Download as pdf or txt
Download as pdf or txt
You are on page 1of 13

Archives of Biochemistry and Biophysics 581 (2015) 98–110

Contents lists available at ScienceDirect

Archives of Biochemistry and Biophysics


journal homepage: www.elsevier.com/locate/yabbi

Review

Correlative microscopy
Céline Loussert Fonta 1, Bruno M. Humbel ⇑
Electron Microscopy Facility, University of Lausanne, Biophore, 1015 Lausanne, Switzerland

a r t i c l e i n f o a b s t r a c t

Article history: In recent years correlative microscopy, combining the power and advantages of different imaging system,
Received 9 March 2015 e.g., light, electrons, X-ray, NMR, etc., has become an important tool for biomedical research. Among all
and in revised form 26 May 2015 the possible combinations of techniques, light and electron microscopy, have made an especially big step
Available online 10 June 2015
forward and are being implemented in more and more research labs.
Electron microscopy profits from the high spatial resolution, the direct recognition of the cellular ultra-
Keywords: structure and identification of the organelles. It, however, has two severe limitations: the restricted field
Correlative microscopy
of view and the fact that no live imaging can be done. On the other hand light microscopy has the advan-
Light and Electron microscopy
STEM scanning transmission electron
tage of live imaging, following a fluorescently tagged molecule in real time and at lower magnifications
microscopy the large field of view facilitates the identification and location of sparse individual cells in a large con-
Mouse brain text, e.g., tissue. The combination of these two imaging techniques appears to be a valuable approach to
GFP green fluorescent protein dissect biological events at a submicrometer level.
Epoxy Light microscopy can be used to follow a labelled protein of interest, or a visible organelle such as mito-
Methacrylate chondria, in time, then the sample is fixed and the exactly same region is investigated by electron micros-
Cryo-fixation and Freeze-substitution copy. The time resolution is dependent on the speed of penetration and fixation when chemical fixatives
Tokuyasu cryo-sectioning
are used and on the reaction time of the operator for cryo-fixation. Light microscopy can also be used to
Immuno-(gold) labelling
identify cells of interest, e.g., a special cell type in tissue or cells that have been modified by either trans-
fections or RNAi, in a large population of non-modified cells. A further application is to find fluorescence
labels in cells on a large section to reduce searching time in the electron microscope. Multiple fluores-
cence labelling of a series of sections can be correlated with the ultrastructure of the individual sections
to get 3D information of the distribution of the marked proteins: array tomography. More and more
efforts are put in either converting a fluorescence label into an electron dense product or preserving
the fluorescence throughout preparation for the electron microscopy.
Here, we will review successful protocols and where possible try to extract common features to better
understand the importance of the individual steps in the preparation. Further the new instruments and
software, intended to ease correlative light and electron microscopy, are discussed. Last but not least we
will detail the approach we have chosen for correlative microscopy.
Ó 2015 Elsevier Inc. All rights reserved.

Introduction advantages of both techniques and also how useful, yet important
it is to combine them to complete the picture in biological
For many years light and electron microscopy were two sepa- research. The first successful correlative light and electron micros-
rate imaging techniques. Either the researcher was a light micros- copy data of a study of microtubules in PtK2 cells were published
copist or an electron microscopist. Also sample preparation in 1978 [1,2].
methods differed a lot from formalin fixation or acetone/methanol Electron microscopy profits from the high spatial resolution and
precipitation to highly sophisticated cryo-preparation schemes. the direct recognition of the cellular components. It, however, has
Even today the division exists and talking about it is not always two severe limitations: the restricted field of view and the fact that
well received. Fortunately, the two groups start to realise the no live imaging can be done. Light microscopy has the advantage of
live imaging, following a tagged molecule in real time. Through
⇑ Corresponding author. Fax: +41 21 6925055. super-resolution the localisation of fluorescent molecules have
E-mail addresses: Celine.Loussert@rdls.nestle.com (C. Loussert Fonta), Bruno. greatly improved [3–6] but there is no high-resolution information
Humbel@unil.ch (B.M. Humbel). of the total cellular ultrastructure as in electron microscopy. On
1
Present address: Nestle Research Center, Vers-chez-les-Blanc, Case postale 44, electron micrographs the reference structures can easily be
1000 Lausanne 26, Switzerland.

http://dx.doi.org/10.1016/j.abb.2015.05.017
0003-9861/Ó 2015 Elsevier Inc. All rights reserved.
C. Loussert Fonta, B.M. Humbel / Archives of Biochemistry and Biophysics 581 (2015) 98–110 99

identified. The immuno-gold labelling of sparse antigens, however,


can be frustrating. What may still give a decent fluorescence signal
can be only a few gold particles on the section leading to no or at
best very doubtful results. Here, the direct combination of the two
information, localisation by (super-resolution) light microscopy
and high resolution morphology by electron microscopy is the
method of choice.
Combining the data provided by these two optical methods
enables live cell imaging, finding a single event in a large popula-
tion of cells and identifying the local surrounding of the event at
high resolution. In the last two decades correlating light and elec-
tron microscopy data has become popular and essential in address-
ing specific questions in cell biology [7]. New preparation methods
but also instruments and software dedicated to correlative micros-
copy have been developed.
In this review we address the current state of successful prepa-
Fig. 1. Fluorescence map. The light micrograph serves the operator as a guide to
ration schemes for the different approaches for correlative micros- find the marked cells in the electron microscope. Using the shape of the section and
copy. Special attention is given to the protocols that preserve the local surrounding of the cell of interest it can be identified in the electron
fluorescence throughout the preparation protocol for electron microscope. In more complicated tissue, the searching process can be very time
microscopy. Here, common denominators are searched and their consuming and sometimes even unsuccessful.
importance is discussed. Further, the newly developed light–elec-
tron microscopes are described and put in the perspective of their
applications. Last but not least we are describing in detail the
approach chosen in our lab, using the Tokuyasu cryo-sectioning
technique in combination with the Maps software.

Methods of correlation

On-section immuno-labelling

There are many methods to prepare a sample for correlative


microscopy depending on the ‘cultural’ background of the lab
and the specific needs. The easiest and most straightforward way
is the labelling of thin sections. The specimen is prepared with a
protocol suitable for immuno labelling (electron) microscopy.
There is a wealth of methods based on chemical fixation and meth-
acrylate embedding [8–17], on cryo-fixation, freeze-substitution
and low-temperature embedding [18–21] or on Tokuyasu
cryo-sections [22–27]. From such prepared material sections of
200 nm but also ultrathin sections can be cut and mounted on glass
coverslips and immuno labelled with fluorescent antibodies [28–
34] for light microscopy investigations and consecutive sections
on grids, labelled with colloidal gold for electron microscopy.
Alternatively, sections can also be mounted directly on electron
microscopy grids or ITO coverslips (coverslips that are coated with
Indium-Tin-Oxide to make the surface electrically conductive,
Optics Balzers AG, Balzers, Liechtenstein) [35] and labelled with
fluorescent and gold conjugates for analysis with light microscopy
and thereafter in a TEM2 [36] or SEM [30].
After imaging in a fluorescence microscope the resulting micro-
graphs are used as a map, either lying next to the electron micro-
scope (Fig. 1) or overlaid with the electron micrograph using a
sophisticated software [37–40], to ease the localisation of the cells
of interest in the electron microscope (Fig. 2).
Often, however, it is desired to observe the fluorescent target in
the living cells before fixation to catch a special event [41–47] or to Fig. 2. Overlay of a fluorescence image with an electron micrograph. The fluores-
prepare a whole sample to find the place of interest, indicated by cence image was loaded into the correlation software Maps (see below) and
the fluorescence label, in a three-dimensional environment matched with the electron micrograph of the same section mounted on an ITO
coverslip. Groups of cells of interest were searched on the fluorescence image, the
[38,48–50]. In the following we describe different approaches that
magnification and position of the microscope were adjusted that the group of cells
was within the recording frame (pink frame in A) and then an electron micrograph
(B) was recorded (adapted from [30]). Bars represent 3 lm (A) and 2 lm (B). (For
2
Abbreviations used: LM, light microscope; EM, electron microscope; TEM, trans- interpretation of the references to colour in this figure legend, the reader is referred
mission electron microscopy; SEM, scanning electron microscopy; STEM, scanning to the web version of this article.)
transmission electron microscopy; ILEM, integrated laser electron microscope; GFP,
green fluorescent protein; miniSOG, mini singlet oxygen generator; RNAi, RNA
interference; ITO, indium tin oxide; CEMOVIS, cryo-electron microscopy of vitreous allow the identification of fluorescently labelled proteins in whole
sections; DAB, 3,30 -diaminobenzidine; GMA, glycol methacrylate. cells by light and electron microscopy.
100 C. Loussert Fonta, B.M. Humbel / Archives of Biochemistry and Biophysics 581 (2015) 98–110

