You are on page 1of 29

MICROSCOPY, CONFOCAL 449

MICROSCOPY, CONFOCAL units, high throughput fiber optics, better thin-film dielec-
tric coatings, and detectors having reduced noise charac-
NATHAN S. CLAXTON teristics (1). In addition, fluorochromes that were more
THOMAS J. FELLERS carefully matched to laser excitation lines were beginning
MICHAEL W. DAVIDSON to be synthesized (13). Coupled to the rapidly advancing
The Florida State University computer processing speeds, enhanced displays, and large-
Tallahassee, Florida volume storage technology emerging in the late-1990s, the
stage was set for a virtual explosion in the number of
INTRODUCTION applications that could be targeted with laser scanning
confocal microscopy.
The technique of laser scanning and spinning disk confocal Modern confocal microscopes can be considered as com-
fluorescence microscopy has become an essential tool in pletely integrated electronic systems where the optical micro-
biology and the biomedical sciences, as well as in materials scope plays a central role in a configuration that consists of
science due to attributes that are not readily available one or more electronic detectors, a computer (for image dis-
using other contrast modes with traditional optical micro- play, processing, output, and storage), and several laser
scopy (1–12). The application of a wide array of new syn- systems combined with wavelength selection devices and a
thetic and naturally occurring fluorochromes has made it beam scanning assembly. In most cases, integration between
possible to identify cells and submicroscopic cellular compo- the various components is so thorough that the entire con-
nents with a high degree of specificity amid nonfluorescing focal microscope is often collectively referred to as a digital or
material (13). In fact, the confocal microscope is often cap- video imaging system capable of producing electronic
able of revealing the presence of a single molecule (14). images (24). These microscopes are now being employed
Through the use of multiply labeled specimens, different for routine investigations on molecules, cells, and living
probes can simultaneously identify several target molecules tissues that were not possible just a few years ago (15).
simultaneously, both in fixed specimens and living cells and Confocal microscopy offers several advantages over con-
tissues (15). Although both conventional and confocal micro- ventional widefield optical microscopy, including the abil-
scopes cannot provide spatial resolution below the diffrac- ity to control depth of field, elimination, or reduction of
tion limit of specific specimen features, the detection of background information away from the focal plane (that
fluorescing molecules below such limits is readily achieved. leads to image degradation), and the capability to collect
The basic concept of confocal microscopy was originally serial optical sections from thick specimens. The basic key
developed by Minsky in the mid-1950s (patented in 1961) to the confocal approach is the use of spatial filtering
when he was a postdoctoral student at Harvard University techniques to eliminate out-of-focus light or glare in speci-
(16,17). Minsky wanted to image neural networks in mens whose thickness exceeds the immediate plane of
unstained preparations of brain tissue and was driven focus. There has been a tremendous explosion in the popu-
by the desire to image biological events as they occur in larity of confocal microscopy in recent years (1–4,6,7), due
living systems. Minsky’s invention remained largely unno- in part to the relative ease with which extremely high
ticed, due most probably to the lack of intense light sources quality images can be obtained from specimens prepared
necessary for imaging and the computer horsepower for conventional fluorescence microscopy, and the growing
required to handle large amounts of data. Following Min- number of applications in cell biology that rely on imaging,
sky’s work, Egger and Petran (18) fabricated a multiple- both fixed and living cells and tissues. In fact, confocal
beam confocal microscope in the late-1960s that utilized a technology is proving to be one of the most important
spinning (Nipkow) disk for examining unstained brain advances ever achieved in optical microscopy.
sections and ganglion cells. Continuing in this arena, In a conventional widefield optical epi-fluorescence
Egger went on to develop the first mechanically scanned microscope, secondary fluorescence emitted by the speci-
confocal laser microscope, and published the first recogniz- men often occurs through the excited volume and obscures
able images of cells in 1973 (19). During the late-1970s and resolution of features that lie in the objective focal plane
the 1980s, advances in computer and laser technology, (25). The problem is compounded by thicker specimens (>2
coupled to new algorithms for digital manipulation of mm), which usually exhibit such a high degree of fluores-
images, led to a growing interest in confocal microscopy (20). cence emission that most of the fine detail is lost. Confocal
Fortuitously, shortly after Minsky’s patent had expired, microscopy provides only a marginal improvement in both
practical laser-scanning confocal microscope designs were axial (z; parallel to the microscope optical axis) and lateral
translated into working instruments by several investiga- (x and y; dimensions in the specimen plane) optical resolu-
tors. Dutch physicist Brakenhoff developed a scanning tion, but is able to exclude secondary fluorescence in areas
confocal microscope in 1979 (21), while almost simulta- removed from the focal plane from resulting images (26–
neously, Sheppard contributed to the technique with a 28). Even though resolution is somewhat enhanced with
theory of image formation (22). Wilson, Amos, and White confocal microscopy over conventional widefield techni-
nurtured the concept and later (during the late-1980s) ques (1), it is still considerably less than that of the trans-
demonstrated the utility of confocal imaging in the exam- mission electron microscope (TEM). In this regard, confocal
ination of fluorescent biological specimens (20,23). The first microscopy can be considered a bridge between these two
commercial instruments appeared in 1987. During the classical methodologies.
1990s, advances in optics and electronics afforded more Illustrated in Fig. 1 are a series of images that com-
stable and powerful lasers, high efficiency scanning mirror pare selected viewfields in traditional widefield and laser

