 Dundee, Scotland, UK www.gla.ac.uk/ibls/II/em/SMG/smgnew.html<br />

Cryo Microscopy Group Meeting November 21 University of Birmingham, UK www.cryomicroscopygroup.org.uk<br />

MRS Fall Meeting 2007 November<br />

26–30<br />

Symposium on Quantitative Electron Microscopy for<br />

Materials Science<br />

November<br />

26–30<br />

Hynes Convention Center and Sheraton<br />

Boston Hotel<br />

Boston, MA, USA<br />

Hynes Convention Center and Sheraton<br />

Boston Hotel<br />

Boston, MA, USA<br />

www.mrs.org/s_mrs<br />

www.mrs.org/s_mrs<br />

American Society for Cell Biology Annual Meeting December 1–5 Washington DC, USA www.ascb.org<br />

4 th International Conference of MRS-Africa December<br />

10–14<br />

Advanced TEM & TOM December<br />

17–18<br />

2008<br />

2008 Winter Conference on Plasma Spectrochemistry<br />

Winter School on Microstructural Characterization<br />

Focused on Electron Microscopy<br />

20 th Australian Conference on Microscopy and<br />

Microanalysis<br />

Dar es Salaam, Tanzania www.mrs.org/s_mrs<br />

Manchester, UK www.rms.org.uk/event_temtom.shtml<br />

January 7–12 Temecula, CA, USA www.uc.edu/plasmachem/taormina/Documents/2008_<br />

Winter_Conference_Information.pdf<br />

January 14–18 Thessaloniki, Greece pam1.physics.auth.gr<br />

pam1@physics.auth.gr<br />

February 9–15 Perth, Western Australia, Australia www.microscopy.org.au/ACMM20<br />

MRS Spring Meeting 2008 March 24–28 Moscone West and San Francisco<br />

Marriott, San Francisco, CA, USA<br />

Electron Backscatter Diffraction Meeting March 31–<br />

April 1<br />

The Microscopic Ice Age – A Course in Cryo<br />

Techniques for Electron Microscopy<br />

www.mrs.org/s_mrs<br />

Sheffield, UK www.rms.org.uk/event_EBSD.shtml<br />

April 7–11 Rothamsted Research, Harpenden, UK www.rms.org.uk/event_cryo08.shtml<br />

Spring School in Electron Microscopy April 14–18 University of Birmingham victoria@rms.org.uk<br />

Jeels 2008 May 14–16 Laboratoire de Métallurgie Physique<br />

Poitiers, France<br />

7 th European Conference on Nonlinear Optical<br />

Spectroscopy (ECONOS 2008)<br />

1 st European Conference on CARS microscopy<br />

(microCARS 2008)<br />

May 25–28 Igls, Austria Igls2008.org<br />

May 25–28 Igls, Austria Igls2008.org<br />

jeels2008.sp2mi.univ-poitiers.fr<br />

ELMI Meeting 2008 May 27-30 Davos, Switzerland elmi08.unibas.ch/index.html<br />

EM2008: 8 th International Conference on Electron<br />

Microscopy of Solids<br />

June 8–11 Cracow-Zakopane, Poland kusinski@uci.agh.edu.pl<br />

MRS International Materials Research Conference June, 9–12 Chongqing, China www.mrs.org/s_mrs<br />

13 th International Conference on Alkali-Aggregate<br />

Reaction in Concrete<br />

June 16–19 Trondheim, Norway www.icaar2008.org


MICROSCIENCE 2008 June 23–26 ExCeL, London, UK www.microscience2008.org.uk<br />

BIAMS 08–9 th International Workshop on Beam<br />

Injection Assessments of Microstructures in Semiconductors<br />

June 29– July 3 Toledo, Spain www.biams08.org<br />

Light Microscopy Summer School July 7–9 University of York www.rms.org.uk/event_lmschool06.shtml<br />

Getting the most from your Confocal July 10 University of York www.rms.org.uk/event_Confocal.shtml<br />

Intern. Conference on Mass Data Analysis of Signals<br />

and <strong>Images</strong> in Medicine, Biotechnology, Chemistry<br />

and Food Industry, MDA<br />

July 14 Leipzig, Germany mda-signals.de<br />

Microscopy & Microanalysis 2008 August 3–7 Albuquerque, New Mexico, USA www.msa.microscopy.com<br />

14 th European Microscopy Congress (EMC 2008) September 1–5 Aachen, Germany www.eurmicsoc.org/emc2008.html<br />

Microscopy of Oxidation 7 September<br />

15–17<br />

The University of Chester, UK www.liv.ac.uk/engdept/conferences/moo_07.htm<br />

APMC9: 9 th Asia-Pacific Microscopy Conference November 2–7 Jeju Island, Korea www.apmc9.or.kr<br />

huchul@snu.ac.kr<br />

ASCB Annual Meeting 2008 December<br />

13–17<br />

THROUGH<br />

YOUR WORLD<br />

IN SECONDS<br />

Bridging the gap between optical and electron microscopy<br />

www.fei.com/phenom<br />

San Francisco, USA www.ascb.org<br />

G.I.T. Imaging & Microscopy 4/2007 •


Lasers Fabricated by Nanoimprint<br />

Lithography<br />

The team of V. Reboud at the Tyndall National Institute,<br />

Cork, Ireland, report on the fabrication and<br />

characterization of two-dimensional polymer photonic<br />

crystal band-edge lasers operating in the visible<br />

range. The components have been fabricated in<br />

a dye chromophore-loaded polymer matrix by nanoimprint<br />

lithography and high-symmetry bandedge<br />

modes are used to generate laser emission.<br />

Their work demonstrates the potential of nanoimprint<br />

lithography for the fabrication of two-dimensional<br />

planar photonic crystal structures in an active<br />

medium in a one-step process.<br />

» Appl. Phys. Lett. 91, 151101<br />

» doi:10.1063/1.2798250<br />

Optical Coherence Computed<br />

Tomography<br />

L. Li and L.V. Wang from the Washington University<br />

in St Louis, Missouri, USA, propose a device to<br />

bridge the gap between diffuse optical tomography<br />

and optical coherence tomography. Both ballistic<br />

and multiple-scattered photons are measured at<br />

multiple source-detection positions by low-coherence<br />

interferometry providing a temporal resolution<br />

smaller than 100 fs. A light-tissue interaction<br />

model was established using the time-resolved<br />

Monte Carlo method. The optical properties were<br />

then reconstructed by solving the inverse transient<br />

radiative transport problem under the first Born approximation.<br />

Absorbing inclusions of 100 µm diameter<br />

were imaged through a 2.6-mm-thick (~ 30<br />

scattering mean-free-paths) scattering medium.<br />

» Appl. Phys. Lett. 91, 141107<br />

» doi:10.1063/1.2793625<br />

Frequency Response of an AFM:<br />

Magnetic Versus Acoustic Excitation<br />

E.T. Herruzo and R. Garcia from the CSIC, Madrid,<br />

Spain, discuss the dynamics of an amplitude modulation<br />

AFM in different environments such as water<br />

and air, and show that the resonance curves depend<br />

on the excitation method used to drive the<br />

cantilever, either mechanical or magnetic. This dependence<br />

is magnified for small force constants<br />

and quality factors, i.e., below 1 N/m and 10, respectively.<br />

They also show that the equation for the<br />

observable, the cantilever deflection, depends on<br />

the excitation method. Under mechanical excitation,<br />

the deflection involves the base and tip displacements,<br />

while in magnetic excitation, the cantilever<br />

deflection and tip displacement coincide.<br />

» Appl. Phys. Lett. 91, 143113<br />

» doi:10.1063/1.2794426<br />

• G.I.T. Imaging & Microscopy 4/2007<br />

NEwS TICkER<br />

Influence of Sample Conductivity on<br />

Oxidation by the Tip of AFM<br />

V. Cambel and J. Soltys at the Slovak Academy of<br />

Sciences, Bratislava, Slovakia analyzed the role of<br />

the electric field distribution in the nano-oxidation<br />

process realized by the tip of AFM experimentally<br />

and theoretically. Authors show the importance of<br />

the sample conductivity and the water bridge in the<br />

process applied to bulk GaAs and Ga[Al]As heterostructures<br />

in both contact and noncontact AFM<br />

modes and the consequences for the lines witten.<br />

They show that the electric field distribution in the<br />

system tip-sample is controlled by the sample conductivity.<br />

In the case of low-conductive samples,<br />

maximum field is located apart from the tip apex<br />

for both contact and noncontact AFM modes.<br />

» J. Appl. Phys. 102, 074315<br />

» doi:10.1063/1.2794374<br />

High-resolution Microscope for Tipenhanced<br />

Optical Processes in UHV<br />

J. Steidtner and B. Pettinger in Berlin, Germany<br />

present an optical microscope based on tip-enhanced<br />

optical processes that can be used for studies<br />

on adsorbates as well as thin layers and nanostructures.<br />

It provides chemical and topographic<br />

informations with a resolution of a few nanometers<br />

and can be employed in ultrahigh vacuum as well<br />

as gas phase. The central idea is to mount, within<br />

an UHV system, an optical platform with all necessary<br />

optical elements to a rigid frame that also carries<br />

the scanning tunneling microscope unit and to<br />

integrate a high numerical aperture parabolic mirror<br />

between the scanning probe microscope head<br />

and the sample. Authors present the first results of<br />

Raman measurements using the device and the experimentally<br />

determined requirements of the parabolic<br />

mirror in terms of alignment accuracy.<br />

» Rev. Sci. Instrum. 78, 103104<br />

» doi:10.1063/1.2794227<br />

An UHV Fast-scanning and Variable<br />

Temperature STM for Large Scale<br />

Imaging<br />

B. Diaconescu and co-workers from the University<br />

of New Hampshire, USA describe the design and<br />

performance of a fast-scanning, variable temperature<br />

scanning tunneling microscope (STM) operating<br />

from 80 to 700 K in ultrahigh vacuum (UHV),<br />

which routinely achieves large scale atomically resolved<br />

imaging of compact metallic surfaces. The<br />

vertical resolution of the instrument was found to<br />

be about 2 pm at room temperature. The total<br />

scanning area is about 8 × 8 µm 2 . The sample tem-<br />

perature can be adjusted by a few tens of degrees<br />

while scanning over the same sample area.<br />

» Rev. Sci. Instrum. 78, 103701<br />

» doi:10.1063/1.2789655<br />

Suppression of Spurious Vibration of<br />

Cantilever in AFM<br />

T. Tsuji and colleagues from Tohoku University, Japan,<br />

developed a simple but effective method for<br />

suppressing spurious response (SR) to improve the<br />

precision of dynamic AFM using cantilever vibration<br />

spectra. The dominant origin of SR was identified<br />

to be the bending vibration of the cantilever<br />

substrate, but while a rigid cover pressing the<br />

whole surface of the substrate suppressed SR, the<br />

utility was insufficient. Then, a method of enhancing<br />

the bending rigidity of the substrate by gluing a<br />

rigid plate (clamping plate, CP) to the substrate<br />

was developed. The CP method will particularly<br />

contribute to improving dynamic-mode AFM, in<br />

which resonance spectra with a low quality factor<br />

are used, such as noncontact mode AFM in liquid or<br />

contact resonance mode AFM.<br />

» Rev. Sci. Instrum. 78, 103703<br />

» doi:10.1063/1.2793498<br />

High Resolution Gamma Ray<br />

Tomography Scanner<br />

U. Hampel and co-workers at Forschungszentrum<br />

Dresden-Rossendorf e.V., Dresden, Germany, report<br />

on the development of a high resolution gamma<br />

ray tomography scanner that is operated with a Cs-<br />

137 isotopic source at 662 keV gamma photon energy<br />

and achieves a spatial image resolution of 0.2<br />

line pairs/mm at 10 % modulation transfer function<br />

for noncollimated detectors. It is primarily intended<br />

for the scientific study of flow regimes and phase<br />

fraction distributions in fuel element assemblies,<br />

chemical reactors, pipelines, and hydrodynamic machines,<br />

but it is applicable to nondestructive testing<br />

of larger radiologically dense objects. They also<br />

built a computed tomography scanner gantry for<br />

measurements at fixed vessels or plant components.<br />

» Rev. Sci. Instrum. 78, 103704<br />

» doi:10.1063/1.2795648<br />

Correction of Axial Geometrical Distortion<br />

in Microscopic <strong>Images</strong><br />

H.J. Van Elburg and colleagues from the University<br />

of Antwerp, Belgium, discuss the extraction of<br />

quantitative data from microscopic volume images<br />

when imperfectly matched immersion and mounting<br />

media result in axial geometrical distortion. Linear<br />

correction of the axial distortion using the


Want to know what your cells are<br />

up to when you’re not looking?<br />

Complete cell surveillance with BioStation CT<br />

A revolutionary approach to cell culture and imaging, BioStation CT combines<br />

incubator, microscope and camera in one easy-to-use unit. Designed for unattended<br />

operation, BioStation CT provides 24/7 automated cell culture and cell surveillance<br />

with remote access to data via Internet /LAN. Providing greater workflow flexibility<br />

and increased productivity, users never need to miss a moment in their cell study!<br />

• Complete variable, monitored environmental control<br />

• Multi-user / multi-experiment system<br />

• Cell & sample traceability with culture & image history<br />

• Touch screen operation<br />

• 24/7 remote operation & monitoring<br />

• Cell-friendly illumination - phase contrast / multi-channel fluorescence<br />

• Easy to set up and use<br />

• No thermal / mechanical drift<br />

• Secure for cells, safe for operators<br />

With no need to remove vessels for observation, contamination risk and cell stress<br />

are minimised. Nikon’s cellogy* technologies control phototoxicity, vibration, and<br />

focus for successful results even in prolonged time-lapse studies! Ideal for cell<br />

biology, stem cell research, embryology and IVF, BioStation meets the most<br />

demanding cell culture and imaging requirements.<br />

Let BioStation take care of your live cell imaging. For more information visit<br />

www.nikoninstruments.eu/biostationct<br />

I M A G I N G S O L U T I O N S F O R Y O U R C E L L S E N V I R O N M E N T<br />

*Cellogy describes Nikon’s ethos of caring for, managing, and getting the<br />

very best results from living cells during imaging<br />

CT<br />

IM<br />

www.nikoninstruments.eu


I & M N e w s T I c k e r<br />

paraxial estimate of the axial scaling factor yields<br />

results that may differ as much as 4 % from the actual<br />

values. From calculations based on a theoretical<br />

expression of the 3-D point-spread function in<br />

the focal region of a high-aperture microscope authors<br />

derived axial scaling factors that result in<br />

quantitative results accurate to better than 1 %.<br />

From a non-linear correction procedure, an improved<br />

formula for the paraxial estimate of the axial<br />

scaling factor is derived.<br />

» Journal of Microscopy 228 (1), 45–54.<br />

» doi:10.1111/j.1365-2818.2007.01822.x<br />

A white Light Confocal Microscope for<br />

Spectrally Resolved Multidimensional<br />

Imaging<br />

J.H. Frank and co-workers from the University of<br />

Cambridge, UK, demonstrate spectrofluorometric<br />

imaging microscopy in a confocal microscope using<br />

a supercontinuum laser as an excitation source and<br />

a custom-built prism spectrometer for detection.<br />

This microscope system provides confocal imaging<br />

with spectrally resolved fluorescence excitation and<br />

detection from 450 to 700 nm, and authors present<br />

the device performances. The speed of the spectral<br />

scans is suitable for spectrofluorometric imaging of<br />

live cells. Effects of chromatic aberration are modest<br />

and do not significantly limit the spatial resolution<br />

of the confocal measurements.<br />

» Journal of Microscopy 227 (3), 203–215.<br />

» doi:10.1111/j.1365-2818.2007.01803.x<br />

In Vivo Imaging of Hydrogen Peroxide<br />

with Chemiluminescent Nanoparticles<br />

D. Lee from the Georgia Institute of Technology, Atlanta,<br />

Georgia, demonstrate that nanoparticles formulated<br />

from peroxalate esters and fluorescent<br />

dyes can image hydrogen peroxide in vivo with<br />

• G.I.T. Imaging & Microscopy 4/2007<br />

high specificity and sensitivity and discuss possible<br />

applications. The peroxalate nanoparticles image<br />

hydrogen peroxide by undergoing a three-component<br />

chemiluminescent reaction between hydrogen<br />

peroxide, peroxalate esters and fluorescent dyes.<br />

The peroxalate nanoparticles have attractive properties<br />

for in vivo imaging, such as tunable wavelength<br />

emission (460–630 nm), nanomolar sensitivity<br />

for hydrogen peroxide and excellent specificity<br />

over other reactive oxygen species.<br />

» Nature Materials 6, 765–769<br />

» doi:10.1038/nmat1983<br />

3-D Imaging of Liquid Crystals by CARS<br />

Microscopy<br />

B.G. Saar and colleagues used Coherent anti-Stokes<br />

Raman scattering (CARS) microscopy to provide<br />

three-dimensional chemical maps of liquid crystalline<br />

samples without the use of external labels.<br />

CARS is an optical imaging technique that derives<br />

contrast from Raman-active molecular vibrations in<br />

the sample, that offers more rapid chemical characterization<br />

without the use of external dyes or contrast<br />

agents. The use of CARS to image chemical<br />

and orientational order in liquid crystals is demonstrated<br />

using several examples, and the limitations<br />

and benefits are discussed.<br />

» Opt. Express 15, 13585–13596<br />

» http://www.opticsinfobase.org/abstract.<br />

cfm?URI=oe-15-21-13585<br />

Direct Measurement of Hydrophobic<br />

Forces on Cell Surfaces Using AFM<br />

D. Alsteens and co-workers at the Université<br />

Catholique de Louvain, Louvain-La-Neuve, Belgium<br />

present chemical force microscopy (CFM) with hydrophobic<br />

tips to measure local hydrophobic forces<br />

on organic surfaces and on live bacteria. On organic<br />

surfaces, they found an excellent correlation<br />

between nanoscale CFM and macroscale wettability<br />

measurements, demonstrating the sensitivity of<br />

the method toward hydrophobicity and providing<br />

novel insight into the nature of hydrophobic forces.<br />

Authors also studied hydrophobic forces associated<br />

with mycolic acids on the surface of mycobacteria,<br />

and discuss the importance of these compounds.<br />

» Langmuir, ASAP Article 10.1021/la702765c<br />

S0743-7463(70)02765-8<br />

Multicolor STORM Imaging<br />

Using photoswitchable fluorescent probes with distinct<br />

colors, M. Bates and his colleagues demonstrate<br />

the feasibility of multicolour stochastic optical<br />

reconstruction microscopy (STORM). The pairing<br />

of different activator dyes with a range of photoswitchable<br />

reporter fluorophores allowed iterative,<br />

color-specific activation of distinct color subsets<br />

and STORM imaging with a resolution of 20–30<br />

nm.<br />

» Science, 317, pp. 1749–1753<br />

Fluorescence Nanoscopy<br />

A. Egner and co-workers report nanoscopy imaging<br />

in intact cells through reversible photoswitching of<br />

individual fluorophores at fast recording speeds<br />

and demonstrate this by imaging the microtubular<br />

network of a mammalian cell at 40 nm resolution.<br />

» Biophys. Journal, 93, pp. 3285–3290<br />

Correlative 3D LM-EM Imaging of<br />

Mitochondria During Apoptosis<br />

M. Sun and colleagues studied the behaviour of mitochondria<br />

during apoptosis using fluorescence microscopy<br />

in combination with 3D electron tomography<br />

of the same cells. After locating apoptotic cells<br />

in the light microscope, they could subsequentially<br />

study the remodelling of the inner mitochondrial<br />

membrane into separate vesicular compartments at<br />

the ultrastructural level.<br />

» Nature Cell Biology, 9, pp. 1057–1065<br />

Even Illumination Field in TIRF Imaging<br />

with a Laser<br />

R. Fiolka and co-workers describe a method to overcome<br />

the problem of the scattering of coherent laser-light<br />

that normally causes uneven illumination<br />

of the image field during TIRF imaging by azimuthal<br />

rotation of the illumination laser beam. The incidence<br />

angle of the laser can still be changed quickly<br />

so that fast switching to epifluorescence illumination<br />

is still possible.<br />

» Microsc. Res. Tech. (advance on-line publication)<br />

Cryo-fluorescence Microscopy<br />

The use of a novel cryo-light microscope stage enabled<br />

C. Schwartz and her colleagues to perform correlative<br />

light and electron microscopy of vitreous<br />

samples prepared for cryo-EM. In addition to the<br />

correlative imaging aspect, photobleaching of the<br />

fluorophores is reduced the cryogenic temperatures<br />

(–140 °C) of the setup.<br />

» J. Microsc., 227, pp. 98–109<br />

white Light Confocal with a Supercontinuum<br />

Laser<br />

J.H. Frank and co-workers describe the use of a supercontinuum<br />

laser as the excitation light source<br />

for a confocal microscope. Throught the use of an<br />

AOTF, up to eight excitation wavelengths can be selected<br />

and be freely positioned along the spectrum,<br />

thus allowing to closely match the excitations to<br />

the spectral requirements of the selected fluorophores.<br />

In combination with spectral detection fluorescence<br />

excitation and emission spectra can be<br />

taklen at every position in the image.<br />

» J. Microsc., 227, pp. 203–215


Understand the<br />

Dynamic Processes of Life.<br />

Reach Out for Experience.<br />

Axio Observer LSM 5 DUO PALM MicroBeam<br />

Carl Zeiss: Living Cells<br />

www.zeiss.de/FluoresScience


Ueli Aebi<br />

EMS President<br />

During the summer months no EMS<br />

Newsletter was published, so we do have<br />

quite a few items to cover in this autumn<br />

issue. As you know, supporting European<br />

microscopy activities is one of the primary<br />

goals of EMS. In this spirit the EMS<br />

Board at its meeting in Prague decided to<br />

offer six scholarships of 500 Euro each to<br />

young researchers to participate at Symposium<br />

C on “Quantitative TEM for Advanced<br />

Materials” that will be held during<br />

the Boston Fall Meeting of the US<br />

Materials Research Society and is organized<br />

by Etienne Snoeck, Rafal Dunun-<br />

Burkowski, Johan Verbeeck and Uli Dahmen.<br />

This support was made possible by<br />

a special 2500 Euro grant from FEI. The<br />

successful applicants are Leonardo LARI<br />

(Liverpool), Wouter van den Broek (Antwerp),<br />

Lang-Yun (Shery) Chang (Cambridge),<br />

Sandra Van Aert (Antwerp),<br />

Florent Houdellier (Toulouse) and Magnus<br />

Garbrecht (Kiel); we wish them all a<br />

very fruitful meeting.<br />

By the closing of the first round of applications<br />

for sponsored events taking<br />

place during the first six months of 2008,<br />

four applications have been received and<br />

will be evaluated by the Board during the<br />

coming weeks. Since in 2008 EMS will<br />

Nick Schryvers<br />

EMS Secretary<br />

EMS Newsletter 20, October 2007<br />

Dear EMS member,<br />

We also like to announce the following<br />

training courses<br />

Network of Excellence (EU-NOE)<br />

for 3D-electron microscopy (3D-EM)<br />

(http://www.3dem-noe-training.org):<br />

Transmission Electron Microscopy in Life Science<br />

February 4 th –8 th , Eindhoven, The Netherlands,<br />

and<br />

N e w s f r o m e m s<br />

Single Particle Analysis, February 25 th –29 th 2008,<br />

Madrid, Spain<br />

10 • G.I.T. Imaging & Microscopy 4/2007<br />

organize the quadrennial European Microscopy<br />

Congress EMC 2008, no EMS<br />

extension was granted. The next Extension<br />

will be held in 2009 with the deadline<br />

for applications being June 30,<br />

2008.<br />

Most exciting for EMS during the past<br />

summer period has been the formal decision<br />

of the Portuguese Microscopy Society<br />

SPMicros to join EMS as an en-bloc<br />

member, which brings the total number<br />

of EMS members to over 5000 and a coverage<br />

of the continent that is close to<br />

complete! The Portuguese Society has<br />

gone through some important organizational<br />

changes lately and we do warmly<br />

welcome SPMicros to the EMS family<br />

hoping that we can be of some assistance<br />

to help increasing the visibility of microscopy<br />

in Portugal and, most importantly,<br />

improving communication channels with<br />

the rest of Europe.<br />

And now a quick update on EMC 2008<br />

in Aachen: during the past three months<br />

chair persons for symposia and regular<br />

sessions have been selected, and at<br />

present names for keynote and invited<br />

speakers are being solicited. Most importantly,<br />

a first flyer has been mailed, and<br />

potential exhibitors have been contacted.<br />

Last but not least, the first few pages of<br />

the EMC 2008 website have recently been<br />

made available at www.emc2008.de.<br />

Please allow us to remind you that we<br />

are presently looking for candidates<br />

poised to organize the 15 th European Microscopy<br />

Congress in 2012. With the<br />

growing interest in microscopy, EMC is<br />

looking for congress sites capable of accommodating<br />

up to 1500 participants<br />

following up to 10 parallel sessions plus<br />

a commercial exhibitor space of around<br />

2000 m 2 (incl. walking space). Applications<br />

have to be sent to the EMS Secre-<br />

tary by January 30 th , 2008, according to<br />

the stipulations listed in point F.2 of the<br />

By-Laws of the Constitution (see www.<br />

eurmicsoc.org). The final venue will be<br />

decided by the General Council which<br />

will meet in Aachen during EMC 2008.<br />

At EMC 2008 the quadrennial FEI<br />

Awards (one in the Life Sciences and one<br />

in Materials Science/Physics) will be presented.<br />

Candidates should be proposed<br />

by a Microscopy Society, a group of scientists,<br />

or an individual scientist. Applications<br />

for these prestigious awards<br />

should be sent to the EMS Secretary by<br />

regular mail and reach him by no later<br />

than April 1 st , 2008. More details on the<br />

qualification criteria and application procedures<br />

can be found on the EMS website<br />

www.eurmicsoc.org under the<br />

header “funding”.<br />

We have just been informed that Dr.<br />

Charles (Chuck) Garber, chairman of SPI<br />

Supplies and one of our early-day ECMA<br />

members, died on September 19 th , 2007.<br />

We all valued Chuck for his candid remarks<br />

and ever constructive suggestions<br />

aimed at improving the performance of<br />

scientists and exhibitors alike, so we will<br />

all miss Chuck at future microscopy<br />

meetings, be this at his booth, in the sessions<br />

or at the social gatherings. The<br />

EMS Board members would like to express<br />

their sincere condolences to Babszy<br />

Garber, his wife, and his family.<br />

Contact:<br />

Prof. Dr. D. Schryvers, Ph.D.<br />

Electron Microscopy for Materials Science (EMAT)<br />

Department of Physics<br />

University of Antwerp, Belgium<br />

Tel.: +32 3 2653247<br />

Fax: +32 3 2653257<br />

nick.schryvers@ua.ac.be


R M S I n F o c u S<br />

The RMS – Access and Progression …<br />

… From the Cradle (Well Almost!)<br />

“ When planning for a year, plant corn.<br />

When planning for a decade, plant trees.<br />

When planning for life, train and educate people.”<br />

Chinese proverb: Guanzi (c.645 B.C.)<br />

The European Commission has recognised<br />

the inherent wisdom of this Chinese<br />

proverb through the instigation this<br />

year of the Lifelong Learning Programme.<br />

Billed as the flagship European funding<br />

programme in the field of education and<br />

training, this is the first time a single programme<br />

will cover learning opportunities<br />

from childhood to old age. It aims to<br />

support projects and activities that foster<br />

interchange, cooperation and mobility<br />

between education and training systems<br />

within the EU, so that they become a<br />

world quality reference.<br />

The Royal Microscopical Society<br />

wholeheartedly endorses this ethos and<br />

is a keen advocate of learning and professional<br />

development. “As a professional<br />

body we see that one of our core rolls is<br />

to provide educational resources, not<br />

only to our members but to the community<br />

as a whole, in order to develop careers<br />

and support a wider understanding<br />

of science and microscopy at all levels”,<br />

explains Rob Flavin, Executive Director<br />

of the RMS.<br />

Training and Events<br />

In support of the above, the RMS publishes<br />

The Journal of Microscopy and a<br />

series of microscopy books, as well as<br />

Date Event Title Location<br />

17–18 December 2007 Aberration Corrected & Quantitative<br />

(S)TEM & Advances in Tomography II<br />

Manchester<br />

31 March–01 April 2008 Electron Backscatter Diffraction Conference<br />

Sheffield<br />

07–11 April 2008 The Microscopic Ice Age – A Course in<br />

Cryo Techniques for Electron Microscopy<br />

Rothamsted Research, Harpenden<br />

14–18 April 2008 Spring School in Electron Microscopy University of Oxford<br />

23–26 June 2008 MICROSCIENCE 2008 – International<br />

Conference and Exhibition<br />

ExCeL, London<br />

07–09 July 2008 Light Microscopy Summer School University of York<br />

10–11 July 2008 Getting the most from your Confocal University of York<br />

12 • G.I.T. Imaging & Microscopy 4/2007<br />

helping young scientists through bursaries.<br />

The Society further demonstrates<br />

its commitment to professional development<br />

by offering qualifications in microscopy,<br />

and organising annual 5 day training<br />

courses in light, electron and confocal<br />

microscopy, flow cytometry and cell imaging.<br />

In addition, it stages its own scientific<br />

meetings that address topics at the<br />

cutting­edge of microscopy.<br />

Financial Assistance in order to support<br />

their continuing education, RMS<br />

members are entitled to a substantial<br />

discount on all course and meetings fees.<br />

Offering further assistance to young scientists<br />

who are members, the RMS encourages<br />

them to attend conferences in<br />

both the UK and abroad through a generous<br />

bursary scheme.<br />

RMS Learning Zone at Microscience<br />

2008<br />

One of the unique features of Microscience<br />

is the RMS Learning Zone, which<br />

provides a free ‘taster’ to RMS courses.<br />

The Learning Zone aims to provide a<br />

friendly environment for absolutely anyone<br />

to drop in and get help with all aspects<br />

of electron and light microscopy.<br />

This makes it an ideal place to visit for<br />

those just starting out and for others<br />

wishing to learn more about unfamiliar<br />

techniques.


