<<

Scanning Confocal MicroscopyREVIEW 127

Principles and Practices of Confocal Stephen W. Paddock*

Abstract The laser scanning confocal (LSCM) is an essential tool for many biomedical imaging appli- cations at the level of the light microscope. The basic principles of and the evolution of the LSCM into today’s sophisticated instruments are outlined. The major imaging modes of the LSCM are introduced including single optical sections, multiple wavelength images, three-dimensional reconstruc- tions, and living cell and tissue sequences. Practical aspects of specimen preparation, image collection, and image presentation are included along with a primer on troubleshooting the LSCM for the novice. Index Entries: Confocal microscopy; laser scanning; light microscopy.

1. Introduction field light microscope (theoretical maximum The major application of confocal microscopy resolution of 0.2 µm), but not as great as that in in the biomedical sciences is for imaging either the transmission (0.1 nm), it fixed or living tissues that have been labeled with has bridged the gap between these two commonly one or more fluorescent probes. When these used techniques. samples are imaged using a conventional light The method of image formation in a confocal microscope, fluorescence in the specimen in focal microscope is fundamentally different from that planes away from the region of interest interferes in a conventional wide-field microscope where with resolution of structures in focus, especially the entire specimen is bathed in light from a mer- for those specimens that are thicker than 2 µm or cury or xenon source, and the image can be so (Fig. 1). The confocal approach provides a viewed directly by eye. In contrast, the illumina- slight increase in both lateral and axial resolution. tion in a confocal microscope is achieved by scan- It is the ability of the instrument to eliminate the ning one or more focused beams of light, usually “out-of-focus” flare from thick fluorescently from a laser, across the specimen. An image pro- labeled specimens that has caused the explosion duced by scanning the specimen in this way is in its popularity recently (1). Most modern con- called an optical section. This term refers to the focal are now relatively easy to noninvasive method of image collection by the operate and have become integral parts of instrument, which uses light rather than physical many multiuser imaging facilities. Since the means to section the specimen. The confocal resolution achieved by the LSCM is a little bet- approach has thus facilitated the imaging of liv- ter than that achieved in a conventional wide- ing specimens, enabled the automated collection

*Author to whom all correspondence and reprint requests should be addressed. Department of Molecular , University of Wisconsin, 1525 Linden Drive, Madison, Wisconsin 53706, USA, E-mail: [email protected]

Molecular Biotechnology 2000 Humana Press Inc. All rights of any nature whatsoever reserved. 1073–6085/2000/16:2/127–149/$15.75

MOLECULAR BIOTECHNOLOGY 127 Volume 16, 2000 128 Paddock

Fig. 1. Conventional epifluorescence image (A) compared with a confocal image (B) of a similar region of a whole mount of a butterfly (Precis coenia) pupal wing stained with propidium iodide. Note the improved resolu- tion of the nuclear detail (B).

of three-dimensional data in the form of Z-series, by Minsky, and all of the modern confocal imag- and improved the images of multiple labeled ing systems employ the principle of confocal specimens (1). imaging that he patented in 1957 (5). Emphasis has been placed on the laser scan- In Minsky’s original confocal microscope the ning confocal microscope (LSCM) throughout point source of light is produced by a pinhole this article because it is currently the instrument placed in front of a zirconium arc source. The of choice for most biomedical research applica- point of light is focused by an objective lens into tions, and it is therefore most likely to be the the specimen, and light that passes through it is instrument first encountered by the novice user. focused by a second objective lens at a second Several alternative designs of confocal instru- pinhole, which has the same focus as the first pin- ments occupy specific niches within the biologi- hole, i.e., it is confocal with it. Any light that cal imaging field (2). Optical sections can be passes through the second pinhole strikes a low- produced using other, nonconfocal, methods. For noise photomultiplier, which produces a signal example, using , which calculates that is directly proportional to the brightness of the out-of-focus information in an image and the light. The second pinhole prevents light from removes it digitally (3), and multiple photon above or below the plane of focus from striking imaging (4), which uses the same method of scan- the photomultiplier. This is the key to the confo- ning as the LSCM but uses a laser that only cal approach, namely the elimination of out-of- excites the fluorochromes that are imaged in the focus light or “flare” in the specimen by spatial optical section itself. filtering. Minsky also described a reflected light version of the microscope that uses a single objec- 2. Evolution of the Confocal Approach tive lens and a dichromatic mirror arrangement. The development of confocal microscopes This is the basic configuration of most modern was driven largely by a desire to image biologi- confocal systems used for fluorescence imaging cal events as they occur in vivo. The invention (Fig. 2). of the confocal microscope is usually attributed In order to build an image, the focused spot of to Marvin Minsky, who built a working micro- light must be scanned across the specimen in scope in 1955 with the goal of imaging neural some way. In Minsky’s original microscope the networks in unstained preparations of living beam was stationary and the specimen itself was brains. Details of the microscope, and of its devel- moved on a vibrating stage. This optical arrange- opment can be found in an informative memoir ment has the advantage of always scanning on the

MOLECULAR BIOTECHNOLOGY Volume 16, 2000 Laser Scanning Confocal Microscopy 129

image quality in his microscope was not very impressive because of the quality of the oscillo- scope display and not because of lack of resolu- tion achieved with the microscope itself. It is clear that the technology was not available to Minsky in 1955 to fully demonstrate the poten- tial of the confocal approach especially for imag- ing biological structures. According to Minsky, this is perhaps a reason why confocal microscopy did not immediately catch on with the biological community, who were, (as they are now) a highly demanding and fickle group when it came to the quality of their images. After all, at the time they could quite easily view and photograph their brightly stained and colorful histological tissue sec- tions using light microscopes with excellent optics. In modern confocal microscopes the image is either built up from the output of a photo- multiplier tube or captured using a digital charge coupled device (CCD) , directly pro- cessed in a computer imaging system, and then displayed on a high-resolution video monitor and recorded on modern hard copy devices, with Fig. 2. The light path in a typical LSCM is based upon that of a conventional reflected-light wide-field spectacular results. epifluorescence microscope, but with pinholes placed The optics of the light microscope have not in front of the light source (the point source is now a changed drastically in decades since the final laser) and in front of the . The pinhole at resolution achieved by the instrument is governed the light source, the focused point in the specimen, by the wavelength of light, the objective lens and and the pinhole in front of the detector are all confocal properties of the specimen itself. However, the with one another. associated technology and the dyes used to add contrast to the specimens have been improved sig- optical axis, which can eliminate any lens defects. nificantly over the past 20 yr. The confocal However, for many biological specimens, move- approach is a direct result of a renaissance in ment of the specimen can cause them to wobble light microscopy that has been fueled largely by and distort, which results in a loss of resolution in advancements in modern technology. Several the image. Moreover, it is impossible to perform major technological advances that would have various manipulations such as microinjection of benefited Minsky’s confocal design are now fluorescently labeled probes when the specimen available (and affordable) to biologists. These is moving. advances include: Finally an image of the specimen has to be pro- 1. Stable multiwavelength for brighter duced. A real image is not formed in Minsky’s point sources of light. original microscope but rather the output from 2. More efficiently reflecting mirrors. the photodetector is translated into an image of 3. Sensitive low-noise . the region-of-interest. In Minsky’s original design 4. Fast microcomputers with image processing the image was built up on the screen of a military capabilities. surplus oscilloscope with no facility for hard 5. Elegant software solutions for analyzing the copy. Minsky admitted at a later date that the images.