Pre-embedding immuno-labelling (photo-oxidation and particulate Methacrylates (comprehensively reviewed in [79]), however,
markers) polymerise by radical chain reaction meaning they can only
cross-link with themselves, leaving the biomolecules untouched
There are many possibilities to label a specific protein in a cell. [73–75]. All of the published recipes of successful preservation
The classical approach is, after chemical fixation with aldehydes, to of GFP fluorescence use methacrylates as resin. The different
permeabilise and immunolabel with a first antibody against the methacrylate formulations have different polarities, where
protein of interest. The permeabilisation step can be omitted when Lowicryl K4M is the most hydrophilic, followed by Lowicryl
surface antigens are labelled [47] or the label is taken up by a nat- K11M, then by LR white, LR gold and Unicryl, and then by
ural process such as endocytosis [42]. In a second step the primary Lowicryl HM20 and the least hydrophilic Lowicryl HM23 [79].
antibody is detected with a fluorescent secondary antibody [51], an Even HM20 is not absolutely apolar, it can tolerate 0.5%
antibody that fluoresces and has a particulate marker like fluoro- (w/w) of water, whereas the hydrophilic K4M tolerates up to
nanogold (Nanoprobes, Inc., Yaphank, NY, USA) [52], 10% (w/w) of water [9,79]. No systematic experiments have
streptavidin-Alexa488-10 nm gold [42] or quantum dots (Life been done to investigate the influence of resin polarity on
Technologies, Grand Island, NY, USA) [53], which fluoresce and the preservation of the fluorescence of GFP or other
are electron dense. fluorochromes.
The cells of interest are identified by fluorescence microscopy,
then the sample is thoroughly fixed with higher concentrations Successful preparation protocols
of glutaraldehyde and the fluorescence signal is transferred into a
diaminobenzidine precipitate [51,54]. 5 nm-sized and larger colloi- In the following we will summarise a few of the successful
dal gold particles or quantum dots are electron dense and directly preparation protocols to preserve fluorescence with the aim to find
visible in the electron microscope. The ultra-small gold particles the common denominator.
generally need to be silver-enhanced [52]. Small quantum dots
are sometimes not easily seen in an electron dense cellular envi- Chemical fixation and resin embedding
ronment this can be remedied by silver-enhancement [55]. After Two protocols are based on chemical fixation. (a) Zebrafish was
the different markers are made sufficiently electron dense, the fixed with 4% formaldehyde, 0.1% glutaraldehyde, 3% sucrose in
sample is post-fixed with osmium tetroxide and embedded into PBS, pH 7.4, then dehydrated with ethanol and embedded in LR
an epoxy resin. Caveat, quantum dots can be routed into a different white [80]. (b) Tobacco petioles were fixed with 4% formaldehyde,
pathway in living cells [53] and osmium tetroxide is not always 2% glutaraldehyde, 1 mM CaCl2 in 50 mM PIPES, (pH not men-
compatible with silver-enhancement [56]. Alternatively, to classi- tioned), dehydrated in 90% ethanol/water and embedded in LR
cal embedding, cryo-fixation and freeze-substitution can be white [81].
applied [42].
More elegantly, clonable markers [57–61] are used to fluores- Cryo-fixation, freeze-substitution and resin embedding
cently tag the protein of interest. After imaging in a light micro- The other protocols rely on cryo-fixation by high-pressure
scope the fluorescence signal is used to polymerise freezing [82–86], freeze-substitution [87] and low-temperature
diaminobenzidine into an insoluble osmophilic precipitate as done embedding [18,20,88–90]. It is, however, not clear if, next to the
with horseradish peroxidase [62]. This method uses the fact that improved structural preservation, the cryo-fixation–freeze-substi
during fluorescence in combination of externally added oxygen, tution approach is also beneficial for the preservation of the fluo-
the singlet oxygen molecules are generated that are needed for rescence of GFP and other fluorescent dyes.
the diaminobenzidine reaction. Hence, the precipitate is closely All freeze-substitution media are based on acetone without
associated with the fluorescence marker. There are methods [48,91] or with 1–5% added water [92–96]. Water addition
described to convert GFP [43,63–65], ReAsh [58] and an improved can improve membrane contrast [97,98], it might also help
system, the miniSOG, mini singlet oxygen generator [57] into the to preserve the fluorescence signal. Except for one [95], they
osmophilic DAB precipitate. all contain uranyl acetate as a fixative and stain. The concen-
As an additional option, areas or volumes of interest can be tration varies from a saturated solution (0.1%) to 0.2%. The
marked during light microscopy investigation with a higher concentration is added from a 20% methanol stock solu-
near-infrared laser, the so called infrared (IR) branding. The mark- tion [92,94], which results in the addition of 4% methanol!
ers are autofluorescent and can be photooxidised to an electron Interestingly, nobody of those using water tried to add the
dense signal for their re-location in the electron microscope [66]. uranyl acetate as an aqueous solution to avoid the negative
Interestingly, although these methods are very potent for cor- effects of methanol on the cellular ultrastructure [99]. In one
relative microscopy they have not yet found routine application case the best results in terms of ultrastructure and preserva-
and the wish remains to preserve the fluorescence signal through- tion of the fluorochromes were obtained with 0.001% osmium
out the preparation for electron microscopy imaging. tetroxide and 0.1% potassium permanganate as fixatives
[95,96].
Resins The freeze-substitution scheme varies only slightly, 30–58 h at
90 °C (85 °C), then warming with or without intermediate stops
For electron microscopy sample preparation, two different to 50 °C to 20 °C. Usually the substitution medium remains
types of resins are available: Epoxies (Epon [67], Araldite [68], unchanged during the whole freeze-substitution programme
Spurr’s low viscosity resin [69], Durcupan [70,71]) and methacy- except in one protocol where the osmium tetroxide and potassium
lates (London Resins [13], Lowicryls [9,11], Unicryl (Bioacryl) permanganate is replaced at 50 °C for 0.1% uranyl acetate [96].
[15], glycol methacrylate (GMA) [72]). After the washing steps with acetone or ethanol, embedding takes
Epoxy resins can co-polymerise with the biological components place into Lowicryl HM20 [48,91,92,94] Lowicryl K4M [94], LR
[73–75], meaning they cross-link with biomolecules [76–78]. The white [94] or GMA [95,96]. Watanabe et al. [95,96] noticed that
epoxy group is very reactive and capable of ring opening reactions some batches of LR white are very acidic and therefore could inter-
to many organic moieties [74], hence also to fluorochromes and fere with the fluorochromes that optimally fluoresce at higher pH
thus it often reduces or even abolishes fluorescence. There is, how- around 8.0–8.5 [100,101]. Peddie et al. [94] use the fast substitu-
ever, a report of successful use of epoxy resin [48]. tion protocol [102] but they do not comment if the fast protocol
C. Loussert Fonta, B.M. Humbel / Archives of Biochemistry and Biophysics 581 (2015) 98–110 101