News from the RMS<br />

RMS endorses university courses:<br />

The first Imaging MSc kicked off at Oxford Brookes<br />

University this September. As it is RMS endorsed,<br />

all students on the course gain free membership.<br />

Any post-graduate microscopy/imaging related<br />

courses are eligible for RMS endorsement and the<br />

Society would be happy to discuss such backing<br />

with any interested universities and institutes.<br />

Call for Microscience 2008 papers:<br />

Building on the success of Microscience 2004 and<br />

2006, London‘s ExCeL will once again host Europe‘s<br />

premier international conference and exhibition<br />

on the science of microscopy, imaging and<br />

analysis on 23–26 June 2008.<br />

First call for papers October 2007 – Deadline for<br />

contributed oral papers 23:00 GMT February 29 th<br />

2008<br />

“Having a good grasp of the basic theories<br />

and practices of microscopy is a vitally<br />

important step to carrying out<br />

meaningful research, and we are especially<br />

keen to support researchers in the<br />

early stages of their careers,” says Debbie<br />

Stokes, RMS Honorary Secretary Science<br />

(Physical). “Our internationally renowned<br />

Learning Zone volunteers will be<br />

on hand to generously pass on their experience<br />

and knowledge to the future<br />

generation of microscopists. This is such<br />

a valuable opportunity and, with the introduction<br />

of Microscience Early Stage<br />

Researcher Bursaries, we hope to see<br />

many academic and industrial researchers<br />

from Europe and around the world.”<br />

Contact:<br />

Allison Winton<br />

Royal Microscopical Society<br />

St Clements, United Kingdom<br />

Tel.: +44 1865 254760<br />

Fax: +44 1865 791237<br />

allison@rms.org.uk<br />

www.rms.org.uk<br />

www.microscience2008.org.ukv<br />

Organizing ELMI 2008 Has Already Started<br />

The next venue for the annual ELMI meeting will be the congress center in Davos, Switzerland from May 27<br />

to 30, 2008. Basis for this decision was not only the nice environment as a promotion for the Swiss countryside<br />

but the very good experience in running the Microscopy Conference late summer in 2005 (see Imaging<br />

& Microscopy November 2005 issue).<br />

In respect to the strong interest of the organizers<br />

that the ELMI meeting will keep<br />

its very special attitude as a workshop<br />

based unique event with a lot of handson<br />

sessions, they invited their partners to<br />

a planning meeting directly at the venue.<br />

The purpose was to inspect the congress<br />

center concerning the needs for appropriate<br />

space for manufacturers system<br />

installations and further infrastructure.<br />

In agreement with the attendant companies,<br />

Carl Zeiss MicroImaging, Leica Microsystems,<br />

Life Imaging Services, Nikon,<br />

and Olympus the whole congress center<br />

will be rent for the 2008 event.<br />

To make your own planning to attend<br />

the conference or to save your company’s<br />

workshop slot we like to recommend to<br />

get in contact with the organizers as listed<br />

in the box below. We are looking forward<br />

to meeting you in springlike Davos.<br />

Martin Friedrich<br />

<strong>GIT</strong> VERLAG, A Wiley Company<br />

Secretary:<br />

Gabriele Gruber<br />

Gabriele.gruber@fmi.ch<br />

Industry Contacts:<br />

Patrick Schwarb<br />

Patrick.schwarb@fmi.ch<br />

ELMI Board Member Contacts:<br />

Jens Rietdorf<br />

jens.rietdorf@fmi.ch<br />

Scientific Committee:<br />

Nathalie Garin<br />

Nathalie.garin@epfl.ch<br />

Gabor Csucs<br />

csucs@bc.biol.ethz.ch<br />

Finances:<br />

Gianni Morson<br />

Gianni.morson@unibas.ch<br />

R M S I n F o c u S<br />

Meeting Contact:<br />

Markus Dürrenberger<br />

markus.duerrenberger@unibas.ch<br />

Media Partner Imaging & Microscopy:<br />

Martin Friedrich<br />

m.friedrich@gitverlag.com<br />

G.I.T. Imaging & Microscopy 4/2007 • 13


C o m pa n y n e w s<br />

Nanosolution Center Opening<br />

Invited by Carl Zeiss AG and the Photonics<br />

BW technology network, German Federal<br />

Minister for Education and Research Dr.<br />

Annette Schavan visited on 7 th of September<br />

Oberkochen. While there, she opened<br />

the Nanosolutions Center, the largest and<br />

most advanced demo center for cuttingedge<br />

microscopy in Germany. During her<br />

address to the 250 guests, she emphasized<br />

the value of optical technologies for the<br />

competitiveness of the location: “These<br />

technologies drive important innovations<br />

and are clearly among the future technologies<br />

of the 21 st century. They provide key<br />

impulses for Germany. The Federal Ministry<br />

of Education and Research (BMBF)<br />

recognized the significance of optical technologies<br />

at an early stage and have specifically<br />

promoted their expansion. These<br />

technologies are a central element in the<br />

government’s high-tech strategy.”<br />

www.smt.zeiss.com<br />

Improving Europe’s Image<br />

The European Science Foundation calls for greater<br />

collaboration across Europe on research in medical<br />

imaging. New imaging technologies will result in<br />

improved and cost-effective healthcare, the ESF<br />

says, but there needs to be closer cooperation between<br />

doctors, scientists and industry if Europe is<br />

to realise the full potential of new developments<br />

and remain competitive globally. The call comes in<br />

a new ESF Science Policy Briefing (SPB) released<br />

this week. The briefing is the result of a workshop<br />

attended by key experts in the field organised by<br />

ESF’s medical section, the European Medical Research<br />

Councils (EMRC). Imaging is one of the fastest<br />

growing areas within medicine.<br />

www.esf.org<br />

Harvard Apparatus Acquires<br />

PanLab<br />

Harvard Apparatus has announced its acquisition of<br />

Panlab, a distributor and manufacturer of products<br />

and software for the life sciences researcher, primarily<br />

in the neuroscience research market. Harvard<br />

Apparatus now offers a full line of products for: activity<br />

and exploration, depression studies, motor<br />

function and coordination, anxiety studies, learning<br />

and memory, pain and inflammation , video tracking,<br />

respiratory metabolism, food and liquid monitoring,<br />

social interactions and phenotyping.<br />

www.harvardapparatus.com<br />

14 • G.I.T. Imaging & Microscopy 4/2007<br />

From left to right: Dr. Hermann Gerlinger, Member of the Board at Carl Zeiss SMT AG, German<br />

Federal Minister for Education and Research Dr. Annette Schavan, President and CEO of Carl Zeiss AG<br />

Dr. Dieter Kurz, and Dr. Dirk Stenkamp, Member of the Board at Carl Zeiss SMT AG.<br />

JPK in Asia-Pacific Region<br />

JPK Instruments has strengthened its marketing position<br />

in the booming Asia-Pacific region: The company<br />

has granted the exclusive distribution rights<br />

for its products in India to Inkarp Instruments, and<br />

in Australia to Scitech. In addition to Canada, Brazil<br />

and the European market, JPK Instruments’ market<br />

presence now covers Australia, India, Taiwan, China,<br />

Korea, Japan, Singapore and Thailand.<br />

www.jpk-instruments.de<br />

Palm Merged with Carl<br />

Zeiss Microimaging<br />

On 24 September 2007 Palm Microlaser Technologies,<br />

a 100 % subsidiary of Carl Zeiss Microimaging,<br />

was merged with Carl Zeiss Microimaging. All current<br />

activities in the field of laser microdissection<br />

will be continued on an ongoing basis. Under the<br />

company name Carl Zeiss Microimaging, all former<br />

contacts can be reached under the same address<br />

data from the Bernried location in the Microdissection<br />

business field. With its Microdissection business<br />

sector the company is now the sole manufacturer<br />

of laser microdissection and micromanipulation<br />

systems using patent protected LMPC technology.<br />

The systems are used both in biomedical and clinical<br />

research as well as in routine applications.<br />

www.zeiss.de/microbeam<br />

Gold Medal for Donal Denvir<br />

Dr. Donal Denvir, co-founder of Andor Technology<br />

has been recognized with a gold medal from the Institute<br />

of Physics. The Business and Innovation<br />

medal of the Institute of Physics is awarded to individuals<br />

for outstanding contributions to the organisation<br />

or application of physics in an industrial or<br />

commercial context. Dr Denvir’s award in recognition<br />

for his role in founding Andor Technology<br />

which manufactures high-performance digital cameras,<br />

and for leading an R&D programme.<br />

www.andor.com


R&D 100 Award for Zeiss Electron Microscope<br />

Carl Zeiss SMT was honoured with a coveted R&D 100 Award for one of the 100<br />

most technologically significant new products in 2007. R&D Magazine recognized<br />

the company’s Scanning Transmission Electron Microscope (STEM) for its<br />

ability to advance the field of materials science and research. The advanced version<br />

of the Libra 200 STM enables scientists and researchers to analyze materials<br />

at the atomic level. Nanostructures can be viewed with imaging resolutions<br />

and analytical capabilities never before possible in one single instrument.<br />

www.smt.zeiss.com<br />

JPK Fastest Growing Nanotech Company<br />

JPK Instruments is the fastest growing company of the nanotech industry on the<br />

Deloitte Technology Fast 50 ranking, which has been established for the fifth<br />

consecutive year. The company develops, produces and markets essential base<br />

technologies enabling unique analytical access at an atomic and molecular level.<br />

The leading companies in Germany’s high-tech industries were determined on<br />

the basis of their compounded percentage sales growth rates of the past five<br />

years. JPK achieved a growth rate of over 970 %.<br />

www.jpk-instruments.de<br />

Leica Selling BioSystems Products in Sweden<br />

Leica Microsystems announced that it has signed a purchase contract for the<br />

rights to sell products for the former Vision BioSystems into the Swedish market<br />

thereby establishing a stronger European sales organization for the efficient<br />

support of ist pathology and diagnostics business. Leica Microsystems and the<br />

Swedish company Immunkemi announced the signing of this purchase contract<br />

on October 1, 2007. For the last seven years, Immunkemi had represented the<br />

former Australian Vision BioSystems as the Swedish distributor for its specimen<br />

preparation instruments as well as reagents and antibodies.<br />

www.leica-microsystems.com<br />

Leica to Sell Expression Pathology’s Slides<br />

Rapid, gentle and contamination free laser microdissection, directly into sample<br />

buffer, is now possible with the combination of Leica Microsystems’ laser microdissection<br />

system Leica LMD6000 and Expression Pathology’s Director glass<br />

slides. A sales agreement between the two companies now gives scientists the<br />

opportunity to buy both the instrument and the slides from Leica Microsystems<br />

worldwide. The slides bring a new level of speed and accuracy to laser microdissection,<br />

allowing collection directly into a vial, while eliminating the need for<br />

plastic membrane-coated slides or sticky caps. These membrane-free slides employ<br />

a proprietary energy transfer coating bonded to a glass support. UV-laser<br />

energy is converted to kinetic energy upon striking the energy transfer coating,<br />

vaporizing it, thereby propelling selected features into the vial.<br />

www.leica-microsystems.com<br />

Image Reconstruction Software Successful<br />

in Hospital<br />

Syncroscopy has announced Auto-Montage Pro, its 3D image reconstruction<br />

software, has been successfully used at a major UK teaching hospital, Addenbrooke’s<br />

Hospital, part of the Cambridge University Hospitals group, (UK), to<br />

help confirm diagnosis of schistosomiasis, a parasitic disease rarely seen in the<br />

UK. Pathologists in the Department of Histopathology used the software as a diagnostic<br />

tool to produce in-focus microscope images of the abnormality within<br />

the brain biopsy from an adult patient who had recently developed focal epileptic<br />

seizures. With the help of these images, the eggs of Schistosoma mansoni,<br />

one of several species of flatworm that cause schistosomiasis, could be identified<br />

and characterised. Schistosomiasis is a common chronic disease in Africa<br />

and Asia, where the patient had extensively traveled.<br />

www.syncroscopy.com<br />

Microscope Automation<br />

at Every Stage<br />

If you demand speed, accuracy and<br />

precision from your microscope, take a<br />

look at Prior’s world leading microscope<br />

automation systems for effective (and<br />

cost-effective) solutions. We design and<br />

manufacture the widest range of<br />

scanning stages, filter wheels, shutters,<br />

automated focus units, motor controllers<br />

and illumination systems for end users,<br />

system integrators and OEM’s worldwide.<br />

For further information visit the web at<br />

www.prior.com or email a brochure<br />

request to uksales@prior.com<br />

Focussed on microscopy<br />

Prior Scientific Instruments Cambridge CB21 5ET UK<br />

Telephone +44 (0)1223 881711 www.prior.com<br />

G.I.T. Imaging & Microscopy 4/2007 • 15


A n n o u n c e m e n t<br />

Focus on Microscopy 2008<br />

April 13–16, Osaka-Awaji, Japan<br />

After the successful 2007 FOM conference this year in Valencia, Spain the next conference in the FOM series<br />

will take place in Osaka, Awaji Island, Japan from Sunday, April 13 to Wednesday, April 16, 2008. It will start<br />

around 6 o‘clock in the afternoon on Sunday the 13 th with a plenary opening session followed by a welcome<br />

reception. The program schedule and general information of the conference can be found at the conference<br />

website: FocusOnMicroscopy.org.<br />

The conference location is the Awaji<br />

Yumebutai International Conference<br />

Center/Resort near Osaka. The location<br />

can easily be reached from Kobe and Osaka<br />

airports. All details around registration,<br />

abstract submission and deadlines<br />

etc. is present or will come shortly available<br />

on this website.<br />

A wide range of microscopy and microscopy<br />

related subjects will be addressed.<br />

These range from the advanced<br />

use of fluorescent probes in – live – cellular<br />

biophysics, specialized spatial image<br />

analysis of the resulting images to the<br />

physics of sub-resolution spatial image<br />

formation.<br />

With its origin and focus in sectioned<br />

confocal/2-photon microscopy the conference<br />

also signals important developments<br />

in neighboring fields.<br />

Topics of the FOM conference series<br />

include:<br />

� Confocal and multiphoton-excitation<br />

microscopies<br />

� 3D and 4D live cell and tissue imaging<br />

16 • G.I.T. Imaging & Microscopy 4/2007<br />

�<br />

�<br />

�<br />

�<br />

�<br />

�<br />

�<br />

�<br />

�<br />

�<br />

Novel illumination and detection strategies<br />

– selective plane extended depth<br />

of focus, 4pi, structured illumination<br />

Fluorescence – new labels, fluorescent<br />

proteins, quantum dots, single molecule,<br />

excitation-emission spectroscopy<br />

Time-resolved fluorescence – FRET,<br />

FRAP, FLIM, FCS<br />

Coherent non-linear microscopies –<br />

SHG, THG, SFG, CARS<br />

Scattering processes: Raman, light<br />

scattering spectroscopy, second harmonic<br />

Multi-dimensional imaging<br />

Sub-wavelength resolution – near field<br />

microscopy, total internal reflection<br />

Laser manipulation, ablation and<br />

microdissection, photoactivation<br />

3D Image processing and visualization<br />

Whole tissue imaging – optical coherence<br />

tomography, endoscopy, whole<br />

animal fluorescence<br />

A technical exhibition will accompany<br />

the Osaka-Awaji conference.<br />

Welcoming you to the FOM2008 conference<br />

and exhibition, on behalf of the<br />

FocusOnMicroscopy society<br />

Satoshi Kawata, Katsumasa Fujita, Osaka<br />

University, RIKEN, Wako City, Japan<br />

Fred Brakenhoff, University of Amsterdam,<br />

The Netherlands<br />

The present Focus on Microscopy<br />

2008 conference incorporates<br />

21st International Conf. on 3D Image<br />

Processing in Microscopy<br />

20th �<br />

� International Conf. on Confocal<br />

Microscopy<br />

Contact:<br />

Satoshi Kawata<br />

Katsumasa Fujita<br />

Osaka, Japan<br />

fom2008@ap.eng.osaka-u.ac.jp<br />

Prof. Dr. G.J. Brakenhoff<br />

Section of Molecular Cytology<br />

Centre for Advanced Microscopy<br />

Swammerdam Institute for Life Sciences<br />

University of Amsterdam, The Netherlands<br />

Tel.: +31 20 525 5189<br />

Fax.: +31 20 525 6271<br />

brakenhoff@science.uva.nl


Accessories for Microscopy<br />

Agar Scientific is a leading international supplier<br />

of accessories and specialist equipment for all<br />

disciplines of microscopy.<br />

• Renowned for its technical advice<br />

and fast, efficient, friendly service<br />

• With a worldwide network of<br />

agents and distributors<br />

Request a catalogue or visit our website to<br />

see our extensive range of products.<br />

www.agarscientific.com<br />

Agar Scientific Limited, 66a Cambridge Road, Stansted, CM24 8DA UK<br />

Telephone: +44(0)1279 813519 Fax: +44(0)1279 815106<br />

Email: sales@agarscientific.com<br />

Sub-nanometer resolution<br />

Low settling time<br />

High-precision flexure guidance<br />

QuickLock adapter<br />

Look Sharp!<br />

Highly compact<br />

PIFOC ® —High Dynamics Piezo Nanofocusing<br />

Systems<br />

These extremely precise focusing systems are unique<br />

for their extra-long travel ranges and sub-nanometer<br />

resolution. With their minimal settling times and outstanding<br />

focus stability, they are winning over users in<br />

Life Sciences and Metrology.<br />

■ Travel Ranges up to 460 μm<br />

■ Resolution < 1 nm<br />

■ Linearity to 0.03 %<br />

You, too, can look sharp:<br />

COMPAMED · Hall 8a · Stand F29<br />

Physik Instrumente(PI) GmbH & Co.KG · Tel. +49-721-4846-0<br />

G.I.T. Imaging & Microscopy 4/2007 • 17


E v E n t R E p o R t<br />

See You Later Alligator<br />

Traditionally, one of the most important events for presenting new developments in high end microscopy instrumentation is the Microscopy & Microanalysis Congress<br />

and Fair in the United States. Reason enough for the Imaging & Microscopy team to join this year’s exhibition at the Broward County Convention Center in<br />

Fort Lauderdale, Florida. In contrast to the very hot temperatures and the incredible humidity outside, the Convention Center provided an appropriate cooled exhibition<br />

floor. Following the challenge to increase visibility in nanostructures multitude up-to-date products in hardware, software and accessories were offered<br />

by 100 manufacturers. In this section we would like to take you on our trip through this tradeshow by introducing some products and the people behind them.<br />

Dr Stefan Scherer, President of Alicona Imaging<br />

presenting the InfiniteFocus; an optical 3D measurement<br />

device for quality assurance in the micro- and<br />

nano range.<br />

Eric C. Ambrose, Carestream Healths Media Product<br />

Manager for Molecular Imaging Systems with the In-<br />

Vivo Imaging System FX. This instrument combines<br />

high-sensitivity Optical Molecular Imaging and high<br />

resolution Digital X-ray to deliver anatomical<br />

localization of molecular and cellular biomarkers.<br />

Mike Sousa and Nicole Lackey from Evex presenting<br />

the LN-free violin-shaped QD Detector to be applied<br />

for spectral acquisition and elemental mapping.<br />

18 • G.I.T. Imaging & Microscopy 4/2007<br />

Alex Vogt (left), President, and Dr Andres Kaech<br />

(sitting), Scientific Director of Life Science Applications<br />

of Bal-Tec introducing the HPM 100, a high<br />

Pressure Freezing work station for cryo fixation.<br />

Stacie G. Kirsch, Managing Director of Diatome U.S.,<br />

showing the Lynx II; an automated tissue processor<br />

for histology and microscopy from Electron Microscopy<br />

Science.<br />

The desktop electron microscope Phenom with touch<br />

screen control from FEI Company introduced by JJ<br />

Blackwood (left), Senior Application Development<br />

Engineer and Matthew Harris (right), Vice President<br />

and General Manager NanoBiology Market Division.<br />

Bruker AXS Microanalysis Marketing Communications<br />

Manager Stefan Langner in front of the poster<br />

of HyperMap/PTS, a software interface for fast<br />

spectral mapping based on “position-tagged<br />

spectrometry” technique.<br />

Christine Meehan, Marketing Communication<br />

Specialist and Mark Massey European Director of<br />

Sales flanking Edax WDS TEXS HP, a parallel beam<br />

spectrometer that employs capillary optics enabling<br />

the spectrometer to have an energy range from<br />

100 eV to 10 keV.<br />

President Paul Fischione (right) and Director of Market<br />

Development Alan Robins (left) from Fischione Instruments<br />

with their 1040 NanoMill device used to create<br />

thin specimens needed for advanced transmission<br />

electron microscopy imaging and analysis.


<strong>Huge</strong> <strong>Images</strong>? No Problem<br />

Introducing Imaris® 6.0<br />

Interactive Visualization, Quantification, Tracing and Tracking<br />

Imaris® 6.0 shatters the barriers that cause other<br />

programs to fail by allowing users to visualize and<br />

process 3D and 4D microscopy data sets of up to<br />

30 GB in size. Through constant innovation, Bitplane<br />

has shaped the way microscopists work with<br />

images for the past 12 years. The feature-filled<br />

Imaris® version 6.0 release will be no exception.<br />

For more information visit: www.bitplane.com<br />

T h e i m a g e r e v o l u t i o n s t a r t s h e r e .<br />

Bitplane‘s suite of software modules provides users<br />

with an opportunity to build a package based on<br />

their specific needs. Imaris® is available for Windows<br />

x32 and x64 systems and both Intel and<br />

PowerPC based Macs. There is no longer a need to<br />

improvise. Bitplane offers customers a complete<br />

solution.


E v E n t R E p o R t<br />

Dr Christel Genoud, 3View Product Specialist and<br />

John Hyun, Marketing Communications Manager<br />

from Gatan in front of application images derived<br />

from their 3View-system. An ultra-microtome, stage<br />

and imaging system, which allows serial block face<br />

scanning electron microscopy within a variable<br />

pressure field emission SEM.<br />

Kenny Witherspoon (right), IXRF Systems Vice<br />

President of Marketing demonstrating their fully<br />

automated EDS systems including quantitative analysis,<br />

high resolution digital imaging, X-ray mapping,<br />

X-ray linescans, and particle analysis.<br />

Ian Lamswood (left), Marketing Manager EM<br />

Products and Ann Korsen (mid), Director of Sales<br />

and Marketing Ultramicrotomy from Leica Microsystems<br />

explaining their automatic microwave tissue<br />

processor for electron microscopy EM AMW to Judi<br />

Stasko (right), National Animal Disease Center,<br />

Ames, IA, USA.<br />

Joel Silfies (left), Senior Applications Manager and<br />

Marty Whitted (right), Sales Representative Industrial<br />

Microscopy & Metrology flanking the AZ100<br />

Multizoom Macro System from Nikon.<br />

20 • G.I.T. Imaging & Microscopy 4/2007<br />

Chris Haig (left), Regional Manager USA Midwest<br />

and Bill Moore (right), Business Manager of System<br />

Division Hamamatsu presenting their electron<br />

multiplier CCD camera ImageEM suitable for high<br />

dynamic range applications to dim fluorescence in<br />

living cells.<br />

Toshiyuki Kanazawa (sitting), Senior SEM Application<br />

Specialist and Vernon E. Robertson (standing), Field<br />

Emission SEM Product Manager from Jeol introducing<br />

the JSM-7500F, an analytical Field Emission SEM<br />

achieving a resolution of 1.4 nm at 1 kV.<br />

Claudia Dunphy, Marketing & Communication<br />

Manager and Brian Graydon, Business Development<br />

Manager Scientific Division from Lumenera showing<br />

the Infinity CCD Camera series for colour and monochrome<br />

images with resolution array from 1,3 to 21<br />

megapixel.<br />

President Dr Thomas Moore (left) and Senior<br />

Mechanical Designer Rocky Kruger (right) from<br />

Omniprobe surrounding OmniGIS the multiple gas<br />

injection device for FIB & SEM.<br />

Michael Dixon, Sales & Business Development<br />

Manager from Hitachi showing the HD-2700, a<br />

STEM which is equipped with a spherical aberration<br />

corrector.<br />

The VHX digital microscope from Keyence containing<br />

54 million pixel 3CCD handheld camera presented<br />

by Thomas Takao (right), Product Sales<br />

Director and Katz Muta (left), Japanese Account<br />

Manager.<br />

Roy Opie (mid), Publisher and Julian Heath (right),<br />

Head Editor from the Wiley Journal Microscopy &<br />

Analysis while talking with Debbie Stokes (left)<br />

from the Royal Microscopical Society.<br />

The 2 k x 2 k pixel side-mounted TEM CCD camera<br />

Veleta presented by Heidi Mills from Olympus Soft<br />

Imaging Solutions Sales Support/Logistics.


Sales Manager Mike Wombell showing Quorum<br />

Technologies PP2000/PP2000T Cryo-SEM System<br />

featuring rapid sample freezing, vacuum transfer,<br />

freeze etching, and sputter coating.<br />

Jack Vermeulen, Head of Sales and Marketing from<br />

Ted Pella offering a broad range of Specimen<br />

Mounts for SEM, including the SEMClip series.<br />

The World’s highest resolution camera for TEMs (8 k,<br />

16 µm, 14 bit) TemCam-F816 presented by TVIPS<br />

President Dr Hans R. Tietz (right) and Dr Matthias<br />

Stumpf (left), Product Development Manager.<br />

Sales Representative Erica Byrd from Wiley USA<br />

offering the broad range of the Publisher’s textbooks<br />

and journals concerning Microscopy and<br />

Microanalysis.<br />

Tim Prusnick, Application and Support Engineer for<br />

raman products introducing fast chemical imaging<br />

using StreamLine technology for Renishaw’s inVia<br />

Raman microscopes<br />

TESCAN’s FESEM series Mira with a Schottky Field<br />

Emission electron gun demonstrated by Tony Owen<br />

(left), and William J. Mershon (right), Application<br />

Manager.<br />

Martin Klein President of Visitec Microtechnik<br />

introducing the large chamber scanning electron<br />

microscope Mira used for non-destructive testing.<br />

Doug D’Arcy, USA Sales Representative from Witec<br />

introducing the Alpha 300 series of scanning nearfield<br />

optical microscopes (SNOM) that combine<br />

SNOM, confocal microscopy and AFM in single<br />

instruments.<br />

E v E n t R E p o R t<br />

Vice President Eugene E. Rodek from SPI Supplies<br />

presenting the Osmium Plasma Coater for SEM and<br />

FESEM samples.<br />

Fran Ebert, Senior Marketing Communication<br />

Specialist Scientific Instruments and David Rohde,<br />

Product Manager X-Ray Microanalysis Scientific<br />

Instruments showing the Thermo Fisher Scientific<br />

UltraDry Silicon Drift Detector.<br />

Confocal Product Manager Layla Billowitz from VTI<br />

Visitech in front of the VT Eye poster. Using Acousto<br />

Optical Deflector (AOD) technology this confocal<br />

point-scanner is designed for Multi-colour acquisition<br />

for FRET, FRAP and FLIP applications.<br />

Nick Economou (left), Board Member from ALIS, a<br />

subsidiary from Carl Zeiss AG and Dr. Peter Fruhstorfer<br />

(right) Director International Sales, Service &<br />

Business Development from Carl Zeiss NTS presenting<br />

the Helium ion microscope Orion.<br />

G.I.T. Imaging & Microscopy 4/2007 • 21


E v E n t r E p o r t<br />

At the Region of Former Ironworks<br />

Microscopy Conference (MC) 2007<br />

At the university campus of Saarbrücken, the capital of Saarland, Germany’s smallest federal state, well<br />

known in the past for its iron producing industry, with the world cultural heritage “Völklinger Hütte”, the<br />

33 rd conference of the German Society of Electron Microscopy (DGE) took place between 2 nd and 7 th of September.<br />

In total 450 delegates were offered to attend<br />

21 oral sessions consisting of 130<br />

talks, four poster sessions with 120 contributions,<br />

one technical lecture including<br />

16 exhibitors presentations and several<br />

workshops. The exhibition floor,<br />

connecting the lecture halls, areas for<br />

poster sessions, coffee bar and registration,<br />

presented 30 companies introduc­<br />

ing their product novelties in Hardware,<br />

Software and consumables to the audience.<br />

Following the welcome and followed<br />

by the first plenary lecture one of the<br />

most prestigious Prizes for outstanding<br />

scientific developments in electron microscopy,<br />

the Ernst Ruska Prize was<br />

awarded this year by Paul Walter (Ulm,<br />

Fig. 1: Ernst Ruska Prize 2007 Awarding. Paul A. Midgley, Hiroshi Jinnai, Richard J. Spontak, and Paul<br />

Walter.<br />

22 • G.I.T. Imaging & Microscopy 4/2007<br />

World cultural heritage “Völklinger Hütte”<br />

Germany) president of the DFE to Richard<br />

J. Spontak (Berkeley, USA), Hiroshi<br />

Jinnai (Kyoto, Japan) and Paul A. Midgley<br />

(Cambridge, UK) (Fig. 1). The Jury<br />

honoured the three Scientists and MC<br />

2007 plenary lecturers for their work on<br />

novel and quantitative uses of electron<br />

tomography in the 3D study of nanostructured<br />

materials. Further plenary<br />

talks were given by:<br />

� Andrew Bleloch, Cambridge, UK (Applications<br />

of Aberration Corrected<br />

STEM: Imaging and EELS)<br />

� Hans Cerva, Munich, Germany (Application<br />

of Selected Electron Microscopy<br />

Methods to Materials Analysis Problems<br />

from an Industrial Environment)<br />

� Marek Cyrklaff, Martinsried, Germany<br />

(Cryo­Tomography of Whole Cells)<br />

� Jacques Dubochet, Lausanne, Switzerland<br />

(Cryo­electron Microscopy of<br />

Vitreous Sections)<br />

� Gareth Griffiths, Heidelberg, Germany<br />

(State of the Art Microscopy for Cell<br />

Biology with a Focus on Mycobacteria<br />

and Phagocytosis)<br />

� Othmar Marti, Ulm, Germany (Nanomechanical<br />

Characterization from<br />

�<br />

Living Cells to the Cytoskeleton)<br />

Andreas Thust, Juelich, Germany<br />

(High­Resolution Transmission Electron<br />

Microscopy Entering the Sub­<br />

Angstrom Era)


�<br />

Roger Wepf, Zurich, Switzerland (Correlative<br />

Microscopy in Focus: Shortcuts,<br />

Prerequisites and Future Needs<br />

in Diagnostic and Analytical Biomedical<br />

Structure Research)<br />

For the social programme delegates were<br />

first invited on September 4 th to the FEI<br />

Company and Carl Zeiss SMT customers<br />

evening event at the Congress Centre<br />

Saarbrücken and “Völklinger Hütte”, respectively.<br />

The following evening was reserved<br />

for the congress dinner at the<br />

“Alte Schmelze” (eng. Old Smeltery) in<br />

St. Ingbert a further monument to former<br />

ironworks close to the venue. But before<br />

the participants could enjoy the delicacies<br />

of the region, a fantastic organ performance<br />

in St. Hildegards Church took<br />

the audience on a trip through compositions<br />

of Johann Sebastian Bach, Charles­<br />

Marie Widor, Louis Vierne, Wolfgang<br />

Amadeus Mozart, and Naji Hakim.<br />

To all who enjoyed this successful and<br />

very informative event hosted by the<br />

DGE, the next Microscopy Conference coorganised<br />

by them is the European Microscopy<br />

Congress in Aachen, Germany<br />

2008 and the “Dreiländertagung” in<br />

Measurement of steep flanks and complex geometries<br />

True color information registered to 3D data<br />

Traceable results even on highly sculptured surfaces<br />

Usable on highly reflective and<br />

inhomogeneous surfaces<br />

Highest resolution across measurement<br />

areas of several mm<br />

Comprehensive roughness<br />

measurement capabilities, conforming<br />

to the latest ISO standards<br />

Usable in the lab and as<br />

Inline measurement sensor<br />

Fig, 2: Heads of local organizing committee: Pedro Mesters-Ventura and Uwe Hartmann<br />

Graz, Austria 2009. Hopefully see you all<br />

there!<br />

Contacts:<br />

Prof. Dr. Uwe Hartmann<br />

Institute of Applied Physics<br />

University of Saarland, Saarbrücken, Germany<br />

Tel.: +49 681 302 3798<br />

Fax: +49 681 302 3790<br />

u.hartmann@mx.uni-saarland.de<br />

OPTICAL MEASUREMENT AND INSPECTION<br />

E v E n t r E p o r t<br />

Prof. Dr. Pedro Mestres-Ventura<br />

Institut für Anatomie und Zellbiologie<br />

Zentrum für Elektronenmikroskopie<br />

Universitätsklinikum, Homburg/Saar, Germany<br />

Tel.: +49 6841 1626141<br />

Fax: +49 6841 1626293<br />

anpmes@uniklinikum-saarland.de<br />

www.dge-homepage.de<br />

www.uni-saarland.de/mc2007<br />

Alicona Imaging GmbH<br />

headquarters<br />

Austria, Teslastraße 8<br />

A-8074 Grambach/Graz<br />

phone: +43 316 4000-700<br />

fax: +43 316 4000-711<br />

e-mail: info@alicona.com<br />

G.I.T. Imaging & Microscopy 4/2007 • 23


E l E c t r o n M i c r o s c o p y<br />

Enabling 360 degree TEM/STEM<br />

of Nanoparticles<br />

Functionalized Holders for 3D Electron Microscopy<br />

A new protocol for functionalizing sample holders has been developed for 360° TEM/STEM observation of<br />

nanoparticles and nanostructures. The three step process includes FIB milling to customize sample stub geometry,<br />

thin film deposition for substrate selection and subsequent chemical functionalization for nanoparticle<br />

adhesion. This protocol was used to determine the morphology and local material properties of individual<br />

Au/SiO 2 core-shell nanoparticles used in a DNA detection assay.<br />

Nanoscience Imaging & Spectroscopy<br />

Tools for characterizing nanoparticles<br />

with complex three-dimensional (3D)<br />

geometrics are required to further nanoscience<br />

research and enable the continuing<br />

development of viable nanotechnologies.<br />

In this manuscript we report a<br />

protocol which combines focused ion<br />

beam milling (FIB), thin film deposition<br />

and solution chemistry to customize a rotation<br />

holder [1] for 3D structural and<br />

chemical analysis of nanoparticles with<br />

transmission, and scanning transmission<br />

electron microscopy (TEM and STEM).<br />

By using this protocol the geometry, sub-<br />

Fig. 1: A. A novel sample<br />

holder allowing for 360°<br />

rotation in a TEM/STEM [6].<br />

The location of the sample<br />

to be analyzed is indicated<br />

by the arrow. B. Illustrated<br />

geometry of rotation<br />

holder sample, incident<br />

electron beam and analytical<br />

detectors.<br />

24 • G.I.T. Imaging & Microscopy 4/2007<br />

strate material and chemistry of the<br />

STEM or TEM rotation holder can be optimized<br />

for the nanoparticle system of interest.<br />

To illustrate this protocol, 3D<br />

STEM imaging rotation holders were<br />

modified for analysis of core-shell nanoparticles<br />

used in a DNA detection assay.<br />

The details of the Au/SiO 2 core-shell nanoparticle<br />

synthesis and resulting increased<br />

DNA detection efficiency are reported<br />

elsewhere [2].<br />

Electron microscopy techniques are<br />

widely used to characterize morphology<br />

and electronic properties of materials at<br />

the near-atomic scale but are not well<br />

suited for visualizing complex structures<br />

Keywords:<br />

nanoscience, nanoanalysis, 3D STEM, functionalized<br />

holder, bionanotechnology<br />

in 3D [3, 4]. A nanoparticle must be either<br />

tilted or rotated with respect to the<br />

electron beam to enable multiazimuthal<br />

observation and 3D analysis. This requires<br />

a “nanoparticle holder” which can<br />

be either rotated or titled in a TEM or<br />

STEM. A 3D rotation holder, with sample<br />

location indicated by the arrow in Figure<br />

1A, allows analysis from a full 360 degrees<br />

as opposed to traditional tomographic<br />

tilt-series based analysis which<br />

suffers from the missing wedge [5]. Unfortunately<br />

the scanning electron microscopy<br />

(SEM) and focused ion beam (FIB)<br />

based nano-manipulation techniques traditionally<br />

used for attaching samples to<br />

such rotation holders are not well suited<br />

for mounting individual nanoparticles<br />

[6–8]. Due to the small particle size and<br />

the practical difficulties of attaching and<br />

depositing individual particles with nanomanipulators<br />

a new technique is<br />

needed.<br />

To enable 3D STEM or TEM analysis<br />

of nanoparticles a three step process was<br />

developed to functionalize the brass needle<br />

stubs used in the rotation holder. In


C U T T I N G E D G E<br />

T E C H N O L O G Y D E L I V E R I N G<br />

U N I Q U E S O L U T I O N S<br />

For further information please visit us at<br />

www.smt.zeiss.com/nts<br />

Enabling the Nano-Age World ®<br />

Carl Zeiss SMT<br />

Nano Technology Systems Division<br />

Carl-Zeiss-Str. 56<br />

73447 Oberkochen<br />

Germany<br />

C a r l Z e i s s S M T – N a n o T e c h n o l o g y S y s t e m s D i v i s i o n<br />

ZEISS CrossBeam ® Solutions<br />

The ultimate 3D research tool that combines FIB and<br />

GEMINI ® column to one extraordinary powerful system<br />

with new dimensions in analytical science.<br />

Combining high precision, unique live imaging and perfect handling.<br />

NEON ® 40, 40EsB, 60 and NVision 40 – unlimited analytical capabilities for all requirements<br />