MOLECULAR BIOTECHNOLOGY Volume 16, 2000 130 Paddock

6. High-resolution video displays and digital of confocal instrumentation have been covered printers. elsewhere (2), but briefly there are two funda- 7. Brighter and more stable fluorescent probes. mentally different methods of beam scanning: multiple-beam scanning or single-beam scan- These technologies have been developed inde- ning. The most popular way is currently single- pendently, and since 1955, they have slowly been beam scanning, which is typified by the LSCM. incorporated into modern confocal imaging sys- Here the scanning is most commonly achieved tems. For example, Shinya Inoue and Robert by computer-controlled galvanometer driven Allen first effectively applied digital image pro- mirrors (1 frame/s), or in some systems, by an cessing to biological imaging in the early 1980s acoustooptical device for faster scanning rates at Woods Hole. Their “video-enhanced micro- (near video rates). The alternative is to scan the scopes” enabled an apparent increase in resolu- specimen with multiple beams (almost real time) tion of structures using digital enhancement of usually using some form of spinning Nipkow images captured using a low-light-level silicon- disc. The forerunner of these systems was the intensified-target (SIT) video camera mounted on tandem scanning microscope (TSM) and subse- a light microscope and connected to a digital quent improvements to the design have become image processor. Cellular structures such as the more efficient for collecting images from flu- microtubules, which are just beyond the theo- orescently labeled specimens. retical resolution of the light microscope, were There are currently two viable alternatives to imaged using differential interference contrast confocal microscopy that produce optical sections (DIC) optics and enhanced using such digital in technically different ways. These are decon- methods as background subtraction and contrast volution (3) and multiple photon imaging (4), and enhancement in real time. These techniques are as with confocal imaging they are based on a reviewed in a landmark book titled Video Micros- conventional light microscope. Deconvolution is copy by Shinya Inoue, which has been recently a computer-based method that calculates and updated with Ken Spring, and provides an excel- removes the out-of-focus information from a fluo- lent primer for the principles and practices of rescence image. The deconvolution algorithms modern light microscopy (6). and the computers themselves are now fast Confocal microscopes are usually classified by enough to make this technique a practical option the method in which the specimens are scanned. for imaging. Multiple photon microscopy uses a Minsky’s original design was a stage scanning scanning system that is identical to that of the system driven by a primitive tuning fork arrange- LSCM but there is no need for the pinhole since ment that was rather slow to build an image. Stage the laser only excites at the point of focus in the scanning confocal microscopes have evolved into specimen. This makes the technique more practi- instruments that are used traditionally for materi- cal for imaging living tissue (4). als science applications such as the microchip industry. Systems based upon this principle have 3. The Laser Scanning Confocal Microscope recently become popular in biomedical applica- The LSCM is built around a conventional light tions for screening arrays of fluorescently labeled microscope, and uses a laser rather than a lamp DNA oligonucleotides on glass wafer supports or for a light source, sensitive “gene chips” (7). detectors (PMTs), and a computer to control the An alternative to moving the specimen is to scanning mirrors and to facilitate the collection scan the beam across a stationary one, which is and display of the images. The images are subse- more practical for imaging biological samples, quently stored using computer media and ana- and is the basis of many systems that have lyzed using a plethora of computer software either evolved into the confocal imaging systems that using the computer of the confocal system or a are so prevalent today. The more technical aspects second computer (Fig. 3).

MOLECULAR BIOTECHNOLOGY Volume 16, 2000 Laser Scanning Confocal Microscopy 131

Fig. 3. Information flow in a laser scanning confocal imaging system.

In the LSCM, illumination and detection are these embryos are impossible to image after the confined to a single, -limited, point in two-cell stage using conventional epifluores- the specimen. This point is focused in the speci- cence microscopy because as cell numbers rise, men by an objective lens, and scanned across it the overall volume of the embryo remains approx- using some form of scanning device. Points of imately the same, which means that increased light from the specimen are detected by a fluorescence from the more and more closely PMT, which is positioned behind a pinhole, and packed cells out of the focal plane of interest the output from the PMT is built into an image interferes with image resolution. by the computer (Fig. 2). Specimens are usually When he investigated the confocal micro- labeled with one or more fluorescent probes, or scopes available to him at the time, White discov- unstained specimens can be viewed when the ered that no system existed that would satisfy his light reflected back from the specimen is detected imaging needs. The technology consisted of the at the PMT. stage scanning instruments, which tended to be One of the more commercially successful slow to produce images (approx 10 s for one full LSCMs was designed by White, Amos, Durbin, frame image), and the multiple-beam micro- and Fordham (8) in order to tackle a fundamental scopes, which were difficult to align at that time problem in , namely, imag- and the fluorescence images were extremely dim, ing specific macromolecules in fluorescently if not invisible without extremely long labeled embryos. Many of the structures inside times! Using available lasers, electronics, and

MOLECULAR BIOTECHNOLOGY Volume 16, 2000 132 Paddock

digital image processors, White and his col- many laboratories are purchasing the systems as leagues designed and built a LSCM that was suit- shared instruments in preference to electron able for conventional epifluorescence microscopy microscopes despite the reduced resolution, which of biological specimens, for example, fluores- is between the conventional light microscope and cently labeled embryos, that has since evolved the electron microscope. The advantage of confo- with improvements in technology into an instru- cal microscopy lies within its great number of ment that is now used in a plethora of biomedical applications and its relative ease for producing applications. extremely high-quality digital images from speci- In a landmark paper that captured the attention mens prepared for the light microscope. of the community by the quality of The first generation LSCMs were tremen- the images published in it (9), White et al. com- dously wasteful of photons in comparison to the pared images collected from the same specimens new microscopes. The early systems worked well using conventional wide-field epifluorescence for fixed specimens but tended to kill living speci- microscopy and their LSCM. Rather than physi- mens unless extreme care was taken to preserve cally cutting sections of multicellular embryos, the viability of specimens on the stage of the their LSCM produced “optical sections” that were microscope by minimizing the exposure to laser thin enough to resolve structures of interest and light. Nevertheless, the microscopes produced were free of much of the out-of-focus fluores- such good images of fixed specimens that confo- cence that had previously contaminated their cal microscopy was fully embraced by the bio- images. This technological advance allowed them logical imagers. to take full advantage of the specificity of immu- Improvements have been made at all stages of nofluorescence labeling to follow changes in the the imaging process in the subsequent generations cytoskeleton in cells of early embryos at a higher of instruments including the addition of more resolution than was possible using conventional stable lasers, more efficiently reflecting mirrors, epifluorescence microscopy. more sensitive low noise photodetectors, and Simply adjusting the diameter of a pinhole in improved digital imaging systems (Fig. 3). The front of the photodetector could vary the thick- new instruments are much improved ergonom- ness of the optical sections (Fig. 2). This optical ically so that alignment is easier to achieve (if at path has proven to be extremely flexible for imag- all necessary), the choice of different filter com- ing biological structures as compared with some binations, which is now controlled by software, other designs that employ fixed diameter pin- motorized filter wheels and electronic filters, is holes. The image can be zoomed with no loss of much easier to achieve, and multiple fluoro- resolution simply by decreasing the region of the chromes can be imaged either simultaneously by specimen that is scanned by the mirrors, and plac- frame or sequentially on a line-by-line basis (1). ing the scanned information into the same size of Furthermore, it is easier to manipulate the images digital memory or framestore. This imparts a using improved, more reliable software that has range of magnifications to a single objective lens, been developed over the ten or so years of experi- and is extremely useful when imaging rare events ence with the LSCM, and using faster computers when changing a lens may risk losing the region with more hard disk space and random access of interest. memory (RAM). This microscope and several other LSCMs 4. Confocal Imaging Modes developed during the same time period, were the forerunners of the sophisticated instruments that 4.1. Single Optical Sections are now available to biomedical researchers The optical section is the basic image unit of from several commercial vendors (10). There has the confocal microscope. Data are collected from been a tremendous explosion in the popularity of fixed and stained samples in single-, double-, confocal microscopy over the past 10 yr. Indeed triple-, or multiple-wavelength modes (Fig. 4).