Encyclopedia of Medical Devices and Instrumentation, Second Edition, edited by John G. Webster
Copyright # 2006 John Wiley & Sons, Inc.
450 MICROSCOPY, CONFOCAL

Figure 1. Comparison of widefield (upper row) and laser scanning confocal fluorescence microscopy
images (lower row). Note the significant amount of signal in the widefield images from fluorescent
structures located outside of the focal plane. (a) and (b) Mouse brain hippocampus thick section
treated with primary antibodies to glial fibrillary acidic protein (GFAP; red), neurofilaments H
(green), and counterstained with Hoechst 33342 (blue) to highlight nuclei. (c) and (d) Thick section of
rat smooth muscle stained with phalloidin conjugated to Alexa Fluor 568 (targeting actin; red),
wheat germ agglutinin conjugated to Oregon Green 488 (glycoproteins; green), and counterstained
with DRAQ5 (nuclei; blue). (e) and (f) Sunflower pollen grain tetrad autofluorescence.

scanning confocal fluorescence microscopy. A thick (16 PRINCIPLES OF CONFOCAL MICROSCOPY


mm) section of fluorescently stained mouse hippocampus
in widefield fluorescence exhibits a large amount of glare The confocal principle in epi-fluorescence laser scanning
from fluorescent structures located above and below the microscope is diagrammatically presented in Fig. 2. Coher-
focal plane (Fig. 1a). When imaged with a laser scanning ent light emitted by the laser system (excitation source)
confocal microscope (Fig. 1b), the brain thick section passes through a pinhole aperture that is situated in a
reveals a significant degree of structural detail. Likewise, conjugate plane (confocal) with a scanning point on the
widefield fluorescence imaging of rat smooth muscle fibers specimen and a second pinhole aperture positioned in front
stained with a combination of Alexa Fluor dyes produce of the detector (a photomultiplier tube). As the laser is
blurred images (Fig. 1c) lacking in detail, while the same reflected by a dichromatic mirror, and scanned across the
specimen field (Fig. 1d) reveals a highly striated topogra- specimen in a defined focal plane, secondary fluorescence
phy when viewed as an optical section with confocal emitted from points on the specimen (in the same focal
microscopy. Autofluorescence in a sunflower (Helianthus plane) pass back through the dichromatic mirror, and are
annuus) pollen grain tetrad produces a similar indistinct focused as a confocal point at the detector pinhole aperture.
outline of the basic external morphology (Fig. 1e), but The significant amount of fluorescence emission that
yields no indication of the internal structure in widefield occurs at points above and below the objective focal plane is
mode. In contrast, a thin optical section of the same grain not confocal with the pinhole (termed out-of-focus light
(Fig. 1f) acquired with confocal techniques displays a rays in Fig. 2) and forms extended Airy disks in the
dramatic difference between the particle core and the aperture plane (29). Because only a small fraction of the
surrounding envelope. Collectively, the image comparisons out-of-focus fluorescence emission is delivered through
in Fig. 1 dramatically depict the advantages of achieving the pinhole aperture, most of this extraneous light is not
very thin optical sections in confocal microscopy. The ability detected by the photomultiplier and does not contribute to
of this technique to eliminate fluorescence emission from the resulting image. The dichromatic mirror, barrier filter,
regions removed from the focal plane offsets it from tradi- and excitation filter perform similar functions to identical
tional forms of fluorescence microscopy. components in a widefield epi-fluorescence microscope (30).
MICROSCOPY, CONFOCAL 451