Tel. +49 73 64 / 20 44 88<br />

Fax +49 73 64 / 20 43 43<br />

info-nts@smt.zeiss.com<br />

www.smt.zeiss.com/nts<br />

EDITORS’ EDITORS’<br />

CHOICE<br />

Best Product<br />

2007


E l E c t r o n M i c r o s c o p y<br />

Fig. 2: A. Brass sample stub before modification.<br />

B. Sample stub after FIB modification.<br />

the first step a FIB is used to customize<br />

the geometry of a brass needle stub.<br />

Then a ion beam sputter-deposition system<br />

(Gatan 682 PECS) is used to coat the<br />

surface of the needle stub with a substrate<br />

material ‘matched’ to the nanoparticle<br />

of interest. In the final step, the<br />

surface chemistry of the stub or nanoparticle<br />

is optimized to control the adhesion<br />

between these two surfaces.<br />

To demonstrate this protocol, functionalized<br />

rotation holders were created<br />

to enable 3D STEM imaging and analysis<br />

of core-shell nanoparticles used in a DNA<br />

detection assay [2]. The functionalized<br />

holders were used to rotate individual<br />

Au/SiO 2 nanoparticles a full 360° with respect<br />

to the electron beam of the STEM<br />

as illustrated in figure 1B. High angular<br />

annular darkfield (HAADF or Z-contrast<br />

(ZC)), bright-field (BF) and secondary<br />

electron (SE) STEM signals were recorded<br />

at regular angular intervals to<br />

study the 3D morphology and composition<br />

of the nanoparticle. Electron energy<br />

loss spectroscopy (EELS) spectrum images<br />

[9] were recorded at several angular<br />

intervals to map optoelectronic and<br />

material properties of the core-shell nanoparticle.<br />

Functionalized Holder Preparation<br />

The sample stub used for 360° rotation in<br />

a STEM/TEM is composed of brass and<br />

measures 3 mm in length along the rotation<br />

axis, ~300 um in diameter and is tapered<br />

to a point at the tip with a 30 um diameter<br />

flat (fig. 2A). The flat at the apex<br />

is well suited for attaching large micronsized<br />

samples with FIB or SEM based<br />

nano-manipulation techniques. However<br />

these techniques are not well suited for<br />

nanoparticles and the roughness of the<br />

30 um flat can easily obscure the nanoparticles<br />

from the electron beam at certain<br />

angles of rotation. To improve the<br />

sample stub geometry, a FIB was used to<br />

customize the shape of the holder apex.<br />

For holding the 100 nm core-shell nanoparticles<br />

used in the DNA assay, the<br />

30 um flat was FIB milled into a 5 x 5 array<br />

of pillars. This array of pillars minimizes<br />

potential shadowing from the substrate<br />

during rotation and increases the<br />

number of potential sites well suited for<br />

TEM/STEM analysis of individual nanoparticles.<br />

The apex of the rotation holder<br />

before and after FIB modification is<br />

shown in figures 2A and B. A variety of<br />

custom holder geometries can created<br />

using FIB modification and application<br />

specific solutions are only limited by creativity.<br />

The nanoparticle system of interest<br />

dictates which substrate materials are<br />

best suited for controlling of the adhesion<br />

and disperion of the nanoparticles.<br />

Fortunately there are a variety of thin<br />

film deposition techniques and materials<br />

which can be used to coat the FIB modified<br />

brass sample stub with a well<br />

‚matched‘ substrate material. For the silica<br />

core-shell particles, a 10–20 nm thick<br />

silica film was deposited on the stub using<br />

a Gatan 682 PECS ion beam sputter<br />

coater. By coating the stub with silica, solution<br />

chemistry could be varied in a<br />

predicatible way to control the adhesion<br />

and dispersion of the core-shell particles<br />

used in the DNA assay.<br />

The nanoparticles were deposited<br />

onto the coated and FIB modified sample<br />

holder by placing a 20 uL droplet of the<br />

Fig. 3: A. and B. STEM SE<br />

and ZC micrographs<br />

showing many nanoparticles<br />

adhered onto a<br />

functionalized sample<br />

holder. The large number<br />

of nanoparticles make<br />

STEM/STEM analysis of an<br />

individual nanoparticle<br />

difficult.<br />

nanoparticles in solution onto the apex of<br />

the customized sample holder. Several<br />

nanoparticle solution chemistries, from<br />

Ref. [2], were tested to determine which<br />

solution resulted in the best adhesion,<br />

dispersion and produced minimal beam<br />

induced carbon deposition. For some solutions,<br />

several layers of nanoparticles<br />

adhered to apex, or analysis region of<br />

the pillars as shown in figures 3A and B.<br />

For other solutions, the nanoparticlesubstrate<br />

adhesion was reduced, and individual<br />

nanoparticles were found near<br />

the apex of several pillars as shown in<br />

Figure 4A. The effect of solution chemistry<br />

on the surface charge of the nanoparticle<br />

and substrate silica is thought to explain<br />

these differences in adhesion and<br />

dispersion, though this has not yet been<br />

independently verified. This example of<br />

functionalizing sample holders for a particular<br />

nanoparticle system can be<br />

adapted for many different types of nanoscience<br />

applications by customizing<br />

the holder geometry, the composition of<br />

the thin film coating and/or by modifying<br />

the surface chemistry of the sample stub<br />

for the system of interest.<br />

360° Rotation STEM Imaging<br />

The ability to rotate individual nanoparticles<br />

360° with respect to the incident electron<br />

beam greatly simplifies nanoscale<br />

structural characterization. The micrographs<br />

of figure 4A-D show a single silica<br />

core-shell nanoparticle which adhered<br />

near the apex of a functionalized 3D<br />

holder. The size, shape, surface features<br />

and chemical composition of this core-sell<br />

nanoparticle was mapped in 3D by acquiring<br />

STEM ZC, BF, and SE images<br />

Fig. 4: A. Functionalized sample holder with individual nanoparticles adheared near the apex, well suited for STEM/TEM analysis. B. Single core-shell nanoparticle<br />

(arrow) attached to apex of functionalized stub (STEM BF). C. STEM SE. D. STEM ZC.<br />

26 • G.I.T. Imaging & Microscopy 4/2007


while rotating this nanoparticle a full<br />

360°. Z-contrast images from another<br />

core-shell nanoparticle, containing two<br />

Au cores, acquired at 20° intervals are<br />

shown in figure 5. The dual core nanoparticle<br />

shown in figure 5A could have been<br />

mistaken for a single core nanoparticle<br />

(fig. 5F) if rotation was not possible. Using<br />

this same approach, images of yet another<br />

nanoparticle were acquired in 5° intervals<br />

and then used to create animations and<br />

tomographic reconstruction (to be published).<br />

EELS spectrum images were also<br />

acquired at several angular increments to<br />

determine the optoelectronic properties<br />

of the sol-gel silica shell. The EELS data<br />

showed the silica to have a band gap of<br />

8.9 eV and a chemical composition ratio<br />

of 1:2 for silicon to oxygen (to be published),<br />

similar values to those reported in<br />

the literature for evaporated microelectronics<br />

grade silica [9, 10].<br />

Summary<br />

A new electron microscopy protocol has<br />

been developed for functionalizing sample<br />

holders to enable 360° TEM/STEM<br />

nanoanalysis of individual nanoparticles.<br />

This protocol can be easily adapted for a<br />

wide variety of nanoparticle systems, and<br />

was demonstrated by preparing and then<br />

using functionalized holders for the analysis<br />

of core-shell nanoparticles used in a<br />

DNA detection assay. The functionalized<br />

holders were used for 360° STEM/EELS<br />

imaging and analysis of individual nanoparticles<br />

cantilevered over vacuum. Local<br />

material properties responsible for<br />

the increased DNA detection efficiency of<br />

these core-shell nanoparticles were<br />

mapped in 3D by combining information<br />

taken at regular angular intervals. By<br />

controlling the sample holder‘s geometry,<br />

coating and surface chemistry this<br />

three-step protocol can be adapted to enable<br />

the 360° TEM/STEM analysis of<br />

many nanoparticle systems.<br />

E l E c t r o n M i c r o s c o p y<br />

Fig. 5: STEM ZC micrograph series showing an individual nanoparticle with a double<br />

Au core at different angles of rotation. A. 0° rotation. B. 20° rotation. C. 40° rotation.<br />

D. 60° rotation. E. 80° rotation. F. 100° rotation.<br />

Acknowledgments<br />

We would like to thank Drs. M. Cerruti<br />

and S. Franzen from North Carolina State<br />

University’s Dept. of Chemistry for synthesis<br />

of the core-shell nanoparticles and<br />

supporting our attempt to elicit the materials<br />

properties responsible for increased<br />

DNA detection efficiency.<br />

References:<br />

[1] Koguchi, M., et al., J. Of Electron Microscopy<br />

50(3), 235–241 (2001).<br />

[2] Cerruti, M. G., et al., Analytical Chemistry 78,<br />

3282–3288 (2006).<br />

[3] deJong, K., et al., Chem. Phys. Chem. 3, 776–<br />

780 (2002).<br />

[4] Holzer, L., et al., J. of Microscopy 216, 84–95<br />

(2004).<br />

[5] Kawase, N., et al., Ultramicroscopy 107, 8–15<br />

(2007).<br />

[6] Kamino, T., et al., J. Of Electron Microscopy<br />

53(6), 583–588 (2004).<br />

[7] Kamino, T., et al., J. Of Electron Microscopy<br />

53(5), 563–566 (2004).<br />

[8] Ozasa, K., et al., Ultramicroscopy 101, 55–61<br />

(2004).<br />

[9] Barranco, A., et al., J. of Applied Physics<br />

97(11), (2005).<br />

[10] Keranen, J., et al., J. of Applied Physics<br />

84(12), 6827–6831 (1998).<br />

Contact:<br />

Dr. Konrad F. Jarausch<br />

Product Development Manager<br />

Hitachi High Technologies America, Inc.<br />

Nanotechnology Systems Division<br />

Pleasanton, CA, USA<br />

Tel.: +1 925 218 2830<br />

konrad.jarausch@hitachi-hta.com<br />

www.hitachi-hta.com<br />

Dr. Donovan N. Leonard<br />

Research Assistant Professor<br />

Appalachian State University<br />

Dept. of Physics & Astronomy<br />

Boone, NC, USA<br />

leonarddn@appstate.edu<br />

I II<br />

j NEW!<br />

Microscopy<br />

Workstation<br />

Ergonomic desks with integrated<br />

active vibration isolation<br />

system. For applications<br />

that require the maximum in<br />

effective damping.<br />

Unbeatable Advantages:<br />

• Virtually no low-frequency resonance<br />

• Exceptionally effective vibration isolation<br />

-25 dB (94.38%) @ 5 Hz<br />

-40 dB (99.00%) @10 Hz and above<br />

• Active isolation in all six degrees<br />

of freedom<br />

• Excellent position stability<br />

• Ergonomic design<br />

• Acoustic Enclosures available<br />

Especially designed for:<br />

• Atomic Force Microscopes<br />

• Life Science AFMs<br />

�<br />

Life Science<br />

AFM isolated<br />

by Microscopy<br />

Workstation<br />

with Acoustic<br />

Enclosure.<br />

Halcyonics GmbH<br />

Tuchmacherweg 12<br />

37079 Goettingen – Germany<br />

Tel.: +49 - 551- 99 90 62-0<br />

Fax: +49 - 551- 99 90 62-10<br />

info@halcyonics.de<br />

www.halcyonics.de<br />

G.I.T. Imaging & Microscopy 4/2007 • 27


Verifying Engineering at the Nanoscale<br />

Electron Microscopy and Drug Delivery<br />

Packaging drugs and genes into nanoparticles enables drug or gene biodistribution to be<br />

favourably altered, with an ultimate therapeutic benefit [1–3]. To acquire such control on<br />

the in vivo fate of drugs and genes requires that such particles be precision engineered and<br />

electron microscopy is one of the techniques used to visualise and confirm the results of<br />

such engineering.<br />

Methods<br />

Pharmaceutical nanosystems in our laboratory<br />

have been prepared from the self<br />

assembly of: a) comb type polymers [4–6]<br />

and b) dendrimers [7, 8] (fig. 1). By exercising<br />

control on the chemistry of these<br />

self assembling molecules, a variety of<br />

functional nanosystems may be prepared.<br />

Applying physical characterisation techniques<br />

including imaging to the resulting<br />

self assemblies and studying their in vivo<br />

behaviour ultimately enables robust correlations<br />

to be made between polymer<br />

chemistry, nature of the self assembly<br />

and drug delivery performance.<br />

Results<br />

E l E c t r o n M i c r o s c o p y<br />

Self assembling comb type polymers have<br />

been prepared from carbohydrates [2,<br />

5], polyamino acids [4] and polyamines<br />

[1, 6] and altering the level of hydropho­<br />

28 • G.I.T. Imaging & Microscopy 4/2007<br />

Fig. 1: Amphiphilic polymers and<br />

dendrimers used to make self<br />

assembling drug and gene delivery<br />

systems respectively.<br />

Fig. 2: Polymeric dense nanoparticles,<br />

vesicles and micelles<br />

produced by cetyl<br />

poly(ethylenimine). Increasing<br />

the level of hydrophobic substitution<br />

(cetyl chains) gives rise, in<br />

turn, to polymeric micelles,<br />

polymeric bilayer vesicles and<br />

dense nanoparticles. The polymer<br />

coat is clearly visible on the<br />

surface of the polymeric vesicles<br />

(arrowed).<br />

Ijeoma F. Uchegbu


Electron microscopes from Hitachi.<br />

www.hitachi-hitec-uk.com<br />

You provide<br />

the challenges<br />

- we’ll help you<br />

find the answers.<br />

Hitachi High-Technologies Corporation<br />

Tel: +44 (0) 800 316 1500


E l E c t r o n M i c r o s c o p y<br />

Fig. 3: The linear relationship between the level of cetyl substitution on cetyl<br />

poly(ethylenimine) and polymeric bilayer vesicle size. Vesicles were prepared<br />

by probe sonicating cetyl poly(ethylenimine) (4 mg mL –1 ) and cholesterol<br />

(2 mg mL –1 ).<br />

Fig. 4: Pharmacodynamic activity (sleep time, mean ± s.d., n ≥ 4) of propofol formulations after tail<br />

vein dosing to male MF1 mice. Mice were dosed as described below. 0.2 mg animals were dosed with<br />

0.2 mg propofol in a 100 µL volume administered as either propofol emulsion (10 mg mL –1 , Fresenius,<br />

Germany) diluted to 2 mg mL –1 with phosphate buffered saline (PBS, pH = 7.4) or a filtered amphiphilic<br />

chitosan formulation (amphiphilic chitosan – 5 mg mL –1 , propofol – 1.9 mg mL –1 in PBS – pH<br />

= 7.4). 0.2 UF animals were dosed with 0.2 mg propofol in a 100 µL volume administered as either<br />

propofol emulsion (10 mg mL –1 , Diprivan) diluted to 2 mg mL –1 in glycerol – 0.24M or an unfiltered<br />

amphiphilic chitosan formulation (amphiphilic chitosan – 5 mg mL –1 , propofol – 1.9 mg mL –1 , lecithin -<br />

2 mg mL –1 , in glycerol – 0.24 M). 0.4 mg animals were dosed with 0.4 mg of propofol in a 100 µL<br />

volume administered as either Diprivan diluted in glycerol (0.24 M – TEM of formulation shown in<br />

inset i) to a concentration of 4 mg mL –1 or a filtered amphiphilic chitosan formulation of propofol<br />

(amphiphilic chitosan – 5 mg mL –1 , propofol – 4.2 mg mL –1 , lecithin - 2 mg mL –1 , in glycerol – 0.24 M –<br />

TEM of formulation shown in inset ii). 0.5 mg animals were dosed with 0.5 mg propofol administered<br />

in a 50 µL volume as either Diprivan (10 mg mL –1 ) or propofol emulsion (Fresenius, 10 mg mL –1 ). A loss<br />

of righting reflex time was only observed in animals receiving a low dose of the commercial emulsion<br />

formulations (0.37 ± 0.19 min in the 0.2 Fresenius animals). No sleep times were recorded in animals<br />

receiving the polymer alone. * = statistically significantly different (p < 0.05).<br />

bic substitution alters the nature of the<br />

self assembly (fig. 2). Additionally we<br />

have identified two chemical features of<br />

these amphiphilic polymers which control<br />

nanosystem size (and ultimately nanosystem<br />

biodistribution) and these are<br />

the molecular weight of the polymer [5]<br />

and the level of hydrophobic substitution<br />

30 • G.I.T. Imaging & Microscopy 4/2007<br />

of the polymer [6], both of which are positively<br />

and linearly correlated with nanosystem<br />

size (e.g. fig. 3).<br />

Carbohydrate micellar clusters, in<br />

which one micelle is linked to another,<br />

presumably via trailing polymer chains<br />

are able to form nanoparticles in the<br />

presence of hydrophobic drugs [2]. These<br />

drug loaded nanoparticles increase drug<br />

bioavailability across the blood brain<br />

barrier by up to ten fold when compared<br />

to the current state of the art commercial<br />

emulsion system (fig. 4) [2]. Amphiphilic<br />

polyamine drug loaded nanoparticles increase<br />

drug absorption via the oral route<br />

by up to three fold when compared to the<br />

drug suspension in water [1]. Additionally<br />

polypropylenimine dendrimers form<br />

colloidal particles with DNA which are<br />

able to transfer an anti­proliferative<br />

gene, the tumour necrosis alpha gene,<br />

into mouse tumours and produce a 100 %<br />

response in all tumours studied, due in<br />

part to the additional anti­proliferative<br />

activity of the dendrimer [3].<br />

Conclusions<br />

It is possible to correlate polymer chemistry<br />

with the nature of the resulting self<br />

assembly and ultimately link a range of<br />

morphologically distinct nano­size self<br />

assemblies with specific drug delivery<br />

function. With these nanosystems, a ten<br />

fold increase in drug activity may be obtained<br />

[2] and an effective anti­cancer<br />

gene medicine is achievable [3].<br />

References:<br />

[1] Cheng, W., et al., Biomacromolecules, 7,<br />

1509–1520 (2006)<br />

[2] Qu, X., et al., Biomacromolecules, 7, 3452–<br />

3459 (2006)<br />

[3] Dufes, C., et al., Cancer Research, 65, 8079–<br />

8084 (2005)<br />

[4] Wang, W., et al., Langmuir, 16, 7859–7866<br />

(2000)<br />

[5] Wang, W., et al., Langmuir, 17, 631–636<br />

(2001)<br />

[6] Wang, W., et al., Macromolecules, 37, 9114–<br />

9122 (2004)<br />

[7] Zinselmeyer, B. H., et al., Pharmaceutical Research,<br />

19, 960–967 (2002)<br />

[8] Schätzlein, A. G., et al., J. Control. Rel., 101,<br />

247–258 (2005)<br />

Contact:<br />

Ijeoma F. Uchegbu<br />

University of London, United Kingdom<br />

Department of Pharmaceutics, School of Pharmacy<br />

Tel.: +44 20 7753 5997<br />

Fax: +44 20 7753 5946<br />

ijeoma.f.uchegbu@pharmacy.ac.uk<br />

www.pharmacy.ac.uk


Cryo Electron Tomography<br />

Unique Capability for Structural Biology Investigations<br />

Cryo electron tomography’s (CET) ability to visualize<br />

three dimensional biological structures – ranging<br />

in size from molecular to cellular – fills a critical<br />

gap between techniques with atomic resolution,<br />

such as x-ray diffraction (XRD) and nuclear magnetic<br />

resonance (NMR), and conventional light microscopy.<br />

However, it is CET’s ability to investigate<br />

biological structures in their unperturbed, native<br />

context that makes it an indispensable tool in the<br />

currently exploding field of structural biology.<br />

Introduction<br />

The recent decoding of the human<br />

genome, and the realization that we can<br />

now read the entire blueprint of the<br />

vastly complex biological machine that is<br />

a living organism-human or any other,<br />

has had a tremendous symbolic and<br />

practical impact on the public consciousness<br />

in general, and the scientific community<br />

in particular. Since that time,<br />

biologists have focused increasing attention<br />

on understanding the structure and<br />

function of the machine’s components –<br />

the proteins encoded by the genetic blueprint.<br />

Fig. 1: The image shows a surface rendered<br />

representation of a segment of the nucleus from<br />

a eukaryotic organism (Dictyostelium discoideum).<br />

The nuclear membranes are in blue, and<br />

the Nuclear Pore Complexes (NPCs) are in red.<br />

The NPCs are 125 nm in diameter. Structures of<br />

individual NPCs have been substituted by the<br />

averaged structure. <strong>Images</strong> courtesy of: Martin<br />

Beck, Friedrich Förster, Mary Ecke, Jürgen M.<br />

Plitzko, Frauke Melchior, Günther Gerisch, Wolfgang<br />

Baumeister and Ohad Medalia, Max-<br />

Planck-Institute of Biochemistry, Martinsried,<br />

Germany. We gratefully acknowledge help with<br />

the design and visualization by Julio Ortiz. Details<br />

published in Science 306:1387–1390, 2004.<br />

The Strength of CET in<br />

Structural Biology<br />

One approach, call it brute force, in many<br />

ways analogous to the approach taken in<br />

the decoding effort itself, simply seeks to<br />

determine the structure of each of the<br />

large but finite number of individual proteins<br />

that constitute the entire proteome,<br />

perhaps one to two million in a human.<br />

Though this basic structural information<br />

is certainly fundamental to a complete<br />

understanding, it is only the lowest level<br />

in the hierarchy of structural complexity.<br />

Few proteins work alone. Most function<br />

in concert with others as components of<br />

supramolecular complexes that may be<br />

persistent or quite transient. At a still<br />

higher level, the cellular context in which<br />

these complexes function cannot be<br />

regarded as a simple sack of cytoplasm<br />

filled with randomly colliding complexes.<br />

It is itself a highly structured environment<br />

throughout which components are<br />

manufactured, regulated, transported<br />

and consumed among at myriad functional<br />

localities.<br />

Clearly, the operation of the machine<br />

can be more easily understood by observations<br />

of the assembled components.<br />

Compare it to building a watch. We have<br />

the parts list, the genetic code. With sufficient<br />

effort we may eventually describe<br />

each component in atomic detail. But<br />

how much easier is it to comprehend the<br />

workings of the entire machine if we can<br />

look at a fully assembled watch. This is<br />

the strength of CET in structural biology.<br />

Alone among experimental techniques, it<br />

permits observations of fully assembled<br />

biological machines from the macromolecular<br />

cogs and pulleys to the cellular<br />

E l E c t r o n M i c r o s c o p y<br />

systems for synthesis and distribution (as<br />

shown in figures 1 – 3).<br />

Vitrification Essential for CET<br />

As its name implies, cryo electron tomography<br />

uses electron images of frozen<br />

samples to construct a three dimensional<br />

model of the imaged volume. The images,<br />

provided by a high resolution transmission<br />

electron microscope (TEM), are two<br />

dimensional projections acquired as the<br />

sample is rotated incrementally about an<br />

axis perpendicular to the viewing direction.<br />

A computer creates the 3D model<br />

from the projections with reconstruction<br />

techniques familiar to most of us from<br />

their use in medical imaging applications<br />

such as CAT scans and MRI. Key to the<br />

value of CET is its use of a cryogenic<br />

freezing process known as vitrification.<br />

The process freezes the hydrated sample<br />

so rapidly that water molecules do not<br />

crystallize, instead forming an amorphous<br />

solid (vitreous ice) that does little<br />

or no damage to delicate molecular<br />

structures.<br />

CET can resolve structures down to a<br />

few nanometers, sufficient for tertiary<br />

and quaternary protein morphology. In<br />

hybrid approaches to structural analysis,<br />

atomic scale structures determined by<br />

XRD and NMR are fitted to larger scale<br />

CET models to enhance the level of structural<br />

detail. CET samples may be pristine<br />

biological material, or selectively stained<br />

with electron dense materials to emphasize<br />

particular features. More specific<br />

emphasis to particular proteins or<br />

domains can be achieved with immuno<br />

labeling techniques using gold or other<br />

high contrast nanoparticles.<br />

G.I.T. Imaging & Microscopy 4/2007 • 31


E l E c t r o n M i c r o s c o p y<br />

Fig. 2: The image shows a 3D rendering of a<br />

mitochondrion from a mouse neuron. Tomograms<br />

obtained from thick mouse brain sections<br />

were selectively segmented to highlight the<br />

mitochondrial membranes (in green), cristae (in<br />

blue) and surrounding neurofilaments and microtubules<br />

(in grey).<br />

Investigation of Morphological<br />

Transformations<br />

The ability of XRD and NMR to achieve<br />

atomic resolution is due in large part to<br />

its use of a composite signal acquired<br />

from a collection of presumably identical<br />

structures. In contrast, CET examines an<br />

individual structure. Since the sample is<br />

quite literally frozen in time, CET can<br />

distinguish morphological differences<br />

among structures with identical atomic<br />

compositions, such as protein molecules,<br />

with identical peptide sequences. By<br />

examining a collection of frozen molecular<br />

instances, CET can be used to investigate<br />

morphological transformations that<br />

32 • G.I.T. Imaging & Microscopy 4/2007<br />

occur as a result of interactions, the<br />

dynamic behavior of flexible proteins,<br />

the formation of transient complexes in<br />

signaling pathways, and more.<br />

CET eliminates XRD’s requirement for<br />

crystalline samples, avoiding the possibility<br />

of structural distortions induced by<br />

crystallization, and allowing the examination<br />

of virtually any specimen, including<br />

flexible and insoluble proteins that<br />

are notoriously difficult to crystallize. It<br />

also does not suffer the increasing difficulty<br />

of NMR analysis with larger molecular<br />

size.<br />

CET does face at least two significant<br />

challenges. Because the signal originates<br />

from a single molecule it is weak relative<br />

to random variations in its intensity (shot<br />

noise). The delicate nature of molecular<br />

structures and the high energy of the<br />

interrogating radiation do not allow<br />

increases in beam intensity or exposure<br />

time without inflicting significant damage<br />

in the sample. Careful dose management<br />

and automated acquisition procedures<br />

can help, but this remains a<br />

fundamental limitation.<br />

Correlative Microscopy Increasingly<br />

Important<br />

The second issue relates to the crowded<br />

environment of the cell. We would like to<br />

look at distinct structures clearly contrasted<br />

against a featureless background.<br />

However, in an intact biological system<br />

there is no empty space and little fundamental<br />

difference between the atoms<br />

that compose the structures of interest<br />

and those that surround it. It is rather<br />

like looking into a very big bag of marbles,<br />

or perhaps like looking for specific<br />

configurations of bubbles in a bucket of<br />

foam. There are techniques that reduce<br />

the difficulty. We have mentioned staining<br />

and immuno labeling. Another area<br />

of growing interest is correlative microscopy,<br />

using an optical technique such as<br />

Fig. 3: Reovirus-Polymerase. Cross-section<br />

of a reovirus shows features down to 7.6-<br />

Ångström resolution, a scale that reveals<br />

the inner features of the viral particle.<br />

Visible for the first time within the virus<br />

are several tiny ‘factories’, shown here in<br />

red, which convert raw materials from a<br />

victim cell’s interior into RNA messages<br />

instructing the cell to begin manufacturing<br />

more viruses. Photo by Purdue University/<br />

Department of Biology, sample courtesy of<br />

Xing Zhang, Stephen B. Walker, Paul R.<br />

Chipman, Timothy S. Baker, Department of<br />

Biological Sciences, Purdue University and<br />

Max L. Nibert, Department of Microbiology<br />

and Molecular Genetics, Harvard<br />

Medical School.<br />

fluorescence microscopy to locate labeled<br />

structures for subsequent high resolution<br />

CET analysis. Finally, the digital<br />

nature of tomographic data lends itself<br />

well to computational search and fit routines<br />

using previously defined templates<br />

to locate specific structures within the<br />

sample volume.<br />

TEM technology has advanced dramatically<br />

in recent years. Perhaps the<br />

greatest development has been the<br />

incorporation of aberration correctors,<br />

which has pushed image resolution well<br />

below the Angstrom barrier – 0.5 Angstrom<br />

resolution was just announced by<br />

the TEAM (Transmission Electron Aberration-corrected<br />

Microscope) project, a<br />

joint effort by the U.S. Department of Energy,<br />

FEI Company and CEOS GmbBH.<br />

Improvements in TEM<br />

A significant contribution to improved<br />

performance has also come from better<br />

instrument stability – mechanical, electronic<br />

and environmental. Sophisticated<br />

automation at all levels from setup and<br />

alignment, through operation, data<br />

acquisition, and sample preparation has<br />

made TEM much faster, easier, more<br />

repeatable and more reliable. Some very<br />

interesting progress has been made in<br />

sample preparation. FEI’s Vitrobot provides<br />

automatic vitrification fluid suspensions<br />

on a TEM sample grid. Focused ion<br />

beam (FIB) based preparation procedures<br />

have greatly improved the ease,<br />

speed, and reliability of preparing thin<br />

samples from bulk specimens, including<br />

frozen material. Another area of interest<br />

is the FIB based preparation of cylindrical<br />

tomography samples that permit the<br />

acquisition of 2D images through a complete,<br />

360 degree range of rotation.<br />

The sheer volume of information<br />

potentially available in a high resolution<br />

tomogram is mind boggling. Theoretically,<br />

one could capture the entire proteome<br />

of a cell, including all of its functional<br />

complexes and higher level<br />

structures. As one researcher put it, one<br />

good tomogram can make a whole career<br />

– or perhaps several. With this kind of<br />

potential, there is little doubt that CET<br />

will play a critical role in the future of<br />

structural biology.<br />

Contact:<br />

Matthew Harris<br />

Vice President NanoBiology Market Division<br />

FEI Company<br />

Eindhoven, NL<br />

Tel.: +31 40 23 56184<br />

Fax: +31 40 23 56634<br />

matthew.harris@fei.com<br />

www.fei.com


SCANNING SECTION


Ultrafast Confocal Raman Imaging<br />

Acquiring Spectra in a Few Milliseconds with Improved Sensitivity<br />

In Confocal Raman imaging the acquisition time for<br />

one Raman spectrum is a crucial value, as it influences<br />

the acquisition time of the image which typically<br />

consists of tens of thousands of Raman spectra.<br />

This article describes how the use of a<br />

spectroscopic EMCCD as the detector can significantly<br />

reduce the acquisition time down to a few<br />

milliseconds per spectrum, as well as tremendously<br />

improve sensitivity.<br />

Introduction<br />

S c a n n i n g S e c t i o n<br />

In confocal Raman imaging, as in the<br />

WITec alpha300 R system which was<br />

used for the experiment described here,<br />

only light from the image focal plane can<br />

reach the detector, which strongly increases<br />

image contrast and slightly increases<br />

resolution. Special filters are<br />

used to suppress the reflected laser light<br />

while enabling the Raman scattered light<br />

to be detected with a spectrometer/CCD<br />

camera combination. To obtain an image,<br />

thousands of spectra are acquired in<br />

a very short time with typically less than<br />

100 ms integration time per spectrum<br />

using a normal CCD camera. In order to<br />

further improve the overall sensitivity of<br />

the system a EM-CCD can be used.<br />

Spectroscopic EMCCD<br />

An electron multiplying CCD (EMCCD) is<br />

a normal CCD with an additional readout<br />

register which is driven with a much<br />

higher clock voltage than a normal CCD<br />

readout register. Due to this high clock<br />

voltage, an electron multiplication<br />

through impact ionization is achieved<br />

with an adjustable total amplification of<br />

the signal of up to 1000 times. With this<br />

setup, it is always possible to amplify the<br />

signal above the readout noise so that<br />

the S/N ratio is always limited by the<br />

Poisson noise of the signal, even if a very<br />

fast readout amplifier is used. As an example,<br />

a 1600 x 200 pixel EMCCD with a<br />

2.5 MHz readout amplifier, as used for<br />

the experiments in this article, can be<br />

read out in only 1.7 ms.<br />

34 • G.I.T. Imaging & Microscopy 4/2007<br />

Fig. 1: a) Confocal Raman Image of a PS-PMMA blend acquired with the WITec Ultrafast Raman Imaging<br />

Option. 120 x 120 pixel = 14,400 spectra. Acquisition time for one spectrum: 2,3 milliseconds; total<br />

acquisition time for the image 67 seconds. Red: PS; green: PMMA. b) Corresponding Raman spectra<br />

The following calculations show the<br />

improvement in S/N that can be expected<br />

for different signals. It is assumed that<br />

the quantum efficiency (QE) of the CCD is<br />

90 % and that the amplification of the<br />

signal is set to a value at which one A/D<br />

count equals the number of electrons of<br />

the readout noise (1 A/D count = 30 electrons<br />

for a 2.5 MHz readout amplifier).<br />

If 100 photons fall on a CCD pixel in a<br />

given integration time, 90 electrons will<br />

be generated and converted to 3 A/D<br />

counts. The readout noise will be 1 A/D<br />

count and the Poisson noise will be 9.5,<br />

which is approximately 0.3 A/D counts.<br />

With these numbers, the S/N ratio is<br />

about 2.6.<br />

In an EMCCD, the signal will be multiplied<br />

by the electron gain factor which<br />

can be as high as 1,000. A smaller amplification<br />

factor would generally be used,<br />

but for the calculation it does not make a<br />

difference. 90 electrons will be amplified<br />

to 90,000 electrons resulting in 3,000 A/<br />

D counts. The Poisson noise is 9,500 electrons<br />

which translates to 317 counts,<br />

while the 1 count readout noise is completely<br />

negligible. S/N is 9.5, which is an<br />

improvement of a factor of 3.6.<br />

If the signal is only 10 photons, this<br />

will result in a signal of only 0.3 counts<br />

for a normal CCD. Poisson noise can be<br />

neglected in this case. With 1 count readout<br />

noise, S/N is 0.3, hardly a detectable<br />

signal.<br />

For an EMCCD, the signal is 333<br />

counts and Poisson noise is 100 counts<br />

which gives a S/N of 3.3, an improvement<br />

of 11 times over a normal CCD.<br />

In reality the electron multiplying<br />

process itself adds an additional, so<br />

called excess noise factor of about 1.4, so<br />

that the real improvements in S/N are reduced<br />

to 2.6 and 7.9 respectively for the<br />

above examples.<br />

For higher signals, in which the signal<br />

intensity is no longer readout limited, the<br />

excess noise factor of the EM process reduces<br />

the S/N ratio of an EMCCD to below<br />

that of a normal CCD. In this case,<br />

the EM register can be switched off and<br />

then the “normal” readout register is<br />

used. Thus, the EMCCD behaves just as a<br />

normal back-illuminated CCD.<br />

Application Examples<br />

Figure 1 a) shows a confocal Raman image<br />

of a PS-PMMA polymer blend spin-<br />

coated onto a glass slide. It was imaged<br />

with the WITec Ultrafast Raman Imaging<br />

Option for the alpha300 R Confocal Raman<br />

Microscope using a spectroscopic<br />

EMCCD as the detector. The scan range<br />

was 50 x 50 µm with 120 x 120 image<br />

pixels = 14,400 spectra. The acquisition<br />

time per spectrum was only 2.3 milliseconds,<br />

resulting in a total acquisition time<br />

for the complete image of 67 seconds (including<br />

retrace at the end of the line).<br />

The image was obtained by evaluating<br />

the different peak characteristics of PS<br />

and PMMA as shown in figure 1b. with<br />

the WITec Project software package. This<br />

results in a color-coded image of the distribution<br />

of the two compounds (red: PS;<br />

green: PMMA).