MOLECULAR BIOTECHNOLOGY Volume 16, 2000 Laser Scanning Confocal Microscopy 133

Fig. 4. Single optical sections collected simultaneously using a single krypton argon laser at three different excitation wavelengths—488 nm, 568 nm, and 647 nm—of a fruit fly third instar wing imaginal disc immunofluorescently labeled with antibody probes for three genes involved with patterning the developing wing; (A) vestigial (fluorescein 496 nm); (B) apterous (lissamine rhodamine 572 nm) and (C) CiD (cyanine 5 649 nm); with a grey-scale image of the three images merged (D).

The resulting images are in register with each 4.2. Time-Lapse and Live Cell Imaging other and portray an accurate representation of Time-lapse imaging refers to the collection of the specimen, as long as an objective lens that is single optical sections at preset points in time, and corrected for chromatic aberration is used. Regis- it uses the improved resolution of the LSCM for tration can usually be relatively easily restored us- studies of the dynamics of living cells (Fig. 5). Im- ing digital methods. The time of image collection aging living tissues is perhaps an order of magni- will also depend on the size of the image and the tude more difficult than imaging fixed ones using speed of the computer; for example, a typical 8- the LSCM (Table 1). For successful live-cell im- bit image of 768 by 512 pixels in size will occupy aging extreme care must be taken to maintain the approx 0.3 MB. Using most LSCMs it takes cells in a healthy state on the stage of the micro- approx 1 s to collect a single optical section, scope throughout the imaging process (11), and to although several such sections are usually digi- use the minimum laser exposure possible because tally averaged in order to improve the signal-to- photodamage from the laser beam can accumulate noise ratio (6). over multiple scans. Antioxidants such as ascorbic

MOLECULAR BIOTECHNOLOGY Volume 16, 2000 134 Paddock

Fig. 5. Time lapse imaging of a living fruit fly embryo injected with Calcium green (A–D). The larger outline of a dividing cell is shown—arrow in (D). One method of showing change in distribution of the fluorescent probe over time on a journal page is to merge a regular image of one time point (F) with a reversed contrast image of a second time point (G) to give a composite image (H). The same technique can be used by merging different colored images from different time points.

acid can be added to the medium to reduce oxygen chamber to maintain the carbon dioxide balance from excited fluorescent molecules, which can in the medium, whereas other cells such as insect cause free radicals to form and kill the cells. cells are usually perfectly happy at room tempera- A series of preliminary control experiments is ture in a relatively large volume of medium. Many usually necessary in order to assess the effects of experimental problems can be avoided by choos- light exposure on the fluorescently labeled cells. ing a cell type that is more amenable to imaging A postimaging test of viability should ideally be with the LSCM. The photon efficiency of most performed; for example, an embryo should con- modern confocal systems has been improved tinue its normal development after imaging. Each significantly over the early models, which were cell type has its own specific requirements for life; tremendously wasteful of photons, and when for example, mammalian cells will require some coupled with the availability of brighter objective stage heating device, and perhaps a perfusion lenses (including high NA water immersion lenses),

MOLECULAR BIOTECHNOLOGY Volume 16, 2000 Laser Scanning Confocal Microscopy 135

Table 1 tems can also be used for imaging fast physiologi- Different Considerations for Imaging Fixed cal events. The multiple-photon approach is now and Living Cells with the LSCM the method of choice for imaging living cells, Fixed Cells Living Cells however (4). Limits of illumination Fading of Phototoxicity and fading 4.3. Z-Series and Three-Dimensional Imaging of dye A Z-series is a sequence of optical sections Antifade reagent Phenylenediamine, NO! etc. collected at different levels from a specimen Mountant Glycerol (n = 1.51) Water (n = 1.33) (Fig. 6). Z-series are collected by correlating the Highest NA lens 1.4 1.2 movement of the fine focus of the microscope Time per image Unlimited Limited by with image collection usually using a computer- speed of controlled stepping motor to move the stage of phenomenon; the microscope by preset distances (Fig. 3). This light is relatively easily accomplished using a macro sensitivity of specimen program that collects an image, moves the micro- Signal averaging Yes No scope stage (focus) by a predetermined distance, Resolution Wave optics Photon statistics collects a second image, moves the microscope stage (focus), and carries on in this way until sev- eral images through the region of interest have been collected. Often two or three images are and brighter and less phototoxic dyes, live-cell extracted from such a Z-series and digitally pro- confocal analysis has become much more of a jected to highlight some specific cells. It is also practical option. relatively easy to display a Z-series as a montage The bottom line is to use the least amount of of images (Fig. 6). These programs are standard laser power possible for imaging and to collect features of most of the commercially available the images quickly in order to reduce the time of imaging systems. exposure of the specimen to laser light. The pin- Z-series are ideal for further processing into a hole may be opened wider than for fixed samples three-dimensional (3D) representation of the to speed up the imaging process and to collect the specimen using volume visualization techniques signal from all of the available photons for later (13). This approach is now used to elucidate the image processing, for example, deconvolution relationships between the 3D structure and func- may be used to improve the images. tion of tissues, because it can be conceptually Many physiological events take place faster difficult to visualize complex interconnected than the image-acquisition speed of most LSCMs, structures from a series of 200 or more optical which is typically on the order of a single frame sections taken through a structure with the LSCM. per second. Faster scanning LSCMs that use an Care must be taken to collect the images at the acoustooptical device and a slit to scan the speci- correct Z-step of the motor in order to reflect the men rather than the slower galvanometer-driven actual depth of the specimen in the image. Since point scanning systems are marketed with physi- the Z-series produced with the LSCM are in per- ological imaging in mind. These designs have the fect register (assuming the specimen itself does advantage of good spatial resolution coupled with not move during the period of image acquisition) good temporal resolution, i.e., full screen resolu- and are in a digital form, they can relatively eas- tion 30 frames per s (near video rate). Using the ily be processed into a 3D representation of the point scanning LSCMs good temporal resolution specimen (Fig. 7). is achieved by scanning a much-reduced area. There is sometimes confusion about what is Here frames at full spatial resolution are collected meant by optical section thickness. This usually more infrequently (12). The disk scanning sys- refers to the thickness of the section of the sample

MOLECULAR BIOTECHNOLOGY Volume 16, 2000 136 Paddock

Fig. 6. A Z-series of optical sections collected from a fruit fly embryo labeled with the antibody designated 22C10, which stains the peripheral nervous system.

collected with the microscope and not to the step 4.4. Four-Dimensional Imaging sizes taken by the stepper motor, which is set up by Time-lapse sequences of Z-series can also be the operator. In some cases these can be the same collected from living preparations using the LSCM however, and may be a source of the confusion. to produce four-dimensional (4D) data sets, i.e., The series of optical sections from a time-lapse three spatial dimensions X, Y, and Z with time as run can also be processed into a 3D representa- the fourth dimension. Such series can be viewed tion of the data set so that time is the Z-axis. This using a 4D-viewer program, stereo pairs of each approach is useful as a method for visualizing time point can be constructed and viewed as a physiological changes during development. For movie or a 3D reconstruction at each time point is example, calcium dynamics have been character- subsequently processed (16,17). ized in sea urchin embryos (14). A simple method for displaying 3D information is by color coding 4.5. X-Z Imaging optical sections at different depths. This can be An X-Z section is a profile of the specimen, and achieved by assigning a color (usually red, green, it can either be produced directly from the speci- or blue) to sequential optical sections collected at men using the LSCM by scanning a single line at various depths within the specimen. The colored different Z depths under the control of the stepper images from the Z-series are then merged and motor (Fig. 8) or indirectly in a 3D reconstruction colorized using an image manipulation program program by extracting the X-Z profile from a such as Adobe Photoshop (15). Z-series of optical sections (13).