Figure 3. Three-channel spectral imaging laser scanning


microscope confocal scan head with SIM scanner laser port. The
Figure 2. Schematic diagram of the optical pathway and SIM laser enables simultaneous excitation and imaging of the
principal components in a laser scanning confocal microscope. specimen for photobleaching or photoactivation experiments. Also
illustrated are ports for a visible, ultraviolet (UV), and infrared (IR)
Refocusing the objective in a confocal microscope shifts the laser, as well as an arc discharge lamp port for widefield observation.
excitation and emission points on a specimen to a new
plane that becomes confocal with the pinhole apertures of In epi-illumination scanning confocal microscopy, the
the light source and detector. laser light source and photomultiplier detectors are both
In traditional widefield epi-fluorescence microscopy, the separated from the specimen by the objective, which func-
entire specimen is subjected to intense illumination from tions as a well-corrected condenser and objective combina-
an incoherent mercury or xenon arc-discharge lamp, and tion. Internal fluorescence filter components (e.g., the
the resulting image of secondary fluorescence emission can excitation and barrier filters and the dichromatic mirrors)
be viewed directly in the eyepieces or projected onto the and neutral density filters are contained within the scan-
surface of an electronic array detector or traditional film ning unit (see Fig. 3). Interference and neutral density
plane. In contrast to this simple concept, the mechanism of filters are housed in rotating turrets or sliders that can be
image formation in a confocal microscope is fundamentally inserted into the light path by the operator. The excitation
different (31). As discussed above, the confocal fluorescence laser beam is connected to the scan unit with a fiber optic
microscope consists of multiple laser excitation sources, a coupler followed by a beam expander that enables the thin
scan head with optical and electronic components, electro- laser beam wrist to completely fill the objective rear aper-
nic detectors (usually photomultipliers), and a computer ture (a critical requirement in confocal microscopy).
for acquisition, processing, analysis, and display of images. Expanded laser light that passes through the microscope
The scan head is at the heart of the confocal system and objective forms an intense diffraction-limited spot that is
is responsible for rasterizing the excitation scans, as well as scanned by the coupled galvanometer mirrors in a raster
collecting the photon signals from the specimen that are pattern across the specimen plane (point scanning).
required to assemble the final image (1,5–7). A typical scan One of the most important components of the scanning
head contains inputs from the external laser sources, unit is the pinhole aperture, which acts as a spatial filter at
fluorescence filter sets and dichromatic mirrors, a galvan- the conjugate image plane positioned directly in front of the
ometer-based raster scanning mirror system, variable pin- photomultiplier (39). Several apertures of varying dia-
hole apertures for generating the confocal image, and meter are usually contained on a rotating turret that
photomultiplier tube detectors tuned for different fluores- enables the operator to adjust pinhole size (and optical
cence wavelengths. Many modern instruments include section thickness). Secondary fluorescence collected by the
diffraction gratings or prisms coupled with slits positioned objective is descanned by the same galvanometer mirrors
near the photomultipliers to enable spectral imaging that form the raster pattern, and then passes through a
(also referred to as emission fingerprinting) followed barrier filter before reaching the pinhole aperture (40).
by linear unmixing of emission profiles in specimens The aperture serves to exclude fluorescence signals from
labeled with combinations of fluorescent proteins or fluor- out-of-focus features positioned above and below the focal
ophores having overlapping spectra (32–38). The general plane, which are instead projected onto the aperture as
arrangement of scan head components is presented in Airy disks having a diameter much larger than those
Fig. 3 for a typical commercial unit. forming the image. These oversized disks are spread over
452 MICROSCOPY, CONFOCAL