Fig 2: a) Overview scan of carbon nanotubes on<br />

a silicon substrate, 25 x 25 µm, 120 x 120 pixel =<br />

14,400 spectra. b) Zoom-in of the marked area.<br />

Scan Range 3.5 x 3.5 µm, 120 x 120 pixel =<br />

14,400 spectra. Total acquisition time of both<br />

images was 96 seconds. c) Typical measured<br />

spectrum obtained by averaging some of the<br />

14,400 spectra which showed a clear signal from<br />

the CNTs.<br />

In a second experiment, carbon nanotubes<br />

(CNTs) on a silicon substrate were<br />

imaged using the WITec Ultrafast Raman<br />

Imaging Option. As the first step, a large<br />

overview scan with a scan range of 25 x<br />

25 µm with 120 x 120 pixels = 14,400<br />

spectra was performed. The acquisition<br />

time was 4 ms per spectrum, so the<br />

measurement required only 96 seconds<br />

to complete the image shown in figure<br />

2 a). It clearly reveals the heterogeneous<br />

distribution of the CNTs on the substrate.<br />

S c a n n i n g S e c t i o n<br />

In a second step, a zoomed-in image of<br />

the marked area with a scan range of 3.5<br />

x 3.5 µm and 120 x 120 pixels was obtained<br />

as shown in figure 2 b). Due to the<br />

optimized readout of the EMCCD, the acquisition<br />

time per spectrum was again 4<br />

ms and 96 seconds for the complete image<br />

(including retrace at the end of the<br />

line). Figure 2 c) shows a typical measured<br />

spectrum of the CNTs also showing<br />

peaks from the Si-Substrate. It was obtained<br />

by averaging some of the 14,400<br />

spectra which showed a clear signal from<br />

the CNTs.<br />

Summary<br />

It was demonstrated that the use of an<br />

EMCCD camera can greatly increase detection<br />

efficiency and speed, especially<br />

for the short integration times necessary<br />

with a confocal Raman microscope. For<br />

very small signals that are dominated by<br />

the CCD’s readout noise, the use of an<br />

EMCCD can improve the S/N ratio by a<br />

factor of 5 10 compared to the best available<br />

standard CCD’s, while for larger signals<br />

the electron multiplying circuit can<br />

simply be switched off and all properties<br />

of a standard (back-illuminated) CCD are<br />

maintained.<br />

Contact:<br />

Olaf Hollricher<br />

Harald Fischer<br />

Andrea Jauss<br />

Thomas Dieing<br />

WITec GmbH<br />

Ulm, Germany<br />

Tel: +49 731 14070 0<br />

Fax: +49 731 14070 20<br />

Harald.Fischer@witec.de<br />

www.witec.de<br />

G.I.T. Imaging & Microscopy 4/2007 • 35


S c a n n i n g S e c t i o n<br />

Ultrasonic Nanofabrication with an AFM<br />

Ultrasound Facilitates Nanolithography and Nanomanipulation<br />

Ultrasonic AFM may improve fabrication technologies on the nanometer scale. In the presence of ultrasonic<br />

vibration, hard surfaces can be indented and scratched with the tip of a soft cantilever, due to its inertia. Ultrasound<br />

reduces or even eliminates friction, and hence modifies the tip-nanoparticle-surface interactions in<br />

AFM manipulation. The subsurface sensitivity of the technique makes feasible the purposed manipulation of<br />

subsurface nanoscale features by ultrasonic actuation.<br />

Ultrasonic Atomic Force Microscopies<br />

Information of ultrasonic vibration on<br />

the nanoscale has recently become<br />

accessible by a new family of Scanning<br />

Probe Microscopy techniques based on<br />

the use of Atomic Force Microscopy<br />

(AFM) with ultrasound excitation [1].<br />

Among them, the techniques of Ultrasonic<br />

Force Microscopy (UFM) [2] and<br />

Heterodyne Force Microscopy (HFM) [3]<br />

rely in the so-called „mechanical-diode“<br />

effect [4], in which a cantilever tip is in<br />

contact with the sample surface and normal<br />

ultrasonic vibration is excited at the<br />

tip-sample contact (see fig. 1). If the<br />

excitation frequency is high enough, or is<br />

not coincident with a high-order cantilever<br />

contact resonance, the cantilever<br />

will not be able to linearly follow the surface<br />

vibration due to its inertia. Nevertheless,<br />

if the ultrasonic excitation am-<br />

Keywords:<br />

atomic force microscopy, ultrasonic force<br />

microscopy, nanomanipulation, nanolithography,<br />

nanoscratching<br />

36 • G.I.T. Imaging & Microscopy 4/2007<br />

plitude is sufficiently high that the<br />

tip-sample distance varies over the nonlinear<br />

tip-sample force interaction<br />

regime, the cantilever experiences a<br />

static force during the time that the<br />

ultrasonic excitation is acting. This force<br />

is the so-called “ultrasonic force”, and<br />

can be understood as the net force that<br />

acts upon the cantilever during a complete<br />

ultrasonic cycle, due to the nonlinearity<br />

of the tip-sample interaction force.<br />

The cantilever behaves then as a<br />

mechanical diode and deflects when the<br />

tip-sample contact vibrates at ultrasonic<br />

frequencies of sufficiently high amplitude.<br />

The magnitude of the ultrasonic<br />

force or of the ultrasonic-force-induced<br />

additional cantilever deflection (UFM signal)<br />

is dependent on the details of the<br />

tip-sample interaction force, and hence<br />

on material properties such as elasticity<br />

and adhesion. Therefore, UFM allow us<br />

to discern nanoscale topographic regions<br />

with distinct elastic contrast, as shown in<br />

figure 2, where images of Sb nanoparticles<br />

on Highly Oriented Pyrolitic Graphite<br />

(HOPG) are displayed. Remarkably, in<br />

M. Teresa Cuberes<br />

this case, the UFM contrast reveals stiffness<br />

variations even within individual Sb<br />

particles [5]. Recently, a novel ultrasonic<br />

AFM mode, namely Mechanical-Diode<br />

Ultrasonic Friction Force Microscopy<br />

(MD-UFFM) based on the lateral<br />

mechanical diode effect has additionally<br />

been proposed for the study of friction<br />

and lubrication on the nanoscale, in the<br />

presence of surface shear ultrasonic<br />

vibration [6].<br />

Ultrasonic Nanofabrication<br />

Ultrasonic AFM techniques provide a<br />

means to monitor ultrasonic vibration at<br />

the nanoscale, and open up novel opportunities<br />

to improve nanofabrication technologies<br />

[7].<br />

In the presence of ultrasonic vibration,<br />

the tip of a soft cantilever can dynamically<br />

indent hard samples due to its inertia. In<br />

addition, it has been demonstrated that<br />

ultrasound reduces or even eliminates<br />

nanoscale friction [8]. Typical top-down<br />

approaches that rely in the AFM are<br />

based on the use of a cantilever tip that


acts as a plow or as an engraving<br />

tool. The ability of the<br />

AFM tip to respond inertially<br />

to ultrasonic vibration excited<br />

perpendicular to the sample<br />

surface and dynamically indent<br />

hard samples may facilitate<br />

the nanoscale machining<br />

of semiconductors or engineering<br />

ceramics in a reduced<br />

time. Figure 3 demonstrates<br />

the machining of nanotrenches<br />

and holes on a silicon sample<br />

in the presence of ultrasonic<br />

vibration. Interestingly, no debris<br />

is found in the proximity<br />

of lithographed areas. Figure<br />

3 (a) refer to results performed<br />

using a cantilever with nominal<br />

stiffness comprised between<br />

28–91 Nm –1 and a diamond-coated<br />

tip. Figure 4 (b)<br />

refer to results achieved using<br />

a cantilever with nominal<br />

stiffness 0.11 Nm –1 and a SiN<br />

tip; in the absence of ultrasound,<br />

it was not possible to<br />

scratch the Si surface using<br />

such a soft cantilever. In the<br />

machining of soft materials,<br />

as for instance plastic coatings,<br />

the ultrasonic-induced<br />

reduction of nanoscale friction<br />

may permit eventual finer<br />

features and improved surface<br />

quality in quasi-static approaches.<br />

In [9], an in-plane<br />

acoustic wave coupled to the<br />

sample support was used to<br />

enhance the intermittent force<br />

exerted by the tip in dynamic<br />

AFM nanomachining of thin<br />

polymer resist films.<br />

In bottom-up approaches,<br />

ultrasound may assist in the<br />

self-assembly or AFM manipulation<br />

of nanostructures [7].<br />

Effects such as sonolubrication<br />

and acoustic levitation<br />

have been studied at the microscale.<br />

These phenomena<br />

may facilitate a tip-induced<br />

motion of nano-objects. In the<br />

manipulation of nanoparticles<br />

(NPs) on surfaces with<br />

the tip of an AFM cantilever,<br />

when ultrasound is excited at<br />

a sample surface both tipparticle<br />

and particle-surface<br />

frictional properties change<br />

[10]. Moreover, the excitation<br />

of NP high-frequency internal<br />

vibration modes may also<br />

modify the NP dynamic re-<br />

sponse, and introduce novel<br />

mechanisms of particle motion.<br />

Some of the opportunities<br />

in ultrasonic-assisted<br />

AFM manipulation are illustrated<br />

in figure 4, which show<br />

images of Au NPs on a silicon<br />

surface. The surface was covered<br />

by poly-l-lysine to prevent<br />

that the Au NPs were<br />

swept away by the AFM tip.<br />

Nevertheless, when scanning<br />

in contact mode in the absence<br />

of ultrasound, most NPs<br />

were inevitably swept away<br />

by the tip. Scanning in the<br />

same conditions that led to a<br />

NP displacement but in the<br />

presence of surface ultrasonic<br />

vibration, with an appropriate<br />

election of the ultrasonic<br />

amplitude, the NPs remained<br />

undisturbed. In figure 4, two<br />

NP were displaced while recording<br />

the images, which<br />

have been pointed out by blue<br />

arrows. A close inspection of<br />

the data in figure 4 reveals<br />

traces of the UFM and LFM<br />

responses from those moving<br />

particles while being in motion<br />

(see areas enclosed by ellipses<br />

and rectangles). The<br />

study of the UFM response of<br />

a moving NP may allow us to<br />

learn about the dynamic<br />

mechanisms of NP displacement<br />

across surfaces. Consistently<br />

with the fact that ultrasound<br />

eliminates friction,<br />

no frictional contrast is distinguished<br />

on the undisturbed<br />

Au NPs in the LFM images.<br />

However, from the comparison<br />

of the traces of the two<br />

moving NPs in the forward<br />

and backward LFM scans,<br />

friction during the tip-induced<br />

NP motion is apparent. Controlled<br />

and accurate measurements<br />

of lateral and ultrasonic<br />

forces exerted by<br />

individual NPs when in motion<br />

under tip ultrasonic actuation<br />

may bring about a wealth of<br />

information about the dissipated<br />

energy, ultrasonic lubrication<br />

effects, NP dynamics,<br />

etc.<br />

Eventually, it should be<br />

pointed out that the sensitivity<br />

of ultrasonic-AFM to subsurface<br />

features makes feasible<br />

to monitor subsurface<br />

The New BioMAT TM<br />

Workstation AFM.<br />

Perfect Imaging<br />

Results for<br />

Opaque Samples.<br />

The BioMAT TM Workstation combines upright optical<br />

microscopy with AFM and opens up a wide range of<br />

applications for the study of non-transparent specimens.<br />

Even in liquids.<br />

www.jpk.com<br />

S c a n n i n g S e c t i o n<br />

G.I.T. Imaging & Microscopy 4/2007 • 37


S c a n n i n g S e c t i o n<br />

Fig. 1: Detection of surface vibration with the tip<br />

of an AFM cantilever. (a) At low frequencies, the<br />

tip follows the surface vibration. (b) In the highfrequency<br />

regime, for sufficiently high vibration<br />

amplitudes, tip experiences an ultrasonic force<br />

F us. (c) Tip­sample force F t­s versus tip­sample distance<br />

d curve.<br />

Fig. 2: Sb NPs on HOPG. (a, b) Simultaneously<br />

recorded contact­mode AFM topography (a) and<br />

UFM image (b). (c, d) Simultaneously recorded<br />

high­resolution images from the areas enclosed<br />

by white squares in (a), (b). Set point: 1 nN; Kc:<br />

0.11 Nm –1 ; UFM parameters: 2.2 MHz, 8 Vpp.<br />

Contrast in UFM indicates stiffness variations.<br />

modifications [7]. We have recently demonstrated<br />

that actuation with an AFM tip<br />

in the presence of ultrasonic vibration<br />

can produce stacking changes of extended<br />

grapheme layers, and induce permanent<br />

displacements of buried dislocations<br />

in Highly Oriented Pyrolytic<br />

38 • G.I.T. Imaging & Microscopy 4/2007<br />

Fig. 3: AFM nanomachining of trenches and holes<br />

on Si(111) in the presence of normal surface<br />

ultrasonic vibration of ~5 MHz. (a) Scratch and<br />

indentation with a diamond­coated cantilever<br />

tip. Rt: 35 nm. Kc: 28­91 Nm –1 . (b) Nanotrenches<br />

formed in 50, 75 and 100 cycles respectively, at<br />

a load of ~40 nN, with a pyramidal SiN cantilever<br />

tip. Kc: 0.11 Nm –1 .<br />

Fig. 4: Displacement of Au NP induced by the tip<br />

of an AFM cantilever in the presence of normal<br />

surface ultrasonic vibration of ~ 2.6 MHz. (a­d)<br />

were simultaneously recorded. (a) Contact­mode<br />

AFM topography. (b) UFM; (c) LFM forward scan;<br />

(d) LFM backward scan<br />

Fig. 5: Lateral displacement<br />

of a subsurface dislocation<br />

in HOPG by ultrasonic tip<br />

actuation (a) Topography of<br />

the HOPG surface in AFM<br />

contact mode: (700 x 700)<br />

nm; Fo=105 nN. (b,c) Ultrasonic­AFM<br />

images recorded<br />

in sequence over nearly the<br />

same surface region:<br />

a=2.15 MHz (b) A=3.5 Vpp<br />

(c) A=2.4 Vpp. (d–f) LFM<br />

forward scan, simultaneously<br />

recorded with (a–b)<br />

respectively, with the same<br />

parameters.<br />

Graphite (HOPG). This effect is illustrated<br />

in figure 5. In the presence of normal<br />

surface ultrasonic vibration, both AFM<br />

and LFM images reveal subsurface features<br />

[1]. Subsurface modification was<br />

brought about in this case by scanning in<br />

contact mode, with high set-point forces,<br />

and high surface ultrasonic excitation<br />

amplitudes [7].<br />

Summary<br />

Ultrasonic AFM techniques provide a<br />

means to monitor ultrasonic vibration at<br />

the nanoscale, and open up novel opportunities<br />

in nanofabrication technologies.<br />

The use of ultrasound may improve both<br />

down-top and bottom-down approaches<br />

in nanofabrication, facilitating the patterning<br />

of nanoscale surface features,<br />

the manipulation or self-assembly of nanostructures,<br />

and possibly the controlled<br />

subsurface manipulation of buried nanoobjects.<br />

Acknowledgments<br />

The author thanks J. J. Martinez and A.<br />

Lusvardi for assistance in the UFM lab.<br />

The samples of Sb NPs were provided by<br />

C. Ritter and U. Schwarz. The Au NPs<br />

were provided by M. A. Gonzalez and<br />

M.P. Morales. Financial support from the<br />

JCCM (Junta de Comunidades de Castilla-La<br />

Mancha) under project PBI-05-<br />

018 is gratefully acknowledged.<br />

References:<br />

[1] Gnecco, E. and Meyer, E. (Eds.), Fundamentals<br />

of Friction and Wear on the nanometer<br />

scale, Springer, 2007, 49-71.<br />

[2] Yamanaka K., et al., Appl. Phys. Lett. 64,<br />

178–180 (1994).<br />

[3] Cuberes, M. T., et al., J. Phys. D.: Appl. Phys.<br />

33, 2347–2355 (2000).<br />

[4] Rohrbeck, W. and Chilla, E., Phys. Status<br />

Solidi (a) 131, 69–71(1992).<br />

[5] Cuberes, M. T., et al., Ultramicroscopy 107,<br />

1053–1060 (2007).<br />

[6] Cuberes, M. T. and Martínez, J. J., J. of Phys.:<br />

Conf. Ser. 62, 224–228 (2007).<br />

[7] Cuberes, M. T., J. of Phys.: Conf. Ser. 61, 219–<br />

223 (2007).<br />

[8] Dinelli, F., et al., Appl. Phys. Lett. 71, 1177–<br />

1179 (1997).<br />

[9] Rubio-Sierra, F. J., et al., Phys. Stat. Sol. (a) 6,<br />

1481–1486 (2006).<br />

[10] Cuberes, M. T., Proc. of the 30th Annual Meeting<br />

of the Adhesion Society, 430–432 (2007).<br />

Contact:<br />

Dr. M. Teresa Cuberes<br />

Engineering School Professor<br />

University of Castilla-La Mancha<br />

Applied Mechanics and Project Engineering<br />

Laboratory of Nanotechnology<br />

Almadén, Spain<br />

Tel.: +34 902 204100 ext. 6045<br />

Fax: +34 926 264401<br />

teresa.cuberes@uclm.es<br />

www.uclm.es


Shhh. It’s the Innova AFM.<br />

No noise. Higher resolution. This kind<br />

of news doesn’t stay quiet for long.<br />

www.veeco.com/innova


S c a n n i n g S e c t i o n<br />

3D Orientation Microscopy<br />

Electron Backscatter Diffraction in a Combined FIB/SEM<br />

Combining electron backscatter diffraction (EBSD), a scanning electron microscope (SEM) and a focused ion<br />

beam (FIB) together into a single instrument enables three dimensional (3D) characterization of microstructure<br />

in crystalline materials. Combining these techniques together has enormous potential in materials science.<br />

Electron Backscatter Diffraction<br />

Traditionally, microstructure refers to<br />

features that are visually evident in an<br />

optical or electron microscope. However,<br />

many critical aspects of microstructure<br />

are not visually evident. For example,<br />

the crystallographic orientation of the<br />

constituent grains can not be observed in<br />

basic microscopic imaging. While some<br />

indirect evidence may be observed, other<br />

techniques are required to gain true<br />

quantitative information. If we consider<br />

the microstructure at the scale revealed<br />

40 • G.I.T. Imaging & Microscopy 4/2007<br />

Keywords:<br />

electron backscatter diffraction (EBSD),<br />

3D microstructure characterization Stuart I. Wright Stefan Zaefferer<br />

in the SEM then the crystallographic orientation<br />

is best characterized using<br />

EBSD. An EBSD pattern is formed when<br />

a stationary electron beam is focused on<br />

a highly-tilted and properly-prepared<br />

sample in the SEM. Electrons are scattered<br />

as the incident electron beam interacts<br />

with the crystal lattice within the<br />

Fig. 1: A schematic showing<br />

how EBSD is used to obtain<br />

orientation from a crystalline<br />

material and an example<br />

of an orientation map generated<br />

from the data.<br />

sample. Electrons satisfying physical<br />

laws governing the interaction of electrons<br />

with the crystal lattice are coherently<br />

diffracted out of the sample. The<br />

diffracted electrons form a pattern on a<br />

phosphor screen positioned near the<br />

sample. The pattern provides information<br />

about the structure of the crystal lattice<br />

within the interaction volume such<br />

as the orientation of the crystal lattice<br />

with respect to the sample reference<br />

frame. Orientation microscopy [1] is a<br />

fully automated technique where the patterns<br />

are imaged using a CCD camera<br />

and then analyzed to determine the corresponding<br />

orientation. The system controls<br />

the electron beam enabling the procedure<br />

to be repeated at each point on a<br />

scan grid superposed over the sample.<br />

By mapping the measured orientations<br />

to various color scales it is possible to<br />

visualize orientation aspects of the micro-


structure. Orientation microscopy has<br />

proven to be a valuable tool for characterizing<br />

crystalline materials as evidenced<br />

by the large number of papers<br />

published where the technique has been<br />

used in the research.<br />

3D Data Acquistion<br />

As with traditional metallographic techniques,<br />

EBSD is usually performed on 2D<br />

planes cut through a sample. 3D microstructural<br />

characterization can be<br />

achieved through serial sectioning. The<br />

technique simply comprises the cutting<br />

of material slices, recording of the structure<br />

of these slices and then reconstructing<br />

the 3D structure by stacking of the<br />

recorded images. Serial sectioning is applicable<br />

to a wide range of materials and<br />

material problems with the only serious<br />

disadvantage that it is destructive. For<br />

serial sectioning a large number of different<br />

methods can be imagined, e.g. mechanical<br />

cutting, grinding or polishing,<br />

chemical polishing, or laser or electrical<br />

discharge ablation. The main challenge<br />

associated with these methods is controlling<br />

the sectioning depth, obtaining flat<br />

and parallel surfaces and correctly redetecting<br />

and aligning the observation<br />

area. Also, these serial sectioning methods<br />

tend to be extremely laborious. A<br />

technique that avoids these problems is<br />

serial sectioning with an ion beam in a<br />

combined FIB-SEM. The impact of the<br />

ion beam onto the sample leads to localized<br />

removal of the target material and<br />

can therefore be used to mill parallel serial<br />

sections through the material a few<br />

nanometers in depth. Irradiating a material<br />

surface in grazing incidence allows<br />

preparation of smooth surfaces that show<br />

little damage [2]. Recent progress has led<br />

to a fully automated technique for alternately<br />

sectioning and subsequent scanning<br />

of the new section using EBSD<br />

[3–5].<br />

Data Visualization<br />

Software for analyzing and visualizing<br />

the wealth of information in the 3D EBSD<br />

data is still in its infancy. Basic functions<br />

based on the analysis of 2D EBSD data<br />

are being extended into 3D [6]. Visualization<br />

software is being built upon existing<br />

tools developed for general 3D rendering.<br />

Functions for rotating and slicing<br />

through the data cube as well as the ability<br />

to extract individual grains from the<br />

3D data have been developed. Grain facets<br />

can be colored according to their orientation<br />

relative to the associated crystal<br />

Fig. 2: Schematic of the instrument combining<br />

the EBSD, SEM and FIB techniques and micrograph<br />

of a milled section.<br />

lattice of the grain [7]. Figures 3 and 4<br />

show examples from steel and nickel.<br />

Conclusions<br />

A system for 3D orientation microscopy<br />

based on fully automated serial sectioning<br />

and EBSD based orientation microscopy<br />

has been developed in a FIB-SEM.<br />

The technique yields the crystal orientation<br />

at each voxel of the measured volume.<br />

A spatial resolution 50 x 50 x 50 nm³<br />

can be achieved. Volumes on the order of<br />

50 x 50 x 50 μm³ can be practically observed.<br />

The technique works on a wide<br />

variety of materials but some exceptions<br />

have been found, namely materials that<br />

adversely react to the Ga+-ion beam irradiation.<br />

The technique has been applied<br />

to several different materials and<br />

has yielded information that gives insight<br />

into microstructures that cannot be realized<br />

without the 3 rd dimension.<br />

References:<br />

[1] Adams, B. L., et al., Met. Trans. A 24, 819–<br />

831 (1986).<br />

[2] Michael, J., et al., Microscopy and Micoranal-<br />

ysis 13, 926–927 (2007).<br />

[3] Mulders, J. J. L., et al., Mat. Sci. Forum 495–<br />

497, 237–242 (2005).<br />

[4] Uchic, M. D., et al., Scripta Mat. 55, 23–28<br />

(2006)<br />

[5] Groeber, M. A., et al., Mat. Char. 57, 259–273<br />

(2006).<br />

[6] Rollett, A. D., et al., Annu. Rev. Mater. Res.<br />

37, 627–658 (2007).<br />

[7] Rowenhorst, D. J., et al., Scripta Mat. 55, 11–<br />

16 (2006).<br />

Further references are available from the au-<br />

thors.<br />

S c a n n i n g S e c t i o n<br />

Fig. 3: A 3D EBSD scan from pearlite structure in<br />

a steel sample showing a 3D data cube reconstructed<br />

from EBSD scans on serial sections.<br />

Fig. 4: A 3D EBSD scan from a nickel sample and<br />

a twinned grain extracted from the data and<br />

rendered in 3D.<br />

Contact:<br />

Stuart I. Wright<br />

Director of Applications Science and Software<br />

Engineering<br />

EDAX-TSL<br />

Draper, Utah, USA<br />

Tel.: +1 801 4952750<br />

Fax: +1 801 4952758<br />

stuart.wright@ametek.com<br />

www.edax.com<br />

Stefan Zaefferer<br />

Group Head – Diffraction and Microscopy<br />

Max-Planck-Institute for Iron Research<br />

Department of Microstructure Physics and Metal<br />

Forming<br />

Düsseldorf, Germany<br />

Tel.: +49 211 6792 803<br />

s.zaefferer@mpie.de<br />

www.mpie.de<br />

G.I.T. Imaging & Microscopy 4/2007 • 41


S c a n n i n g S e c t i o n<br />

Combining Optical Upright<br />

Microscopy and AFM<br />

Working with the Biomaterial Workstation BioMAT<br />

The atomic force microscope (AFM) is a flexible instrument that can be used for imaging, measuring forces<br />

and elastic properties and manipulating a variety of samples, at high resolution. The applicability of AFM is<br />

further extended in combination with light microscopy as optics deliver more bulk details and by fluorescence,<br />

compositional contrast. To date, the combination of AFM and light microscopy has been limited to<br />

samples on transparent substrates, where AFM has top-down access to the sample and an inverted light microscope<br />

has bottom-up access to the same area [1, 2].<br />

A Combination of Techniques<br />

However, there are many areas of research<br />

in which the combination of AFM<br />

with optical microscopy on opaque samples<br />

would be a powerful tool. Topics<br />

such as bacterial growth on metallic surfaces,<br />

bionics and surface chemistry, fluorescent<br />

polymers and coatings, i.e. areas<br />

from both life and material science<br />

could be addressed with such a combination<br />

of techniques.<br />

The main problem limiting the effective<br />

combination of these techniques on<br />

non-transparent surfaces has been providing<br />

access for both techniques at the<br />

same spot on the sample surface. To reach<br />

the full capabilities of optical microscopy,<br />

objective lenses with an extremely short<br />

working distance are required, leaving no<br />

space for AFM access to the same position.<br />

JPK Instruments has developed a solution<br />

to allow integration of upright optical<br />

microscopy and AFM, named the<br />

BioMAT Workstation (fig. 1).<br />

42 • G.I.T. Imaging & Microscopy 4/2007<br />

Portable Shuttle Stage<br />

The workstation spatially separates the<br />

upright optical microscope from the AFM<br />

to assure that neither of the two techniques<br />

is compromised. The key element<br />

of the BioMAT Workstation design is the<br />

portable shuttle stage on which the sample<br />

is loaded. The transfer of this shuttle<br />

stage from the upright optical microscope<br />

to AFM and vice versa allows precise<br />

positioning of the sample on both<br />

microscopes such that the same area is<br />

imaged by both systems. This transfer<br />

can be repeated as often as necessary,<br />

allowing the sequential measurement of<br />

optics and AFM.<br />

For combining upright light microscopy<br />

with AFM first the BioMAT Workstation<br />

has to be aligned, matching the<br />

AFM scan rage to the field of view of the<br />

optical microscope. To do this, a transparent<br />

reference sample displaying a<br />

cross-hair structure, which can be imaged<br />

with both systems is used (fig. 2).<br />

With the workstation’s integrated inverted<br />

optics, the AFM tip can be coarsely<br />

aligned with respect to the cross hair on<br />

the reference sample (fig. 3). This coarse<br />

alignment involves adjusting the position<br />

of the AFM head on top of the BioMAT<br />

Workstation so that the AFM tip position<br />

matches the center of the reference cross.<br />

By exchanging the reference sample with<br />

the sample of interest, the same area of<br />

the sample can be imaged sequentially<br />

with the two separate microscopes.<br />

Applications<br />

One area of research where combining<br />

light microscopy and AFM on opaque<br />

substrates would be useful is the investigation<br />

of bacteria with metal surfaces.<br />

Thiobacteria can leach mineral sulfides<br />

from various metals. During this process<br />

of bioleaching the bacteria, which mostly<br />

belong to species of thiobacillus ferrooxidans<br />

are in close contact to the mineral<br />

sulfides, forming a monolayered biofilm.<br />

AFM is particularly suitable for investigation<br />

of such biofilms as it allows the<br />

visualization and characterization of biological<br />

samples under physiological conditions<br />

with high spatial resolution. The<br />

option to combine AFM with fluorescence<br />

microscopy is a powerful means to correctly<br />

interpret and validate the topographic<br />

images obtained by AFM with


Fig. 1: NanoWizard II integrated into the BioMAT Workstation<br />

the help of corresponding images of fluorescently<br />

labelled structures. Since almost<br />

all sulphur containing minerals are<br />

opaque such samples are the perfect application<br />

for the BioMAT Workstation.<br />

Here, thiobacillus ferrooxidans was<br />

grown on a piece of compressed sulphur<br />

(sample courtesy Prof. Sand, University<br />

Duisburg-Essen). For fluorescence microscopy<br />

the bacterial DNA was stained<br />

using DAPI. Fluorescence images were<br />

acquired with a Zeiss AxioImager A1m<br />

equipped with a 100 x Acroplan water<br />

immersion objective. After imaging on<br />

the AxioImager, the specialized sample<br />

holder was transferred to the BioMAT<br />

Workstation and AFM imaging was conducted<br />

in intermittent contact mode in<br />

fluid using the NanoWizard II.<br />

As contrast in AFM is based on structure,<br />

the images of bacteria on the surface<br />

of the elementary sulphur are complex.<br />

One can see steps and structures in<br />

the sulphur substrate, as well as groups<br />

of bacteria on the surface. By comparing<br />

the AFM image with the fluorescence image<br />

of the same area it is clear which<br />

surface features correspond to bacteria<br />

(fig 4). When it is clear exactly which region<br />

of the surface contains bacteria the<br />

surface properties of the metal and the<br />

biofilm can be characterised in high resolution<br />

with the AFM.<br />

Conclusion<br />

With such a tool many new possibilities<br />

for combined imaging of biological samples<br />

on opaque surfaces are now possi-<br />

Fig. 2: To demonstrate imaging of the same area<br />

a mixture of 80 nm fluorescence PMMA and<br />

1 µm silica beads were first imaged in the upright<br />

microscope (left) and then with the AFM<br />

(right).<br />

Fig. 3: The cantilever of the AFM can be aligned<br />

with the chrome reference cross using the alignment<br />

optics integrated into the BioMAT workstation.<br />

This allows the user to establish common<br />

reference points in both AFM and optical<br />

space that can be applied to a sample of interest.<br />

ble. The investigation of structural and<br />

elastic properties of various types of cells<br />

on patterned surfaces can provide valuable<br />

information about the interaction of<br />

cells with potential implant surfaces. As<br />

seen here, the effect of biofilm formation<br />

on a metal or crystalline surface can be<br />

investigated. Polymer formation on<br />

opaque surfaces can also be investigated,<br />

using both fluorescence microscopy and<br />

AFM. Such access to a sample with the<br />

two forms of microscopy further extends<br />

the applicability of the AFM for characterization<br />

of samples on opaque surfaces.<br />

References:<br />

S c a n n i n g S e c t i o n<br />

Fig. 4: Imaging of bacteria on an opaque sulphur<br />

surface. In (a) DAPI stained bacteria have been<br />

imaged using fluorescent microscopy. The<br />

marked regions correspond to the AFM images<br />

in (b) and (c). While the fluorescent image makes<br />

it clear where the bacteria are located, high<br />

resolution structural information can only be<br />

derived from the corresponding AFM images.<br />

[1] Chiantia, S., Kahya, N., Schwille, P., Lang-<br />

muir. 21(14) 6317–23 (2005).<br />

[2] Poole, K., Müller, D., Br J Cancer. 92(8):1499–<br />

505 (2005).<br />

Contact:<br />

Jörg Barner<br />

application scientist<br />

JPK Instruments AG<br />

Berlin, Germany<br />

Tel.: +49 30 5331 12070<br />

Fax: +49 30 5331 22555<br />

nanowizard@jpk.com<br />

www.jpk.com<br />

G.I.T. Imaging & Microscopy 4/2007 • 43


M i c r o s c o p y Fa c i l i t i e s<br />

Cancer Research in Glasgow<br />

The Beatson Advanced Imaging Resource (BAIR)<br />

Cancer Research UK is a major funder of biomedical research in Europe. In 2006/2007 they spent £315 million<br />

to support over 500 research groups in the UK through a variety of funding mechanisms including research<br />

institutes, clinical centers, and grants. The Beatson Cancer Research Institute is one of four corefunded<br />

CR-UK research institutes. Recently, CR-UK invested £22 million towards a new building for the<br />

Beatson (Fig. 1) located on the University of Glasgow campus and scheduled for occupation in December of<br />

2007. This expansion will double the size of the institute and is associated with substantial new investment<br />

in research infrastructure.<br />

New Facility of the Beatson Cancer<br />

Research Institute<br />

The Beatson Advanced Imaging Resource<br />

(BAIR) is the new imaging facility of the<br />

Beatson Cancer Research Institute. The<br />

Bair occupies 280 m 2 of space in eleven<br />

rooms located primarily in the basement<br />

of the new building, with two additional<br />

rooms located in the biomedical unit to<br />

facilitate mouse in vivo imaging (see below).<br />

Smooth operation is ensured by two<br />

full time scientific officers who train users,<br />

assist with advanced applications,<br />

troubleshoot equipment, and perform<br />

quality control. Staff also maintain an internal<br />

website which includes tutorials,<br />

operating procedures, protocols, and<br />

equipment specifications in addition to<br />

the scheduling database through which<br />

both equipment and assistants are<br />

booked.<br />

The BAIR is managed by Dr Kurt I.<br />

Anderson, who brought to Glasgow the<br />

experience of having set up the light mi-<br />

44 • G.I.T. Imaging & Microscopy 4/2007<br />

croscopy facility of the Max Planck Institute<br />

for Cell Biology and Genetics in Dresden.<br />

Dr. Anderson also leads a research<br />

group at the Beatson Institute, which investigates<br />

actin dynamics in cell migration<br />

using advanced imaging techniques<br />

including TIRF, FRAP, photo-activation/<br />

-switching (PA/PS), and FLIM-FRET. The<br />

rational behind this joint appointment is<br />

to promote the use of advanced imaging<br />

techniques among Beatson research<br />

groups by introducing opportunities for<br />

substantive collaborations which might<br />

not be possible within the context of a<br />

service alone.<br />

Imaging Systems<br />

The imaging systems are roughly grouped<br />

according to applications (Fig. 2). At the<br />

south end of the facility (room 122) are<br />

three systems for medium throughput,<br />

high content, long term time lapse microscopy<br />

consisting of a Nikon TE2000 stand<br />

with Perfect Focus, ASI stage with linear<br />

Fig. 1: The new Beatson Institute for Cancer<br />

Research in Glasgow<br />

encoder, Sutter illumination system, and<br />

okolab incubator, all driven by Meta-<br />

Morph software. BAIR staff maintain<br />

FACS analyzers throughout the building,<br />

in addition to a BD FACSAria in room 123<br />

with 561 nm diode laser for use with red<br />

fluorescent proteins. Three confocal microscopes<br />

are housed in room 124, including<br />

a Leica TCS-SP2 with AOBS and<br />

Olympus FV1000 with 405 nm diode and<br />

SIM scanner. The latter system is especially<br />

powerful for FRAP and the use of<br />

PA/PS probes. A second TCS-SP2 is fitted<br />

with a Becker and Hickl module (SPC-<br />

730) and pulsed IR laser for time domain<br />

detection of FLIM-FRET. A small room<br />

(room 125) is centrally located for the<br />

placement of microscope laser modules,<br />

which promotes operational stability of<br />

the equipment and a more comfortable<br />

user environment. A workshop is located<br />

in room 126 along with a demo space for<br />

testing of new equipment. Room 127<br />

houses several custom systems for live<br />

cell imaging. A system for TIRF microscopy<br />

has been built based on a Nikon<br />

TE2000 stand and custom condensor incorporating<br />

405 and 473 nm diode lasers,<br />

which enables FRAP and PA/PS in<br />

TIRF. Integration of a 561 nm diode laser<br />

is currently under way, which will enable<br />

simultaneous GFP/RFP imaging in conjunction<br />

with an Optical Insights Dual-<br />

View and Roper 512 x 512 EMCCDFT


camera. A frequency domain system for<br />

FLIM-TIRF is under development together<br />

with Lambert Instruments based<br />

on their LIFA system, which will offer enhanced<br />

time resolution compared to the<br />

TCSPC approach and sensitive detection<br />

of protein-protein interactions at the cell<br />

surface due to the optical sectioning of<br />

TIRF. A LIFA system coupled to a<br />

Yokogawa CSU22 spinning disk scanhead<br />

is also planned. Room 130 houses several<br />

upright stands for imaging fixed<br />

specimens, including histological stains,<br />

immunofluorescence, and FISH. Offline<br />

Beatson Advanced Imaging Resource (BAIR)<br />

rm 122 rm 123 rm 124<br />

rm 125 / rm 126 rm 127 rm 130 rm 129 rm 131<br />

C<br />

D E<br />

E<br />

Fig. 2: Floor plan of the Beatson Advanced Imaging Resource (see text for details)<br />