has any influence on the stability and emission efficiency of the


fluorochromes.
As an additional note: uranyl acetate binds to phosphate groups
and can thus cross-link phospholipids and nucleic acids [103].
Osmium tetroxide at high concentration and room temperature
can act as a protease [104,105]. At low concentrations, e.g.,
0.02%, antigenicity is not completely abolished but reduced by
50–75% for a particular epitope (1987, unpublished observation),
used for chemical fixation during freeze-substitution osmium
tetroxide works more like a fixative [106,107] and the effect of
1–2% of osmium tetroxide on the antigenicity is comparable to
1–2% of glutaraldehyde [108].
Summarising the observations, we can conclude that the cellu-
lar ultrastructure is better preserved by cryo-fixation and
freeze-substitution; methacrylates are the preferred resins, how-
ever, LR white might be too acidic to retain high fluorescence effi-
ciency. 0.1–0.2% uranyl acetate or low concentrations of osmium
tetroxide and potassium permanganate do not abolish fluorescence
of GFP, mCherry and mTomato. The results give no clue on the
importance of added water during the substitution process and
washing steps, although an aqueous environment seems to be
important for maintaining GFP fluorescence [59]. It is advised to
store the resin blocks in the cold and dark to preserve the GFP sig-
nal and the sections need to be imaged as fast as possible. There is
one report on a reduction of fluorescence from living cells to resin
embedded cells of up to 70% [109].

Tokuyasu cryo-sectioning
An aqueous environment, at least a certain number of water
molecules close to the chromophore, seems to be important to
maintain fluorescence of GFP [59]. Therefore we decided to
setup a correlative approach based on the cryo-sectioning
method developed by Kyoteru Tokuyasu [22–24,110] and
refined by the group of Slot and Geuze [26,27]. During the
Fig. 3. Finding the area of interest. The whole 300 lm thick slice is imaged to find whole preparation process, until the final drying step, for imag-
the fluorescent neuron. The area containing the neuron is cut out and infiltrated ing in the electron microscope, the proteins are kept in an
with sucrose. Bars represent 200 lm. aqueous environment.

Fig. 4. CorrSight light microscope. In (A), the top of the light microscope is visible, the screen with the open software, depicting the fluorescence signal of a section, the lamp
in the cap for bright field imaging and the setup of the microscope slide holder. The large part with the handles but also the inner part (C) can easily be removed and replaced
by other setups. (B) The special holder for the pin of the microtome with the container to be filled with ice to keep the sample cold and in a moist atmosphere during
recording. (C) Mounting of the special setup.
102 C. Loussert Fonta, B.M. Humbel / Archives of Biochemistry and Biophysics 581 (2015) 98–110

Fig. 5. 3D acquisition of a neuron. In the wide-field microscope fluorescence images are recorded while stepping through the sample block at 10 lm per step in Z. The neuron
is in focus from image 8 to image 24, i.e., the fluorescent neuron begins at 80 lm below the block surface and ends at 240 lm.

Cryo-fixation and imaging of the frozen-hydrated sections Instrumentation


Another option is to keep the sample in its native water sur-
rounding. As water will evaporate in high vacuum it needs to Correlative light and electron microscopy can be done with any
be kept frozen. Already in the early 80s biological material was fluorescence and electron microscope. The fluorescence image is
immobilised by cryo-fixation, sectioned and imaged at very low used as a guide during electron microscopy to find the correspond-
temperatures [111–114]. This preparation method has been ing cell (Fig. 1). To ease and speed-up the correlation between light
improved considerably by the group of Prof. Jacques Dubochet and electron microscopy new instruments and software packages
[115–117] and today with modern equipment CEMOVIS have been and are being developed and are discussed in the
(cryo-electron microscopy of vitreous sections) has become less following.
exclusive. The next step was to develop a cryo-stage for a light
microscope to image the fluorescence within the
frozen-hydrated section and record the X, Y positions of the area Light and electron microscopes as one instrument
of interest for high-resolution analysis in the electron microscope
[39,118]. A further step forward was the development of a com- The idea of using a combined light and electron microscope is
bined light–electron microscope [37,119] where the fluorescence not new, already in 1982 an instrument was built for easy diagnos-
can directly be imaged in the TEM with an attached light micro- tics in routine surgical pathology [125]. This instrument, however,
scope, before imaging with electrons, thus omitting one was never commercialised and got forgotten.
cryo-transfer step. As an additional benefit it has been noted that The idea was picked-up by the groups of Prof. Arie Verkleij and
the fluorescence is more stable in vacuum, probably due to the Prof. Hans Gerritsen of Utrecht University and a laser light micro-
absence of singlet oxygen molecules, and under cryogenic condi- scope (ILEM) was constructed. The ILEM could be attached, inte-
tions [120]. With this approaches correlative light and electron grated, at a side-entry port, 90° to the rod of the sample holder,
microscopy has entered the realm of true cryo-microscopy into the twin lens of a Tecnai electron microscope (FEI Company,
[40,121–124]. Eindhoven, The Netherlands) [37]. Fluorescently labelled sections
C. Loussert Fonta, B.M. Humbel / Archives of Biochemistry and Biophysics 581 (2015) 98–110 103

membrane that serves as the final electron lens. Depending on


the nature of the detectors, they are mounted in the vacuum of
the microscope, e.g., the BSE detector, or outside under atmo-
spheric conditions. The airSEM can be coupled with any light
microscopy system for easy shuttling of the sample between the
imaging units [128].
A similar approach has been chosen by JEOL Ltd for their
ClairScope™ [129,130]. The scanning electron microscope with a
tungsten emitter is mounted up-side-down under a table. The col-
umn is sealed with a SiN window, 250 lm  250 lm and 100 nm
thick. The BSE detector is mounted in the vacuum of the SEM col-
umn. The sample is cultured and manipulated in the dish with the
SiN window above the pole piece of the electron column. Opposite
the electron beam a light microscope is attached to image fluores-
cence or cathodoluminescence [129,130].
Thirdly, there is also an inverted version of the ClairScope setup
described [131]. The handling of the cells with this instrument
seems a bit more tricky as they are hanging up-side down under
the SiN window and cannot be manipulated during imaging.
Fourthly, there is an inverted light microscope, the
Simultaneous Correlative Light–Electron Microscope (SCLEM), that
can be installed in any scanning electron microscope as an attach-
ment, [132,133]. The objective of the light microscope is mounted
in the vacuum chamber of the scanning electron microscope
directly under the electron beam. The light source and CCD camera
are outside. The sample is mounted on a coverslip above the light
optics. The advantage of this system is that high-numeric aperture
immersion-oil objectives, up to 1.4 NA, can be used; the electron
beam can be aligned with the photon beam at very high accuracy,
within 10 nm; and last but not least any light microscope system,
e.g., PALM, can be attached and fed into the light path [132]. The
SCLEM is produced and marketed by DELMIC BV, Delft, The
Netherlands.
The published electron micrographs of all those SEM systems,
however, are of minor quality [128–130,132,133] they do not make
use of the resolution power of the SEM. Effort in designing accurate
Fig. 6. Observation of the area of interest in the ultramicrotome. The area of
interest, here the Lucifer yellow injected neuron (arrow head), is always visible sample treatment and preparation is needed to be able to fully
during microtomy and the block can be precisely trimmed. Then ultrathin sections profit from the theoretical resolution of the instruments. Using
containing the neuron can be cut. thin sections of fluorescently labelled cells, comparable to the
TEM setups, the DELMIC system can provide stunning results
[134]. In addition this system has the potential to automate array
with or without additional gold label on grids are loaded into the tomography [135,136].
electron microscope, tilted 90° to the electron beam and the fluo- Next to dedicated microscopes there are special holders, shut-
rescence is imaged in vacuum with the ILEM. The coordinates of tles, developed to ease the transfer of the samples between a ded-
the interesting cells are stored, the holder is tilted back into the icated light to a dedicated electron microscope. Using individual
electron beam and the positions are recalled to image the same systems has the advantage that no compromises need to be done
areas by electron microscopy [36,37,126]. The fluorescence needs in the preparation of the sample. The fluorescence signal can be
to be imaged before the section is observed in the electron beam kept under optimal conditions and high-numeric aperture objec-
as after a certain dose of electrons the observed area starts to auto- tives can be used. Further any light microscopy imaging mode from
fluoresce and the specific fluorescence on the section can no longer wide-field to confocal to super-resolution can be applied. After the
be observed. A commercial version of this instrument, however, fluorescence micrographs are taken, the sample can be further pro-
with wide-field illumination, iCorr™, is available from FEI cessed, e.g., by immuno-gold labelling, and contrasted with heavy
Company, Eindhoven, The Netherlands. metal solutions for optimal imaging conditions in the electron
The group of Prof. Kuniaki Nagayama had the same idea. In their microscope.
construction they keep the sample in the imaging position of the
electron microscope and added a mirror at 45° with a central hole Calibrated transfer shuttles
for the electrons to pass and image the fluorescence also with the
side-mounted light microscope [119,127]. This instrument is mar- Carl Zeiss Microscopy GmbH has developed the ‘Shuttle & Find’
keted by JEOL Ltd., Tokyo, Japan. system that uses coverslips with 3 fiducial markers to calibrate the
There are 4 descriptions of correlative microscopes that use a positions. Images are taken with any kind of light microscope, e.g.,
scanning electron microscope for the high-resolution imaging the super-resolution light microscope ZEISS ELYRA PS.1, using a
along with a light microscope. 1.46 NA objective and the coordinates of the images are stored in
Firstly, the airSEM™ that can image samples kept under atmo- relation to the fiducials of the coverslip. After preparation for elec-
spheric conditions [128]. The microscope consists of a field emis- tron microscopy the samples are transferred to a scanning electron
sion scanning electron microscope that is vacuum-sealed with a microscope and the location of the fluorescence images is automat-
membrane at the pole piece. The sample is 50–200 lm below the ically retrieved by the ‘Shuttle & Find’ system [49,137,138]. A similar
104 C. Loussert Fonta, B.M. Humbel / Archives of Biochemistry and Biophysics 581 (2015) 98–110