this manner, the area of interest on the specimen in a


single focal plane is excited by laser illumination from the
scanning unit.
As each scan line passes along the specimen in the lateral
focal plane, fluorescence emission is collected by the objec-
tive and passed back through the confocal optical system.
The speed of the scanning mirrors is very slow relative to the
Figure 4. Widefield versus confocal microscopy illumination speed of light, so the secondary emission follows a light path
volumes, demonstrating the difference in size between point along the optical axis that is identical to the original excita-
scanning and widefield excitation light beams. tion beam. Return of fluorescence emission through the
galvanometer mirror system is referred to as descanning
(40,42). After leaving the scanning mirrors, the fluorescence
a comparatively large area so that only a small fraction of emission passes directly through the dichromatic mirror
light originating in planes away from the focal point passes and is focused at the detector pinhole aperture. Unlike
through the aperture. The pinhole aperture also serves to the raster scanning pattern of excitation light passing over
eliminate much of the stray light passing through the optical the specimen, fluorescence emission remains in a steady
system. Coupling of aperture-limited point scanning to a position at the pinhole aperture, but fluctuates with respect
pinhole spatial filter at the conjugate image plane is an to intensity over time as the illumination spot traverses the
essential feature of the confocal microscope. specimen producing variations in excitation.
When contrasting the similarities and differences Fluorescence emission that is passed through the pin-
between widefield and confocal microscopes, it is often hole aperture is converted into an analog electrical signal
useful to compare the character and geometry of specimen having a continuously varying voltage (corresponding to
illumination utilized for each of the techniques. Traditional intensity) by the photomultiplier. The analog signal is
widefield epi-fluorescence microscope objectives focus periodically sampled and converted into pixels by an
a wide cone of illumination over a large volume of the analog-to-digital (A/D) converter housed in the scanning
specimen (41), which is uniformly and simultaneously unit or the accompanying electronics cabinet. The image
illuminated (as illustrated in Fig. 4a). A majority of the information is temporarily stored in an image frame buffer
fluorescence emission directed back toward the microscope card in the computer and displayed on the monitor. Note
is gathered by the objective (depending on the numerical that the confocal image of a specimen is reconstructed,
aperture) and projected into the eyepieces or detector. The point by point, from emission photon signals by the photo-
result is a significant amount of signal due to emitted multiplier and accompanying electronics, yet never exists
background light and autofluorescence originating from as a real image that can be observed through the micro-
areas above and below the focal plane, which seriously scope eyepieces.
reduces resolution and image contrast.
The laser illumination source in confocal microscopy is
first expanded to fill the objective rear aperture, and then LASER SCANNING CONFOCAL MICROSCOPE
focused by the lens system to a very small spot at the focal CONFIGURATION
plane (Fig. 4b). The size of the illumination point ranges
from 0.25 to 0.8 mm in diameter (depending on the Basic microscope optical system characteristics have
objective numerical aperture) and 0.5 to 1.5 mm deep at remained fundamentally unchanged for many decades
the brightest intensity. Confocal spot size is determined by due to engineering restrictions on objective design, the
the microscope design, wavelength of incident laser light, static properties of most specimens, and the fact that
objective characteristics, scanning unit settings, and the resolution is governed by the wavelength of light (1–10).
specimen (41). Figure 4 presents a comparison between the However, fluorescent probes that are employed to add
typical illumination cones of a widefield (Fig. 4a) and point contrast to biological specimens and, and other technolo-
scanning confocal (Fig. 4b) microscope at the same numer- gies associated with optical microscopy techniques, have
ical aperture. The entire depth of the specimen over a wide improved significantly. The explosive growth and develop-
area is illuminated by the widefield microscope, while the ment of the confocal approach is a direct result of a renais-
sample is scanned with a finely focused spot of illumination sance in optical microscopy that has been largely fueled by
that is centered in the focal plane in the confocal microscope. advances in modern optical and electronics technology.
In laser scanning confocal microscopy, the image of an Among these are stable multiwavelength laser systems
extended specimen is generated by scanning the focused that provide better coverage of the uv, visible, and near-
beam across a defined area in a raster pattern controlled by IR spectral regions, improved interference filters (includ-
two high speed oscillating mirrors driven with galvan- ing dichromatic mirrors, barrier, and excitation filters),
ometer motors. One of the mirrors moves the beam from sensitive low noise wide-band detectors, and far more
left to right along the x lateral axis, while the other powerful computers. The latter are now available with
translates the beam in the y direction. After each single relatively low cost memory arrays, image analysis software
scan along the x axis, the beam is rapidly transported back packages, high resolution video displays, and high quality
to the starting point and shifted along the y axis to begin a digital image printers. The flow of information through a
new scan in a process termed flyback (42). During the modern confocal microscope is presented diagrammati-
flyback operation, image information is not collected. In cally in Fig. 5 (2).
MICROSCOPY, CONFOCAL 453

computer, which also controls the scanning mirrors and/or


other devices to facilitate the collection and display of
images. After a series of images (usually serial optical
sections) has been acquired and stored on digital media,
analysis can be conducted utilizing numerous image pro-
cessing software packages available on the host or a sec-
ondary computer.