MOLECULAR BIOTECHNOLOGY Volume 16, 2000 Laser Scanning Confocal Microscopy 137

Fig. 7. A (A) single optical section compared with a (B) Z-series projection of a fruit fly peripheral nervous system stained with the 22C10 antibody.

4.6. Reflected-Light Imaging method of imaging has the advantage that photo- Unstained preparations can also be viewed with bleaching is not a problem especially for living the LSCM using reflected- (backscattered) light samples. Some of the probes tend to attenuate imaging. This is the mode used in all of the early the laser beam, and in some LSCMs there can be confocal instruments (Fig. 9). In addition, speci- a reflection from optical elements in the micro- men can be labeled with probes that reflect light scope. The problem can be solved using polar- such as immunogold or silver grains (18). This izers or by imaging away from the artifact, and

MOLECULAR BIOTECHNOLOGY Volume 16, 2000 138 Paddock

Fig. 8. X-Z imaging; the laser is scanned across a single line at different Z depths—black line in (A) and an X- Z image of all the scanned lines was constructed by the confocal imaging system (B). Note that the butterfly wing epithelium is made up of two epithelial layers, and note that the fluorescence intensity drops off deeper into the specimen.

off the optical axis. The reflection artifact is not the spatial and temporal components of the migra- a problem using the slit- or multiple-beam scan- tion of labeled cells within an unlabeled population ning systems. of cells can be mapped over hours or even years (19). A real-color transmitted-light detector has been 4.7. Transmitted Light Imaging introduced into those systems that scan three Any form of light-microscope image, includ- channels simultaneously. The detector collects ing phase contrast, DIC, polarized light or dark the transmitted signal in the red, the green, and field, can be collected using a transmitted light the blue channels through red, green, and blue fil- detector (Fig. 8). This is a device that collects the ters, respectively, and builds the real-color image light passing through the specimen and the signal in a similar way to some color digital (6). from it is transferred to one of the PMTs in the This device is useful to pathologists who are used scan head via a fiber optic. Since confocal epi- to viewing real colors in bright-field mode of their fluorescence images and transmitted-light images stained preparations, and also to combine them are collected simultaneously using the same exci- tation beam, image registration is preserved, so with confocal images taken of the same region of that the precise localization of labeled cells within the specimen in fluorescence mode. the tissues is preserved when the images are merged using digital methods. 4.8. Correlative Microscopy It is often informative to collect a transmitted, The aim of correlative or integrated microscopy nonconfocal image of a specimen and to merge such is to collect information from the same region of a a transmitted light image with one or more confocal specimen using more than one microscopic tech- fluorescence images of labeled cells. For example, nique. Confocal microscopy can be used in tandem

MOLECULAR BIOTECHNOLOGY Volume 16, 2000 Laser Scanning Confocal Microscopy 139

Fig. 9. Reflected-light and transmission imaging: Interference reflection microscopy in the LSCM demon- strates cell substratum contacts in black around the cell periphery (A); confocal systems are used extensively in the materials sciences - here the surface of an audio CD is shown (B); and (C) through (E) is an in situ hybridiza- tion of HIV infected blood cells. The silver grains can be clearly seen in the reflected light confocal image (C) and in the transmitted light dark-field image (D) and bright-field image (E). Note the false positive from the dust particle—arrow in (D).

with transmission electron microscopy (TEM). For conventional wide field microscope. A good start- example, the distribution of microtubules within ing point for the development of a new protocol fixed tissues has been imaged using the LSCM, and for the confocal microscope therefore is with a the same region was imaged in the TEM using protocol for preparing the samples for conven- eosin as a fluorescence marker in the LSCM and as tional light microscopy, and to later modify it to an electron-dense marker in the electron micro- the confocal instrument if necessary. Most of the scope (20). Reflected light imaging and the TEM methods for preparing specimens for the conven- have also been used in correlative microscopy to tional light microscope were developed to reduce image cell substratum contacts (21). the amount of out-of-focus fluorescence. The con- focal system undersamples the fluorescence in a 5. Specimen Preparation and Imaging thick sample as compared with a conventional More details of specimen preparation and the epifluorescence light microscope, with the result confocal methods used to image them are avail- that samples may require increased staining times able elsewhere (22–25). Most of the protocols for or concentrations for confocal analysis, and may confocal imaging are based upon those developed appear to be overstained in the light microscope. over many years for preparing samples for the Moreover, whole mounts may be imaged success-

MOLECULAR BIOTECHNOLOGY Volume 16, 2000 140 Paddock

fully in the LSCM where it was only practical to relatively transparent and therefore the laser beam view sections using a conventional wide field will penetrate further into it (approx 200 µm) than light microscope. into unfixed skin (approx 10 µm), which is rela- The illumination in a typical laser scanning tively opaque and therefore scatters more light. The confocal system is extremely bright, although tissue can act as a neutral density filter and attenu- millions of points are scanned per second. For ate the laser beam. Many fixation protocols incor- example, a typical scan speed is one point per 1.6 porate some form of clearing agent, which will µs so that the average illumination at any one increase the transparency of the specimen. point is relatively moderate, and generally less If problems do occur with depth penetration of than a conventional epifluorescence light micro- the laser light into the specimen, then thick sections scope. Many protocols include an antibleaching can be cut using a microtome; usually fixed speci- agent that protects the fluorophore from the mens but also slices of living brain have been cut bleaching effects of the laser beam. It is advisable using a vibratome, and imaged successfully. The to use the lowest laser power that is practical for specimen can also be removed from the slide, imaging in order to protect the fluorochrome, and inverted, and remounted, although this is often it may be possible to omit the antibleaching agents messy, and usually not too successful. Dyes that are when using many of the more modern instruments. excited at longer wavelengths, for example, cya- The major application of the confocal micro- nine 5, can be used to collect images from a little scope is for improved imaging of thicker spe- deeper within the specimen than those dyes excited cimens although the success of the approach at shorter wavelengths (26). Here the resolution depends on the specific properties of the speci- is slightly reduced in comparison to that attained men. Some simple ergonomic principles apply: with images collected at shorter wavelengths. for example, the specimen must physically fit on Multiple photon imaging allows images to be col- the stage of the microscope and the area of inter- lected from more deeply within specimens (4). est should be within the working distance of the lens. For example, a high-resolution lens such as 5.1. The Objective Lens a 60× NA 1.4 has a working distance of 170 µm, The choice of objective lens for confocal inves- whereas a 20× NA 0.75 has a working distance of tigation of a specimen is extremely important 660 µm. This means that resolution may have to (27). This is because the (NA) be compromised in order to reach the region of of the lens, which is a measure of its light collect- interest and to prevent squashing the specimen ing ability, is related to optical section thickness and risking damage to the lens. and to the final resolution of the image. Basically, It is often necessary to take steps to preserve the higher the NA, the thinner the optical section. the 3D structure of the specimen for confocal The optical section thickness for the 60× (NA analysis using some form of spacer between the 1.4) objective with the pinhole set at 1 mm (closed) slide and the coverslip, for example, a piece of is on the order of 0.4 µm, and for a 16× (NA 0.5) coverslip or plastic fishing line. When living objective, again with the pinhole at 1 mm, the specimens are the subject of study, it is usually optical section thickness is around 1.8 mm. Open- necessary to mount them in some form of cham- ing the pinhole (or selecting a pinhole of increased ber that will provide all of the essentials for life diameter) will further increase the optical section on the stage of the microscope, and will also allow thickness (Table 2). The vertical resolution is access to the specimen by the objective lens for never as good as lateral resolution (Fig. 8). For imaging without deforming the specimen. example, for a 60× NA 1.4 objective lens the hori- Properties of the specimen such as opacity and zontal resolution is around 0.2 µm and the verti- turbidity can influence the depth that the laser cal resolution is around 0.5 µm. Additional factors beam may penetrate into it. For example, unfixed include chromatic aberration especially for imag- and unstained corneal epithelium of the eye is ing multilabel specimens at different wavelengths