Fig. 7. A serial section of mouse brain tissue with eGFP labelled astrocytes. Serial Tokuyasu cryo-sections were collected on a Formvar-carbon coated copper finder grid and
imaged, sandwiched between a coverslip and microscope slide, using oil immersion objectives. The numbers indicate the areas imaged afterwards by electron microscopy.
The arrows indicate the cell of interest in the consecutive section that was not imaged as it is lying on a grid bar. Bar represents 500 lm.

Fig. 8. Bright field and fluorescence images in CorrSight. Bright field and fluorescence images at different magnification are recorded with Maps in the CorrSight light
microscope. All images are already linked to each other. (A) is a montage of a bright field overview of the whole grid and the fluorescence image. Note the central hole with
the arrow and the number 1 in the rim of the grid. (B) A higher resolution fluorescence image of the Tokuyasu serial section is shown. Bars represent 1 mm (A) and 300 lm
(B).

approach has been developed by JEOL and Nikon, the MiXcroscopy Institute in Munich [39,118], that is now marketed by FEI
correlative imaging solution. The same specimen holder is used for Company. There is also a version from the University of Colorado
both light microscopy (Nikon) and SEM (JEOL). The specimen regis- at Boulder [139] and the stage of Linkham Inc., was engineered
tration is fully controlled by the MiXcroscopy software. at the University of Leiden [140]. After imaging by cryo-light
For correlative cryo-microscopy several cryo-stages for light microscopy the calibrated reference data are fed into the correla-
microscopy were developed. The first one by the Max-Planck tive software controlling the TEM.
C. Loussert Fonta, B.M. Humbel / Archives of Biochemistry and Biophysics 581 (2015) 98–110 105

Fig. 9. Alignment and correlation of the light and electron micrographs. A secondary electron image of the central hole and the number 1 is recorded and the light micrograph
is loaded into the Maps software. With 2 points, e.g., the very centre of the hole (green) and one corner of the number 1 (blue), the light micrograph is rotated and stretched to
match the electron micrograph. Bars represent 50 lm (A), 30 lm (B) and 400 lm (C).

Fig. 10. Result of coarse alignment. The precision of the coarse alignment of the fluorescence and electron micrograph is within 1.5–5 lm. In the example shown, the
fluorescently labelled cell is displaced by 4.97 lm (green line) in respect to the electron micrograph. Bar represents 7 lm.

Correlation and imaging software TESCAN Orsay holding. With CORAL any image can be correlated
with TESCAN SEM systems.
The Maps software (FEI Company) is a specially designed soft- In this section we describe our approach for correlative electron
ware to correlate light and electron microscopy for FEI CorrSight microscopy, the preparation scheme and in detail the use of the
light microscopes and scanning electron microscope systems Maps software.
[30,38]. In addition, Maps software can read any type of images The examples given are from brain tissue: one that expresses
obtained by any type of light microscope. A similar software, eGFP in astrocytes [141] and one where one neuron was stained
CORAL (correlative microscopy for life science) was developed by with Lucifer yellow. Fresh brain tissue was cut with a vibratome
106 C. Loussert Fonta, B.M. Humbel / Archives of Biochemistry and Biophysics 581 (2015) 98–110

Fig. 11. Fine alignment. With the radio buttons in the lower left corner the light
micrograph can be shifted and rotated to perfectly match the electron micrograph. Fig. 12. Fluorescence to identify the cell of interest. The neuron was labelled by
Due to local distortion this alignment might be repeated for distant structures. Bar Lucifer yellow during physiological experiments. Correlating the fluorescence with
represents 4 lm. the electron micrograph identifies the cell that has been studied. Note: the intenser
fluorescence signal over the crease. Bar represents 9 lm.

Fig. 13. Tileset setup. For large overview at high resolution the area of interest is covered with a set of tiles defined by the pixel resolution and the frame size. There is an
overlap of 10% for subsequent stitching. At 3 points the focus is fixed (white circles, WD Points 1–3) to calculate the focus for each tile and the set is recorded. Bar represents
20 lm.
C. Loussert Fonta, B.M. Humbel / Archives of Biochemistry and Biophysics 581 (2015) 98–110 107

Fig. 14. Tileset of a brain section. (A) A part of a large stitched tileset of 44.9 lm by 41 lm with 1 nm pixel resolution. When gradually zooming in (B and C) the individual
gold particles indicating astrocytes become visible (C). Note that the cellular ultrastructure in the astrocyte is remarkably well preserved by the Tokuyasu preparation
method. Bar represents 2 lm.
108 C. Loussert Fonta, B.M. Humbel / Archives of Biochemistry and Biophysics 581 (2015) 98–110