ADVANTAGES AND DISADVANTAGES OF CONFOCAL


MICROSCOPY

The primary advantage of laser scanning confocal micro-


scopy is the ability to serially produce thin (0.5–1.5 mm)
optical sections through fluorescent specimens that have a
thickness ranging up to 50 mm or more (48). The image
series is collected by coordinating incremental changes in
Figure 5. Confocal microscope configuration and information the microscope fine focus mechanism (using a stepper
flow schematic diagram. motor) with sequential image acquisition at each step.
Image information is restricted to a well-defined plane,
Although many of these technologies have been devel- rather than being complicated by signals arising from
oped independently for a variety of specifically targeted remote locations in the specimen. Contrast and definition
applications, they have been incorporated gradually into are dramatically improved over widefield techniques due to
mainstream commercial confocal microscopy systems. In the reduction in background fluorescence and improved
current microscope systems, classification of designs is signal to noise (48). Furthermore, optical sectioning elim-
based on the technology utilized to scan specimens (7). inates artifacts that occur during physical sectioning and
Scanning can be accomplished either by translating the fluorescent staining of tissue specimens for traditional
stage in the x, y, and z directions while the laser illumina- forms of microscopy. The noninvasive confocal optical sec-
tion spot is held in a fixed position, or the beam itself can tioning technique enables the examination of both living
be raster-scanned across the specimen. Because three- and fixed specimens under a variety of conditions with
dimensional (3D) translation of the stage is cumbersome enhanced clarity.
and prone to vibration, most modern instruments employ With most confocal microscopy software packages, opti-
some type of beam-scanning mechanism. cal sections are not restricted to the perpendicular lateral
In modern confocal microscopes, two fundamentally (x–y) plane, but can also be collected and displayed in
different techniques for beam scanning have been devel- transverse planes (1,5–8,49). Vertical sections in the x–z
oped. Single-beam scanning, one of the more popular and y–z planes (parallel to the microscope optical axis) can
methods employed in a majority of the commercial laser be readily generated by most confocal software programs.
scanning microscopes (43), uses a pair of computer-con- Thus, the specimen appears as if it had been sectioned in a
trolled galvanometer mirrors to scan the specimen in a plane that is perpendicular to the lateral axis. In practice,
raster pattern at a rate of approximately one frame per vertical sections are obtained by combining a series of x–y
second. Faster scanning rates (to near video speed) can be scans taken along the z axis with the software, and then
achieved using acoustooptic devices or oscillating mirrors. projecting a view of fluorescence intensity as it would
In contrast, multiple-beam scanning confocal microscopes appear should the microscope hardware have been capable
are equipped with a spinning Nipkow disk containing an of physically performing a vertical section.
array of pinholes and microlenses (44–46). These instru- A typical stack of optical sections (often termed a z
ments often use arc-discharge lamps for illumination series) through a Lodgepole Pine tree pollen grain reveal-
instead of lasers to reduce specimen damage and enhance ing internal variations in autofluorescence emission wave-
the detection of low fluorescence levels during real-time lengths is illustrated in Fig. 6. Optical sections were
image collection. Another important feature of the multiple- gathered in 1.0 mm steps perpendicular to the z axis
beam microscopes is their ability to readily capture images (microscope optical axis) using a laser combiner featuring
with an array detector, such as a charge-coupled device an argon ion (488 nm; green fluorescence), a green helium–
(CCD) camera system (47). neon (543 nm; red fluorescence), and a red helium–neon
All modern laser scanning confocal microscope designs (633 nm; fluorescence pseudocolored blue) laser system.
are centered on a conventional upright or inverted Pollen grains from this and many other species range
research level optical microscope. However, instead of between 10 and 40 mm in diameter and often yield blurred
the standard tungsten–halogen or mercury (xenon) arc- images in wide-field fluorescence microscopy (see Fig. 1c),
discharge lamp, one or more laser systems are used as a which lack information about internal structural details.
light source to excite fluorophores in the specimen. Image Although only 12 of the >36 images collected through this
information is gathered point by point with a specialized series are presented in the figure, they represent indivi-
detector, such as a photomultiplier tube or avalanche dual focal planes separated by a distance of 3 mm and
photodiode, and then digitized for processing by the host provide a good indication of the internal grain structure.
454 MICROSCOPY, CONFOCAL