MOLECULAR BIOTECHNOLOGY Volume 16, 2000 Laser Scanning Confocal Microscopy 141

Table 2 Table 3 Optical Section Thickness (in µm) Important Properties of Microscope Objectives for Different Objective Lenses Using for Confocal Imaginga The BioRad MRC600 Laser Scanning Property Objective 1 Objective 2 Confocal Microscope Design plan-apochromat CF-fluor DL Objective Pinhole Magnification 60 20 Closed Open Numerical aperture 1.4 0.75 Magnification NA (1 mm) (7 mm) Coverslip thickness 170 µm 170 µm 60× 1.40 0.4 1.9 Working distance 170 µm 660 µm 40× 1.30 0.6 3.3 Tube length 160 mm 160 mm 25× 0.80 1.4 7.8 Medium oil dry 20× 0.75 1.8 10.0 Contrast technique None or DIC Ph3 4× 0.20 20.0 100.0 Color correction Best Good Flatness of field Best Fair UV transmission None Excellent and flatness of field are of importance when aAn aid for choosing the correct lens for imaging. Objec- choosing an objective lens for confocal imaging. tive 1 would be more suited for high-resolution imaging of The lenses with the highest numerical apertures fixed cells, whereas Objective 2 would be better for imaging a are generally the highest magnifications (and the living preparation. most expensive), so that a compromise is often struck between the area of the specimen to be (Fig. 10). However, when possible, a higher NA scanned and the maximum achievable resolution lens should be used for the best resolution. for the area (Table 3). For example, when imag- Many instruments have an adjustable pinhole. ing Drosophila embryos and imaginal discs a 4× Opening the pinhole gives a thicker optical sec- lens (NA 0.2) is used to locate the specimen on tion and reduced resolution but it is often neces- the slide, a 16× (NA 0.5) lens for imaging whole sary to give more detail within the specimen or to embryos, and a 40× (NA 1.2) or 60× (NA 1.4) allow more light to the photodetector. As the pin- lens for resolving individual cell nuclei within hole is closed down the section thickness and embryos and imaginal discs. In contrast for but- brightness decreases, and resolution increases up terfly work where the structures of interest are to a certain pinhole diameter, when resolution much larger, the 4× lens is extremely useful for does not increase but brightness continues to imaging whole wing imaginal discs in a single decrease. This point is different for each objec- image, and for cellular resolution a 40× or a 60× tive lens (28). objective lens is used (Fig. 10). Some micro- scopes have the facility to view large fields at high 5.2. Probes For Confocal Imaging resolution using an automated X-Y stage that can The synthesis of novel fluorescent probes for move around the specimen collecting images into improved localization con- a montage. Such montages can also be built tinues to influence the development of confocal manually and pasted together digitally. instrumentation (29). Fluorochromes have been A useful feature of most LSCMs is the ability introduced over the years that have their excita- to zoom an image with no loss of resolution using tion and emission spectra more closely matched the same objective lens. This is achieved simply to the wavelengths delivered by the lasers sup- by decreasing the area of the specimen scanned plied with most commercial LSCMs (Table 4). by the laser by controlling the scanning mirrors Improved probes that can be conjugated to anti- and by placing the information from the scan into bodies continue to appear. For example, the cya- the same area of frame store or computer memory. nine dyes are alternatives to more established In this way, several magnifications are imparted dyes; cyanine 3 is a brighter alternative to rhod- onto a single lens without moving the specimen amine and cyanine 5 is useful in triple-label strat-

MOLECULAR BIOTECHNOLOGY Volume 16, 2000 142 Paddock

Fig. 10. Different objective lenses and zooming using the same lens. The 4× lens (A) is useful for viewing the entire butterfly fifth instar wing imaginal disc although the 16× lens (B) gives more nuclear detail of the Distal- less stain. The 40× lens gives even more exquisite nuclear detail (C), and zoomed by progressive increments (D, E, and F). This is achieved by scanning smaller regions of the same area of the specimen, and in this way, several magnifications are imparted onto the same objective lens.

egies. More recently, the brighter Alexa dyes have developed for imaging specimens prepared by been introduced by Molecular Probes (29). FISH, for example, the tyramide amplification Fluorescence in situ hybridization (FISH) is system (30). In addition, brighter probes are now an important approach for imaging the distri- available as counterstains for imaging the total bution of fluorescently labeled DNA and RNA DNA in nuclei and isolated chromosomes using sequences in cells. Improved protocols have been the LSCM. For example, the dimeric nucleic acid

MOLECULAR BIOTECHNOLOGY Volume 16, 2000 Laser Scanning Confocal Microscopy 143

Table 4 gies in order to locate antigens of interest to spe- Peak Excitation and Emission Wavelengths cific compartments in the cell (24). For example, of Some Commonly Used using a combination of phalloidin and a nuclear and Nuclear Counterstains dye with the antigen of interest in a triple-label- Excitation Emission ing scheme, the labeled protein can be mapped to Dye Max (nm) Max (nm) a specific cellular compartment. In addition, anti- Coumarin 350 440 bodies to proteins of known distribution or func- GFP 470 508 tion in cells, for example, antitubulin, is a useful FITC 496 518 tool for mapping in multilabel experiments. Bodipy 503 511 When imaging living cells, it is most important CY3 554 568 Tetramethylrhodamine 554 576 to be aware of the effects of adding fluorochromes Lissamine rhodamine 572 590 to the system. Such probes can be toxic to living CY3.5 581 588 cells especially when they are excited with the Texas red 592 610 laser. Adding ascorbic acid to the medium can CY5 652 672 reduce such effects. The region of the cell labeled CY5.5 682 703 can also affect its viability; for example, nuclear CY7 755 778 stains tend to have a more deleterious effect than Nuclear Dyes vital staining in the cytoplasm. One trick is to use Hoechst 33342 346 460 a fluorescent dye, which does not enter the cells DAPI 359 461 at all, but rather fluoresces in the medium sur- Acridine orange 502 526 rounding the cells. Here an outline or negative Propidium iodide 536 617 ToPro 642 661 image of the sample is produced. Probes that distinguish between living and dead cells are also available. Most of these assays rely on the fact that the membranes of dead cells are perme- dyes TOTO-1 and YOYO-1, dyes such as Hoechst able to many dyes that cannot cross them in the 33342 and DAPI, have excitation spectra (346 nm living state. Such probes include acridine or- and 359 nm) that are too short for most of the ange, and various “cell-death” kits are available lasers and mirrors that are supplied with the com- commercially (29). mercially available LSCMs, although these dyes Dyes are available that change their fluores- can be imaged using a HeNe laser/UV system (31) cence characteristics in the presence of ions such or multiple photon microscopy. The latter tech- as calcium. These dyes can be imaged using vis- nique does not require specialized UV mirrors and ible wavelengths used in the LSCM, for exam- lenses, because red light from a pulsed Ti-Sap- ple, fluo-3 or rhod-2 (14,23,24). New reporter phire laser is used for illumination (4). probes for imaging gene expression have been Many fluorescent probes are now available that introduced. For example, the jellyfish green stain specific cellular organelles and structures in fluorescent protein (GFP) allows gene expres- relatively simple protocols (24). This includes a sion and protein localization to be observed in plethora of dyes that stain nuclei (Fig. 11), mito- vivo. For example, GFP has been used to moni- chondria, the Golgi, and the endoplasmic reticu- tor gene expression in many different cell types lum, and also dyes such as the fluorescently including living Drosophila oocytes, in mamma- labeled phalloidins that label polymerized lian cells, and in plants using the 488 nm line of in cells (29). Phalloidin is used to image cell out- the LSCM for excitation (32,33). Moreover, lines in developing tissues, because the periph- spectral mutants of GFP are now available for eral actin meshwork appears as bright fluorescent multilabel experiments and are also useful for rings in the optical section (Fig. 11). These dyes avoiding problems with of liv- are extremely useful in multiple-labeling strate- ing tissues.