�<br />

������� �� ���������������������<br />

����������������������������������������<br />

�������������������������������<br />

�������������������������<br />

����������������������<br />

�������������������������������������������������<br />

��������������������������<br />

��������������������������������<br />

B<br />

A<br />

workstations for image processing and<br />

analysis are located in room 129, including<br />

Volocity, MetaMorph, the Olympus<br />

and Leica confocal packages, and of<br />

course ImageJ. Finally, staff share office<br />

space in room 128.<br />

Focus at the Beatson<br />

rm 128<br />

The development and use of murine cancer<br />

models to better understand and treat<br />

human disease is an important focus at<br />

the Beatson. Mouse in vivo imaging involves<br />

a wide range of magnification, sen-<br />

A<br />

2 m<br />

M i c r o s c o p y Fa c i l i t i e s<br />

sitivity, and resolution depending on the<br />

experiment. For imaging groups of fluorescent<br />

cells down to the level of single<br />

cells an Olympus OV100 is used. With this<br />

system tumor progression can be monitored<br />

in a variety of tissues, including skin,<br />

colon and pancreas. For high resolution<br />

imaging of cells and sub-cellular structure<br />

is currently used an Olympus FV1000 confocal<br />

microscope. However a multi-photon<br />

TRIM scope from LaVision Biotec is under<br />

installation. This system incorporates both<br />

a Coherent Chameleon laser for excitation<br />

of GFP and an APE optical parametric oscillator<br />

for excitation of red fluorescent<br />

proteins. Finally, an Ivis 50 is used for detection<br />

of chemi-luminescence.<br />

The Beatson Institute looks forward to<br />

introducing the BAIR and their new research<br />

facility to the imaging community<br />

by hosting the European Light Microscopy<br />

Initiative meeting in Glasgow together<br />

with the University of Strathclyde in<br />

2009.<br />

Contact:<br />

Dr. Kurt I. Anderson<br />

Beatson Institute for Cancer Research<br />

Glasgow, Scotland, United Kingdom<br />

Tel.: +44 141 330 2864<br />

Fax: +44 141 942 6521<br />

k.anderson@beatson.gla.ac.uk<br />

��������������������������������<br />

� � ������� ��<br />

� ��������������������������<br />

����������� �� ���������������������������������������������<br />

�����������������������������������������������������������<br />

����������������������������������������������������������������<br />

����������������������������������������������������������������<br />

������������������������������������������������������������<br />

����������������������������<br />

����������� �� ������������������������������������������������������<br />

�������������������������������������������������������������<br />

��������������������������������������������������������������������<br />

��������������������������������������������������������������<br />

������������������������������������������������ �� �����������������<br />

������������������������������������������<br />

��������������������������������������������������������������<br />

������������������������������������������������������������������<br />

�����������������������������������������������������������������<br />

�������������������������������������������������������������<br />

�����������������������������������������<br />

� � � � � � � � � � � � � � � � � � � � � � � � � � � � � �<br />

G.I.T. Imaging & Microscopy 4/2007 • 45


C o v e r S t o ry<br />

The “F” Words<br />

FRET, FRAP, and FISH – Technology and Techniques<br />

The use of fluorescence microscopy in the life sciences<br />

runs the gamut from basic photodocumentation<br />

to dynamic single-molecule fluorescence (SMF)<br />

studies. Recent advances in digital imaging technology<br />

are helping to expand the utility and popularity<br />

of many fluorescence microscopy techniques,<br />

including Förster resonance energy transfer (FRET),<br />

fluorescence recovery after photobleaching (FRAP),<br />

and fluorescence in situ hybridization (FISH). The<br />

newly created Microimaging Applications Group,<br />

which comprises Photometrics, Media Cybernetics,<br />

QImaging, Gatan, and MAG Biosystems, offers a<br />

broad range of innovative imaging solutions designed<br />

to enhance life science research capabilities.<br />

FRET Imaging<br />

Förster resonance energy transfer is a<br />

phenomenon in which nonradiative<br />

transfer of energy occurs between donor<br />

and acceptor molecules in close proximity<br />

(2–7 nm). Since FRET efficiency decays<br />

as a function of the inverse sixth<br />

power of the distance between the donor<br />

and acceptor, this phenomenon can be<br />

leveraged to provide solid evidence of an<br />

interaction between the donor and acceptor<br />

in a FRET pair.<br />

In FRET, the donor molecule is returned<br />

to a ground state without fluorescence<br />

emission while the acceptor molecule<br />

is raised to an excited state. Upon<br />

decay of the acceptor’s excited state, fluorescence<br />

emission may be witnessed.<br />

Thus, an increase in FRET between label<br />

molecules will result in a decrease in donor<br />

emission and a simultaneous increase<br />

in acceptor emission. Using FRET detection,<br />

interactions between molecules can<br />

be monitored in subcellular compartments<br />

and tracked as a function of time.<br />

FRET applications include evaluating the<br />

structure of proteins, determining the<br />

spatial distribution and assembly of protein<br />

complexes, monitoring receptor/ligand<br />

interactions, and sensing the presence<br />

of small molecules in living cells.<br />

FRET experiments are often performed<br />

using standard ratio imaging<br />

46 • G.I.T. Imaging & Microscopy 4/2007<br />

techniques. Depending on the application,<br />

FRET is used to qualitatively or<br />

quantitatively investigate experimental<br />

phenomena. While qualitative experiments<br />

focus on simply determining the<br />

presence or absence of FRET as an indicator<br />

of interaction, quantitative experiments<br />

require a more methodical strategy.<br />

To help ensure accurate results,<br />

next-generation Photometrics electronmultiplying<br />

CCD (EMCCD) cameras provide<br />

exceptionally high quantum efficiency,<br />

quantitative stability across 16<br />

bits, and linear EM gain up to 1000x.<br />

Even with superior camera performance,<br />

sequential imaging techniques<br />

(e.g., using an emission filter wheel or<br />

switching microscope filter cubes) can<br />

make proper data calibration and correction<br />

very difficult, if not impossible,<br />

when dynamic samples are used. Therefore,<br />

many quantitative FRET applications<br />

require that the donor and acceptor<br />

emissions be simultaneously imaged. To<br />

meet this criterion, MAG Biosystems offers<br />

easy-to-use instrumentation that<br />

splits the incident beam from the microscope<br />

into independent beams. Each of<br />

the resultant emission channels is projected<br />

onto a region of a CCD or EMCCD<br />

array. A precision optical and mechanical<br />

design allows subpixel image registration<br />

and minimizes light loss for simultaneous<br />

multichannel acquisition.<br />

FRAP Imaging<br />

Fluorescence recovery after photobleaching<br />

is useful for examining intracellular<br />

molecular variables such as nuclear<br />

protein complex dynamics,<br />

diffusional mobility of membrane proteins,<br />

and cytoskeletal dynamics. FRAP is<br />

a powerful mode of fluorescence light<br />

microscopy in which a specialized illumination<br />

strategy is implemented in order<br />

to permit perturbation of the steady-state<br />

fluorescence distribution by bleaching<br />

fluorescence in selected regions of a sample.<br />

After the bleaching step, researchers<br />

can observe and analyze how the fluorescence<br />

distribution returns to the<br />

steady state. Because the photobleaching<br />

of fluorophores is permanent, changes in<br />

the fluorescence intensity in both the<br />

bleached and unbleached regions are attributable<br />

to the exchange of bleached<br />

and unbleached fluorescent molecules<br />

between those regions. FRAP microscopy<br />

is typically geared towards dynamic, lowlight<br />

endeavors.<br />

Recently, MAG Biosystems introduced<br />

a widefield imaging system designed to<br />

study the intracellular dynamics of proteins<br />

and other macromolecular complexes<br />

via FRAP and iFRAP (inverse<br />

FRAP) experiments in 2D plus time and<br />

3D plus time. Photoactivation and photo-


conversion studies with fluorescent proteins<br />

such as PA-GFP, EOS, KFP, Kaede,<br />

and Dronpa can be performed.<br />

Several technological innovations, including<br />

Burst mode and a custom optical<br />

path, provide a combination of speed,<br />

sensitivity, and ease of use not found in<br />

other FRAP systems. When run in Burst<br />

mode, the delay between the end of the<br />

bleach pulse and the first recovery image<br />

is minimized, enabling fast dynamic<br />

analyses. The FRAP-3D system also lets<br />

researchers photobleach-on-the-fly by<br />

simply clicking within a live image display<br />

window to bleach the region appearing<br />

under the cursor.<br />

FRAP-3D includes a galvanometerbased<br />

FRAP head, an advanced laser<br />

launch module, high-speed I/O circuitry<br />

to control all system components, acquisition<br />

and analysis software with an intuitive<br />

graphical user interface (GUI), and a<br />

configured workstation. The head can be<br />

mounted to many inverted microscopes<br />

through the epi-illumination port in order<br />

to enable simultaneous laser and widefield<br />

illumination. A versatile optical design<br />

allows researchers to switch seamlessly<br />

between FRAP studies and standard<br />

widefield applications without reconfiguring<br />

the system’s hardware.<br />

To meet user-specific quantum efficiency,<br />

spatial resolution, and frame rate<br />

requirements, the FRAP-3D system utilizes<br />

high-performance quantitative CCD<br />

and EMCCD cameras from Photometrics.<br />

To ensure the utmost instrumentation<br />

utility, FRAP-3D allows future upgrades<br />

for spinning-disk confocal microscopy<br />

and other imaging modalities.<br />

FISH Imaging<br />

Fluorescence in situ hybridization is a biochemical<br />

means of labeling specific nucleic<br />

acid sequences in cell preparations<br />

for the purposes of confirming the presence<br />

of certain genes and for spatial localization<br />

of sequences of interest within<br />

a cell or on chromosomes. Essentially,<br />

FISH provides a way to visualize and map<br />

genetic material in single cells. FISH has<br />

been instrumental in elucidating a variety<br />

of chromosomal abnormalities and<br />

genetic anomalies. The technique is used<br />

heavily in the basic research arena as<br />

well as the clinical arena. The use of<br />

FISH continues to grow quickly in such<br />

areas as genetics, cytogenetics, prenatal<br />

research, and tumor biology.<br />

The first step in FISH is the production<br />

of sequence-specific probes, which<br />

is accomplished by synthesizing antisense<br />

strands to sequences of interest<br />

and conjugating these antisense strands<br />

to fluorescent probes so that they can be<br />

detected using fluorescence microscopy.<br />

The power of FISH is greatly enhanced<br />

by the simultaneous use of multiple fluorescent<br />

probes. By using a multiplexing<br />

strategy, numerous nucleic acid sequences<br />

of interest can be detected and<br />

mapped.<br />

There are three basic types of FISH<br />

probes: (1) locus-specific probes, (2) alphoid<br />

or centromeric repeat probes, and<br />

(3) whole-chromosome probes. Locusspecific<br />

probes, which bind to a particular<br />

region of a chromosome, are useful<br />

for determining which chromosome a<br />

gene is located on once a small sequence<br />

of a particular gene has been isolated.<br />

Centromeric repeat probes, which are<br />

generated from repetitive sequences<br />

found in the middle of each chromosome,<br />

are utilized to determine whether a cell<br />

has the correct number of chromosomes.<br />

Whole-chromosome probes, which are<br />

collections of genetic sequences common<br />

to a particular chromosome, can be used<br />

to map individual chromosomes as well<br />

as to identify different chromosomes in<br />

respect to one another.<br />

Many researchers utilize two- to fourcolor<br />

FISH analysis on fixed samples.<br />

The main requirement for imaging of this<br />

kind is an ultra-high-resolution detector<br />

that allows all of the spatial information<br />

to be preserved under high magnification.<br />

Since the preparations most often<br />

contain fixed cells, more intense illumination<br />

can be used to produce stronger<br />

signals. A midrange-performance camera<br />

may be deemed adequate for fixedsample,<br />

moderate-light FISH studies,<br />

provided it offers sufficiently high spatial<br />

resolution.<br />

With the maturation of techniques<br />

such as spectral imaging, many researchers<br />

are now enhancing their FISH experiment<br />

capabilities by using a far greater<br />

number of fluorescent probes at one time.<br />

Spectral imaging enables the identification<br />

of probes based on their spectral<br />

curves, allowing differentiation of closely<br />

overlapping fluorophores. When a camera<br />

is utilized in conjunction with a spectral<br />

imaging system, a specimen’s fluorescent<br />

emission can be split into<br />

component wavelengths prior to reaching<br />

the detector. Thus, excellent camera performance<br />

and sensitivity become critical.<br />

To facilitate the use of FISH, QImaging<br />

has engineered a wide selection of easyto-operate<br />

CCD cameras that address<br />

myriad resolution, speed, and sensitivity<br />

requirements. QImaging also offers several<br />

color options, including solutions<br />

that combine the use of Bayer mask CCDs<br />

and innovative data interpolation meth-<br />

ods to deliver high-fidelity color rendition.<br />

Software Considerations<br />

Fluorescence microscopy techniques are<br />

evolving at a rapid pace. New fluorescent<br />

probes, camera technologies, and optics<br />

offer researchers an expansive set of investigative<br />

capabilities. The importance<br />

of using intelligent software for image<br />

acquisition, analysis, and management<br />

cannot be overstated.<br />

Powerful packages designed specifically<br />

for life science experiments, such as<br />

the Media Cybernetics line of software<br />

solutions, provide researchers the latest<br />

image analysis tools for object tracking,<br />

3D rendering, image deconvolution, and<br />

a broad diversity of fluorescence-based<br />

techniques. These programs integrate<br />

extensive hardware automation support<br />

and flexible image acquisition and<br />

processing into an easy-to-use GUI.<br />

Media Cybernetics also offers an image-asset<br />

management solution that lets<br />

life science researchers store, query, and<br />

share a large number of images (as well<br />

as data) via a client-server database application<br />

or the internet. Simple clickand-choose<br />

tools streamline archiving,<br />

searching, displaying, customizing, and<br />

reporting.<br />

Future Trends<br />

C o v e r S t o ry<br />

As imaging instrumentation simultaneously<br />

becomes more sophisticated yet<br />

simpler to use, the list of applications for<br />

FRET, FRAP, and FISH gets longer every<br />

day. Other fluorescence-based techniques<br />

benefiting from these technological advances<br />

include total internal reflection<br />

fluorescence (TIRF), fluorescence lifetime<br />

imaging microscopy (FLIM), and fluorescence<br />

anisotropy and polarization.<br />

Instrumentation providers such as the<br />

Microimaging Applications Group will<br />

continue to introduce high-performance<br />

cameras, versatile software, and complete<br />

imaging systems that offer life science<br />

researchers increasingly powerful<br />

and efficient capabilities for fluorescence<br />

microscopy.<br />

Contact:<br />

Karl Garsha<br />

Applications Scientist<br />

Microimaging Applications Group<br />

Photometrics<br />

Tucson, AZ, USA<br />

Tel.: +1 520 5472704<br />

Fax: +1 520 5731944<br />

kgarsha@photomet.com<br />

www.magworldwide.com<br />

G.I.T. Imaging & Microscopy 4/2007 • 47


Systematic Analysis of FRAP Experiments<br />

An Approach for the Evaluation of Spatially Resolved Data<br />

Introduction<br />

L i g h t M i c r o s c o p y<br />

Sebastian Seiffert<br />

Fluorescence Recovery after Photobleaching (FRAP)<br />

is a versatile technique to study dynamic phenomena.<br />

Performing FRAP on a confocal laser scanning<br />

microscope documents the recovery process with<br />

high spatial resolution. This enables a consistent<br />

determination of the diffusion coefficient and the<br />

dimensionality of diffusion in calibration-free manner.<br />

Moreover, experiments representing multi-component<br />

diffusion can be analyzed as well, thus<br />

yielding the distribution of diffusion coefficients.<br />

In the last 30 years, Fluorescence Recovery<br />

after Photobleaching (FRAP) has<br />

evolved as a versatile technique to determine<br />

diffusion coefficients of suitably labeled<br />

species in fields like pharmaceutical<br />

research, biophysics or polymer<br />

chemistry [1]. Basically, a FRAP experiment<br />

is realized by bleaching a certain<br />

area of a sample by short and intense laser<br />

irradiation. Afterwards, the diffusion<br />

of unbleached molecules from the surroundings<br />

leads to a temporal recovery<br />

of fluorescence intensity in the bleached<br />

region that is monitored with a highly attenuated<br />

beam. The diffusion coefficient<br />

can be deduced from the rate of recovery<br />

after suitable calibration. This procedure<br />

forms the basis for a variety of classical<br />

approaches to evaluate FRAP experiments<br />

[1].<br />

Analysis of Spatially Resolved FRAP –<br />

Theoretical Background<br />

When FRAP is performed on a confocal<br />

laser scanning microscope (CLSM), the<br />

recovery process can be followed with<br />

high spatial resolution on a µm-scale besides<br />

temporal resolution. Utilizing this<br />

aspect for a systematic analysis of FRAP<br />

48 • G.I.T. Imaging & Microscopy 4/2007<br />

data enables the estimation of the diffusion<br />

coefficient and the dimensionality of<br />

the diffusion process without any need<br />

for calibration [2].<br />

Basically, each FRAP experiment<br />

makes use of the diffusion equation,<br />

which is also known as Fick’s second<br />

law:<br />

(1)<br />

where C represents the concentration of<br />

the substance under consideration, while<br />

D is the translational diffusion coefficient.<br />

Eq. (1) describes diffusion phenomena<br />

in an isotropic medium for the one-,<br />

two- or three-dimensional case. Solutions<br />

can be derived for certain initial<br />

and boundary conditions [3]. We focus<br />

on the following simple case which is relevant<br />

to FRAP experiments on a CLSM:<br />

(2)<br />

Herein, r represents the generalized (radial)<br />

coordinate, and M denotes the total<br />

amount of the diffusing species in the<br />

three-dimensional case, while in the two-<br />

or one-dimensional case, it stands for the<br />

amount of substance per unit length or<br />

unit area, respectively. d symbolizes the<br />

diffusion dimension.<br />

The shapes of the functions according<br />

to Eq. (2) are Gaussians with an e- 1/2 -radius<br />

of . They broaden and become<br />

shallower with increasing time. Since the<br />

decrease of the prefactor with time depends<br />

on the dimensionality and the<br />

amount of diffusing substance, it is suited<br />

to estimate either of these parameters.<br />

On the other hand, the diffusion coefficient<br />

can be derived from the course of<br />

the widths.<br />

If we consider FRAP processes, the<br />

situation is in essence just inversed, since<br />

bleaching takes away a certain amount<br />

of the fluorescent molecules, while the<br />

considerations above dealt with a local<br />

excess of a substance. This means no<br />

more than a change of sign combined<br />

with a baseline shift.<br />

By utilizing a CLSM and an objective<br />

with a low NA value, it is readily possible<br />

to bleach geometries into the sample that<br />

correspond closely to the cases of diffusion<br />

from a plane (one-dimensional) or a<br />

line source (two-dimensional). Hence,<br />

the solutions of Fick’s second law are applicable<br />

to FRAP processes too. One obtains


eady for<br />

����<br />

����������� ���� �� �� ��� ������� ����� ���������<br />

������� ������ �������� ������������ �� ����� ���<br />

������������� �������� �� �������� ��� �����<br />

�������������� ��������� ��� �����������<br />

���������������� ���� ������������� �������� �����<br />

��������������� ��� ����� �������� �����<br />

������������ ��������������� ��� ���������������<br />

������� ���� ����������� �������� ���� �� ������� ����<br />

���� ������ ��� ������ ��� ���� �� ����������<br />

��� ���� ���� �� �� ���������������������<br />

��� ������������<br />

����������������������<br />

���������������������


L i g h t M i c r o s c o p y<br />

where I represents the fluorescence intensity<br />

at the position r and the time t after<br />

bleaching. M now is formally a fluorescence<br />

intensity (per length or area for<br />

d = 2 or 1) corresponding to the amount<br />

of fluorophore destroyed by bleaching. w<br />

is the e- 1/2 -radius of the Gaussian function<br />

and I 0 denotes the background intensity<br />

at r → ∞.<br />

The quantity D appears in the prefactor<br />

and the exponent of the Gaussian,<br />

hence both terms can be used to determine<br />

it. Comparison of the exponential<br />

terms of Eq. (3) yields<br />

(4)<br />

and therewith the possibility to deduce<br />

the diffusion coefficient by plotting w² vs.<br />

t for a series of intensity profiles obtained<br />

from images taken during the recovery<br />

process. A straight line with slope 2D<br />

passing through the origin should be expected.<br />

On the other hand, we obtain an algebraic<br />

decay for the time dependence of<br />

A(t):<br />

(5)<br />

As an alternative, Eq. (5) can be written<br />

in logarithmic form:<br />

(6)<br />

This means that a plot of log A versus log<br />

t should give a straight line with a slope<br />

of –d/2, forming the basis for the experimental<br />

determination of the dimensionality<br />

of the diffusion process.<br />

Analysis of Spatially Resolved FRAP –<br />

Practical Realization<br />

Figure 1 shows some intensity profiles<br />

taken from FRAP measurements on solutions<br />

of rhodamine B in glycerol. One observes<br />

Gaussians as expected from Eq.<br />

(3). Plots of the fit parameters w² vs. t<br />

thus yield straight lines. However, they<br />

do not pass through the origin but show<br />

a notable intercept as illustrated in figure<br />

2a. Moreover, double-logarithmic<br />

plots of A vs. t do not give straight lines<br />

but curves as depicted in figure 2b.<br />

These findings can be addressed to<br />

the fact that in a real experiment, the initial<br />

conditions used so far to obtain ex-<br />

50 • G.I.T. Imaging & Microscopy 4/2007<br />

(3)<br />

Fig. 1: <strong>Images</strong> and corresponding intensity profiles obtained from FRAP experiments<br />

on rhodamine B in glycerol for one-dimensional diffusion after bleaching a line (a)<br />

and two-dimensional diffusion after bleaching a point (b) into the focal plane. They<br />

show the sample before bleaching and at 1s (�), 30 s (�), and 90 s (�) after bleaching,<br />

respectively. The image dimensions are about 80 × 80 μm² in each case. Full<br />

lines in the diagrams represent fits to Gaussians like Eq. (3). Adapted from [2].<br />

Fig. 2: Plots of w² vs. t and log A vs. log t for two examples of FRAP experiments on<br />

rhodamine B in glycerol. �: one-dimensional line-bleaching, �: two-dimensional<br />

point-bleaching. (a) & (b) represent the real case accompanied by deviations from<br />

ideality. Introduction of an appropriate time shift t 0 transforms the situation into<br />

the ideal case as illustrated in (c) & (d). Adapted from [2].<br />

act solutions of the diffusion equation are<br />

never met perfectly. Instead of starting<br />

from a Dirac function (in one, two, or<br />

three dimensions) for the concentration<br />

profile at t = 0, the initial state will be<br />

characterized by some finite spatial<br />

width. Furthermore, some time is required<br />

to achieve bleaching. This both<br />

means that it is not possible to exactly<br />

define the zero point of the time scale.<br />

Since in an experiment the initial conditions<br />

are only approximated, one should<br />

be aware that deviations from Eq. (2) are<br />

expected when such approximations cannot<br />

be neglected. That is the case when<br />

the diffusion occurs fast in comparison<br />

with the duration of the bleach pulse, or<br />

when concentration profiles are analyzed<br />

on a length scale comparable to the spatial<br />

width of the bleaching beam.<br />

However, with the aid of a procedure<br />

described recently [2] one is able to take<br />

the deviations from ideality into account<br />

by a proper shift of the experimental<br />

time scale. Advantageously, the two independent<br />

criteria requiring<br />

i the intercept of the plot of w² vs. t to<br />

vanish<br />

ii the curvature of the plot of log A vs.<br />

log t to be minimized<br />

can be consistently employed to determine<br />

the appropriate shift t 0 . For convenience,<br />

we proceed in two steps: after<br />

deriving a first estimate for t 0 from an<br />

extrapolation of the w² vs. t plot (crite-


Fig. 3: D-distributions estimated from mixtures of differently sized<br />

fluorescent polystyrene microspheres (Molecular Probes, Eugene)<br />

in aqueous suspension. The shading below the signals visualizes<br />

their intensity in an alternative form.<br />

rion i), fine-tuning is achieved<br />

by plotting log A vs. log (t + t 0)<br />

and varying t 0 iteratively with<br />

respect to fulfill criterion ii.<br />

Therewith, the graphs depicted<br />

in figure 2a and 2b<br />

turn into the ones shown in<br />

figure 2c and 2d, which now<br />

correspond to the expectations<br />

according to Eq. (4) and<br />

(6), i.e., the ideal case.<br />

A number of experiments<br />

analyzed by the method<br />

shows that the diffusion coefficient<br />

and the expected dimensionality<br />

are obtained<br />

with high accuracy. Some results<br />

from measurements on<br />

rhodamine B in glycerol as<br />

well as aqueous suspensions<br />

of differently sized fluorescent<br />

microspheres are compiled<br />

in table 1 and 2, illustrating<br />

that both, D and d, can<br />

be estimated in consistent<br />

manner. Note that these analyses<br />

do not require any kind<br />

of previous calibration.<br />

Multi-component FRAP<br />

Processes<br />

Besides the possibilities for<br />

the evaluation of simple FRAP<br />

experiments as discussed<br />

above, the usage of temporal<br />

and spatial information also<br />

enables the estimation of multiple<br />

diffusion coefficients<br />

from experiments where<br />

more than one species is involved.<br />

The principle for this<br />

is to superimpose Eq. (3) for<br />

many independently diffusing<br />

components with diffusion coefficients<br />

D i and corresponding<br />

fractions M i and to fit the<br />

entire dataset of fluorescence<br />

intensity profiles as a function<br />

of time and space I(r,t).<br />

This yields a set of D i;M i-pairs,<br />

i.e., the distribution of diffusion<br />

coefficients. Figure 3 depicts<br />

several examples for<br />

such distributions as derived<br />

from mixtures of the differently<br />

sized spherical probes<br />

mentioned above (cf. table 2).<br />

Ratios of 10:90, 30:70, 50:50,<br />

70:30 and 90:10 were employed<br />

in this respect. The results<br />

illustrated show that one<br />

accurately finds both, the Dvalues<br />

of the pure compounds<br />

as well as the correct compositions<br />

when analyzing the<br />

mixtures. Note that these<br />

types of analyses can again<br />

be performed without any<br />

need for calibration. Moreover,<br />

they also allow one to determine<br />

the accompanying<br />

distributions of dimensionalities<br />

(results not shown) and to<br />

account for effects that sometimes<br />

penetrate a FRAP experiment,<br />

e.g., a continuous<br />

loss of fluorescence intensity<br />

due to bleaching during the<br />

scanning process.<br />

Closing Remark<br />

All procedures for the systematic<br />

evaluation of spatially<br />

resolved FRAP data mentioned<br />

here are fully implemented<br />

in a MATLAB software,<br />

which is available upon<br />

request from the authors.<br />

Note that in principle, one<br />

could also imagine to modify<br />

them with regard to allow for<br />

the analysis of anisotropic<br />

FRAP experiments, what is<br />

presently in progress in our<br />

group.<br />

References:<br />

[1] Meyvis, T. K. L., et al., Pharm.<br />

Res. 16, 1153–1162 (1999).<br />

[2] Seiffert, S., Oppermann W., J.<br />

Microsc. 220, 20–30 (2005).<br />

[3] Crank, J., The Mathematics of<br />

Diffusion, Clarendon Press, Ox-<br />

ford, 1975.<br />

L i g h t M i c r o s c o p y<br />

experiment D/(μm s 2–1 ) d<br />

line-bleaching (one-dimensional diffusion) 0.58 ± 0.04 1.02 ± 0.03<br />

point-bleaching (two-dimensional diffusion) 0.57 ± 0.11 2.00 ± 0.09<br />

Tab. 1: Translational diffusion coefficients (D) and dimensionalities (d) obtained from<br />

FRAP measurements on solutions of rhodamine B in glycerol. Adapted from [2].<br />

method r (24 nm microspheres)/nm r (100 nm microspheres)/nm<br />

FRAP 17.2 50.2<br />

DLS 16.7 49.5<br />

UA 14.0 51.7<br />

AFM 12.2 49.1<br />

Tab. 2: (Hydrodynamic) radii of fluorescently labeled microspheres with diameters of<br />

about 24 nm and 100 nm as estimated by FRAP in comparison with dynamic light<br />

scattering (DLS), ultracentrifugal analysis (UA) and atomic force microscopy (AFM).<br />

������� ������� ���<br />

����������� ��������� ��� ����<br />

�� �� � ����<br />

A publication focusing on the analy-<br />

sis of multi-component FRAP exper-<br />

iments is presently in preparation.<br />

Contact:<br />

Sebastian Seiffert<br />

Gerald I. Hauser<br />

Wilhelm Oppermann<br />

Clausthal University of Technology<br />

Institute of Physical Chemistry<br />

Clausthal-Zellerfeld, Germany<br />

Tel.: +49 5323 72 3642<br />

Fax.: +49 5323 72 2863<br />

sebastian.seiffert@tu-clausthal.de<br />

� ������������ ���������<br />

�������� ��� ������ � ����������� ������<br />

� ����������� �������� ��� ����� �����������<br />

��� ��������� ������������ ����� ��������������<br />

� ��� �� �������� ������� ��������<br />

� ���������� ������<br />

��� ���� ����������� ��� ������ ������� �����<br />

�����������������<br />

G.I.T. Imaging & Microscopy 4/2007 • 51


L i g h t M i c r o s c o p y<br />

Wide-field CARS-Microscopy<br />

Functional Imaging at a Glimpse<br />

Coherent anti-Stokes Raman scattering (CARS) microscopy is a branch of nonlinear microscopy that allows<br />

chemical imaging of targeted vibrational transitions in unstained samples. A resonantly enhanced blueshifted<br />

CARS signal is generated from NIR or visible light, thus the method is more sensitive than normal Raman<br />

microscopy and offers better resolution than IR microscopy. CARS microscopes are mostly set up as<br />

confocal scanning microscopes, but wide-field approaches are possible as well.<br />

Brief Introduction to CARS-microscopy<br />

The beating between two laser beams<br />

overlapping at a sample in space and<br />

time can excite vibrations of molecules in<br />

the sample, if their difference in frequency<br />

matches a vibrational transition.<br />

In this case inelastic coherent anti-Stokes<br />

Raman scattering (CARS) takes place and<br />

Fig.1: Fluorescence-label-free differentiation of<br />

polymer beads by wide-field CARS microscopy.<br />

52 • G.I.T. Imaging & Microscopy 4/2007<br />

a blue-shifted (anti-Stokes) signal beam<br />

is emitted. As this is a nonlinear optical<br />

process, high intensities, i.e. pulsed lasers,<br />

are required. The CARS signal grows<br />

proportional to the Stokes intensity and<br />

quadratically with the pump intensity and<br />

the number of resonant scatterers. CARSmicroscopy<br />

allows labelling-free mapping<br />

of spatial distributions of organic molecules<br />

as shown in figure 1. As no staining<br />

with fluorescent dyes is necessary, one<br />

avoids problems of photobleaching and<br />

phototoxicity alltogether.<br />

Since pioneering work in the early<br />

eighties [1], over the last 10 years CARS<br />

microscopy has developed into a powerful<br />

new microscopic tool, triggered especially<br />

by the work of Zumbusch, Holtom,<br />

and Xie [2] in the late nineties. Many<br />

technical variations of CARS microscopy<br />

have been developed (see [3] for an overview)<br />

most of them motivated by the necessity<br />

to increase the signal to noise ratio,<br />

which is often affected by a nonlinear<br />

CARS background that is not chemically<br />

selective.<br />

Keywords:<br />

coherent anti-Stokes Raman scattering, vibrational<br />

imaging, labelling-free imaging, nonlinear<br />

microscopy, wide-field microscopy<br />

Setup and Instrumentation<br />

Most CARS microscopes are based on<br />

confocal scanning microscopy, working<br />

with tightly focussed beams of NIR laser<br />

pulses in the pico- or femtosecond regime<br />

and with oil-immersion objectives of high<br />

numerical aperture (NA). The confocal<br />

(CF) setup has the advantage of high spatial<br />

resolution, but requires scanning of<br />

the sample, which is avoided in wide-field<br />

(WF) CARS microscopy where one can<br />

take an image of the whole sample “all at<br />

once”. In contrast to scanning CARS-microscopy,<br />

where the tight focussing of the<br />

lasers leads to large momentum uncertainty<br />

and thus makes momentum conservation<br />

rather uncritical, in wide-field<br />

CARS the phase-matching condition imposes<br />

a restriction that has to be met. In<br />

our case a special excitation geometry,<br />

which satisfies momentum conservation<br />

among the beams in the sample, enables<br />

efficient build-up of a CARS signal from<br />

the scatterers [4–5]. The idea for the implementation<br />

of wide-field CARS in the


“extremely-folded box-CARS geometry”<br />

is schematically shown in figure 2. We<br />

use nanosecond pulses and wavelengths<br />

in the NIR, for example 1064 nm for the<br />

Stokes pulse and a wavelength around<br />

800 nm for the pump pulse which is tuned<br />

by an optical parametric oscillator (OPO).<br />

L i g h t M i c r o s c o p y<br />

Fig. 2: Schematic sketch of a wide-field CARS-microscope: The excitation laser beams distribute the<br />

intensity of ns pulse trains homogeneously over the whole sample region of interest. The pump beam is<br />

coupled through a high numerical aperture dark field condenser delivering a cone of light from above,<br />

while the Stokes beam is guided in from below through the objective of the inverted microscope. This<br />

gives rise to an anti-Stokes signal beam that counter-propagates with respect to the Stokes beam, and<br />

which can conveniently be read out through the microscope objective. Note that the high numerical<br />

aperture of the dark-field condenser provides a narrow “sheet of light” illumination which reduces the<br />

non-resonant CARS background.<br />

Fig. 3: Selective imaging of a mix of polymer beads at two CARS resonances. The 1,44 µm large particles<br />

are excited at two different CARS resonances which are selected by tuning the NIR pump beam.<br />

The PS and the PMMA CARS images were overlayed in the image on the left, the measured Raman<br />

spectra of the Raman-active resonances that were used are shown on the right.<br />