Figure 6. Lodgepole pine (Pinus contorta) pollen grain optical


sections. Bulk pollen was mounted in CytoSeal 60 and imaged with
a 100 oil immersion objective (no zoom) in 1 mm axial steps. Each
image in the sequence (1–12) represents the view obtained from Figure 7. Three-dimensional volume renders from confocal
steps of 3 mm. microscopy optical sections. (a) Autofluorescence in a series of
sunflower pollen grain optical sections was combined to produce
a realistic view of the exterior surface. (b) Mouse lung tissue thick
In specimens more complex than a pollen grain, complex (16 mm) section. (c) Rat brain thick section. These specimens were each
interconnected structural elements can be difficult to dis- labeled with several fluorophores (blue, green, and red fluorescence)
cern from a large series of optical sections sequentially and the volume renders were created from a stack of 30–45 optical
acquired through the volume of a specimen with a laser sections. (d) Autofluorescence in a thin section of fern root.
scanning confocal microscope. However, once an adequate
series of optical sections has been gathered, it can be further pollen grain through 3608 for examination. Similarly, thick
processed into a 3D representation of the specimen using sections (16 mm) of lung tissue and rat brain are presented in
volume-rendering computational techniques (50–53). This Fig. 7b and 7c, respectively. These specimens were each
approach is now in common use to help elucidate the labeled with several fluorophores (blue, green, and red
numerous interrelationships between structure and func- fluorescence) and created from a stack of 30–45 optical
tion of cells and tissues in biological investigations (54). In sections. Autofluorescence in plant tissue was utilized to
order to ensure that adequate data is collected to produce a produce the model illustrated in Fig. 7d of a fern root section.
representative volume image, the optical sections should be In many cases, a composite or projection view pro-
recorded at the appropriate axial (z step) intervals so that duced from a series of optical sections provides important
the actual depth of the specimen is reflected in the image. information about a 3D specimen than a multidimensional
Most of the software packages accompanying commer- view (54). For example, a fluorescently labeled neuron
cial confocal instruments are capable of generating com- having numerous thin, extended processes in a tissue
posite and multidimensional views of optical section data section is difficult (if not impossible) to image using
acquired from z-series image stacks. The 3D software wide-field techniques due to out-of-focus blur. Confocal
packages can be employed to create either a single 3D thin sections of the same neuron each reveal portions of
representation of the specimen (Fig. 7) or a video (movie) several extensions, but these usually appear as fragmented
sequence compiled from different views of the specimen streaks and dots and lack continuity (53). Composite views
volume. These sequences often mimic the effect of rotation created by flattening a series of optical sections from the
or similar spatial transformation that enhances the appre- neuron will reveal all of the extended processes in sharp
ciation of the specimen’s 3D character. In addition, many focus with well-defined continuity. Structural and func-
software packages enable investigators to conduct measure- tional analysis of other cell and tissue sections also benefits
ments of length, volume, and depth, and specific parameters from composite views as opposed to, or coupled with, 3D
of the images, such as opacity, can be interactively altered to volume rendering techniques.
reveal internal structures of interest at differing levels Advances in confocal microscopy have made possible
within the specimen (54). multidimensional views (54) of living cells and tissues that
Typical 3D representations of several specimens exam- include image information in the x, y, and z dimensions as
ined by serial optical sectioning are presented in Fig. 7. A a function of time and presented in multiple colors (using
series of sunflower pollen grain optical sections was com- two or more fluorophores). After volume processing of
bined to produce a realistic view of the exterior surface individual image stacks, the resulting data can be dis-
(Fig. 7a) as it might appear if being examined by a scanning played as 3D multicolor video sequences in real time. Note
electron microscope (SEM). The algorithm utilized to that unlike conventional widefield microscopy, all fluoro-
construct the 3D model enables the user to rotate the chromes in multiply labeled specimens appear in register
MICROSCOPY, CONFOCAL 455