into 300 lm slices then they where immediately fixed in 2% form- be identified without the need of an electron microscopy marker
aldehyde and 0.2% glutaraldehyde. In a first step, the fluorescently (Fig. 12).
labelled neuron is located (Fig. 3). For large image acquisition, tilesets covering the entire area of
Tissue blocks fixed with aldehydes, were impregnated with interest with the desired pixel resolution, typically 1 nm, are
2.3 M sucrose and then mounted on a sample support pin of the drawn (Fig. 13). Our usual imaging conditions are: 30 keV,
cryo-ultramicrotome. The block on the pin is kept humid and cool 800 pA, 5 mm working distance, 6144  4096 pixels per frame,
either by a self-made support dish [38] for imaging with a Zeiss 1–3 ls dwell time, using the STEM III detector (FEI Company) in
fluorescence microscope (Imager.Z2, Carl Zeiss Microimaging the high-angle annular-dark field mode [142,143]. Up to now the
GmbH, Germany) or a special holder (Fig. 4) designed for the 3 point focus regime was the most reliable. At 3 points (Fig. 13,
CorrSight light microscope (former Till Photonics, now FEI white circles) close to the area of interest the section is focused
Company, Munich). at higher magnification, then brightness and contrast are adjusted
A 3D stack is recorded to define the Z-position of the area of at the same magnification and imaging conditions the tileset will
interest, e.g., eGFP expressing astrocytes or a fluorescently labelled be recorded. After all these adjustments are done the recording
neuron (Fig. 5) in brain tissue with the Zeiss Axiovision rel.4.8 soft- can be started. With the specified settings, typical recording time
ware (Carl Zeiss) or the LA software (Live Acquisition, Till is about 1 h for a tileset of 8  11 images. The overlapping zone
Photonics) integrated into the Maps software (FEI Company). of the tiles often is seen as a brighter strip, which is rather disturb-
After freezing, the pin is mounted in a cryo-ultramicrotome ing. To avoid this effect, the whole area of interest, when possible
equipped with a fluorescence stereo microscope [38] and the block the whole mesh is pre-irradiated with a high current, 13–26 nA;
is trimmed until the area of interest, using the 3D information large frame size, 6144  4096 pixels; and short dwell time,
obtained in the light microscope and the observation of the fluo- 50 ns; for about 30 min.
rescence in the microtome (Fig. 6). The tiles can be stitched by the software and exported as tiff
The sections, often serial sections (Fig. 7), are collected on files or raw files when they become too big. An other useful option
carbon-coated electron microscopy grids. is the export as an HD view file (http://research.microsoft.com/en-
The grids are sandwiched between a microscope slide and a us/um/redmond/groups/ivm/HDView/) that, under the Windows
coverslip in PBS and sealed with nail polish. With a light micro- operating system, allows viewing the whole recorded field and to
scope low resolution, 20 objective NA 0.5, the whole grid is zooming into the individual areas of interest (Fig. 14).
imaged using a montage option in bright field (Fig. 8A) and fluores-
cence and then fluorescence of the section at higher resolution,
Outlook
40 oil objective NA 1.3 (Figs. 7 and 8B). These maps are then used
to find the same areas in the electron microscope. Any fluorescence
Correlative light and electron microscopy has become a stan-
microscope can be used but the CorrSight has an added advantage
dard technique for many investigations in biomedical research.
that it operates with the correlative Maps software and all light
With it, the micro- and nanoscopic dimensions in imaging are cov-
micrographs of one particular project are stored with their respec-
ered. Light microscopy can image areas in millimetre in X, Y and
tive coordinates and are automatically linked to each other.
200–300 lm in thickness at about 200 nm lateral resolution and
After light microscopy imaging is completed, the project is
electron microscopy in the micrometre range and thickness in
transferred on the SEM computer and opened in the Maps software
the 100 nm range down to subnanometre resolution.
installed on that system. The grids are recovered from the light
In biology there are many more dimensions to be covered.
microscopy setup and further treated, e.g., by immuno gold label-
Further, the morphological information can be complemented with
ling, post-stained with uranyl acetate and embedded in methyl cel-
chemical information. Presently, soft X-ray microscopy stands
lulose before loading into the STEM holder of the scanning electron
between light an electron microscopy. The thickness of a sample
microscope [38]. An electron micrograph is recorded with the
that can be analysed is about 15 lm and the achieved resolution
Everhart–Thornley secondary electron detector. We usually take
below 50 nm [144]. Soft X-ray microscopy is usually associated
an image of the central hole with the arrow and the number 1 at
with a synchrotron [144,145] but laboratory based models are
the border of the grid, but any two points that are clearly visible
upcoming [146]. Soft X-ray imaging in addition can provide chem-
on the light and electron micrograph can be selected (Fig. 9). In
ical information [147]. An other exciting development is combin-
the 2 points alignment scheme, we select the very centre of the
ing NMR chemical data with electron microscopy and localisation
hole and a corner of the number 1. In case the images are mirrored,
of an isotope-labelled precursors by nanoSIMS technology [148].
the light micrograph can be flipped to match the orientation of the 13
C labelled glucose was fed to animals and the metabolites could
grid in the electron microscope.
be identified by NMR. After high-resolution mapping using elec-
Then the software rotates and stretches the light micrograph to
tron microscopy the distribution of the isotope was localised
match the 2 points of the electron micrograph, which aligns the
within cells of brain and liver tissue to study the glycogen metab-
light micrograph (or the entire project when imaged in the
olism [148].
CorrSight) with the current grid position in the SEM. After the
The ideal situation would be to detect a diseased area in the
coarse alignment there might be a displacement in the range of
body of a living person, e.g., by MRI or X-ray computed tomogra-
1.5–5.5 lm (Fig. 10). To fine align an individual section to compen-
phy, take a biopsy and narrow down the area by light microscopy
sate for small shifts and distortions that may occur during the dry-
until high resolution imaging by electron microscopy. The morpho-
ing process of the section, the alignment can be repeated at higher
logical data could be complemented by functional data gathered by
magnification. The software also has a 1 point alignment option for
NMR, nanoSIMS or mass spectroscopy.
X, Y translational shifts or a 3 point alignment to better compen-
sate for distortions.
For fine alignment the layers can be shifted and rotated with the Acknowledgments
radio buttons in the ‘fine alignment’ window (Fig. 11, lower left
corner) until precise overlap is achieved. The authors would like to thank Dr. J.M. Mateos and Dr. J. Ster
With the overlay of the fluorescence signal and the electron for brain tissue with Lucifer yellow labelled neurons and Dr. A.
micrograph the fluorescently labelled neuron could unequivocally Volterra for brain tissue with GPF labelled astrocytes. W.
Blanchard provided the images in Figs. 1 and 4. We are especially
C. Loussert Fonta, B.M. Humbel / Archives of Biochemistry and Biophysics 581 (2015) 98–110 109