MOLECULAR BIOTECHNOLOGY Volume 16, 2000 144 Paddock

Fig. 11. Examples of dyes used for labeling cellular features. Cell outlines can be labeled with fluorescently labeled phalloidin (A) or nuclei using ToPro (B). Both samples are whole mounts of butterfly (Precis coenia) pupal wing imaginal discs.

5.3. Autofluorescence to use time-resolved . Autofluores- Autofluorescence can be a major source of cence can also be removed digitally by image sub- increased background when imaging some tis- traction. Although it is more often a problem, sues. Tissue autofluorescence occurs naturally in tissue autofluorescence can be utilized as a low most cell types, and especially in yeast and in level background signal for imaging overall cell plant cells, for example, chlorophyll fluoresces in morphology in multiple-labeling schemes (Fig. 4). the red. Some reagents, especially glutaraldehyde 5.4. Collecting the Images fixative, are sources of autofluorescence, and it can be reduced using borohydride treatment. The novice user can experience of confocal Autofluorescence can be avoided by using a imaging from several sources. The manual pro- wavelength for excitation that is out of the range vided with the confocal imaging system will usu- of natural autofluorescence. For example, cyanine ally include a series of simple exercises necessary 5 excitation is often out of the range of the for getting started. The person responsible for run- autofluorescence of many tissues. ning the instrument will usually be only too happy An idea of the amount of autofluorescence can to provide a short orientation session, and in most be gained by viewing an unstained specimen at multiuser facilities, the manager will usually different wavelengths, and taking note of the PMT require a short training session and demonstration settings of gain and black level together with the of a certain competence level before solo imaging laser power (Fig. 12). A relatively simple solu- is allowed. The novice should pay particular atten- tion is to collect the experimental images at set- tion to the house rules of the facility. Other useful tings above those recorded for autofluorescence. sources of information are the training courses run Autofluorescence may also be bleached out using by the confocal companies, workshops on light a quick flash at high laser power or flooding the microscopy, and various publications (24). specimen with the mercury lamp to bleach it out It is essential to be familiar with the basic opera- where the fluorescence of labeled structures is tion of the imaging system before working with ex- protected by antifade reagents. A more sophisti- perimental slides. It is usually recommended, for the cated method of dealing with autofluorescence is novice at least, to start imaging with a relatively easy

MOLECULAR BIOTECHNOLOGY Volume 16, 2000 Laser Scanning Confocal Microscopy 145

Fig. 12. Example of tissue autofluorescence in pollen grains. Note that different types of pollen fluoresce at different excitation wavelengths and some at different intensities in different channels—these images were col- lected simultaneously at the same settings of gain, black level and pinhole diameter.

specimen rather than with a more difficult experi- been granted. This is because the beam can be lost mental one. Some good test samples include paper completely, and in some instruments it may soaked in one or more fluorescent dyes or a prepara- require a service visit to rectify the situation. tion of fluorescent beads, which are both bright, and The basic practices of light microscopic tech- relatively easy specimens to image with the confo- nique should be followed at all times (6). For cal microscope. A particular favorite of mine is a example, all glass surfaces should be clean, be- slide of mixed pollen grains that autofluoresce at cause dirt and grease on coverslips and objective many different wavelengths (Fig. 12). Such slides lenses is a major cause of poor images. Care are available from biological suppliers such as Caro- should be taken to mount the specimen so that it lina Biological (www.Carolina.com) or can be eas- is within the working distance of the objective ily prepared from pollen collected from garden lens. The refractive index between the objective plants. These specimens tend to have some interest- lens and the specimen should be matched. For ing surface features and hold up well in the laser example, a coverslip of correct thickness for the beam. A relatively reliable test specimen for living objective lens should be used, especially for studies can be prepared from onion epithelium or higher-power lenses, which will require a #1 or the water plant Elodea sp., using autofluorescence #1.5 coverslip, and not a #2 coverslip. The cover- or staining with DiOC6. slip should be sealed to the slide in some way, The aim should always be to gain the best pos- and mounted flat—use nail polish for fixed speci- sible performance from the instrument, and this mens making sure that it’s dry before imaging, starts with optimal alignment, especially when and some form of nontoxic sealing agent for live imaging with older model confocal instruments. specimens, for example, a Vaseline, beeswax, and The alignment routine depends on the specific lanolin mixture works well although for viewing instrument, and is usually best performed by the living cells some form of specimen chamber is person responsible for the instrument. Alignment much preferred to a coverslip/slide arrangement. should definitely not be attempted before training Taking great care with the simple basics of clean- and permission from the microscope owner has liness at this stage can save a lot of time and effort.

MOLECULAR BIOTECHNOLOGY Volume 16, 2000 146 Paddock

One of the secrets of successful confocal imag- rest of the experimental images before applying ing is in mastering the interplay between lens NA, the equalization routine. If space on the hard disk pinhole size, and image brightness using the low- allows, it is often a good strategy to save raw est laser power possible to achieve the best image. unprocessed images alongside any processed ones. The new user should play with these parameters The image is usually saved to the hard disk of using the test specimen and several different the computer and later backed up onto a mass- objective lenses of different magnifications and storage device, for example, onto CDs. In general numerical apertures in order to get a feel for the it is advisable to collect as many images as pos- capabilities of the instrument before progressing to sible during an imaging session, and, if necessary, the experimental specimens. Try zooming using to cull out the bad ones in a later review session. the zoom function and compare these images with It is quite surprising how a seemingly unneces- those obtained using a different objective lens. sary image at first sight suddenly becomes valued The specific imaging parameters of the micro- at a later date after further review—especially scope should be set up away from the region of with one’s peers! It is much harder to prepare interest to avoid photobleaching of valuable another specimen, and often harder still to repro- regions of the specimen. This usually involves duce the exact parameters of previously prepared setting the gain and the black levels of the pho- specimens, and hard disk space should be more tomultiplier detectors together with the pinhole plentiful. size in order to achieve a balance between accept- A strategy for labeling image files should be able resolution and adequate contrast using the mapped out before imaging, and during imaging lowest laser power possible to avoid excessive many notes should be taken or placed on the photobleaching. Many instruments have color image file along with the image if this facility is tables that aid in setting the correct dynamic range available. Users should conduct a test to ensure within the image. Such look-up tables are designed that the information is accessible after imaging so that the blackest pixels, around zero, are and remember that it can be lost when the images pseudocolored green and the brightest pixels, are subsequently transferred to image manipu- around 255 in an 8-bit system, are colored red. lation programs such as NIH Image or Adobe The gain and black levels (and the pinhole) are Photoshop on other computers. It is hard to adjusted so that there are a few red and green pix- replace a well-ordered note book preferably els in the image thus ensuring the full dynamic using a table of image file names with facility range from 0 to 255 is utilized. These adjustments for comments and details of the objective lens can also be made by eye. It is not always practical and the zoom factor for calculating scale bars at to collect an image at full dynamic range because a later date. Many modern systems incorporate full laser power cannot be used or the specimen an image database that will keep track of file has uneven fluorescence so that a bright region names, location of the files and include a thumb- may obscure a dimmer region of interest in the nail of the images (34). frame. As the specimen is scanned, an image-averag- 5.5. Troubleshooting ing routine is usually employed that filters out ran- A protocol will sometimes inexplicably cease dom noise from the photomultiplier and enhances to work, and there is often an initial reflex to the constant features from labeled structures in the blame the instrument rather than the sample. A image. An image equalization routine can be applied good test is to view the sample with a conven- directly after collection of the images so that the tional epifluorescence microscope, and if some image is scaled to the full dynamic range. This fluorescence is visible by eye, then the signal routine should not be applied if measurements of should be very bright on the confocal system. If fluorescence intensity are to be made unless a the sample is visible by eye but not detected with control image is included in the same frame as the the LSCM, it might be prudent to run through