using the confocal microscope. Temporal data can be col- secondary emission gathered by the objective can be accom-
lected either from time-lapse experiments conducted over plished by several classes of photosensitive detectors (59),
extended periods or through real-time image acquisition in including photomultipliers, photodiodes, and solid-state
smaller frames for short periods of time. The potential for CCDs. In confocal microscopy, fluorescence emission is
using multidimensional confocal microscopy as a powerful directed through a pinhole aperture positioned near the
tool in cellular biology is continuing to grow as new laser image plane to exclude light from fluorescent structures
systems are developed to limit cell damage and computer located away from the objective focal plane, thus reducing
processing speeds and storage capacity improves. the amount of light available for image formation, as
Additional advantages of scanning confocal microscopy discussed above. As a result, the exceedingly low light
include the ability to adjust magnification electronically by levels most often encountered in confocal microscopy neces-
varying the area scanned by the laser without having to sitate the use of highly sensitive photon detectors that do
change objectives. This feature is termed the zoom factor, not require spatial discrimination, but instead respond
and is usually employed to adjust the image spatial resolu- very quickly with a high level of sensitivity to a continuous
tion by altering the scanning laser sampling period flux of varying light intensity.
(1,2,8,40,55). Increasing the zoom factor reduces the speci- Photomultipliers, which contain a photosensitive sur-
men area scanned and simultaneously reduces the scanning face that captures incident photons and produces a stream
rate. The result is an increased number of samples along a of photoelectrons to generate an amplified electric charge,
comparable length (55), which increases both the image are the popular detector choice in many commercial con-
spatial resolution and display magnification on the host focal microscopes (59–61). These detectors contain a criti-
computer monitor. Confocal zoom is typically employed to cal element, termed a photocathode, capable of emitting
match digital image resolution (8,40,55) with the optical electrons through the photoelectric effect (the energy of an
resolution of the microscope when low numerical aperture absorbed photon is transferred to an electron) when
and magnification objectives are being used to collect data. exposed to a photon flux. The general anatomy of a photo-
Digitization of the sequential analog image data col- multiplier consists of a classical vacuum tube in which a
lected by the confocal microscope photomultiplier (or similar glass or quartz window encases the photocathode and a
detector) facilitates computer image processing algorithms chain of electron multipliers, known as dynodes, followed
by transforming the continuous voltage stream into discrete by an anode to complete the electrical circuit (62). When
digital increments that correspond to variations in light the photomultiplier is operating, current flowing between
intensity. In addition to the benefits and speed that accrue the anode and ground (zero potential) is directly propor-
from processing digital data, images can be readily prepared tional to the photoelectron flux generated by the photo-
for print output or publication. In carefully controlled cathode when it is exposed to incident photon radiation.
experiments, quantitative measurements of spatial fluores- In a majority of commercial confocal microscopes, the
cence intensity (either statically or as a function of time) can photomultiplier is located within the scan head or an
also be obtained from the digital data. external housing, and the gain, offset, and dynode voltage
Disadvantages of confocal microscopy are limited pri- are controlled by the computer software interface to the
marily to the limited number of excitation wavelengths detector power supply and supporting electronics (7). The
available with common lasers (referred to as laser lines), voltage setting is used to regulate the overall sensitivity of
which occur over very narrow bands and are expensive to the photomultiplier, and can be adjusted independently of
produce in the UV region (56). In contrast, conventional the gain and offset values. The latter two controls are
widefield microscopes use mercury- or xenon-based arc- utilized to adjust the image intensity values to ensure that
discharge lamps to provide a full range of excitation wave- the maximum number of gray levels is included in the
lengths in the UV, visible, and near-IR spectral regions. output signal of the photomultiplier. Offset adds a positive
Another downside is the harmful nature (57) of high or negative voltage to the output signal, and should be
intensity laser irradiation to living cells and tissues, an adjusted so that the lowest signals are near the photomul-
issue that has recently been addressed by multiphoton and tiplier detection threshold (40). The gain circuit multiplies
Nipkow disk confocal imaging. Finally, the high cost of the output voltage by a constant factor so that the max-
purchasing and operating multiuser confocal microscope imum signal values can be stretched to a point just below
systems (58), which can range up to an order of magnitude saturation. In practice, offset should be applied first before
higher than comparable widefield microscopes, often limits adjusting the photomultiplier gain (8,40). After the signal
their implementation in smaller laboratories. This problem has been processed by the analog-to-digital converter, it is
can be easily overcome by cost-shared microscope systems stored in a frame buffer and ultimately displayed on the
that service one or more departments in a core facility. The monitor in a series of gray levels ranging from black (no
recent introduction of personal confocal systems has com- signal) to white (saturation). Photomultipliers with a
petitively driven down the price of low end confocal micro- dynamic range of 10 or 12 bits are capable of displaying
scopes and increased the number of individual users. 1024 or 4096 gray levels, respectively. Accompanying
image files also have the same number of gray levels.
However, the photomultipliers used in a majority of the
CONFOCAL MICROSCOPE LIGHT DETECTORS commercial confocal microscopes have a dynamic range
limited to 8 bits or 256 gray levels, which in most cases,
In modern widefield fluorescence and laser scanning con- is adequate for handling the typical number of photons
focal optical microscopy, the collection and measurement of scanned per pixel (63).
456 MICROSCOPY, CONFOCAL