indebted to B. Lich, C. Mathisen, Dr. M. Langhorst, Dr. A. de Marco [41] A. Manninen, P. Verkade, S. Le Lay, J. Torkko, M. Kasper, J. Füllekrug, K.
Simons, Mol. Cell. Biol. 25 (2005) 10087–10096.
and Dr. L. Hekking, I. Gestmann, FEI Company, for the fruitful col-
[42] P. Verkade, J. Microsc. 230 (2008) 317–328.
laboration with the Maps software. Dr. H. Schwarz and Dr. C. [43] M. Grabenbauer, W.J.C. Geerts, J. Fernadez-Rodriguez, A. Hoenger, A.J. Koster,
Wurm are acknowledged for their collaboration on on-section T. Nilsson, Nat. Methods 2 (2005) 857–862.
labelling (Fig. 2) and M. van der Werff and S. Gerrits for their sup- [44] A.A. Mironov, R.S. Polishchuk, G.V. Beznoussenko, Methods Cell Biol. 88
(2008) 83–95.
port during the writing of the article. We also thank P. Brey for crit- [45] A.A. Mironov, G.V. Beznoussenko, J. Microsc. 235 (2009) 308–321.
ical reading the manuscript. The STEM work was made possible by [46] C.-L. Forestier, C. Machu, C. Loussert, P. Pescher, G.F. Späth, Cell Host Microbe
the financial support of the Faculty of Biology and Medicine of the 9 (2011) 319–330.
[47] C. Loussert, C.-L. Forestier, B.M. Humbel, Methods Cell Biol. 111 (2012) 59–73.
University of Lausanne, Switzerland, and the Swiss National [48] S.S. Biel, K. Kawaschinski, K.-P. Wittern, U. Hintze, R. Wepf, J. Microsc. 212
Science Foundation, R’Equip grant 316030_128692. (2003) 91–99.
[49] M.S. Lucas, M. Günthert, P. Gasser, F. Lucas, R. Wepf, Methods Cell Biol. 111
(2012) 325–356.
References [50] K. Wilke, K. Wick, F.J. Keil, K.-P. Wittern, R. Wepf, S.S. Biel, Exp. Dermatol. 17
(2008) 73–80.
[1] M. Osborn, R.E. Webster, K. Weber, J. Cell Biol. 77 (1978) R27–R34. [51] T.J. Deerinck, M.E. Martone, V. Lev-Ram, D.P.L. Green, R.Y. Tsien, D.L. Spector,
[2] R.E. Webster, M. Osborn, K. Weber, Exp. Cell Res. 117 (1978) 47–61. S. Huang, M.H. Ellisman, J. Cell Biol. 126 (1994) 901–910.
[3] S.W. Hell, M. Dyba, S. Jakobs, Curr. Opin. Neurobiol. 14 (2004) 599–609. [52] B.M. Humbel, M.D.M. de Jong, W.H. Müller, A.J. Verkleij, Microsc. Res. Tech. 42
[4] R. Henriques, C. Griffiths, E.H. Rego, M.M. Mhlanga, Biopolymers 95 (2011) (1998) 43–48.
322–331. [53] E. Brown, P. Verkade, Protoplasma 244 (2010) 91–97.
[5] E. Betzig, G.H. Patterson, R. Sougrat, O.W. Lindwasser, S. Olenych, J.S. [54] R.E. Pagano, M.A. Sepanski, O.C. Martin, J. Cell Biol. 109 (1989) 2067–2079.
Bonifacino, M.W. Davidson, J. Lippincott-Schwartz, H.F. Hess, Science 313 [55] Y.-D. Stierhof, in: A. Cavalier, D. Spehner, B.M. Humbel (Eds.), Handbook for
(2006) 1642–1645. Cryo-Preparation Methods for Electron Microscopy, CRC Press Inc., Boca
[6] M.J. Rust, M. Bates, X. Zhuang, Nat. Methods 3 (2006) 793–796. Raton, USA, 2008, pp. 588–616.
[7] S. Modla, K. Czymmek, Micron 42 (2011) 773–792. [56] K. Pohl, Y.-D. Stierhof, Microsc. Res. Tech. 42 (1998) 59–65.
[8] J. Roth, M. Bendayan, E. Carlemalm, W. Villiger, M. Garavito, J. Histochem. [57] X. Shu, V. Lev-Ram, T.J. Deerinck, Y. Qi, E.B. Ramko, M.W. Davidson, Y. Jin,
Cytochem. 29 (1981) 663–671. M.H. Ellisman, R.Y. Tsien, PLoS Biol. 9 (2011) e1001041.
[9] E. Carlemalm, R.M. Garavito, W. Villiger, J. Microsc. 126 (1982) 123–143. [58] G. Gaietta, T.J. Deerinck, S.R. Adams, J. Bouwer, O. Tour, D.W. Laird, G.E.
[10] B.L. Armbruster, E. Kellenberger, E. Carlemalm, W. Villiger, R.M. Garavito, J.A. Sosinsky, R.Y. Tsien, M.H. Ellisman, Science 296 (2002) 503–507.
Hobot, R. Chiovetti, J.D. Acetarin, in: J.P. Revel, T. Barnard, G.H. Haggis (Eds.), [59] R.Y. Tsien, Annu. Rev. Biochem. 67 (1998) 509–544.
The Science of Biological Specimen Preparation 1983, SEM Inc., AMF O’Hare, [60] R.Y. Tsien, Integr. Biol. 2 (2010) 77–93.
IL 60666, 1983, pp. 77–81. [61] M.G. Paez-Segala, M.G. Sun, G. Shtengel, S. Viswanathan, M.A. Baird, J.J.
[11] J.D. Acetarin, E. Carlemalm, W. Villiger, J. Microsc. 143 (1986) 81–88. Macklin, R. Patel, J.R. Allen, E.S. Howe, G. Piszczek, H.F. Hess, M.W. Davidson,
[12] B.G. Timms, Am. J. Anat. 175 (1986) 267–275. Y. Wang, L.L. Looger, Nat. Methods (2014), http://dx.doi.org/10.1038/
[13] G.R. Newman, J.A. Hobot, J. Histochem. Cytochem. 35 (1987) 971–981. nmeth.3225.
[14] J.A. Hobot, G.R. Newman, Scanning Microsc. Suppl. 5 (1991) S27–S41. [62] J. Stinchcombe, H. Nomoto, D. Cutler, C. Hopkins, J. Cell Biol. 131 (1995)
[15] C. Scala, G. Cenacchi, C. Ferrari, G. Pasquinelli, P. Preda, G.C. Manara, J. 1387–1401.
Histochem. Cytochem. 40 (1992) 1799–1804. [63] M. Grabenbauer, Methods Cell Biol. 111 (2012) 117–138.
[16] J.J. Bogers, H.A. Nibbeling, A.M. Deelder, E.A. Van Marck, J. Histochem. [64] C. Meißlitzer-Ruppitsch, C. Röhrl, J. Neumüller, M. Pavelka, A. Ellinger, J.
Cytochem. 44 (1996) 43–48. Microsc. 235 (2009) 322–335.
[17] G. Goping, G.A.J. Kuijpers, R. Vinet, H.B. Pollard, J. Histochem. Cytochem. 44 [65] C. Meißlitzer-Ruppitsch, M. Vetterlein, H. Stangl, S. Maier, J. Neumüller, M.
(1996) 289–295. Freissmuth, M. Pavelka, A. Ellinger, Histochem. Cell Biol. 130 (2008) 407–419.
[18] B. Humbel, T. Marti, M. Müller, Beitr. Elektronenmikroskop. Direktabb. Oberfl. [66] D. Bishop, I. Nikić, M. Brinkoetter, S. Knecht, S. Potz, M. Kerschensteiner, T.