MOLECULAR BIOTECHNOLOGY Volume 16, 2000 Laser Scanning Confocal Microscopy 147

some checks of the confocal system. It is advis- for example, if the beam from a krypton/argon able to use a known test specimen rather than the laser appears blue and not white when scanning experimental one for testing the system. A digital on a multiple-label setting, then this suggests that file of an image of the test specimen should be the red line is weak. If this is the case, then the accessible to all users together with all of the anode voltage will usually be high, and can usu- parameters of its collection including laser power, ally be reduced to an acceptable level by adjust- gain, black level, pinhole diameter, zoom, and ing the mirrors on the laser (often called “walking objective lens used. the beam”—this should usually be left to the per- It is advisable to seek help from an expert who son responsible for the instrument). If it is not pos- may have prior experience of the problem. If all sible to reduce the voltage, a new or refurbished else fails, do not panic, each of the confocal com- laser may be required. panies should have a good help line whose num- Sometimes the antibody probes may have ber will usually be posted close to the microscope. degraded or need to be cleaned (36). Older speci- The golden rule is, if you are not sure of some- mens may have increased background fluo- thing, ask or at least step back from the problem rescence and bleed through caused by the before attempting to fix something. fluorochrome coming off the secondary antibody Problems with the protocols themselves are and diffusing into the tissue. Always view freshly usually caused by degradation of reagents, and a prepared specimens if at all possible. Changing series of diagnostic tests should be performed. It the concentration and/or the distribution of the is usually best to make up many of the reagents fluorochromes often helps. For example, if fluo- fresh yourself or to your recipe, or at least, “bor- rescein bleeds into the rhodamine channel, then row” them from a trusted lab mate! Antibodies switch the fluorochromes so that rhodamine is on should be aliquoted in small batches from the fro- the stronger channel because the fluorescein exci- zen stock, and stored in the refrigerator. They tation spectrum has a tail that is excited in the should only be re-used if absolutely necessary, rhodamine wavelengths. The concentration of the although this is sometimes unavoidable with secondaries can be reduced in subsequent experi- expensive or rare reagents, and often does not ments. The use of an acoustooptical tunable filter present a problem. for the excitation beams can help to fine tune the Bleed through can occur from one channel strength of each channel independently (37). into another in multiply labeled specimens. This can result from properties of the specimen 5.6. Image Processing and Publication itself or from problems with the instrument. The Confocal images are collected as digital com- causes and remedies of bleed through have been puter files, and they can usually be manipulated reviewed in much detail elsewhere (35). A good using the proprietary software provided with the test of the instrument is to view a test sample with confocal imaging system or using secondary soft- known bleed through properties using both the ware, for example, Adobe Photoshop (38). One multiple-labeling settings and the single-label set- of the most dramatically improved features of the tings. It is advisable to collect an image of the test LSCM has been in the display and analysis of specimen and record the settings of laser power, confocal images. This part of the process is gain, black level, and pin hole diameter so that extremely important because it is fine to achieve when problems do occur, one can return to these improved resolution using the LSCM but this settings with a test sample and compare the improvement is of little value if it cannot be dis- images collected with those of the stored test played and reproduced in hard copy format. images collected when the instrument was oper- Even 5 yr ago most laboratories used dark- ating in an optimal way. rooms and chemicals for their final hard copy. Additional tests include a visual inspection of Color images were even harder to reproduce the laser color and the anode voltage of the laser, because an independent printer who had little idea

MOLECULAR BIOTECHNOLOGY Volume 16, 2000 148 Paddock

of the correct color balance usually printed them. sues by collecting optical sections of the same For hard copies, images are now exported to a region or Z-series (4D imaging) in a time-lapse slide maker, a color laser printer, or a dye subli- mode (39), and the dimension of position in the mation printer for publication quality prints. Pho- specimen where several locations are imaged tographs were taken directly from the screen of sequentially using a motorized stage that can be the video monitor. Moreover, movie sequences controlled by the computer. can be published on the World Wide Web. The technology continues to improve at a rapid The quality of published images has also rate as evidenced by the introduction of practical improved drastically as most of the journals are able multiple photon imaging, which gives improved via- to accept digital images for publication. This means bility of living cells, deeper penetration into intact that the resolution displayed by the computer of the specimens, and the possibility of multiple wave- confocal imaging system is more faithfully repro- length imaging (40). Instruments are now available duced in the final published article. Many journals with several laser sources so that confocal and mul- publish their articles in CD ROM format or they are tiple photon imaging modes can be used, and when available on the World Wide Web. This means that combined with improved probes, gives a powerful means of imaging the spatial distribution and behav- the images viewed by the reader should be exactly ior of macromolecules in cells (41). the same as those collected using the confocal microscope. These technological advances are espe- Acknowledgments cially useful for color images where the intended I would like to thank Jim Williams (Fig. 4), resolution and color balance can be accurately repro- Grace Panganiban and Duc Dong (Fig. 7), Jane duced by the journals, and, theoretically, at a much Selegue (Fig. 8), Dorothy Lewis (Fig. 9 C,D,E) lower cost to the author. and Julie Gates (Fig. 11) for allowing me to image 6. Conclusions their beautiful specimens, and Sean Carroll for our continued collaborations. The LSCM has found a niche in many biomed- ical imaging laboratories, because it produces References improved images of all specimens prepared for fluo- 1. Paddock, S. W. (1999) Confocal laser scanning rescence microscopy including those labeled by microscopy. BioTechniques 27, 992–1004. immunofluorescence and FISH (24). The confocal 2. Pawley, J. B (1995) Handbook of Biological Confo- systems currently available, while they are still cal Microscopy, 2nd ed., Plenum, New York. 3. Chen, H., Hughes, D. D., Chan, T. A., Sedat, J. W., and based on Minsky’s long-expired patent and a con- Agard, D. A. (1996) IVE (Image Visualization Envi- ventional wide-field epifluorescence microscope, ronment): a software platform for all three-dimensional have been developed into sophisticated digital imag- microscopy applications. J. Struct. Biol. 116, 56–60. ing systems that are relatively easy to operate espe- 4. Potter, S. M. (1996) Vital imaging: two photons are better than one. Curr. Biol. 6, 1595–1598. cially if the user has some experience using an 5. Minsky, M. (1988) Memoir on inventing the confo- epifluorescence microscope and a microcomputer. cal scanning microscope. Scanning 10, 128–138. Today’s instruments are largely computer- 6. Inoue, S. and Spring, K. S. (1997) Video Microscopy: controlled, and they have greatly facilitated The Fundamentals, 2nd ed. Plenum, New York. the collection of multidimensional images from 7. DeRisi, J., Penland, L., Brown, P. O., Bittner, M. L., Meltzer, P. S., Ray, M. et al. (1996) Use of a cDNA fluorescently labeled specimens. These dimen- microarray to analyze gene expression patterns in sions include the X and the Y dimension of the human . Nature 14, 457– 460. optical section, the Z dimension of a through 8. White, J. G., Amos, W. B., Durbin, R., and Fordham, focus series of optical sections (Z-series), the M. (1990) Development of a confocal imaging sys- dimension of wavelength so that multiple fluoro- tem for biological epifluorescence application, in Optical Microscopy For Biology, Wiley-Liss, New chromes are imaged either simultaneously or York, pp. 1–18. sequentially in the same region of the specimen 9. White, J. G., Amos, W. B., and Fordham, M. (1987) (38), the dimension of time for imaging living tis- An evaluation of confocal versus conventional imag-