Fig. 4: Snapshot image of an olive-oil droplet that was imaged with 3 ns, with a single pair of nanosecond<br />

pulses.<br />

This gives access to the aliphatic stretching<br />

vibrations between 2850 cm –1 and<br />

2950 cm –1 , which are abundant in organic<br />

molecules and give a particularly<br />

strong CARS signal. Other non-scanning<br />

apporaches [6] are based on collinear geometries<br />

and rely on the scattering struc-<br />

Image processing and analysis<br />

in microscopy<br />

System solutions for industry,<br />

medicine and life science<br />

Platform independent Image<br />

Management Tools<br />

Consulting for Complete<br />

Digital Management Solutions<br />

Universal drivers for digital<br />

cameras<br />

Interfaces to customer<br />

applications<br />

www.imagic-imaging.com


tures themselves to redirect the beams to<br />

satisfy phase-matching. As phase-matching<br />

is not satisfied in the bulk solvent, the<br />

nonlinear background from the solvent is<br />

effectively prevented.<br />

Performance<br />

L i g h t M i c r o s c o p y<br />

As a consequence of the photon energy<br />

conservation ω_ aS = 2ω_ P – ω_ S, the anti-<br />

Stokes wavelength can be considerably<br />

shorter than the beams exciting the sample.<br />

The combination of NIR excitation and<br />

detection at shorter wavelength makes it<br />

possible to have deep penetration depth<br />

into the sample volume and good optical<br />

resolution at the same time. The lateral<br />

spatial resolution of a WF-CARS microscope<br />

is on the same order as for an ordinary<br />

WF-microscope. The NA of the<br />

microscope objective and the CARS-wavelength<br />

determine the diffraction-limit of<br />

the spatial resolution, whereas the NA of<br />

the dark-field condensor is adapted to fulfil<br />

phase-matching, and thus determines<br />

the efficiency, rather than the resolution.<br />

The actually achieved imaging performance<br />

depends not only on the wavelengths<br />

and the NA of the microscope objective<br />

and the dark-field condensor, but also on<br />

the details of the coherent nonlinear interaction<br />

in the sample (e.g.on the local refractive<br />

index and its influence on the<br />

phase-matching). In practice, we have<br />

been able to image spherical polymer particles<br />

as small as 500 nm in diameter. Axially<br />

one can achieve optical sectioning,<br />

since the region where all interacting<br />

beams have sufficiently high intensity is<br />

only a few µm wide. The nonlinear dependence<br />

on the pump field favors optical<br />

sectioning, as in other types of nonlinear<br />

microscopes. We have measured the optical<br />

sectioning power of our system to be<br />

on the order of 3–4 µm.<br />

The sensitivity of the imaging depends<br />

on the coupling strength of the trageted<br />

Raman-active resonance and quadratically<br />

on the number of scattereres in the<br />

sample. Presently the sensitivity of CF-<br />

CARS microscopy lies around 10 5 vibrating<br />

molecules per focal volume for probing<br />

the symmetric CH 2 stretching mode<br />

[7], which allows to image single lipid bilayers<br />

and cellular membranes [8].<br />

Figure 3 examplifies the typical performance<br />

of our wide-field system. A mix<br />

of two species of 1.44 µm polymer beads<br />

in water is imaged. For polystyrol (PS)<br />

the frequency difference between Stokes<br />

and pump beam was tuned to a CARS<br />

resonance at 3052 cm –1 shown in the upper<br />

spectrum on the right in figure 3. Another<br />

image was recorded while selec-<br />

54 • G.I.T. Imaging & Microscopy 4/2007<br />

Fig. 5: WF-CARS images of small pulmonary surfactant vesicles inside living lung cells from rats.<br />

tively exciting the polymethyl methacrylate<br />

(PMMA) beads at 2943 cm –1 (see lower<br />

spectrum). The beads are spatially and<br />

chemically well resolved, with the spectral<br />

resolution of our WF-CARS microscope<br />

being limited by the bandwidth of<br />

the OPO, which is about 5 cm –1 .<br />

A specialty of our WF-CARS microscope<br />

is the option to take snapshots with<br />

only a single pair of pump- and probe<br />

pulses of 3 ns duration (see fig. 4). This<br />

enables real-time imaging of micrometer-sized<br />

structures, which could be useful<br />

to study transport phaenomena in biological<br />

systems. However, the speed of<br />

acquisition is limited by the repetition<br />

rate of the OPO which is typically less<br />

than 50 Hz.<br />

Applications<br />

Lipids give a particularly strong CARS<br />

signals, thus CARS-microscopy may become<br />

an established tool in lipid metabolism<br />

research, which is a very important<br />

topic at present. Many groups are currently<br />

setting up CARS experiments with<br />

living cells, e.g. trying to visualize<br />

changes in the location, distribution, or<br />

concentration of fatty acids.<br />

Our group is interested in functional<br />

imaging of phospholipid-rich vesicles inside<br />

living alveolar cells. These cells,<br />

called alveolar type II cells, are specialized<br />

to generate cell vesicles (the “lamellar<br />

bodies”) that contain pulmonary surfactant.<br />

These vesicles are released from<br />

the cells by exocytosis to provide the<br />

monomolecular lipid film of surfactant<br />

that is required to lower the surface tension<br />

in the alveoli. Figure 5 shows CARS<br />

images where these small intracellular<br />

vesicles selectively light up inside living<br />

alveolar cells.<br />

Since CARS-microscopy is a labellingfree<br />

method it may prove the ideal<br />

method for risk assesment of nanoparticles,<br />

which in modern nanomedicine are<br />

becoming increasingly popular, e.g. as<br />

contrast agents or for drug delivery. To<br />

date there often exists only limited or unsatisfactory<br />

information on the “fate” of<br />

the nanoparticles in the human body. Be-<br />

ing an optical method, diagnostics deep<br />

within the body is impossible – but CARS<br />

microscopy can provide information on<br />

the up-take or clearance of the nanoparticles<br />

and their degradation products in<br />

cell or tissue studies, for instance,<br />

whether they build aggregates anywhere<br />

in the cells or tissues. The fact that the<br />

no prior marking of the particles with<br />

fluorescent dyes is necessary – which<br />

might change the behavior in the tissue<br />

– could be of special importance.<br />

We expect CARS-microscopy to reach<br />

its full potential in the next years, developing<br />

a user-friendly and compact optical<br />

tool that provides answers for many<br />

of tomorrow‘s urgent questions in the life<br />

and material sciences.<br />

Acknowledgements<br />

This work was supported by the Austrian<br />

Science Fund (FWF-Project Nr. P16658_<br />

N02).<br />

References:<br />

[1] Duncan, M., et al., Opt. Lett. 7, 350–51<br />

(1982).<br />

[2] Zumbusch, A., et al., Phys. Rev. Lett. 82,<br />

4142–45 (1999).<br />

[3] Volkmer, A., J. Phys. D.: Appl. Phys. 38, R59–<br />

81 (2005).<br />

[4] Heinrich, C., et al., Appl. Phys. Lett. 84, 816–<br />

18 (2004).<br />

[5] Heinrich, C., et al., New J. Phys. 8, 36 (2006).<br />

[6] Toytman, I., et al., Opt. Lett. 32, 1941–43<br />

(2007).<br />

[7] Potma, E. O., et al., Optics Letters 31, 241–<br />

243 (2006).<br />

[8] Potma, E. O., Xie, X. S., Journal of Raman<br />

Spectroscopy 34, 642–50 (2003).<br />

Contact:<br />

Prof. Dr. M.A.M. Ritsch-Marte<br />

Chair of Biomedical Physics<br />

Prof. Dr. S. Bernet<br />

Dr. C. Heinrich<br />

Innsbruck Medical University<br />

Innsbruck, Austria<br />

Tel./Fax: +43 512 9003 70870<br />

monika.ritsch-marte@i-med.ac.at<br />

www2.i-med.ac.at/medphysik/


Next Generation Light Sources for Imaging<br />

Fibre Lasers – Compact, Cost-Effective, Turnkey Solutions<br />

Lasers continue to be increasingly important components within biological imaging and analysis systems –<br />

from Argon-Ion and HeNe lasers used within flow cytometry and confocal microscopy, to femtosecond Ti:<br />

Sapphire and DPSS femtosecond sources in multi-photon microscopy. However, many commercial imaging<br />

systems are still limited in performance by the availability of suitable laser sources – both in the limited<br />

availability of wavelengths and in the size, cost and reliability of conventional laser technologies. Ultrafast<br />

fibre lasers offer turnkey, compact and reliable solutions for next generation biomedical imaging systems.<br />

Here we introduce the basic concept of the ultrafast fibre laser and describe some of the applications and<br />

benefits of this technology within biophotonics research and imaging systems.<br />

Ultrafast fibre lasers, with their compact<br />

form, inherent reliability and low cost of<br />

ownership are becoming the laser of<br />

choice for many biomedical imaging applications,<br />

challenging the Ti:Sapphire<br />

laser within multi-photon excitation microscopy<br />

and diode, HeNe and Ar-Ion lasers<br />

within fluorescence imaging.<br />

The Master Oscillator, Power Amplifier<br />

(MOPA) architecture of Fianium’s ultrafast<br />

fibre lasers provides simplicity<br />

and flexibility by design [fig. 1]. The<br />

MOPA comprises two independent modules<br />

– a low-power femtosecond or picosecond<br />

fibre laser followed by a high<br />

power diode-pumped fibre amplifier. By<br />

independently changing the parameters<br />

of the oscillator or the amplifier modules,<br />

one can achieve very different performance<br />

parameters from the laser, tailored<br />

to a given application. This approach has<br />

quickly enabled ultrafast fibre lasers to<br />

meet the growing demands of a wide<br />

range of applications.<br />

The master oscillator is an all-fibre,<br />

passively modelocked laser which is both<br />

turnkey operated and self-starting without<br />

the need for any adjustment. The<br />

pulse repetition rates of the laser (from<br />

less than 1MHz to several hundred MHz)<br />

are ideally suited to quasi-cw laser applications<br />

and also for lifetime imaging applications.<br />

For applications within microscopy, ultrafast<br />

lasers are typically associated<br />

with multi-photon fluorescence microscopy,<br />

where femtosecond Ti:Sapphire lasers<br />

have been the historic laser of choice.<br />

While not offering the broad wavelength<br />

tunability of the Ti:Sapphire, Fianium’s<br />

FP1060-s femtosecond lasers do offer a<br />

low-cost, compact solution at discrete<br />

wavelengths from 980 nm to 1100 nm<br />

and offer potential for incorporation into<br />

any existing microscope system.<br />

Delivering average powers up to 5<br />

Watts and with pulse durations shorter<br />

than 250 femtoseconds, the long wave-<br />

L i g h t M i c r o s c o p y<br />

John Clowes<br />

lengths of the FP1060, extending beyond<br />

the tuning limits of most Ti:Sapphire<br />

sources, offer many benefits for Two Photon<br />

Fluorescence (TPF) and Second Harmonic<br />

Generation (SHG) microscopy [1, 2].<br />

The FP1060 high power lasers from<br />

Fianium operate within the picosecond<br />

or femtosecond regime and can provide<br />

pulse energies from a few pico-joules to<br />

ten microJoules, particularly important<br />

for materials processing, both of devices<br />

and of tissues.<br />

Extension of the FP1060 laser source<br />

from the near Infra Red to the visible and<br />

UV region of the spectrum, is achieved<br />

through nonlinear frequency conversion<br />

techniques including harmonic generation<br />

to 532 nm, 355 nm and 266 nm and<br />

in the generation of ultra broad band<br />

“supercontinuum” spectra – a phenomenon<br />

that will have a huge impact on next<br />

generation biomedical imaging systems.<br />

The supercontinuum fibre laser is<br />

based on a high power picosecond source<br />

G.I.T. Imaging & Microscopy 4/2007 • 55


L i g h t M i c r o s c o p y<br />

Fig. 1: The all-fibre MOPA (Master Oscillator Power Amplifier) – flexibility to meet<br />

the demands of a huge application space.<br />

(FP1060) and highly nonlinear photonic<br />

crystal fibre. The nonlinear interaction<br />

between the high intensity pulsed optical<br />

field within the tight confinement of a silica<br />

optical fibre waveguide, results in the<br />

generation of a continuous spectrum<br />

spanning from the visible (below 400 nm)<br />

extending in the IR to beyond 2 um.<br />

Fianium’s SC400 and SC450 supercontinuum<br />

fibre lasers, utilising high<br />

pulse repetition rates in the MHz range,<br />

deliver high spectral brightness in the<br />

range of several milli-Watts per nm<br />

across the entire optical spectrum. Filtration<br />

of several nm of this spectrum<br />

can deliver tens of mW optical power at<br />

any wavelength required – a vast improvement<br />

over the discrete wavelengths<br />

offered by conventional diode and HeNe<br />

laser sources.<br />

Furthermore, the repetition rates of<br />

the supercontinuum make them ideally<br />

suited to fluorescence imaging applications<br />

requiring either a quasi-continuous<br />

wave or a pulsed regime for time-resolved<br />

measurements.<br />

The remainder of this article focuses<br />

on examples of the use of Fianium SC400<br />

and SC450 supercontinuum fibre lasers<br />

within fluorescence imaging.<br />

Single Source Solution for Fluorescence<br />

Imaging<br />

Flow cytometers (or fluorescent activated<br />

cell sorters) are critical tools for biomedical<br />

research. These complex instruments<br />

measure the properties of individual<br />

cells, often by detecting fluorescent<br />

molecules (fluorophores) attached to<br />

their surface. The fluorescent molecules<br />

act as probes for analyzing the identity of<br />

cells for studying the immune system,<br />

identifying cancer cells, and diagnosing<br />

disease.<br />

56 • G.I.T. Imaging & Microscopy 4/2007<br />

Flow cytometers rely almost exclusively<br />

on lasers for exciting fluorophores.<br />

While the coherence and power level of<br />

lasers makes them ideal light sources for<br />

illuminating individual cells, their discrete<br />

wavelengths limits the types of fluorescent<br />

probes that can be analyzed by<br />

flow cytometry.<br />

Although solid state laser technology<br />

has increased the variety of discrete laser<br />

wavelengths available, there are still significant<br />

gaps in the excitation capabilities<br />

that put limitations of the fluorescent<br />

probes used for biomedical analysis.<br />

Confocal fluorescence microscopy has<br />

similar laser source requirements to<br />

those of flow cytometry and is one of the<br />

most powerful techniques for probing a<br />

wide range of phenomena within biological<br />

sciences.<br />

Almost all commercially available<br />

confocal microscope systems use a combination<br />

of lasers to excite fluorescence<br />

at a few discrete wavelengths within the<br />

visible region of the spectrum. As with<br />

flow cytometry, only a small number of<br />

excitation wavelengths are available by<br />

using fixed wavelength lasers. Many<br />

fluorophores are therefore unusable because<br />

of this limitation.<br />

A recent development, which further<br />

aids fluorophore excitation, is the combination<br />

of multi-channel AOTF’s (acoustooptic-tunable-filters)<br />

with the supercontinuum<br />

fibre laser. An SC400<br />

supercontinuum laser in conjunction<br />

with an 8-channel AOTF, enables the delivery<br />

of up to 8 laser lines within the visible<br />

region of the spectrum, each individually<br />

tunable across the entire spectrum<br />

and all within the same, diffraction limited<br />

collinear beam.<br />

The supercontinuum and AOTF provides<br />

the flexibility required for optimal<br />

excitation and detection of a wide range<br />

Fig. 2: The visible spectrum of the SC400 spatially dispersed<br />

through a transmission diffraction grating demonstrates the continuous<br />

nature of the spectrum from violet to near Infra-Red.<br />

of fluorophores within flow cytometers<br />

or fluorescence confocal microscopes.<br />

The laser can be tuned precisely to the<br />

excitation peak of the fluorescent probes<br />

needed for analysis.<br />

Furthermore, the supercontinuum not<br />

only improves performance but is also<br />

cost-effective. For example, with the exception<br />

of the 355 nm frequency tripled<br />

Nd:YVO4 laser, all laser sources and<br />

beam combination optics installed within<br />

the flow cytometer platform shown in<br />

fig. 3 can be replaced by a single SC400-<br />

AOTF. Furthermore, with continued advances<br />

in supercontinuum fibre laser<br />

technology, it is only a matter of time before<br />

the spectrum is further expanded<br />

down to the UV, enabling the replacement<br />

of all lasers within even the highest<br />

specification imaging systems.<br />

While commercial vendors of confocal<br />

microscopes will soon incorporate supercontinuum<br />

fibre lasers within their imaging<br />

systems, it is often research groups<br />

who are first to investigate the technology.<br />

Dr. Clemens Kaminski and his group<br />

at the Laser Analytics Group in Cambridge<br />

(UK) have recently incorporated a<br />

fibre supercontinuum laser within an Olympus<br />

Fluoview microscope scanning<br />

unit to demonstrate 2-D, 3-D and livecell<br />

imaging.<br />

Figure 4 demonstrates an example of<br />

2-D high resolution fluorescent confocal<br />

imaging using an SC450 supercontinuum<br />

fibre laser and AOTF system. In this example<br />

a Convallaria Majallis (Lily-of-the-<br />

Valley) specimen is stained with Safranin<br />

and Fast Green, having peak excitation<br />

wavelengths of 530 nm and 620 nm respectively.<br />

Since the two dyes have affinities<br />

to different regions of the sample,<br />

scanning the excitation wavelength highlights<br />

different regions of the sample.<br />

Fast Green stains the cellulosic cell walls


present over the whole sample (620 nm<br />

and 640 nm images) where the Safranin<br />

dye stains lignin – present within the endodermis<br />

and xylem, highlighted within<br />

[figure 3] at 540 nm to 560 nm excitation<br />

wavelengths.<br />

Looking Forward<br />

The need for suitable illumination<br />

sources across a wide range of biomedical<br />

applications including Flow Cytometry,<br />

Optical Coherence Tomography,<br />

CARS Microscopy and various guises of<br />

Confocal Fluorescence Microscopy continues<br />

to drive the development of laser<br />

technology.<br />

Over the past 5 years, ultrafast fibre<br />

lasers have started to challenge solidstate,<br />

diode and gas lasers across numerous<br />

markets and we expect the ongoing<br />

rapid development in performance and<br />

L i g h t M i c r o s c o p y<br />

Fig. 3: Example high-spec. flow cytometry development platform showing up to 8 discrete laser lines<br />

and associated beam combination optics – can be replaced by a single supercontinuum laser. Image<br />

courtesy of Dr. William Telford, National Institute of Health / National Cancer Institute, USA.<br />

Fig. 4: Image courtesy of the Laser Analytics Group, University of Cambridge, UK2-D Fluorescent confocal<br />

imaging of the rhizome of Convallaria Majalis (Lily-of-the-Valley) stained with Safranin and Fast<br />

Green dyes (peak excitation wavelengths of 530 nm and 620 nm respectively).<br />

reduction in laser cost to further establish<br />

this technology as the illumination<br />

source for next generation imaging systems.<br />

References:<br />

[1] Sacconi, L., et al., PNAS, V.103, No.9, 3124–<br />

3129 (2006)<br />

[2] Dombeck, D. A. et al., J Neurophysiology,<br />

3628–3636, 94 (2005)<br />

[3] Kapoor, V. et al., Nature Methods, V.4, N.9,<br />

678–679 (2007)<br />

[4] Frank, J. H. et al., Jnl of Microscopy, V.227,<br />

Issue 3, pp. 203, (2007)<br />

Contact:<br />

John Clowes, PhD<br />

R & D Manager<br />

Fianium Ltd<br />

Hamble, Southampton, UK<br />

Tel.: +44 2380 458776<br />

Fax: +44 2380 458734<br />

john.clowes@fianium.com<br />

G.I.T. Imaging & Microscopy 4/2007 • 57


Fundamental Knowledge<br />

Part 1<br />

D i g i ta l M at e r i a l s a n a ly s i s<br />

Ever since scientists started investigating our world,<br />

two aims have had the highest priority. One simply<br />

understands how everything around us is organised.<br />

The other is using this knowledge to make our<br />

lives more productive and comfortable. Much of<br />

our mental potential is developed by applying our 5<br />

senses in direct day-to-day practice. Of the 5 senses,<br />

sight is a critical tool with very high potential and<br />

bandwidth for extracting information making it a<br />

powerful interface for gathering knowledge.<br />

“I saw it with my own eyes!” – think<br />

about the significance of that expression.<br />

It provides a hint as to why optical microscopy<br />

is such a popular method for<br />

exploring the structures of materials. All<br />

other information received by instrumental<br />

sensory technologies in materials science<br />

research enters our minds via some<br />

secondary visualisation process. Though<br />

optical microscopy itself makes increasing<br />

use of integrated digital technology<br />

for interfacing image information, it is<br />

still closest to the actual process of seeing<br />

with the naked eye – i.e., receiving<br />

image information directly.<br />

How do people actually receive information<br />

about their surroundings? How<br />

does a person receive the image information<br />

offered via a microscope? The information-bearing<br />

medium is visible light<br />

and the information it carries is as a result<br />

of its various interactions with physical<br />

matter. The electromagnetic radiation<br />

of visible light is the point of<br />

departure for light microscopy in general,<br />

as well as in the metallography<br />

field. It is necessary to understand terms<br />

such as reflection, polarization and interference,<br />

alongside fundamental quantities<br />

closely linked to them such as amplitude,<br />

radiation and phase shift. This<br />

facilitates grasping the various light-microscopical<br />

contrast and imaging procedures<br />

for metallography, which involves<br />

a good deal of fundamental knowledge<br />

on the physical nature of light. This part<br />

of the chapter is from one of our “Digital<br />

Materials Analysis” series, and therefore,<br />

deals with the optical fundamentals of<br />

light and the interaction with a sample.<br />

Subsequently, the light microscopy that<br />

is commonly used in metallography is introduced<br />

and explained in part two. The<br />

58 • G.I.T. Imaging & Microscopy 4/2007<br />

Fig. 1: Schematic display of the visible spectrum of electromagnetic radiation and the resulting colour<br />

ranges.<br />

final segment deals with the contrasts<br />

that play an important role in imaging<br />

metallographic samples.<br />

Optical Fundamentals<br />

The fundamentals described below on<br />

light properties focus on the information<br />

needed to understand the material presented<br />

and are not exhaustive. For more<br />

advanced detail and more sophisticated<br />

theoretical fundamentals, we would advise<br />

interested readers to consult the extensive<br />

web pages of Florida State University,<br />

for example, on microscopy and<br />

the related physical background information<br />

(www.microscopy.fsu.edu/primer/<br />

index.html).<br />

Visible Light<br />

In everyday language, light is understood<br />

as the range of electromagnetic radiation<br />

that we can see with the naked eye. This<br />

comprises a wavelength spectrum of ca.<br />

400 nm–750 nm. The colours of light that<br />

can be seen by the eye occur due to varying<br />

wavelengths and their blending (fig.<br />

1). The adjoining spectra from the deepred<br />

and infrared range (to 1200 nm) as<br />

well as the near ultraviolet light (200–<br />

380 nm) are used in light microscopy.<br />

Even though these segments of the spectrum<br />

are not visible to our eyes, these<br />

segments can be acquired using suitable<br />

detectors, CCD cameras for example, and<br />

made available via a monitor for imaging<br />

or analysis.<br />

Wavelength, Frequency, Energy<br />

The smallest distance between two points<br />

of the same phase of a wave is referred<br />

to as wavelength λ (fig. 2). The two points<br />

are precisely in the same phase when<br />

they have the same deflection, ie, amplitude<br />

(A), and move in the same direction<br />

through time. The wavelength of light<br />

has a direct correspondence with the frequency,<br />

as does any other form of electromagnetic<br />

radiation (ν): i.e. it is inversely<br />

proportional. When entering or<br />

traversing a medium, the speed and<br />

wavelength of the light are changed. The<br />

frequency, however, remains unchanged.<br />

Alongside its wave properties, light is<br />

quantified in photons. A photon (quantum<br />

of electromagnetic energy) is considered<br />

the smallest amount of energy of<br />

any frequency of electromagnetic radiation.<br />

The energy of a photon is directly<br />

proportional to frequency and thus indirectly<br />

proportional to wavelength. This<br />

means that energy increases as frequency<br />

increases and wavelength shortens.<br />

This is why shortwave radiation is<br />

dangerous, since it has high energy in a<br />

narrow range. It can have dangerous effects<br />

on organisms, for example there is<br />

a well known correlation between UV<br />

light and skin cancer.<br />

The brightness of light perceived by<br />

an animal depends on the number of<br />

photons that reach the eye per unit time.<br />

It can be described, albeit simply, as the<br />

amount of wave deflection or amplitude<br />

(fig. 2). Other factors are the colour or<br />

wavelength and contrast (to the background)<br />

as well as its expansion. The<br />

eye‘s sensitivity to brightness is greatest<br />

with green-yellow colour tones (500–<br />

560 nm). In this case, small points of<br />

light are interpreted as brighter than<br />

larger ones of other colours, even though<br />

they have the same physical light intensity.<br />

The eye can discern 50–60 levels of


Fig. 2: Schematic diagram of waves with the<br />

wavelength (λ), of the smallest distance of two<br />

points of the same phase of a wave and of the<br />

deflection height (amplitude: A). The lower<br />

example shows how a wave of light is phase<br />

delayed when passing through an optically<br />

denser medium (eg, glass or a cell). The amplitude<br />

remains practically the same.<br />

Fig. 3: Reflection is where electromagnetic<br />

radiation is deflected and “bounced back“. This<br />

takes place either at the boundary between two<br />

media (surface reflection) or within the interior<br />

of a medium (volume reflection). Transmission is<br />

the expansion of electromagnetic radiation<br />

passing through a medium. Diffusion can take<br />

place in both phenomena. Diffusion is where a<br />

ray of light with a certain dispersive direction is<br />

deflected in many different directions (diffuse<br />

reflection or diffuse transmission). If there is no<br />

scattering upon reflection or transmission, the<br />

ray of light is deflected in a certain direction in<br />

accordance with the physical laws of optics<br />

(directed light reflection or transmission). Reflection,<br />

transmission and scattering do not alter<br />

the frequency of a wave. Absorption is the<br />

conversion of radiation into a different form of<br />

energy (usually heat) that occurs upon the interaction<br />

of radiation with matter.<br />

brightness in gray-scale images. A computer<br />

monitor on the other hand shows<br />

images with 256 gray scales (8 bits) and<br />

digital cameras, depending on the model,<br />

can acquire up to 4096 gray scales (12<br />

bits) – inasmuch these are actually<br />

present in the image. These numbers<br />

show how digital image acquisition exponentially<br />

expands what image analysis<br />

can do – especially compared with viewing<br />

a sample with the naked eye alone.<br />

Light – Object Interactions<br />

As soon as light encounters an object,<br />

three phenomena can take place: reflection<br />

(ρ), absorption (α) and transmission<br />

(τ). These processes, and their respective<br />

interrelationships, vary depending on the<br />

object’s parameters – such as material<br />

(refraction index n), surface, thickness<br />

(absorption coefficient µ) and colour. Important<br />

effects such as polarization or interference<br />

are direct results of these phenomena,<br />

which are decisive for contrast<br />

formation, imaging and material detection<br />

in the incident light microscope used<br />

for metal microscopy.<br />

Absorption and Scatter<br />

In our day-to-day lives we experience<br />

how colourless light, also frequently referred<br />

to as white light, can diminish in<br />

brightness due to absorption. This explains<br />

how much brighter a room looks<br />

after the windows have been cleaned.<br />

When a wave is absorbed by an absorbent,<br />

homogeneous material, the absorption<br />

probability (per unit of length) is the<br />

same at any penetration depth. According<br />

to the Beer-Lambert law, the amount<br />

of photons non-absorbed or scattered in<br />

the process depends on the layer-thick-<br />

D i g i ta l M at e r i a l s a n a ly s i s<br />

Fig. 4: When photons encounter a surface there are various interactions with the electrons<br />

and the material. Fluorescence may occur, which is the absorption of light of a particular<br />

wavelength (excitation light) involving various molecules and the simultaneous emission of<br />

light at longer wavelengths.<br />

ness of the material. This explains why<br />

the sky seems darker and the sun appears<br />

brighter in the mountains.<br />

But it’s not just the intensity that decreases.<br />

The light absorption of surfaces<br />

is usually dependent upon frequency and<br />

also varies in strength according to the<br />

surface colour. This kind of frequencydependent<br />

absorption alters the remaining<br />

wavelength characteristic of the light,<br />

which results in us seeing colours.<br />

Absorption is caused by scatter, i.e.<br />

the interactions of the light quanta with<br />

the free and bonded electrons of the material.<br />

Elastic and inelastic scattering are<br />

of decisive significance here. Elastic scattering<br />

is where the sum of energy of the<br />

incident photons is too small to excite the<br />

atom. The energy of the scattered photon<br />

remains unchanged but its direction<br />

changes (fig. 4). Inelastic scattering is<br />

where the photon loses a part of its energy<br />

as excitation energy of an atom or<br />

during ionization processes (fig. 4). After<br />

scatter, the scattered radiation usually<br />

has an altered diffusional direction. Inelastic<br />

scattering also means changed energy<br />

(frequency ν) and sometimes altered<br />

polarization direction.<br />

Table 1 describes how materials science<br />

visual effects are linked to both<br />

elastic and inelastic scattering.<br />

Refraction and Refraction Index<br />

Various transparent media such as air,<br />

water and glass slow down the light passing<br />

through them (phase shift) to differ-<br />

G.I.T. Imaging & Microscopy 4/2007 • 59


D i g i ta l M at e r i a l s a n a ly s i s<br />

Table 1: Overview of interaction of photons and matter<br />

Visual effect Typical materials Physical origin <strong>Images</strong><br />

Total transparency accompanied<br />

by gradual reflection<br />

Most gasses, some liquids, some<br />

mineral substances like glass (SiO 2),<br />

quartz and other crystalline structures<br />

of similar physical behaviour with<br />

smooth surfaces and homogeneous<br />

structure.<br />

Depending on their refractive index<br />

they are used for optical imaging<br />

lenses.<br />

Diffuse transparency As above, but contaminated with<br />

scattering substances or diffused<br />

interfaces.<br />

Total reflectance All metals and other materials whose<br />

physical behaviour does not bind<br />

electrons by strong individual forces to<br />

an atom or molecule.<br />

Electrons behave like a free moving<br />

gas only weakly bound to the entire<br />

material structure.<br />

(Depending on their reflectance<br />

behaviour these materials are used for<br />

producing mirror devices for optical<br />

instruments.)<br />

Remittance Non-polished, clean, metal surface<br />

without oxidation contamination.<br />

Absorption Most organic and inorganic materials<br />

show strong absorption and are<br />

termed opaque.<br />

To obtain information about their<br />

interaction with photons, these materials<br />

can be prepared as thin locally<br />

homogeneous layers so they still<br />

transmit relevant numbers of photons<br />

to describe the absorption behaviour.<br />

Fluorescence/<br />

Luminescence<br />

Complex organic molecules and semiconducting<br />

crystalline structures<br />

(quantum dots) are typically used for<br />

showing fluorescence.<br />

Polarization Clean, smooth, metal surfaces show a<br />

dependency on photon interaction<br />

(dependent on their polarization).<br />

The parameter is the angle of the<br />

photon incidence towards the surface.<br />

Most natural crystalline minerals show<br />

anisotropy in absorbing or transmitting<br />

to the photon polarization as well.<br />

(birefringent materials such as quartz<br />

or calcite are familiar examples).<br />

60 • G.I.T. Imaging & Microscopy 4/2007<br />

Elastic scattering of photons in interaction with<br />

electrons, which are bound by electromagnetic<br />

forces in an atomic /molecular system.<br />

The system structure must either be completely<br />

homogeneous or of high crystalline order.<br />

Smooth interfaces to other matter or vacuum, like<br />

polished glass to air, cause almost no irregular<br />

effect on interference.<br />

Interfaces between media with different refraction<br />

indexes result in correspondingly clear reflectivity.<br />

Colourless, transparent matter with statistically<br />

irregular interfaces of different refractive indices<br />

(changes the speed of light) cause elastic scattering<br />

in all spatial directions due to interference effects<br />

Elastic scattering of photons in interaction with<br />

electrons which form a common electrical field. The<br />

electrons are just weakly bound to an atomic or molecular<br />

structure; this explains their absorbent<br />

behaviour towards the photon, though the photon<br />

is immediately remitted (repelled). Surfaces need to<br />

be perfectly smooth.<br />

Same as in clear reflectance but with a surface<br />

design which causes diffuse reflection due to<br />

interference effects.<br />

Various effects of inelastic scattering of photons in<br />

interaction with electrons, whereby the absorbed<br />

photon energy typically dissipates inside matter as<br />

thermodynamic energy.<br />

Absorption is typically energy dependent, whereas it<br />

is often that just bands of photon energy are absorbed<br />

by individual matter.<br />

Specific inelastic scattering effects may leave photons<br />

with reduced energy (Raman scattering).<br />

Inelastic scattering of photons which leads to<br />

energy exited status of electrons in matter. Depending<br />

on the type of molecule the absorbed photon<br />

energy is released after some characteristic time<br />

period in form of lower energy photons which are<br />

remitted or absorbed in secondary processes.<br />

Portions of absorbed photon energy may dissipate<br />

to the matter as thermodynamic energy.<br />

The most tricky, as the photon force acts dynamically,<br />

changing symmetrically along an axis perpendicular<br />

to the propagation direction and kept in this<br />

state as long as the photon exists.<br />

Any anisotropy of matter in terms of polarity in<br />

interacting with electromagnetic forces will determine<br />

whether one of the interactions named above<br />

with a polarized photon will take place at all.