16 (1983) 585–594. Misgeld, Nat. Methods 8 (2011) 568–570.
[19] B. Humbel, M. Müller, in: M. Müller, R.P. Becker, A. Boyde, J.J. Wolosewick [67] J.H. Luft, J. Biophys. Biochem. Cytol. 9 (1961) 409–414.
(Eds.), The Science of Biological Specimen Preparation 1985, SEM Inc., AMF [68] A.M. Glauert, R.H. Glauert, J. Biophys. Biochem. Cytol. 4 (1958) 191–194.
O’Hare, 1986, pp. 175–183. [69] A.R. Spurr, J. Ultrastruct. Res. 26 (1969) 31–43.
[20] B.M. Humbel, H. Schwarz, in: A.J. Verkleij, J.L.M. Leunissen (Eds.), Immuno- [70] H. Kushida, J. Electron Microsc. 12 (1963) 71–73.
Gold Labeling in Cell Biology, CRC Press, Boca Raton, 1989, pp. 115–134. [71] W. Stäubli, J. Cell Biol. 16 (1963) 197–201.
[21] H. Schwarz, B.M. Humbel, Scanning Microsc. Suppl. 3 (1989) 57–64. [72] M. Rosenberg, P. Bartl, J. Leško, J. Ultrastruct. Res. 4 (1960) 298–303.
[22] K.T. Tokuyasu, J. Cell Biol. 57 (1973) 551–565. [73] B.E. Causton, Proc. RMS 16 (1981) 265–267.
[23] K.T. Tokuyasu, S.J. Singer, J. Cell Biol. 71 (1976) 894–906. [74] B.E. Causton, in: J.M. Polak, I.M. Varndell (Eds.), Immunolabelling for Electron
[24] K.T. Tokuyasu, Histochem. J. 12 (1980) 381–403. Microscopy, Elsevier Science Publishers, Amsterdam, 1984, pp. 29–36.
[25] G. Griffiths, K. Simons, G. Warren, K.T. Tokuyasu, Methods Enzymol. 96 (1983) [75] B.E. Causton, in: M. Müller, R.P. Becker, A. Boyde, J.J. Wolosewick (Eds.), The
466–485. Science of Biological Specimen Preparation 1985, SEM Inc., AMF O’Hare, 1986,
[26] W. Liou, H.J. Geuze, J.W. Slot, Histochem. Cell Biol. 106 (1996) 41–58. pp. 209–214.
[27] J.W. Slot, H.J. Geuze, Nat. Protoc. 2 (2007) 2480–2491. [76] I.R. Gibbons, Nature 184 (1959) 375–376.
[28] T. Kurth, H. Schwarz, S. Schneider, P. Hausen, Cell Tissue Res. 286 (1996) 1– [77] N. Matsko, M. Müller, J. Struct. Biol. 152 (2005) 92–103.
12. [78] L. Wang, B.M. Humbel, E.W. Roubos, Brain Res. Protoc. 15 (2005) 155–163.
[29] H. Schwarz, in: H.A. Calderón Benavides, M.J. Yacamán, L.F. Jiménez, J.B. [79] G.R. Newman, J.A. Hobot, Resin Microscopy and On-
Kouri (Eds.), Electron Microscopy 1998, ICEM 14, Institute of Physics Section Immunocytochemistry, Springer-Verlag, Berlin, Heidelberg, New
Publishing, Bristol, Philadelphia, 1998, pp. 865–866. York, 2001.
[30] H. Schwarz, B.M. Humbel, Methods Mol. Biol. 1117 (2014) 559–592. [80] K. Luby-Phelps, G. Ning, J. Fogerty, J.C. Besharse, J. Histochem. Cytochem. 51
[31] J.M. Robinson, T. Takizawa, A. Pombo, P.R. Cook, J. Histochem. Cytochem. 49 (2003) 271–274.
(2001) 803–808. [81] K. Bell, S. Mitchell, D. Paultre, M. Posch, K. Oparka, Plant Physiol. 161 (2013)
[32] T. Takizawa, J.M. Robinson, Placenta 24 (2003) 557–565. 1595–1603.
[33] T. Takizawa, J.M. Robinson, J. Nippon Med. Sch. 71 (2004) 306–307. [82] U. Riehle, Über die Vitrifizierung verdünnter wässriger Lösungen, Federal
[34] G. Vicidomini, M.C. Gagliani, M. Canfora, K. Cortese, F. Frosi, C. Santangelo, P.P. Institute of Technology (ETH), Zürich, 1968.
Di Fiore, P. Boccacci, A. Diaspro, C. Tacchetti, Traffic 9 (2008) 1828–1838. [83] M. Müller, H. Moor, in: J.P. Revel, T. Barnard, G.H. Haggis (Eds.), Science of
[35] H. Pluk, D.J. Stokes, B. Lich, B. Wieringa, J.A.M. Fransen, J. Microsc. 233 (2009) Biological Specimen Preparation 1983, SEM Inc., AMF O’Hare, 1984, pp. 131–
353–363. 138.
[36] M.A. Karreman, A.V. Agronskaia, A.J. Verkleij, F.F.M. Cremers, H.C. Gerritsen, [84] H. Moor, in: R.A. Steinbrecht, K. Zierold (Eds.), Cryotechniques in Biological
B.M. Humbel, Biol. Cell 101 (2009) 287–299. Electron Micoscopy, Springer-Verlag, Berlin, Heidelberg, 1987, pp. 175–191.
[37] A.V. Agronskaia, J.A. Valentijn, L.F. van Driel, C.T.W.M. Schneijdenberg, B.M. [85] D. Studer, M. Michel, M. Wohlwend, E.B. Hunziker, M.D. Buschmann, J.
Humbel, P.M.P. van Bergen en Henegouwen, A.J. Verkleij, A.J. Koster, H.C. Microsc. 179 (1995) 321–332.
Gerritsen, J. Struct. Biol. 164 (2008) 183–189. [86] D. Studer, W. Graber, A. Al-Amoudi, P. Eggli, J. Microsc. 203 (2001) 285.
[38] C. Loussert Fonta, A. Leis, C. Mathisen, D.S. Bouvier, W. Blanchard, A. Volterra, [87] A. Van Harreveld, J. Crowell, S.K. Malhotra, J. Cell Biol. 25 (1965) 117–137.
B. Lich, B.M. Humbel, J. Struct. Biol. 189 (2015) 53–61. [88] H. Fernández-Morán, Science 129 (1959) 1284–1285.
[39] A. Sartori, R. Gatz, F. Beck, A. Rigort, W. Baumeister, J.M. Plitzko, J. Struct. Biol. [89] H. Fernández-Morán, Ann. N. Y. Acad. Sci. 85 (1960) 689–713.
160 (2007) 135–145. [90] M. Müller, T. Marti, S. Kriz, in: P. Brederoo, W. de Priester (Eds.), Proc. 7th Eur.
[40] F.G.A. Faas, M. Bárcena, A.V. Agronskaia, H.C. Gerritsen, K.B. Moscicka, A.A. Congr. Electron Microsc., The Hague, 1980, pp. 720–721.
Diebolder, L.F. Van Driel, R.W.A.L. Limpens, E. Bos, R.B.G. Ravelli, R.I. Koning, [91] W. Kukulski, M. Schorb, S. Welsch, A. Picco, M. Kaksonen, J.A.G. Briggs,
A.J. Koster, J. Struct. Biol. 181 (2013) 283–290. Methods Cell Biol. 111 (2012) 235–257.
110 C. Loussert Fonta, B.M. Humbel / Archives of Biochemistry and Biophysics 581 (2015) 98–110