MOLECULAR BIOTECHNOLOGY Volume 16, 2000 Laser Scanning Confocal Microscopy 149

ing of biological structures by fluorescence light 25. Spector, D. L., Goldman, R., and Leinwand, L. (1998) microscopy. J. Cell Biol. 105, 41–48. Cells: A Laboratory Manual, vol. II Light Microscopy 10. Pawley, J. B. (1995) Light paths of current commer- and Cell Structure. Cold Spring Harbor Press, Cold cial confocal microscopes for biology, in Handbook Spring Harbor, NY. of Biological Confocal Microscopy, 2nd ed. (Pawley 26. Cullander, C. (1994) Imaging in the far-red with elec- J. B., ed.) Plenum, New York, pp. 581–598. tronic light microscopy: requirements and limitations. 11. Terasaki, M. and Dailey, M. E. (1995) Confocal J. Microsc. 176, 281–286. microscopy of living cells, in Handbook of Biologi- 27. Keller, H. A. (1995) Objective lenses for confocal cal Confocal Microscopy, 2nd ed. (Pawley J. B., ed.), microscopy, in Handbook of Biological Confocal Mi- Plenum, New York, pp. 327–346. croscopy, 2nd ed. (Pawley, J. B., ed.), Plenum, New 12. Cheng, H., Lederer, W. J., and Cannell, M. B. (1993) Cal- York, pp. 111–126. cium sparks: elementary events underlying excitation-con- 28 Wilson, T. (1995) The role of the pinhole in confocal traction coupling in heart muscle. Science 262, 740–744. imaging system, in Handbook of Biological Confocal 13. White, N. S. (1995) Visualization systems for multi- Microscopy, 2nd ed. (Pawley, J. B., ed.), Plenum, dimensional CLSM, in Handbook of Biological Con- New York, pp. 167–182. focal Microscopy, 2nd ed. (Pawley J.B., ed.) Plenum, 29. Haugland, R. P. (1999) Handbook of Fluorescent New York, pp. 211–254. Probes and Research Chemicals. 7th ed. Molecular 14. Stricker, S. A., Centonze, V. E., Paddock, S. W., and Probes Inc., Eugene, Oregon. (www.probes.com). Schatten, G. (1992) Confocal microscopy of fertili- 30. Wilkie, G. S. and Davis, I. (1998) High resolution and zation-induced calcium dynamics in sea urchin eggs. sensitive mRNA in situ hybridization using fluores- Dev. Biol. 149, 370–380. cent tyramide amplification. Tech. Tips Online t01458 15. Paddock, S. W., Hazen, E. J., and DeVries, P. J. (www.biomednet.com). (1997) Methods and applications of three color con- 31. Bliton, C., Lechleiter, J., and Clapham, D. E. (1993) focal imaging. BioTechniques 22, 120–126. Optical modifications enabling simultaneous confo- 16. Thomas, C. F., DeVries, P., Hardin, J., and White, J. cal imaging with dyes excited by ultra-violet and vis- G. (1996) Four dimensional imaging: computer visu- ible-wavelength light. J. Microsc. 169, 15–26. alization of 3D movements in living specimens. Sci- 32. Chalfie, M. and Kain, S. (1998) Green Fluorescent ence 273, 603–607. Protein: Properties, Applications and Protocols. 17. Mohler, W. A. and White, J. G. (1998) Stereo-4-D Wiley-Liss, New York. reconstruction and animation from living fluorescent 33. Sullivan, K. F. and Kay, S. A. (1998) Green fluorescent pro- specimens. Biotechniques 24, 1006–1012. teins. Meth. Cell Biol. Vol. 58. Academic, San Diego, CA. 18. Paddock, S. W., Mahoney, S., Minshall, M., Smith, 34. Karten, H. J. (1998) Information management of confo- L. C., Duvic, M., and Lewis, D. (1991) Improved de- cal microscopy images. Meth. Mol. Biol. 122, 403–420. tection of in situ hybridization by laser scanning con- 35. Brelje, T. C., Wessendorf, M. W., and Sorenson, R. focal microscopy. BioTechniques 11, 486–494. L. 1993. Multicolor laser scanning confocal immun- 19. Serbedzija, G. N., Bronner-Fraser, M., and Fraser, S. ofluorescence microscopy: practical applications and (1992) Vital dye analysis of cranial neural crest cell mi- Meth. Cell Biol. 38, gration in the mouse embryo. Development 116, 297–307. limitations. 98–177. 20. Deerinck, T. J., Martone, M. E., Lev-Ram, V., Green, 36. Brown, N. L. (1998) Imaging gene expression using D. P. L., Tsien R. Y., Spector, D. L., et al. (1994) antibody probes. Meth. Mol. Biol. 122, 75–91. Fluorescence photooxidation with eosin: a method 37. Sharma, D. (1999) The use of an AOTF to achieve high for high resolution immunolocalisation and in situ quality simultaneous multiple label imaging. BioRad hybridization detection for light and electron micros- Tech. Note 04. www.microscopy.bio-rad.com38. copy. J. Cell Biol. 126, 901–910. 38. Halder, G. and Paddock, S. W. (1998) Presentation of 21. Paddock, S. W. and Cooke, P. (1988) Correlated confocal images. Meth. Mol. Biol. 122, 373–384. confocal laser scanning microscopy with high-voltage 39. Bornfleth, H., Edelmann, P., Zink, D., Cremer, T., electron microscopy of focal contacts in 3T3 cells stained and Cremer. C. (1999) Quantitative motion analysis with Napthol Blue Black. EMSA Abs 46, 100–101. of subchromosomal foci in living cells using four- 22. Sheppard, C. J. R. and Shotten, D. M. (1997) Confo- dimensional microscopy. Biophys. J. 77, 2871–2886. cal laser scanning microscopy. Royal Microscopical 40. Centonze, V. E. and White, J. G. (1998) Multiphoton Society Handbook Series #38, Bios scientific pub- excitation provides optical sections from deeper lishers, Oxford, UK. within scattering specimens than confocal imaging. 23. Matsumoto, B. (2000) Cell Biological Applications Biophys. J. 75, 2015–2024. of Confocal Microscopy, 2nd ed. Methods In Cell 41. Fan, G. Y., Fujisaki, H., Miyawaki, A, Tsay, R.-K., Biology, Academic Press, San Diego, CA, in press. Tsien R. Y., and Ellisman, M. H. (1999) Video-rate 24. Paddock, S. W. (1998) Protocols In Confocal Micros- scanning two-photon excitation fluorescence micro- copy. Methods In Mol. Biol. 122. (J. Walker, ed.) scopy and ratio imaging with cameleons. Biophys. J. Humana, Totowa, NJ. 76, 2412–2420.

MOLECULAR BIOTECHNOLOGY Volume 16, 2000