Fig. 5: Diagram of the refraction of light. Light that enters into an<br />

optically denser medium at an angle is refracted toward the perpendicular.<br />

When it exits, it is subject to the reverse deflection and is<br />

thus parallel shifted.<br />

ing extents, dependent on their optical<br />

density. When light from a medium (e.g.,<br />

air) enters into another medium with a<br />

higher optical density (e.g., glass) at an<br />

angle, the light is not only slowed down,<br />

but also refracted. This means that the<br />

light is deflected in an angle specific to<br />

the two media. When the light exits and<br />

encounters a medium with a lower density<br />

(e.g., air) the speed is once again specific<br />

to this medium and it is subject to a<br />

reverse deflection. The ray of light in this<br />

example is thus parallel shifted (fig. 5).<br />

There is a value that indicates the extent<br />

of refraction for transparent media<br />

– the refractive index n. This is 1 for air<br />

and increases with the optical density of<br />

the medium (e.g., water n = 1.33). Oils<br />

that are used in microscopy for high-resolution<br />

or powerful magnifying objectives<br />

(for immersion of the frontal lens)<br />

have a refraction index of ca. 1.51.<br />

Reflection<br />

If electromagnetic waves are deflected<br />

back from the material, this is termed reflection.<br />

With smooth surfaces, the light<br />

reflects according to the physical principle:<br />

entry angle equals exit angle. This<br />

means that every ray of light is reflected<br />

in precisely one direction. Boundary layers<br />

with significant roughness relative to<br />

the wavelength, reflect diffusely. If the<br />

material contains many scatter centres,<br />

the reflection follows the Beer-Lambert<br />

Law, whereby the main back scatter takes<br />

place perpendicular to the material, independent<br />

of the incident direction. Thus, if<br />

a ray of light encounters a rough surface,<br />

it is reflected in all directions spatially.<br />

Interference<br />

Various waves of light can interact with<br />

one another. These waves can also overlap<br />

each other. If the waves‘ frequencies<br />

and wavelengths are roughly the same, a<br />

new wave emerges. The new wave is amplified<br />

or weakened in comparison to the<br />

original waves – depending on the phase<br />

correspondence between the overlapping<br />

waves. This phenomenon is called interference.<br />

The waves are amplified when<br />

the phase shift corresponds to an entire<br />

wavelength. This effect is referred to as<br />

constructive interference. Wave traits<br />

are erased at a phase shift of half a wavelength<br />

(destructive interference). Any<br />

wave capable of interference is designated<br />

as a coherent wave. These are<br />

wavelengths with a constant phase ratio<br />

and equal frequency.<br />

Polarization<br />

Polarization is a property of transverse<br />

waves, and thus of the electromagnetic<br />

waves, which describe the direction of<br />

the amplitude vector. No polarization<br />

phenomenon, on the other hand, can occur<br />

with longitudinal waves, whose oscillation<br />

takes place in the direction of dispersion.<br />

A transverse wave has two<br />

directions. Firstly, the wave vector, which<br />

points in the direction of dispersion – secondly,<br />

the amplitude vector. This is always<br />

perpendicular to the wave vector in<br />

transverse waves. The third degree of<br />

freedom in three-dimensional space is<br />

rotation around the wave vector.<br />

There are three distinct kinds of polarization.<br />

They differ in direction and<br />

magnitude of the amplitude vector (at a<br />

constant point in space):<br />

linear polarization: The amplitude<br />

vector always points in a constant direction<br />

and the deflection changes its magnitude<br />

and its sign periodically (with constant<br />

amplitude) with the progression of<br />

the wave.<br />

circular polarization (also referred to<br />

as rotational polarization): The amplitude<br />

vector rotates at constant angle<br />

D i g i ta l M at e r i a l s a n a ly s i s<br />

Fig. 6: The law of reflection applies to smooth surfaces. This states that the incident<br />

ray, the perpendicular of incidence and the reflected ray are all within one<br />

incident plane and the incident angle is always exactly the same as the reflection<br />

angle. Boundaries with a greater roughness relative to the wavelength, on the<br />

other hand, reflect diffusely.<br />

speed around the wave vector and does<br />

not change its magnitude with the progression<br />

of the wave.<br />

elliptical polarization: The amplitude<br />

vector rotates around the wave vector<br />

and alters its magnitude periodically. The<br />

peak of the field vector follows an ellipse<br />

in this instance.<br />

Polarization methods facilitate the application<br />

of highly qualitative contrast<br />

methods such as Differential Interference<br />

Contrast (DIC). Polarization microscopy<br />

is of critical importance in microscopy<br />

of double refractive sample<br />

structures such as crystals.<br />

Diffraction<br />

Light that shines through a small aperture<br />

generates a pattern of bright rings<br />

referred to as a diffraction pattern. Patterns<br />

can range from a centrally bright<br />

segment (direct, unrefracted light or<br />

main maximum), followed by a number<br />

of rings with significantly less brightness<br />

(refracted light or secondary maxima).<br />

This occurs due to series of destructive<br />

and constructive interferences. According<br />

to the Abbé theory of resolution, image<br />

formation only takes place once, at<br />

least one secondary maximum interacts<br />

with the main maximum in the intermediary<br />

image plane. The more secondary<br />

maxima contributing to image formation,<br />

the higher the resolution.<br />

Contact:<br />

Esther Ahrent<br />

Department Manager Marketing Communication<br />

Microscopy and Diagnostics<br />

Olympus Life and Material Science Europa GmbH<br />

Hamburg, Germany<br />

Tel.: +49 40 23773 5426<br />

Fax: +49 40 23773 4647<br />

esther.ahrent@olympus-europa.com<br />

www.olympus-europa.com<br />

G.I.T. Imaging & Microscopy 4/2007 • 61


Photo: Pixelio<br />

I m a g e P r o c e s s I n g<br />

Air Quality Control for Hazardous<br />

Bio-Material<br />

Automatic Probe Handling, Image Acquisition and Image Analysis<br />

Airborne microorganisms are ubiquitously present in various indoor and outdoor environments. The potential<br />

implication of fungal contaminants in bio-aerosols on occupational health is recognized as a problem in<br />

several working environments. There is a concern on the exposure of workers to bio-aerosols especially in<br />

composting facilities, in agriculture, and in municipal waste treatment. The European Commission has therefore<br />

guiding rules protecting employees in the workplace from airborne biological hazards. In fact, there are<br />

an increasing number of incidents of building-related sickness, especially in offices and residential buildings.<br />

Some of these problems are attributed to biological agents, especially in relation to airborne fungal spores.<br />

However, the knowledge of health effects of indoor fungal contaminants is still limited. One of the reasons<br />

for this limitation is that appropriate methods for rapid and long-time monitoring of airborne microorganisms<br />

are not available.<br />

Fig. 1: Top view of the mechanical unit for moving<br />

object slides, indicating also the position of<br />

the cover-glass storage, the dosing pump for<br />

lactophenol, the slit impactor or air collector,<br />

and the storage for the object slides. The numbers<br />

1–5 indicate the sequences of the movements;<br />

axis No. 6 is not shown.<br />

62 • G.I.T. Imaging & Microscopy 4/2007<br />

Airborne Microorganisms<br />

Besides the detection of parameters relevant<br />

to occupational and public health,<br />

in many controlled environments the<br />

number of airborne microorganisms has<br />

to be kept below the permissible or recommended<br />

values, e.g. in clean rooms, in<br />

operating theatres, and in domains of the<br />

food and pharmaceutical industry. Consequently,<br />

the continuous monitoring of<br />

airborne biological agents is a necessity<br />

for the detection of risks of human health<br />

as well as for the flawless operation of<br />

technological processes.<br />

At present a variety of methods are<br />

used for the detection of fungal spores.<br />

The culture-based methods depend on<br />

the growth of spores on an agar plate<br />

and on the counting of colony-forming<br />

Dr. Petra Perner<br />

units. Culture-independent methods are<br />

based on the enumeration of spores<br />

under a microscope, the use of a polymerase<br />

chain reaction or on DNA hybridization<br />

for the detection of fungi. However,<br />

all these methods are limited by timeconsuming<br />

procedures of sample preparation<br />

in the laboratory.<br />

Automated Detection of Dangerous<br />

Bio-Substances<br />

We have developed an automated imageacquisition<br />

and probe handling unit of<br />

biologically dangerous substances and<br />

the automated analysis and interpretation<br />

of microscope images of these substances.<br />

In the system contaminated air<br />

containing bio-aerosols is collected in a<br />

defined volume via a carrier agent. They


are recorded by an image-acquisition<br />

unit, counted, and classified. Their nature<br />

is determined by means of an automated<br />

image-analysis and interpretation<br />

system. Air samples are automatically<br />

acquired, prepared and transferred by a<br />

multi-axis servo-system to an imageacquisition<br />

unit based on a standard<br />

optical microscope with a digital color<br />

camera. By a novel image analysis methods<br />

fungi spores are recognized in the<br />

image, described by features, and classified<br />

in one of the different fungi spore<br />

classes. The following parameters are<br />

calculated by the image analysis:<br />

�<br />

�<br />

�<br />

�<br />

�<br />

�<br />

�<br />

�<br />

�<br />

�<br />

Total number of airborne particles<br />

Classification of all particles according<br />

to the calculated image features<br />

Classification of biological particles,<br />

e.g. spores, fragments of fungal mycelia,<br />

and fragments of insects<br />

Number of respirable particles<br />

Total number of airborne particles of<br />

biological origin<br />

Number of dead particles of biological<br />

origin<br />

Number of viable and augmentable<br />

particles of biological origin<br />

Identification of species or geni<br />

Proportion of airborne abiotic and<br />

biotic particles<br />

Proportion of dead and viable airborne<br />

microorganisms.<br />

The probe handling and image acquisition<br />

unit works as follows: In the slit<br />

impactor the air (fig. 3), potentially containing<br />

airborne germs, is guided on the<br />

sticky area of the object slide by the air<br />

stream generated by an air pump. After<br />

a few tens of seconds which can be<br />

adjusted accordingly, the pump is<br />

switched off and the object slide is transported<br />

to the pipetting unit driven by the<br />

dosing pump. To this aim it has to change<br />

its transporting axis and thus its direction<br />

of movement. From a thin nozzle<br />

one drop of lactophenol is deposited on<br />

the sticky area of the object slide which<br />

is afterwards transported via the axis<br />

crossing to the cover-slip gripper unit.<br />

This gripper acts as a low-pressure<br />

sucker and takes one cover glass from<br />

the deposit and puts it with one edge first<br />

on the object slide. Then the cover glass<br />

falls down on the object slide and flattens<br />

the drop so that it will be distributed all<br />

over the sticky area forming a thin layer.<br />

In this way the airborne germs collected<br />

in the sticky layer are immersed in the<br />

lactophenol. In lactophenol living germs<br />

get a blue color. The object slide is then<br />

transported back to an axis crossingpoint<br />

where it again changes its direction<br />

of movement by 90° and is transported to<br />

Fig. 2: System architecture<br />

Fig. 3: Recognized objects in the image<br />

the xy-table of the microscope which<br />

takes over the slide and transports it<br />

directly under the lens. After the object<br />

slide has reached the image acquisition<br />

position, the microscope camera then<br />

grabs the images at the programmed<br />

slide positions after auto-focusing of the<br />

I m a g e P r o c e s s I n g<br />

Fig. 4: Screenshot of<br />

the final system<br />

microscope lens at each position. After<br />

having finished the imaging sequence,<br />

the slide is transported away from the<br />

xy-table with a special arm and falls into<br />

a box. When the image grabbing procedure<br />

by the microscope unit is still under<br />

way, the object-slide preparation unit<br />

G.I.T. Imaging & Microscopy 4/2007 • 63


I m a g e P r o c e s s I n g<br />

already starts with the preparation of a<br />

new object slide. Once an image has been<br />

taken it is given to the image-analysis<br />

unit for further processing.<br />

Objects are recognized in the microscopic<br />

image by a novel case-based<br />

object-recognition unit. This unit has a<br />

case-base of shapes for fungi spores and<br />

determines on a similarity-based inference<br />

if there are objects in the image that<br />

have a similar shape as the ones stored<br />

in the case base. In this case the objects<br />

get labeled and are transferred for further<br />

processing to the feature-extraction<br />

unit. To ensure proper performance of<br />

this unit, the general appearance of the<br />

shapes of the fungi spores have to be<br />

learned. To this end we have developed a<br />

semi-automated procedure that allows<br />

one to acquire the shape information<br />

from the raw image data and to learn<br />

groups of shape-cases and general<br />

shape-cases. The feature-extraction procedures<br />

are based on the knowledge of<br />

an expert. Note that a particular application<br />

requires special feature descriptors.<br />

Therefore not all possible feature-extraction<br />

procedures can be implemented into<br />

c o m Pa n y P r o f I l e<br />

Established in 1972, Agar Scientific has<br />

grown into an internationally respected<br />

supplier of consumables and accessories<br />

for all fields of microscopy, with agents<br />

worldwide and an enviable reputation<br />

for service and quality. The extensive<br />

experience and knowledge of<br />

our staff enables us to provide<br />

technical support and advice to<br />

microscopists from all disciplines.<br />

It is this unique understanding of<br />

the needs of our customers that<br />

has enabled us to grow our product<br />

range to over 4000 items, offering<br />

a single supplier for most<br />

needs. We offer products manufactured<br />

onsite by our skilled technicians.<br />

We also provide products from<br />

well-established suppliers such as Kodak,<br />

British Biocell International, Dumont<br />

and Fischione Instruments.<br />

High quality sample preparation is essential<br />

which is why we market a wide<br />

range of instruments, tools, accessories<br />

and consumables. In addition, we offer<br />

such a system from the beginning. Our<br />

aim was to develop a special vocabulary<br />

and the associated feature-extraction<br />

procedures for application on fungi identification.<br />

Application in Health Monitoring and<br />

Process Control<br />

Based on the feature description, the second<br />

case-based reasoning unit decides<br />

about the type of the fungi spore. This<br />

unit employs a novel prototype-based<br />

classifier. It starts its performance on<br />

prototypical cases that were selected or<br />

created by the expert. It can learn with<br />

time the different appearances of the<br />

fungi spores. The special features of this<br />

unit ensure its proper performance. It<br />

can learn the relevant prototypes from<br />

the subjectively selected set of prototypes,<br />

as well as create new prototypes.<br />

It can also learn the importance of the<br />

features of the cases. The final result of<br />

the system will be the identification of<br />

the fungi spores that appear in the image<br />

and the number of these spores. This is<br />

shown on the display of the system and<br />

<strong>Images</strong> courtesy of David McCarthy, School of<br />

Pharmacy, University of London; John Runions &<br />

Chris Hawes, Oxford Brooks University<br />

in a file, together with the date and the<br />

time when the data were acquired.<br />

The system allows automatic on-line<br />

monitoring of environments. The final<br />

information can be used to determine its<br />

contamination with biological hazardous<br />

material. It can be used for health monitoring<br />

as well as for process control.<br />

References:<br />

Sklarczyk, Ch., et al., In: P. Perner and O. Salvetti,<br />

Mass Data Analysis of Signals and <strong>Images</strong> in Med-<br />

icine, Biotechnology, and Chemistry MDA,<br />

Springer <strong>Verlag</strong> 2007.<br />

Contact:<br />

Dr. Petra Perner<br />

Director<br />

Institute of Computer Vision and Applied Computer<br />

Sciences, Leipzig, Germany<br />

Tel. +49 341 8612 273<br />

Fax: +49 341 8612 275<br />

pperner@ibai-institut.de<br />

www.ibai-institut.de<br />

www.mda-signals.de<br />

Agar Scientific for all your microscopy<br />

accessories and consumable needs<br />

64 • G.I.T. Imaging & Microscopy 4/2007<br />

the Agar branded range of vacuum coating<br />

systems and add-ons including targets,<br />

carbon rods and film thickness<br />

monitors. This ensures that users have<br />

a single source for all their sample<br />

preparation needs.<br />

A core part of our business is the<br />

manufacture and supply of replacement<br />

filaments and specialist<br />

apertures for the majority of electron<br />

microscopes. We also produce<br />

grids, support films and calibration<br />

standards. Furthermore,<br />

we have the capability to provide<br />

customised solutions.<br />

Full details of our extensive range<br />

may be found on our website. Register<br />

for a free copy of our catalogue.<br />

Contact:<br />

Agar Scientific Limited<br />

Stansted, Essex, UK<br />

Tel: +441279 813519<br />

sales@agarscientific.com<br />

www.agarscientific.com


4th generation XFlash ® silicon drift detectors (10, 30 and 40 mm2 �<br />

)<br />

� LN2 free, vibration free<br />

� Unmatched acquisition speed<br />

� Excellent results at both low and high count rates<br />

� Precise light element analysis (FWHM C-Kα = 48 eV)<br />

� New HyperMapping function (high speed PTS)<br />

� Modular, customizable ESPRIT software<br />

Technical Specifications<br />

sales-ma@bruker-axs.de<br />

www.bruker-axs-microanalysis.com<br />

� Long lifetime filament (guaranteed 1000 hrs) running at 5kV<br />

� Specially designed Backscattered Electron Detector (BSD) allowing compositional<br />

and topographical imaging<br />

� High depth of focus with an maximum image resolution of 30nm<br />

� Fast time-to-image; optical image < 10 seconds and electron image<br />

< 30 seconds<br />

� Robust design; no risk of damaging the lens due to sample-container design<br />

www.fei.com/phenom<br />

infophenom@fei.com<br />

I & M S H o w c a S e<br />

QUANTAX – fast, reliable and<br />

convenient microanalysis<br />

Bruker AXS Microanalysis’ QUANTAX EDS system delivers fast<br />

reliable analyses across a broad range of applications for all<br />

kinds of samples ranging from powders and metals to coatings.<br />

Its LN 2-free XFlash ® Silicon Drift Detectors (SDD) are vibration<br />

and maintenance free, while delivering unbeatable energy resolution<br />

at high count rates (125 eV Mn Kα 100,000 cps).<br />

The SDD technology makes real time spectrometry for instant<br />

element preview, high speed element mapping or Colorscan<br />

possible. Just one click and Colorscan turns the black and white<br />

SEM image into a full color image. While the SEM is scanning<br />

the image is superimposed in color with the element information<br />

gathered by the detector. In this way the user can assess<br />

interesting areas and the homogeneity of a sample at a glance.<br />

Especially noteworthy is the QUANTAX HyperMap function. It<br />

stores a complete spectrum at every pixel of an element map.<br />

This “database” can be reanalyzed anytime later for additional<br />

elements or features of interest. Only the XFlash ® detector‘s<br />

high count rate detection capability (>750,000 cps for the<br />

XFlash ® 4010 and >3,000,000 cps for the XFlash ® QUAD<br />

4040) allows for efficient and fast HyperMapping.<br />

PHENOM; Closing the Imaging<br />

Gap between Optical and Electron<br />

Microscopy<br />

The Phenom is a new tabletop scanning electron microscope (SEM)<br />

which combines the high magnification of electron microscopy with the<br />

ease of use of optical microscopy to improve performance in a benchtop<br />

instrument.The Phenom, a tabletop or benchtop SEM provides useful<br />

magnifications up to 20,000 x and is easy to use as the typical laboratory-grade<br />

optical microscopes. Operating an optical microscope<br />

requires little more than placing the sample on the stage and focusing<br />

the image and is usually accomplished in a matter of seconds. In conventional<br />

SEMs, the time required to get an image can easily become<br />

many minutes to hours. The Phenom cuts away the time, difficulty, and<br />

expense of the conventional SEM. The operator simply places the sample<br />

in the specially designed holder on the microscope. The automatically<br />

focused image is displayed in less than 30 seconds later, with the<br />

resolution and depth of focus typical belonging to SEMs. Some of the<br />

key features of the Phenom are;<br />

� Imaging power: Up to 20,000 x magnification with superior depth<br />

of focus and superb picture quality<br />

� Ease of use: Intuitive control system and interactive touch screen<br />

reduces operator training and increases the number of users<br />

� Fastest Time to Image: Through patented vacuum technology<br />

and never lost navigation<br />

� Low cost of ownership: Easy to install and maintain and no<br />

special operator training needed<br />

G.I.T. Imaging & Microscopy 4/2007 • 65


I & M S H o w c a S e<br />

Features:<br />

Completely integrated system comprising upright optical microscope and JPK NanoWizard ® �<br />

II AFM system<br />

� Developed for the investigation of opaque samples in life and materials sciences<br />

� 100 % performance of both techniques, without any limitations<br />

� Unique capability to investigate the ROI precisely with optics and AFM<br />

� Outstanding reproducibility of the focussed position (ROI) with both systems<br />

� Also perfect during operation in liquids<br />

� Wide range of applications: Biochips, cell chips or patterned substrates for cell adhesion, nanostructured<br />

surfaces from PDMS or other imprints, lipid bilayers, biomarkers such as quantum dots, rods, CNT ...<br />

� Flexible concept with shuttle stage<br />

� Both techniques can be operated even in different laboratory rooms<br />

Technical Specifications<br />

� Lateral resolution 90 nm (full width half maximum)<br />

� Easy access to STED resolution by a mouse click<br />

� STED fully integrated into TCS SP5 platform<br />

� All Leica TCS SP5 systems upgradable to STED<br />

� Full functionality of Leica TCS SP5 Multiphoton included<br />

66 • G.I.T. Imaging & Microscopy 4/2007<br />

christine.ludwig@leica-microsystems.com<br />

www.confocal-microscopy.com<br />

BioMAT Workstation<br />

– Uncompromising AFM and<br />

Optical Imaging of Opaque<br />

Samples<br />

JPK Instruments introduces a new technical breakthrough that<br />

enables the combination of upright microscopy with atomic<br />

force microscopy (AFM) on the same sample spot – the BioMAT<br />

Workstation. This clears the way for a new range of applications<br />

where the full capabilities of both AFM and advanced optical<br />

imaging can be realized even on opaque samples. A patented<br />

calibration procedure ensures that the region of interest can be<br />

found and re-imaged with micron precision, so exactly the same<br />

area can be investigated optically and with the AFM.<br />

Beyond the Limits!<br />

nanowizard@jpk.com<br />

www.jpk.com<br />

Superresolution Microscope Leica TCS STED<br />

The new Leica TCS STED opens up the access to resolve the finest<br />

structures beyond the diffraction limit – without any image<br />

computation!<br />

With the integration of the groundbreaking STED concept into<br />

the approved broadband confocal platform Leica TCS SP5 a new<br />

class of microscope has been created. The superresolution<br />

capacity of the Leica TCS STED allows confocal imaging with a<br />

resolution 2 to 3 times higher than could ever be achieved in a<br />

conventional scanning microscope – without compromising on<br />

usability. This means: superresolution at a mouse click!<br />

The Leica TCS STED breaks the diffraction limited resolution of a<br />

light microscope by downsizing the diameter of the fluorescence<br />

spot, that determines the resolution, by a doughnut shaped<br />

pulsed depletion laser beam, tightly synchronized with the excitation<br />

light pulse.<br />

With this technique it is possible to resolve protein complexes<br />

smaller than 90 nm and to gain new information for instance<br />

about the construction of synapses and membrane domains.


‘Cellogy’, a term coined by Nikon, describes the company’s strategic<br />

move into Complete Cell Care:<br />

� Maintaining live cells in an optimum environment, aiding researchers to obtain a true<br />

picture of what is happening in vivo<br />

� Developing optical hardware and analysis software to help researchers accurately track<br />

and minimise stress imposed on cells, reduce phototoxicity and extend cell life<br />

� Focusing R&D activity on ways of integrating imaging solutions with other advanced<br />

technologies to simplify experimental protocols and enhance results<br />

info@nikoninstruments.eu<br />

www.nikoninstruments.eu<br />

Moving further into<br />

‘complete live cell care’<br />

BioStation CT represents next stage in Nikon’s<br />

product development plans<br />

In a major extension of its traditional involvement in cellular<br />

imaging, Nikon Instruments is expanding further into the field of<br />

live cell care with the launch of the new BioStation CT. By combining<br />

the precise environmental control capabilities of a highperformance<br />

incubator with the advanced optics needed for<br />

drift-free live-cell imaging, the BioStation CT creates conditions<br />

that are as close to in vivo as possible.<br />

Nikon’s commitment to Live Cell Care was originally founded on<br />

the TE2000-PFS system for time lapse recording, the C1si microscope<br />

for spectral confocal applications, and the BioStation IM,<br />

a bench-top integrated incubation and monitoring system, but<br />

has recently been enhanced by the introduction of Controlled<br />

Light Exposure Microscopy (CLEM) and the LiveScan Swept Field<br />

Confocal (SFC) Microscope. CLEM reduces photobleaching and<br />

enhances cell survival thus allowing time-lapse studies of protein<br />

interactions to be undertaken. In addition to reduced crosstalk<br />

and noise, the fast scan speed of the LiveScan SFC microscope<br />

dramatically minimises phototoxicity and photobleaching<br />

during image acquisition and is ideal for live samples.<br />

Olympus FV1000: Taking cLSM<br />

to another level<br />

Flexible confocal microscopy<br />

Confocal laser scanning microscopy provided scientists with the<br />

tools to see a much greater level of detail in cells. The Olympus<br />

FV1000 has taken this to an even higher level, with the added<br />

functionality and flexibility of the multi-laser combiner and SIM<br />

scanner.<br />

� Multiple lasers: The multi-laser combiner enables the easy integration and more flexible use of an unmatched number of lasers, providing a wider range of<br />

available wavelengths for high-resolution, confocal live cell imaging. The system supports up to six diode and gas lasers, enabling the selection of a large<br />

number of wavelengths from near UV to far red. Presently, laser diodes provide 405, 440, 473, 559 and 635 nm and the gas lasers provide 457, 488,<br />

514.5, 543 and 594 nm.<br />

� SIM scanner: The FV1000 incorporates 2 laser scanners in a single compact design for simultaneous confocal fluorescence observation and independent<br />

laser light stimulation. Synchronization of these two functions ensures that rapid cellular reactions that occur during or immediately following stimulation<br />

are not overlooked.<br />

� Regions of interest: Any region of interest can be specified for stimulation and scanning independently, with unrestricted control of variations in timing,<br />

duration and intensity. The circular “Tornado” scan provides highly efficient photobleaching and photoactivation in contrast to standard raster-scan patterns.<br />

� Application flexibility: The SIM scanner and multi-laser combiner make the FV1000 the most suitable confocal microscope for a variety of applications,<br />

including FRAP, FLIP, photoactivation, photoconversion, uncaging, laser ablation and many others.<br />

microscopy@olympus-europa.com<br />

www.microscopy.olympus.eu<br />

I & M S H o w c a S e<br />

G.I.T. Imaging & Microscopy 4/2007 • 67


N o t e s f r o m N i k o N<br />

Controlled Light Exposure Microscopy<br />

Minimising Phototoxicity and Photobleaching in Live Cell Fluorescence Imaging<br />

The labelling of cellular targets with fluorescent probes is used widely in live cell imaging to identify and<br />

monitor intracellular events. Excitation of probes with light, however, can result in phototoxicity and photobleaching<br />

– factors that can seriously compromise fluorescence time­lapse recordings especially in confocal<br />

microscopy. Laser­induced phototoxicity and photobleaching can be reduced with an innovative technology<br />

known as ‘Controlled Light Exposure Microscopy (CLEM)’. Here, photobleaching and cell survival are compared<br />

when imaged with a Nikon C1 confocal system configured with, and without, CLEM.<br />

Fluorescence Imaging in Living Cells<br />

The ability to observe dynamic events in<br />

living cells is one the greatest challenges<br />

in biological research. Fluorescent<br />

probes, such as green fluorescent protein<br />

(GFP) and its derivatives, help researchers<br />

view specific cell targets and<br />

track protein localisation and distribution<br />

in living cells. The variety of probes<br />

now available (Miyawaki, 2004) allows<br />

researchers to study interactions between<br />

proteins tagged with different fluorescent<br />

proteins over time. However,<br />

problems associated with light-induced<br />

probe bleaching and phototoxicity can<br />

seriously limit time-lapse experiments<br />

especially when laser light is used. Controlled<br />

Light Exposure Microscopy<br />

(CLEM) regulates laser illumination so<br />

that photobleaching is reduced and cell<br />

survival increased (Hoebe et al., 2007).<br />

Imaging System<br />

In a conventional laser scanning confocal<br />

microscope system every pixel is illumi-<br />

Author’s<br />

background<br />

Maarten Balzar is product manager for Nikon<br />

Instruments Europe BV. He is responsible for<br />

biological research applications such as TIRF,<br />

confocal and advanced fluorescence microscopy.<br />

His current activities are focussed on the<br />

bioscience product line. Maarten joined Nikon<br />

in 1999 after finishing his PhD at the Department<br />

of Pathology, Leiden University Medical<br />

Centre (LUMC), The Netherlands.<br />

68 • G.I.T. Imaging & Microscopy 4/2007<br />

nated individually by laser light (typically<br />

in the micro-second range) and fluorescence<br />

emission detected simultaneously.<br />

A confocal image consists of many of<br />

these individual pixels and typically displays<br />

high (bright) and low (dim) intensity<br />

pixels. Generally, each pixel is acquired<br />

with the same laser power and<br />

detector sensitivity setting. In contrast,<br />

when CLEM is used, exposure to laser<br />

light is determined on a per pixel basis<br />

(i.e. when and where required). Excitation<br />

light is reduced using two strategies:<br />

1. The first is based on the principle<br />

that if there is no signal, then no illumination<br />

is required (for example, the background).<br />

2. The second detects whether there is<br />

sufficient signal to acquire an image. If<br />

so, illumination can be stopped.<br />

A schematic of the CLEM principle is<br />

shown in figure 1A. In “non-CLEM microscopy”<br />

an object is illuminated uniformly<br />

and, consequently, the detected<br />

image is a direct representation of the<br />

object. In “CLEM” microscopy the nonuniform<br />

illumination is controlled by the<br />

detection signal via a feedback system.<br />

The final image is created by combining<br />

the illumination image and the detection<br />

image. Figure 1B shows CLEM configured<br />

with the Nikon C1 confocal system.<br />

CLEM electronics use the pixelclock and<br />

the detector signal as inputs. An acousto<br />

optical modulator (AOM) is used as a fast<br />

shutter. Modulation of a solid-state (diode)<br />

laser may also be used.<br />

The Investigations<br />

Three investigations were carried out:<br />

1. CLEM was used firstly to image fluorescence<br />

in pollen grains to ensure that<br />

there were no differences in cell mor-<br />

phology when imaged with and without<br />

CLEM.<br />

2. BY-2 tobacco cells expressing GFP-<br />

MAP4 associated with microtubules were<br />

used in a time-lapse experiment to determine<br />

the effect of CLEM on photobleaching.<br />

Identical settings were used for the<br />

time-lapse experiment (a 3D acquisition<br />

in time) in the presence and absence of<br />

CLEM.<br />

3. To assess the impact of CLEM on<br />

cell survival, HeLa cells expressing GFPtagged<br />

histone-2B were plated on a Petridish<br />

and multiple areas on the dish imaged<br />

in a multi-dimensional time-lapse<br />

experiment in the presence or absence of<br />

CLEM. Both transmitted light and (GFP)<br />

fluorescence images were acquired to<br />

obtain optimal morphological information<br />

on the cells.<br />

Results<br />

The morphology of the pollen grain is<br />

similar with both non-CLEM and CLEM<br />

imaging (fig. 2). However, exposure of<br />

the sample to light in the absence of<br />

CLEM is much higher than in the presence<br />

of CLEM.<br />

In the absence of CLEM, the microtubule-associated<br />

GFP-MAP4 expressed by<br />

BY-2 tobacco cells is bleached to 50 % of<br />

its original fluorescence intensity (arbitrary<br />

units) after just five 3D scans (fig.<br />

3A). In contrast, in the presence of CLEM,<br />

the GFP-MAP4 fluorescence intensity is<br />

reduced by 50 % after twenty five 3D<br />

scans (fig. 3B).<br />

Time-lapse acquisition of HeLa cells<br />

expressing GFP tagged histone-2B in the<br />

presence or absence of CLEM shows that<br />

the intensity levels of the GFP tagged histone-2B<br />

remain approximately equal in<br />

both cases (fig. 4). However, in the absence<br />

of CLEM, the HeLa cells reveal<br />

membrane blebbing at an early stage followed<br />

by cell death (A: top panel). Cell<br />

survival is greatly prolonged in the presence<br />

of CLEM (B: bottom panel).<br />

It has been shown that CLEM reduces<br />

photobleaching and phototoxicity two- to<br />

tenfold, depending on the fluorophore<br />

distribution in the sample (Hoebe et al.,


Fig. 1: A) Comparison of non-<br />

CLEM and CLEM microscopy.<br />

B) Schematic overview of the<br />

C1 system equipped with the<br />

CLEM module.<br />

Fig. 3: (A) Time-lapse acquisition (XYZ in time) of BY-2 tobacco cells expressing<br />

GFP-MAP4 associated with microtubules. The XY image at the middle<br />

of the image stack is presented in time. The sequential images were acquired<br />

in the absence of CLEM. (B) Time-lapse acquisition (XYZ in time) of<br />

BY-2 tobacco cells expressing GFP-MAP4 associated with microtubules. The<br />

XY image at the middle of the image stack is presented in time. The sequential<br />

images were acquired in the presence of CLEM.<br />

2007). By spatially controlling the lightexposure<br />

time, CLEM reduces the excitation-light<br />

dose without compromising image<br />

quality. CLEM directly affects the<br />

pixel-by-pixel laser excitation light dose.<br />

While CLEM was created for qualitative<br />

live cell imaging, it may also have an impact<br />

on quantitative studies. The observation<br />

that CLEM reduces photobleaching<br />

and phototoxicity means that results<br />

are less likely to be affected by artefacts<br />

related to the processes of cell death.<br />

Since the illumination image, detection<br />

image and CLEM image are all available;<br />

it should be possible to completely correct<br />

the CLEM image for accurate quantitative<br />

imaging. As bright pixels are only<br />

illuminated for a limited time, it should<br />

also be possible to extrapolate the detected<br />

fluorescence emission intensity.<br />

This may contribute to greatly increased<br />

dynamic range and provides an additional<br />

advantage in addition to reduced<br />

photobleaching and improved cell viability.<br />

Conclusions<br />

Cells expressing fluorescent protein are<br />

sensitive to photobleaching and phototoxicity.<br />

In live cell imaging, it is important<br />

to minimise these effects to prolong<br />

cell survival especially for time-lapse<br />

studies. The use of CLEM in a confocal<br />

Fig. 2: Confocal image of pollen grain autofluorescence<br />

in the absence / presence of CLEM.<br />

Top panel shows the illumination image (left)<br />

and confocal image (right) in the absence of<br />

CLEM. Bottom panel shows the illumination<br />

image (left), detected image (middle) and CLEM<br />

image (right).<br />

microscope system can reduce the effects<br />

of both photobleaching and phototoxicity.<br />

CLEM is, therefore, an essential tool for<br />

live cell imaging studies.<br />

Fig. 4: Time-lapse acquisition of HeLa cells expressing GFP tagged histone-<br />