[92] S.J. Nixon, R.I. Webb, M. Floetenmeyer, N. Schieber, H.P. Lo, R.G. Parton, Traffic [123] C. Hagen, S. Werner, S. Carregal-Romero, A.N. Malhas, B.G. Klupp, P.
10 (2009) 131–136. Guttmann, S. Rehbein, K. Henzler, T.C. Mettenleiter, D.J. Vaux, W.J. Parak, G.
[93] W. Kukulski, M. Schorb, S. Welsch, A. Picco, M. Kaksonen, J.A.G. Briggs, J. Cell Schneider, K. Grünewald, Ultramicroscopy 146 (2014) 46–54.
Biol. 192 (2011) 111–119. [124] Y. Fukuda, N. Schrod, M. Schaffer, L.R. Feng, W. Baumeister, V. Lucic,
[94] C.J. Peddie, K. Blight, E. Wilson, C. Melia, J. Marrison, R. Carzaniga, M.-C. Ultramicroscopy 143 (2014) 15–23.
Domart, P. O’Toole, B. Larijani, L.M. Collinson, Ultramicroscopy (2014), http:// [125] S. Jones, S.K. Chapman, P.R. Crocker, G. Carson, D.A. Levison, J. Clin. Pathol. 35
dx.doi.org/10.1016/j.ultramic.2014.1002.1001i. (1982) 425–429.
[95] S. Watanabe, A. Punge, G. Hollopeter, K.I. Willig, R.J. Hobson, M.W. Davis, S.W. [126] M.A. Karreman, A.V. Agronskaia, E.G. van Donselaar, K. Vocking, F. Fereidouni,
Hell, E.M. Jorgensen, Nat. Methods 8 (2011) 80–84. B.M. Humbel, C.T. Verrips, A.J. Verkleij, H.C. Gerritsen, J. Struct. Biol. 180
[96] S. Watanabe, E.M. Jorgensen, Methods Cell Biol. 111 (2012) 283–306. (2012) 382–386.
[97] P. Walther, A. Ziegler, J. Microsc. 208 (2002) 3–10. [127] H. Iijima, H. Minoda, Y. Arai, Y. Kaneko, K. Nagayama, An electron-photon
[98] C. Buser, P. Walther, J. Microsc. 230 (2008) 268–277. hybrid microscope for on-line CLEM, Gordon Research Conference on 3DEM,
[99] D. Studer, M. Chiquet, E.B. Hunziker, J. Struct. Biol. 117 (1996) 81–85. Il Ciocco, Italy, 2010.
[100] D.A. Lennette, Am. J. Clin. Pathol. 69 (1978) 647–648. [128] I. Solomonov, D. Talmi-Frank, Y. Milstein, S. Addadi, A. Aloshin, I. Sagi, Sci.
[101] J. Rodriguez, F. Deinhardt, Virology 12 (1960) 316–317. Rep. 4 (2014) 5987. 5910.1038/srep05987.
[102] K.L. McDonald, R.I. Webb, J. Microsc. 243 (2011) 227–233. [129] T. Kinoshita, Y. Mori, K. Hirano, S. Sugimoto, K.-I. Okuda, S. Matsumoto, T.
[103] W. Bernhard, J. Ultrastruct. Res. 27 (1969) 250–265. Namiki, T. Ebihara, M. Kawata, H. Nishiyama, M. Sato, M. Suga, K.
[104] E.J. Behrman, in: J.P. Revel, T. Barnard, G.H. Haggis (Eds.), The Science of Higashiyama, K. Sonomoto, Y. Mizunoe, S. Nishihara, C. Sato, Microsc.
Biological Specimen Preparation 1983, SEM Inc., AMF O’Hare, IL 60666, 1984, Microanal. 20 (2014) 469–483.
pp. 1–5. [130] H. Nishiyama, M. Koizumi, K. Ogawa, S. Kitamura, Y. Konyuba, Y. Watanabe,
[105] P. Maupin-Szamier, T.D. Pollard, J. Cell Biol. 77 (1978) 837–853. N. Ohbayashi, M. Fukuda, M. Suga, C. Sato, Ultramicroscopy 147 (2014) 86–
[106] D.L. White, S.B. Andrews, J.W. Faller, R.J. Barrnett, Biochim. Biophys. Acta 436 97.
(1976) 577–592. [131] N. Liv, I. Lazić, P. Kruit, J.P. Hoogenboom, Ultramicroscopy 143 (2014) 93–99.
[107] B.M. Humbel, in: A. Cavalier, D. Spehner, B.M. Humbel (Eds.), Handbook for [132] N. Liv, A.C. Zonnevylle, A.C. Narvaez, A.P.J. Effting, P.W. Voorneveld, M.S.
Cryo-Preparation Methods for Electron Microscopy, CRC Press Inc., Boca Lucas, J.C. Hardwick, R.A. Wepf, P. Kruit, J.P. Hoogenboom, PLoS ONE 8 (2013)
Raton, USA, 2008, pp. 320–341. e55707.
[108] Y.-D. Stierhof, E. van Donselaar, H. Schwarz, B.M. Humbel, in: A. Cavalier, D. [133] A.C. Zonnevylle, R.F.C. van Tol, N. Liv, A.C. Narvaez, A.P.J. Effting, P. Kruit, J.P.
Spehner, B.M. Humbel (Eds.), Handbook for Cryo-Preparation Methods for Hoogenboom, J. Microsc. 252 (2013) 48–70.
Electron Microscopy, CRC Press Inc., Boca Raton, USA, 2008, pp. 343–365. [134] C.J. Peddie, N. Liv, J.P. Hoogenboom, L.M. Collinson, Methods Cell Biol. 124
[109] M. Perkovic, M. Kunz, U. Endesfelder, S. Bunse, C. Wigge, Z. Yu, V.-V. (2014) 363–389.
Hodirnau, M.P. Scheffer, A. Seybert, S. Malkusch, E.M. Schuman, M. [135] K.D. Micheva, S.J. Smith, Neuron 55 (2007) 25–36.
Heilemann, A.S. Frangakis, J. Struct. Biol. 186 (2014) 205–213. [136] D. Oberti, M.A. Kirschmann, R.H.R. Hahnloser, Front. Neurosci. 5 (2011) 1–8.
[110] K.T. Tokuyasu, A.H. Dutton, B. Geiger, S.J. Singer, Proc. Natl. Acad. Sci. U.S.A. 78 [137] W.-J. Wang, H.G. Tay, R. Soni, G.S. Perumal, M.G. Goll, F.P. Macaluso, J.M.
(1981) 7619–7623. Asara, J.D. Amack, M.-F.B. Tsou, Nat. Cell Biol. 15 (2013) 591–601.
[111] K. Zierold, Scanning Electron Microsc. (1982) 1205–1214. [138] M. Husain, C. Thomas, M. Kirschmann, D. Oberti, R. Hahnloser, Microsc.
[112] J. Dubochet, A.W. McDowall, B. Menge, E.N. Schmid, K.G. Lickfeld, J. Bacteriol. Microanal. 17 (2011) 242–243.
155 (1983) 381–390. [139] C.L. Schwartz, V.I. Sarbash, F.I. Ataullakhanov, J.R. McIntosh, D. Nicastro, J.
[113] P. Frederik, W.M. Busing, W.M.A. Hax, J. Microsc. 126 (1982) RP1–RP2. Microsc. 227 (2007) 98–109.
[114] M.F. Wendt-Gallitelli, P. Stohr, H. Wolburg, W. Schlote, Scanning Electron [140] L.F. Van Driel, J.A. Valentijn, K.M. Valentijn, R.I. Koning, A.J. Koster, Eur. J. Cell
Microsc. (1980) 499–509. Biol. 88 (2009) 669–684.
[115] A. Al-Amoudi, L.P.O. Norlen, J. Dubochet, J. Struct. Biol. 148 (2004) 131–135. [141] C. Nolte, M. Matyash, T. Pivneva, C.G. Schipke, C. Ohlemeyer, U.-K. Hanisch, F.
[116] B. Zuber, M. Haenni, T. Ribeiro, K. Minnig, F. Lopes, P. Moreillon, J. Dubochet, J. Kirchhoff, H. Kettenmann, Glia 33 (2001) 72–86.
Bacteriol. 188 (2006) 6652–6660. [142] M.T. Otten, D.J. Stenzel, D.R. Cousens, B.M. Humbel, J.L.M. Leunissen, Y.-D.
[117] J. Dubochet, A. Al-Amoudi, C. Bouchet-Marquis, M. Eltsov, B. Zuber, in: A. Stierhof, W.M. Busing, Scanning 14 (1992) 282–289.
Cavalier, D. Spehner, B.M. Humbel (Eds.), Handbook for Cryo-Preparation [143] A.E. Yakushevska, M.N. Lebbink, W.J.C. Geerts, L. Spek, E.G. van Donselaar,
Methods for Electron Microscopy, CRC Press Inc., Boca Raton, USA, 2008, pp. K.A. Jansen, B.M. Humbel, J.A. Post, A.J. Verkleij, A.J. Koster, J. Struct. Biol. 159
255–289. (2007) 381–391.
[118] A. Sartori, R. Gatz, F. Beck, A. Kossel, A.P. Leis, W. Baumeister, J.M. Plitzko, [144] E.A. Smith, B.P. Cinquin, M. Do, G. McDermott, M.A. Le Gros, C.A. Larabell,
Microsc. Microanal. 11 (2005) 16–17. Ultramicroscopy 143 (2014) 33–40.
[119] H. Iijima, Y. Fukuda, Y. Arai, S. Terakawa, N. Yamamoto, K. Nagayama, J. [145] R. Carzaniga, M.-C. Domart, E. Duke, L.M. Collinson, Methods Cell Biol. 124
Struct. Biol. 185 (2014) 107–115. (2014) 151–178.
[120] W.E. Moerner, M. Orrit, Science 283 (1999) 1670–1676. [146] D.B. Carlson, J. Gelb, V. Palshin, J.E. Evans, Microsc. Microanal. 19 (2013) 22–
[121] M. Gruska, O. Medalia, W. Baumeister, A. Leis, J. Struct. Biol. 161 (2008) 384– 29.
392. [147] S. Roudeau, A. Carmona, L. Perrin, R. Ortega, Anal. Bioanal. Chem. 406 (2014)
[122] A. Rigort, F.J.B. Bäuerlein, A. Leis, M. Gruska, C. Hoffmann, T. Laugks, U. Böhm, 6979–6991.
M. Eibauer, H. Gnaegi, W. Baumeister, J.M. Plitzko, J. Struct. Biol. 172 (2010) [148] Y. Takado, G. Knott, B.M. Humbel, S. Escrig, M. Mojgan, A. Meibom, A.
169–179. Comment, Nanomed. NBM 11 (2015) 239–245.

You might also like