2B. The transmitted light and fluorescence images were simultaneously<br />

acquired in the absence (A) or presence (B) of CLEM.<br />

References:<br />

[1] Hoebe, R. A., Van Oven, C. H., Gadella, T. W. Jr,<br />

Dhonukshe, P. B., Van Noorden, C. J., Manders,<br />

E. M., Nat. Biotechnol. 25(2) 249–53 (2007).<br />

[2] Miyawaki, A., Nat. Biotechnol. 22, 1374–1376<br />

(2004).<br />

N o t e s f r o m N i k o N<br />

Contact:<br />

Maarten Balzar (PhD)<br />

Application manager biological microscopes<br />

Nikon Instruments Europe BV<br />

Badhoevedorp, The Netherlands<br />

Tel.: +31 2044 96273, Fax: +31 2044 96298<br />

balzar@nikonbv.nl<br />

G.I.T. Imaging & Microscopy 4/2007 • 69


P r o d u c t s<br />

Creating Own Filtersets<br />

Optical filters made by the<br />

hardcoating technique from<br />

AHF Analysentechnik show<br />

extrem high transmission,<br />

high thermal stability and are<br />

easy to handle. The company<br />

Nikon Instruments Europe<br />

and Cool-LED have agreed<br />

that Nikon will offer the “Precis<br />

Excite” LED light-source<br />

with their fluorescence microscopes<br />

in Europe. Following<br />

product evaluation at<br />

their European headquarters<br />

in Amsterdam, Nikon identified<br />

the benefits and potential<br />

of the light-source to provide<br />

stable, homogeneous control<br />

of fluorescence excitation.<br />

Applications such as Confocal,<br />

time-lapse and live cell<br />

imaging all benefit from the<br />

high level of control. The<br />

modular light source enables<br />

the user to select the wavelength<br />

of excitation and control<br />

the intensity and period<br />

presents a wide program also<br />

of single filters and beamsplitters<br />

which can be combined to<br />

individual filtersets, avoiding<br />

high costs for custom specification.<br />

The company offers to<br />

handle the detailed requests<br />

of difficult optical setups.<br />

Wherever possible, Demofilters<br />

will be offered for testing<br />

at the customers’ samples.<br />

AHF Analysentechnik AG<br />

Tel.: +49 7071 970901-0<br />

info@ahf.de<br />

www.ahf.de<br />

LED Light-source Used for Microscopes<br />

Catalog Released<br />

Omega Optical has released a<br />

new catalog of fluorescence<br />

filter sets. The 2007 catalog,<br />

Precision Optical Filters for<br />

Fluorescence Microscopy, includes<br />

fifteen high performance<br />

sets for fluorescent proteins,<br />

and ten new sets for<br />

Fret applications. These products<br />

complement an extensive<br />

selection of dye-specific filter<br />

sets for all single and multilabel<br />

microscopy applications,<br />

including Quantum Dots, M-<br />

FISH, Pinkel, Sedat, Ratio Imaging,<br />

Confocal, and Multiphoton.<br />

In addition, there are<br />

resources such as a fluorophore<br />

reference table, light<br />

source and detector spectral<br />

70 • G.I.T. Imaging & Microscopy 4/2007<br />

of excitation very precisely.<br />

As new wavelengths are required,<br />

and intensity of LEDs<br />

continues, additional LED Array<br />

Modules can be added<br />

and interchanged in the unit.<br />

CoolLED<br />

Tel.: +44 1264 320989<br />

sales@coolled.com<br />

www.precisexcite.com<br />

data, an explanation of filter<br />

nomenclature, guidelines for<br />

choosing the optimal filter<br />

sets, and a list of components<br />

organized by wavelength.<br />

Omega Optical, Inc.<br />

Tel.: +1 802251 7342<br />

rjiraskova@omegafilters.com<br />

www.omegafilters.com<br />

Optical Filters<br />

Semrock has announced new<br />

filters for the efficient isolation<br />

of individual lines of highpower<br />

mercury arc lamps.<br />

Each filter obtains maximum<br />

throughput of the desired<br />

mercury line through passband<br />

transmission and optimization<br />

of the filter design.<br />

The non-absorbing blocking<br />

and fused silica substrate ensures<br />

that undesired mercury<br />

The Fusion Microslides from<br />

BTX Harvard Apparatus are<br />

designed to fit on a microscope<br />

and allow observation<br />

of the dimer formation during<br />

electrofusion. They are composed<br />

of a glass slide and two<br />

strips of stainless steel (wire<br />

or bar) and are available in<br />

0.5 mm, 1.0 mm, 3.2 mm and<br />

10 mm gap sizes. The slides<br />

work with the company’s ECM<br />

2001 Electrofusion device.<br />

lines and other background<br />

light are effectively eliminated<br />

without the risk of filter<br />

solarization. The new Max-<br />

Lamp family has been<br />

launched with mercury arc<br />

lamp filters to isolate the popular<br />

365 nm i-line and the<br />

critical 254 nm UV line. These<br />

bandpass filters offer an average<br />

transmission of > 93 %<br />

and > 65 % over the 365 nm<br />

and 254 nm bands respectively,<br />

yet with higher and<br />

wider out-of-band blocking<br />

than incumbent filters.<br />

Semrock, Inc.<br />

Tel.: +1 585 594 7003<br />

amacdonald@semrock.com<br />

www.semrock.com<br />

Microslides for Hybridoma Production<br />

Machine Vision Illuminator<br />

Dolan-Jenner Industries has<br />

introduced its Fiber-Lite<br />

DC950H Machine Vision Fiber<br />

Optic Illuminator for machine<br />

vision integrators. This is a<br />

150-watt quartz halogen regulated<br />

illuminator, which has<br />

been improved to produce a<br />

higher output with greater<br />

reliability, and is now RoHS<br />

compliant. It features DC regulated<br />

output, fast lamp response,<br />

and a 0-5 VDC remote<br />

intensity control interface<br />

with linear voltage adjustment<br />

(8 bit D/A module available).<br />

The instrument can be<br />

operated remotely and offers<br />

remote notification of lamp<br />

failure to decrease downtime.<br />

It is made of stackable heavy<br />

duty steel housing with a<br />

BTX Harvard Apparatus<br />

Tel.: +1 800 272 2775<br />

Techsupport@btxonline.com<br />

www.btxonline.com<br />

mounting capability. The front<br />

panel can be accessed easily<br />

for fast lamp changes. Several<br />

illuminating options are<br />

available, including color filtering,<br />

manual iris, and an<br />

analog or digital remote interface.<br />

Dolan-Jenner<br />

Tel.: +1 800 626 8324<br />

bgagnon@donal-jenner.com<br />

www.dolan-jenner.com


Confocal Raman Imaging<br />

Option<br />

Witec introduces the „Ultrafast Raman<br />

Imaging Option“ for the Alpha300 R Confocal<br />

Raman Microscope. With this option<br />

the acquisition time for a single Raman<br />

spectrum can be as low as 1.7 milliseconds.<br />

As a Confocal Raman image typically<br />

consists of tens of thousands of spectra,<br />

the option reduces the total acquisition<br />

time for a complete image to only a few<br />

minutes. For example, a complete hyperspectral<br />

image consisting of 250 x 250<br />

pixels = 62,500 Raman spectra can be recorded<br />

in less than two minutes. The latest<br />

spectroscopic EMCCD detector technology<br />

combined with the high throughput<br />

optics featured system are the keys to this<br />

improvement. The new option reduces<br />

the overall experiment duration and delivers<br />

more valuable Raman data in a<br />

given time, thereby reducing the total cost<br />

of ownership of the system.<br />

Witec GmbH<br />

Tel.: +49 731 140 700<br />

info@witec.de<br />

www.witec.de<br />

Photo-Bleaching and Photo-<br />

Activation<br />

Andor Technology launches a new tool<br />

for its Revolution laser microscopy solutions<br />

aimed at live cell imaging. Revolution<br />

Frappa is the company’s latest innovation<br />

in laser microscopy using a<br />

computer-steered laser beam to photobleach<br />

or photo-active a user-defined region<br />

in a live cell specimen. It is a photobleaching<br />

module using a dual<br />

galvanometer scan head. It can be configured<br />

in line with a CSU and/or camera.<br />

Under Andor iQ software control, the<br />

user commands the instrument to bleach<br />

or activate regions of interest with userdefined<br />

times, laser lines and powers.<br />

Laser switching is tightly synchronized<br />

using the company’s proprietary laser<br />

combiner multi-port switch (MPS).<br />

Andor Technology<br />

Tel.: +44 289023 7126<br />

www.andor.com<br />

Continuous Wave Laser<br />

Newport Corporation’s Spectra-Physics<br />

Laser Division introduced the two newest<br />

members of its Excelsior low power, continuous<br />

wave (CW) laser family: The Excelsior-1064<br />

OEM-laser with 500 mW of<br />

output power, and the Excelsior-1064<br />

P r o d u c t s<br />

Scientific laser with both 500 and 800<br />

mW of output power. Using Vanadate as<br />

the gain medium, these two lasers operate<br />

at the 1064 nm wavelength. The company<br />

has showcased the latest technology<br />

advancements in its 488 nm Excelsior<br />

and Cyan laser product lines during the<br />

“Laser” show in Munich. These demonstrations<br />

included a 100 mW Excelsior-<br />

488, a 75 mW Cyan laser, and a 20 mW<br />

Cyan power adjustable laser that customers<br />

can test on the show floor.<br />

Newport Spectra-Physics GmbH<br />

Tel.: +49 6151 708 0<br />

germany@newport.com<br />

www.newport.com<br />

Image Asset Management<br />

Solution<br />

Media Cybernetics announced the release<br />

of IQbase 2.5 scientific image management<br />

software. This image management<br />

software enables organizations to<br />

effectively store, query, and share large<br />

numbers of images and related data. Users<br />

can collaborate within their organization<br />

and with outside partners by sharing<br />

image data through their network or<br />

via the Internet. With the software, users<br />

can perform powerful image searches<br />

based on keywords or using more detailed<br />

context sensitive search parameters.<br />

<strong>Images</strong> and data can be easily<br />

shared with others through the use of<br />

automatic PDF report templates, onestep<br />

export to PowerPoint tools, and webbased<br />

image searching and downloading.<br />

The software allows users to further<br />

explore their images with qualitative visualization<br />

tools like image overlays and<br />

annotations, as well as with quantitative<br />

graphs and charts for interactive data<br />

analysis.<br />

Media Cybernetics, Inc.<br />

Tel.: +1 301 495 3305 260<br />

www.mediacy.com<br />

� � � � � � � ����� � � � � � � � �<br />

����������<br />

�������<br />

Interference 3D microscope<br />

and vibrometer<br />

�<br />

�<br />

�<br />

�<br />

�<br />

�<br />

Fast and accurate optical 3D profiling<br />

Phase shifting (PSI) and white light<br />

vertical scanning (VSI) interferometry<br />

Sub-nanometer rougness analysis<br />

MEMS option for oscillating surfaces<br />

up to 3 MHz, also Laser Doppler<br />

Vibrometer module<br />

Optional vacuum/pressure chamber<br />

Top quality at affordable price<br />

We also supply<br />

Confocal and other 3D microscopes, AFM’s<br />

for research, industry and teaching, Nano-/<br />

Micro- Indenter and Tribology Tester,<br />

Nanoparticle size measurement, LEED/AES<br />

Contact your nearest office<br />

Germany · info@schaefer-tec.com · +496103300980<br />

France · info@schaefer-tech.com · +33164496350<br />

Italy · italia@schaefer-tec.com · +39 0425460218<br />

Switzerland · ch@schaefer-tec.com · +41344237070<br />

www.schaefer-tec.com<br />

G.I.T. Imaging & Microscopy 4/2007 • 71


P r o d u c t s<br />

Intuitive Smart Camera/Software Package<br />

– Without Programming<br />

The Matrox Iris-E with Design<br />

Assistant is a powerful<br />

smart camera/software package<br />

and allows OEMs and systems<br />

integrators to develop<br />

vision and imaging applications<br />

without programming.<br />

In the Matrox Design Assistant<br />

environment, users create<br />

a flow chart of the application<br />

that instructs the<br />

Matrox Iris E-Series camera<br />

to grab, process and display,<br />

perform measurements, analyze<br />

image data, read machine<br />

codes, and more. The<br />

intuitive nature of the Matrox<br />

Iris E-Series with Design Assistant<br />

eliminates the need<br />

for programming and scripting.<br />

Users benefit from simplified<br />

application development<br />

that offers the same<br />

reliability and robustness as<br />

the field-proven Matrox Imaging<br />

Library. The Matrox<br />

Digital Pathology Solution<br />

Image Solutions has launched<br />

“Tissue-Mine”, a user friendly<br />

web based digital pathology<br />

solution. It has been designed<br />

by pathologists to automate<br />

the pathology workflow for<br />

research and development.<br />

Improving efficiency, accelerating<br />

the workflow and minimising<br />

bottlenecks in an otherwise<br />

manual process. It is<br />

now possible to reduce analysis<br />

time from weeks to minutes<br />

without the process being<br />

subject to user variability.<br />

The system provides a supe-<br />

TEM Camera at M&M<br />

At this year’s Fort Lauderdale<br />

M&M show, Olympus Soft Imaging<br />

Solutions is launching<br />

the Cantega G2, the 2 nd generation<br />

Olympus Soft Imaging<br />

Solutions 2k x 2k, bottommounted<br />

CCD TEM camera<br />

for all TEM brands. The fiberoptically<br />

coupled and watercooled<br />

camera is equipped<br />

with an optimized YAG scin-<br />

72 • G.I.T. Imaging & Microscopy 4/2007<br />

Iris-E is a smart camera that<br />

support the flexibility of PCbased<br />

machine vision systems,<br />

and features hardware<br />

for image sensing as well as<br />

software for image capture,<br />

processing, and analysis. It is<br />

powered by an Ultra Low<br />

Power (ULP) Celeron, an embedded<br />

Intel architecture<br />

processor, and runs the Windows<br />

CE .NET real-time operating<br />

system.<br />

Rauscher<br />

Tel.: +49 8142 44841 0<br />

info@rauscher.de<br />

www.rauscher.de<br />

rior, highly customisable alternative<br />

to the traditional<br />

manual analysis of tissue<br />

slides using a microscope. I<br />

scan, which is included in the<br />

package, is a new high speed<br />

tissue slide scanner which<br />

can acquire images with great<br />

speed and image quality capable<br />

of auto loading up to<br />

160 slides.<br />

Image Solutions (UK) Ltd.<br />

Tel.: +44 1772663 140<br />

andy@imsol.co.uk<br />

www.imsol.co.uk<br />

Board Level Camera<br />

Hamamatsu Photonics introduce<br />

the C10000 TDI (time<br />

delay integration) board level<br />

CCD camera featuring the latest<br />

high speed, high sensitivity,<br />

back thinned CCD image<br />

sensor. The newly developed<br />

CCD sensor features 2048<br />

pixel length by 128 pixel<br />

height, with 90 % peak quantum<br />

efficiency, high UV sensitivity<br />

and a very high speed<br />

data readout of up to 50 KHz<br />

line rates. Ordinarily high<br />

speed imaging systems suffer<br />

from a lack of available light<br />

and special illumination systems<br />

need to be constructed.<br />

However, using the TDI image<br />

acquisition method the<br />

C10000 board camera synchronises<br />

charge transfer<br />

with the moving object. This<br />

Excellent Test Results<br />

In a test commissioned by IDS<br />

and conducted according to<br />

the EMVA 1288 Standard, the<br />

two cameras UI-1220-M and<br />

UI-2220-M from the U-Eye<br />

series of the German machine<br />

vision specialist achieved excellent<br />

results. The cameras<br />

under test were the model UI-<br />

1220-M, a monochrome camera<br />

with a 1/3“ CMOS sensor<br />

and a resolution of 752 x 480<br />

pixels, and the UI-2220-M, a<br />

monochrome camera with a<br />

1/2“ CCD sensor and a 768 x<br />

576 pixel resolution. With an<br />

excess housing temperature<br />

of only 3 °C and 4 °C, respectively,<br />

the cameras shine with<br />

measurement values that are<br />

among the lowest achieved so<br />

far. The design as well as the<br />

tillator. On customer request,<br />

the company supplies a phosphor<br />

scintillator as well. The<br />

highly sensitive CCD chip provides<br />

2048 x 2048 pixel resolution<br />

with a 14-bit dynamic<br />

range. With a pixel size of<br />

14 µm x 14 µm, the full frame<br />

CCD offers an active area of<br />

28.7 mm x 28.7 mm. The CCD<br />

sensor possesses a uniquely<br />

gives 128 times the image integration<br />

of a conventional<br />

line sensor, and when combined<br />

with the high sensitivity<br />

of the back thinned CCD chip<br />

results in a greater magnitude<br />

output of two or three<br />

orders compared to a regular<br />

line scan camera.<br />

Hamamatsu Photonics UK Ltd.<br />

Tel.: +800 80080088<br />

Europe@hamamatsu.com<br />

www.sales.hamamatsu.com<br />

optical density inside the Cmount<br />

lens connector, which<br />

effectively eliminates stray<br />

light in the flange, are exemplary<br />

and the dark noise is<br />

very low.<br />

IDS Imaging Development<br />

Systems GmbH<br />

Tel.: +49 7134 96196 0<br />

t.schmidgall@ids-imaging.de<br />

www.ids-imaging.de<br />

high quantum efficiency. The<br />

camera supports different<br />

frame rates.<br />

Olympus Soft Imaging Solutions<br />

GmbH<br />

Tel.: +49 251 79800 160<br />

Manfred.Kaessens@olympus-sis.<br />

com<br />

www.olympus-sis.com


Ultra-high Resolution<br />

Fei Company has added the<br />

Titan3 80-300 to its Titan<br />

product family. Aptly named<br />

the Titan “Cubed” because of<br />

its fully enclosed profile, the<br />

system is designed to deliver<br />

the highest stability and performance<br />

in a commercial<br />

scanning/transmission electron<br />

microscope (S/TEM). The<br />

ultra-high resolution S/TEM<br />

performance of this new system<br />

is achieved by its all-new<br />

design that allows the combination<br />

of two C s-abberation<br />

correctors and a monochromator<br />

on a single instrument.<br />

The system’s enclosure significantly<br />

reduces environmental<br />

interference providing<br />

greater stability and eliminating<br />

the need for many expensive<br />

lab improvements.<br />

FEI Company<br />

Tel.: +1 503 726 2695<br />

dzenka@fei.com<br />

www.fei.com<br />

Omniprobe Acquires Assets of Ascend<br />

Instruments<br />

Omniprobe announced that<br />

they have acquired the assets<br />

of Ascend Instruments. This<br />

company provides hardware<br />

and software for sample preparation<br />

and manipulation in<br />

the Focused Ion Beam (FIB)<br />

Microanalysis Software<br />

Edax has introduced the latest<br />

generation of its Genesis<br />

EDS microanalysis software.<br />

“The 5.2 version of the software<br />

includes features such<br />

as Auto Shape – an automatic<br />

collection routine with a free<br />

drawing capability,” explains<br />

Del Redfern, Product Marketing<br />

Manager. “The Auto<br />

Shape is an automatic spectrum<br />

collection routine that<br />

allows the user to select<br />

points or shapes from an image<br />

to collect spectrum. The<br />

microscope. In the near term,<br />

its product line will be manufactured<br />

at Omniprobe‘s Dallas,<br />

Texas, facility. In addition,<br />

Omniprobe has been<br />

selected by Entrepreneur<br />

magazine for their “Hot 500”<br />

Fastest Growing Companies<br />

in the U.S. The Hot 500 rankings<br />

are based on company<br />

performance and sales<br />

growth. The listing appears in<br />

the August 2007 issue of Entrepreneur<br />

magazine.<br />

Omniprobe, Inc.<br />

Tel.: +1 214 572 6800<br />

www.omniprobe.com<br />

software saves and labels the<br />

spectrum collected from the<br />

multiple positions along with<br />

their position that is then laid<br />

over the image.” The new features<br />

also include a software<br />

interface for the company’s<br />

Apollo 40 SDD and Apollo 10<br />

SDD silicon drift detectors.<br />

Edax, Inc.<br />

Tel.: +1 201 529 4880<br />

info.edax@ametek.com<br />

www.edax.com<br />

Transmission Electron Microscope<br />

The Nano Technology Systems<br />

Division (NTS) at Carl<br />

Zeiss SMT will introduce the<br />

new Centra 100, a transmission<br />

electron microscope with<br />

up to 100 kV accelerating<br />

voltage, at the MC Saarbrücken.<br />

Specially designed<br />

as a sophisticated “imaging<br />

system”, the highly compact<br />

and robust instrument offers<br />

maximum resolution down to<br />

PELCO ® Silicon Nitride Membranes<br />

Next Generation Si 3N 4TEM Support Films with<br />

many advantages:<br />

• Durable and chemically<br />

inert planar 50nm<br />

substrate<br />

• 3.0mm circular frame<br />

compatible with standardTEM<br />

holders<br />

• EasyGrip micro rough<br />

edges for ease of handling<br />

• Free from debris - no broken edges<br />

• Large area support film: up to 0.5 x 1.5mm<br />

• Complimented with Aperture Frames and Blank<br />

Disks for nanotech experiments<br />

Aperture<br />

Frame<br />

P r o d u c t s<br />

0.2 nm. The ease-of-use and<br />

fast specimen exchange capability<br />

make this microscope<br />

particularly well-suited for<br />

biomedical or clinical laboratory<br />

environments. A key<br />

technical feature of the system<br />

is the option of choosing<br />

between two different imaging<br />

modes: high resolution<br />

and high contrast. This is particularly<br />

important for investigating<br />

low-contrast biological<br />

specimens. The specially<br />

developed mini-lens design<br />

leads to a very compact size<br />

where the electron-optical<br />

lens elements exhibit a minimum<br />

aberration level only.<br />

Carl Zeiss SMT AG<br />

Tel.: +49 736420 2194<br />

info@zeiss.de<br />

www.smt.zeiss.com<br />

Blank Disks<br />

TED PELLA, INC.<br />

Microscopy Products for Science and Industry<br />

+1-530-243-2200 www.tedpella.com<br />

G.I.T. Imaging & Microscopy 4/2007 • 73


P r o d u c t s<br />

New Stereo Microscope<br />

Carl Zeiss Microimaging introduces<br />

the Stereo Discovery<br />

V20 stereomicroscope, designed<br />

for dissection and documentation,<br />

research and development,<br />

and quality<br />

assurance applications in a<br />

number of industries. The optical<br />

performance of the instrument<br />

boasts the largest<br />

available field of view (23 mm<br />

at 10x), the highest available<br />

zoom range (20 to 1) and the<br />

highest available resolution<br />

in the industry, combined in<br />

one stereomicroscope. These<br />

features allow users to visualize<br />

both large specimen and<br />

their small details on a single<br />

microscope without needing<br />

to change objectives or eyepieces.<br />

Thanks to the step<br />

motor control of the zoom op-<br />

LED light sources<br />

Olympus offers a range of<br />

LED sources for all fluorescent<br />

work, from entry level<br />

single colour units to advanced<br />

high speed multi-colour<br />

systems. It provides the<br />

right illumination solution,<br />

even for ‘in field’ fluorescence.<br />

LEDs now offer users<br />

an excellent alternative to arc<br />

burners since they have lifetimes<br />

in excess of 10,000<br />

hours, much longer than even<br />

the most advanced arc burner<br />

system. The Fluo-LED product<br />

range introduces additional<br />

flexibility to fluorescence<br />

microscopy. Each of the<br />

three models is designed to fit<br />

to the company’s CX microscopes<br />

making them ideal for<br />

routine as well as educational<br />

purposes. Further extending<br />

this application area is the<br />

ability to power the LEDs by<br />

74 • G.I.T. Imaging & Microscopy 4/2007<br />

tics, the stereomicroscope enables<br />

continuous increases in<br />

magnification with precise<br />

zoom levels to create a welldefined,<br />

high contrast image<br />

throughout the entire zoom<br />

range.<br />

Carl Zeiss MicroImaging GmbH<br />

Tel.: +49 551 5060 276<br />

mikro@zeiss.de<br />

www.zeiss.com/micro<br />

battery or even solar power<br />

ensuring that fluorescence<br />

microscopy can be conducted<br />

‘in field’.<br />

Olympus Life and Material<br />

Science Europa GmbH<br />

Tel.: +49 4023773 5426<br />

microscopy@olympus-europa.<br />

com<br />

www.olympus-europa.com<br />

Inverted Microscope for Live Cell Imaging<br />

Leica Microsystems presents<br />

the new generation Leica<br />

DMI3000 B inverted microscope,<br />

specifically designed<br />

for live cell research applications.<br />

It offers convenience<br />

and configuration possibilities<br />

that are unparalleled in<br />

this class of manual microscope.<br />

The company’s new,<br />

integrated incident light fluorescence<br />

axis produces brilliant<br />

images for all fluorescence<br />

techniques. The<br />

microscope also offers integrated<br />

modulation and phase<br />

contrast methods that do not<br />

require the use of special objectives.<br />

It is ideal for all manual<br />

fluorescence techniques.<br />

The system features a 5-position<br />

fluorescence turret for<br />

the fluorescence filter cubes.<br />

The company’s Fluorescence<br />

Intensity Manager (FIM) reg-<br />

Live Cell Care<br />

Nikon has launched<br />

its BioStation CT. By<br />

combining the precise<br />

environmental control<br />

capabilities of a highperformanceincubator<br />

with the advanced<br />

optics needed for driftfree<br />

live-cell imaging,<br />

it removes the need<br />

for culture dishes to<br />

be transported from<br />

one location to another for observation,<br />

and represents a<br />

‘hands-free’ approach to managing,<br />

observing and recording<br />

cells in culture. The company’s<br />

commitment to Live<br />

Cell Care was originally<br />

founded on the TE2000PFS<br />

system, for time lapse recording,<br />

the C1si microscope, for<br />

spectral confocal applications,<br />

and the BioStation IM, a single<br />

ulates the illumination, as<br />

well as the aperture and field<br />

diaphragm and their centering.<br />

Leica Microsystems GmbH<br />

Tel.: +49 6441 29 2550<br />

kirstin.henze@leica-<br />

microsystems.com<br />

www.leica-microsystems.com<br />

user, single experiment incubator,<br />

but has recently been<br />

enhanced by the introduction<br />

of Controlled Light Exposure<br />

Microscopy (CLEM) and the<br />

LiveScan Swept Field Confocal<br />

(SFC) Microscope.<br />

Nikon Instruments Europe<br />

Tel.: +44 208247 1718<br />

info@nikoninstruments.eu<br />

www.nikoninstruments.eu


Company page<br />

Agar Scientific 17, 64<br />

AHF Analysentechnik 70, 74<br />

Alicona Imaging 23<br />

Andor Technology 14, 71<br />

Bitplane 19<br />

Bruker-AXS Microanalysis 65<br />

BTX Harvard Apparatus 14, 70<br />

Coolled 70<br />

CRC Beatson Laboratories Garscube Estate 44<br />

Dhs Solution 51<br />

Digital Surf 8<br />

Dolan-Jenner Industries 70<br />

EDAX 40, 73<br />

European Science Foundation 14<br />

FEI Company 5, 31, 65, 73, Outside Back Cover<br />

Fianium 55<br />

Ges. f. Biotechnologische Forschung (GBF) 73<br />

Halcyonics 27<br />

Hamamatsu Photonics 72<br />

Hitachi High Technol. 24, 29<br />

IBaI Solutions Dr. Petra Perner GbR 62<br />

IDS Imaging Development Systems 72<br />

Image Solutions 72<br />

Imagic Bildverarbeitung 53<br />

JPK Instruments 14, 15, 37, 42, 66<br />

Leibniz-Institut für Altersforschung 15<br />

Leica Microsystems 15, 66, 74, Inside Front Cover<br />

Media Cybernetics 71<br />

MPI f. Eisenforschung 40<br />

Imprint<br />

Published by<br />

Head Office:<br />

<strong>GIT</strong> VERLAG GmbH & Co. KG<br />

Roesslerstrasse 90 · 64293 Darmstadt, Germany<br />

Tel.: +49 6151 80 90 0 · Fax: -144<br />

E-mail: imaging@gitverlag.com<br />

Managing Director<br />

Dr. Michael Schön, Bijan Ghawami<br />

Head of Sales & Marketing<br />

Anna Seidinger<br />

Head Journal Management, Advertising<br />

Dr. Martin Friedrich<br />

Tel.: +49 6151 80 90 171 · Fax: -168<br />

E-mail: m.friedrich@gitverlag.com<br />

Editor<br />

Dr. Thomas Matzelle<br />

Tel./Fax: +49 2845 981196<br />

E-mail: t.matzelle@gitverlag.com<br />

Advertising<br />

Dr. Stefanie Krauth<br />

Tel.: +49 6151 80 90 191<br />

E-mail: s.krauth@gitverlag.com<br />

US Office<br />

Dr. Michael Reubold<br />

John Wiley & Sons, Inc., Dept. <strong>GIT</strong> VERLAG<br />

111 River Street, 8th Floor, #8-033<br />

Hoboken, NJ 07030<br />

Tel.: +1 201 748 8810 · Fax: +1 201 748 6313<br />

m.reubold@gitverlag.com<br />

Editorial Assistance<br />

Tina Weber<br />

Tel.: +49 6151 80 90 261 · Fax: -176<br />

E-mail: t.weber@gitverlag.com<br />

Angela Seibert-Weck<br />

Tel.: +49 6151 80 90 131 · Fax: -183<br />

E-mail: a.weck@gitverlag.com<br />

Production Managers<br />

Dietmar Edhofer<br />

Tel.: +49 6151 80 90 122 · Fax: -145<br />

E-mail: production@gitverlag.com<br />

Sales Administrator<br />

Claudia Vogel<br />

Tel.: +49 6151 80 90 159 · Fax: -145<br />

E-mail: c.vogel@gitverlag.com<br />

Layout | Litho<br />

Ruth Herrmann | Elke Palzer, Ramona Rehbein<br />

Reprints<br />

Christine Mühl<br />

Tel.: +49 6151 80 90 169<br />

c.muehl@gitverlag.com<br />

I n d e x / I M P R I n T<br />

Company page<br />

Newport Spectra- Physics 71<br />

Nikon Instruments Europe 7, 67, 68, 74<br />

Olympus Life and Material Science Europa 3, 58, 67, 74<br />

Olympus Soft Imaging Solutions 45, 72<br />

Omega Optical 70<br />

Omniprobe 73<br />

PCO 35<br />

Photometrics 46, 49, Front Cover<br />

Physik Instrumente (PI) 17<br />

PicoQuant 57<br />

Prior Scientific Instr. 15<br />

Rauscher 72<br />

Royal Microscopical Society 12<br />

Schaefer- Tec 71<br />

Semrock 70<br />

SkyScan 11<br />

Syncroscopy 15<br />

Techn. Univers. Clausthal 48<br />

Ted Pella 73<br />

UCLM Laborat. de Nanotecnicas 36<br />

Unesco-Kommission 74<br />

Univers. Amsterdam 16<br />

Univers. Innsbruck 52<br />

Univers. Klinikum des Saarlandes 22<br />

Univers. of Antwerpen 10<br />

Univers. of London 28<br />

Veeco Instruments 39<br />

WiTec Wissenschaftl. Instr. u. Technologie 34, 71<br />

Carl Zeiss MicroImaging 9, 14, 74<br />

Carl Zeiss NTS 14, 15, 25, 73<br />

Advisory Board:<br />

Prof. A. Diaspro, Univ. of Genoa, Italy<br />

Dr. C. Durkan, Univ. of Cambridge, UK<br />

Dr. M. Dürrenberger, Univ. of Basel, Switzerland<br />

Dr. R. Fleck, NIBSC, Herts, UK<br />

Prof. M. Gu, Swinburne Univ., Australia<br />

Prof. B. Hecht, Univ. of Wuerzburg, Germany<br />

Prof. F.-J. Kao, Nat. Sun Yat-Sen Univ., Taiwan<br />

Prof. N. Kruse, Univ. of Brussels, Belgium<br />

Priv.-Doz. M. Hegner, Univ. of Basel, Switzerland<br />

Dr. D. Nicastro, Brandeis Univ., MA, USA<br />

Dr. J. Rietdorf, FMI, Basel, Switzerland<br />

Dr. P. Schwarb, FMI, Basel, Switzerland<br />

Prof. G. A. Stanciu, Univ. of Bucharest, Romania<br />

Prof. G. Valdré, Univ. of Bologna, Italy<br />

Dr. T. Zimmermann, ORG, Barcelona, Spain<br />

Printed by<br />

Frotscher Druck<br />

Riedstrasse 8 · 64295 Darmstadt, Germany<br />

Circulation<br />

18,000 copies, 4 times per annum<br />

Advertising price list from October 1 st 2007<br />

Subscription<br />

Four issues 33 €, single copy 12,60 € ca. plus postage.<br />

Pupils and students receive a discount of 50 % at sight of a valid<br />

certificate. Subscription orders can be revoked within 1 week in<br />

writing. Dispatch complaints are possible only within 4 weeks<br />

after publishing date. Subscription cancellations are accepted<br />

6 weeks before end of year.<br />

Bank account<br />

Dresdner Bank Darmstadt, Germany<br />

Account No. 0171550100<br />

Routing No. 508 800 50<br />

Specially identified contributions are the responsibility of the<br />

author. Manuscripts should be addressed to the editorial office.<br />

We assume no liability for unsolicited, submitted manuscripts.<br />

Reproduction, including excerpts, is permitted only with the<br />

permission of the editorial office and with citation of the source.<br />

The publishing house is granted the exclusive right, with regard<br />

to space, time and content to use the works/ editorial contributions<br />

in unchanged or edited form for any and all purposes any<br />

number of times itself, or to transfer the rights for the use of<br />

other organizations in which it holds partnership interests, as<br />

well as to third parties. This right of use relates to print as well<br />

as electronic media, including the Internet, as well as databases/<br />

data carriers of any kind.<br />

All names, designations or signs in this issue, whether referred<br />

to and/or shown, could be trade names of the respective owner.<br />

Printed in Germany<br />

ISSN 1439-4243<br />

www.imaging-git.com<br />

<strong>GIT</strong><br />

Laboratory<br />

Imaging<br />

BusinessPartner<br />

The purchasing Section for direct contact<br />

Dr. K. Hollborn & Söhne<br />

GmbH & Co. KG<br />

Brahestraße 13<br />

D-04347 Leipzig<br />

Tel: +49-(0)341-2334405<br />

Fax: +49-(0)341-2334406<br />

www.hollborn.de<br />

Medizinchemie@hollborn.de<br />

biology, dermatica, microscopy, pharmacy,<br />

reagents, stains<br />

Laboratory<br />

HWL Scientific Instruments GmbH<br />

Georgstrasse 11 · D-72119 Ammerbuch<br />

Tel: +49 7073/916796 · Fax: +49 7073/916798<br />

hwl@hwlscientific.com · www.hwlscientific.com<br />

Active Vibration Isolation for high resolution<br />

microscopy and measurement. Very effective<br />

against low frequenzy ambient vibrations.<br />

Nikon Instruments Europe B.V.<br />

Schipholweg 321<br />

1171 PL Badhoevedorp<br />

The Netherlands<br />

Tel: 0031 20 44 96 222<br />

Fax: 0031 20 44 96 298<br />

info@nikon-instruments.com<br />

www.nikon-instruments.com<br />

Optical microscopes, digital cameras for<br />

high resolution image capture<br />

Imaging<br />

OLYMPUS EUROPA GMBH<br />

Microscopes Int. Division<br />

Wendenstr. 14–18<br />

D-20097 Hamburg, Germany<br />

Tel.: +49 40 23773-326 · Fax: +49 40 23773-647<br />

microscopy@olympus-europa.com<br />

www.olympus-europa.com<br />

Optical microscopy systems for life science<br />

and material science, BioAnalytics products<br />

for cytomics and single molecule detection.<br />

Imaging<br />

This Space<br />

could be yours!<br />

Contact:<br />

Dr. Martin Friedrich<br />

Tel.: +49 6151 8090-171<br />

m.friedrich@gitverlag.com<br />

www.gitverlag.com


������������<br />

��������������������������������������<br />

���������������� �������� ��������� ��������� ����������<br />

��� ���� ������ ���� ����� ���� �������� �������� ���� �����<br />

����������� ���� ����� ��� ���� ���� ����� ���� ��� �����<br />

����������������������������������������������������������<br />

�������������������������������������������������������<br />

���� ����� ������� ����� ������ �������������� ��� ���������<br />

���������������������������������������������������������<br />

�����������������������<br />

������������������������������������������������������������<br />

������� ���� ����� ������� ���� ������������������ �������<br />

���������� ������� ��� ���� ������� ����������� ���� ���������<br />

��������������������������������������������������������<br />

������������������������������������������������������������<br />

����������������������������������������������������<br />

�������������������������������������������������������<br />

��� ��������� ���� ������� ��� ��������� ��� ����� ������� ����<br />

������������ ��� ��� ����������� ��������� ���������� ��� ����<br />

�����������������������������������������������<br />

��������������<br />

������������������<br />

������������������������������������������������������������������������������<br />

�����������<br />

��� �����<br />

��������������

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!