Changes to the photomultiplier gain and offset levels The photomultiplier gain adjustment is utilized to elec-
should not be confused with postacquisition image proces- tronically stretch the input signal by multiplying with a
sing to adjust the levels, brightness, or contrast in the final constant factor prior to digitization by the analog-to-digital
image. Digital image processing techniques can stretch converter (40). The result is a more complete representa-
existing pixel values to fill the black-to-white display tion of gray level values between black and white, and an
range, but cannot create new gray levels (40). As a result, increase in apparent dynamic range. If the gain setting is
when a digital image captured with only 200 out of a increased beyond the optimal point, the image becomes
possible 4096 gray levels is stretched to fill the histogram grainy, but this maneuver is sometimes necessary to cap-
(from black to white), the resulting processed image ture the maximum number of gray levels present in the
appears grainy. In routine operation of the confocal micro- image. Advanced confocal microscopy software packages
scope, the primary goal is to fill as many of the gray levels ease the burden of gain and offset adjustment by using a
during image acquisition and not during the processing pseudocolor display function to associate pixel values with
stages. gray levels on the monitor. For example, the saturated
The offset control is used to adjust the background level pixels (255) can be displayed in yellow or red, while black-
to a position near 0 V (black) by adding a positive or evel pixels (0) are shown in blue or green, with intermedi-
negative voltage to the signal. This ensures that dark ate gray levels displayed in shades of gray representing
features in the image are very close to the black level of their true values. When the photomultiplier output is
the host computer monitor. Offset changes the amplitude of properly adjusted, just a few red (or yellow) and blue (or
the entire voltage signal, but since it is added to or sub- green) pixels are present in the image, indicating that the
tracted from the total signal, it does not alter the voltage full dynamic range of the photomultiplier is being utilized.
differential between the high and low voltage amplitudes Established techniques in the field of enhanced night
in the original signal. For example, with a signal ranging vision have been applied with dramatic success to photo-
from 4 to 18 V that is modified with an offset setting of 4 V, multipliers designed for confocal microscopy (63,64). Sev-
the resulting signal spans 0–14 V, but the difference eral manufacturers have collaborated to fabricate a head-
remains 14 V. on photomultiplier containing a specialized prism system
Figure 8 presents a series of diagrammatic schematics that assists in the collection of photons. The prism operates
of the unprocessed and adjusted output signal from a by diverting the incoming photons to a pathway that
photomultiplier and the accompanying images captured promotes total internal reflection in the photomultiplier
with a confocal microscope of a living adherent culture of envelope adjacent to the photocathode. This configuration
Indian Muntjac deer skin fibroblast cells treated with increases the number of potential interactions between the
MitoTracker Red CMXRos, which localizes specifically in photons and the photocathode, resulting in an increase in
the mitochondria. Figure 8a illustrates the raw confocal quantum efficiency by more than a factor of 2 in the green
image along with the signal from the photomultiplier. After spectral region, 4 in the red region, and even higher in the
applying a negative offset voltage to the photomultiplier, IR (59). Increasing the ratio of photoelectrons generated to
the signal and image appear in Fig. 8b. Note that as the the number of incoming photons serves to increase the
signal is shifted to lower intensity values, the image electrical current from the photomultiplier, and to produce
becomes darker (upper frame in Fig. 8b). When the gain a higher sensitivity for the instrument.
is adjusted to the full intensity range (Fig. 8c), the image Photomultipliers are the ideal photometric detectors for
exhibits a significant amount of detail with good contrast confocal microscopy due to their speed, sensitivity, high
and high resolution. signal/noise ratio, and adequate dynamic range (59–61).

Figure 8. Gain and offset control in confocal microscopy


photomultiplier detection units. The specimen is a living
adherent culture of Indian Muntjac deer skin fibroblast
cells treated with MitoTracker Red CMXRos. (a) The raw
confocal image (upper frame) along with the signal from
the photomultiplier. (b) Signal and confocal image after
applying a negative offset voltage to the photomultiplier.
(c) Final signal and image after the gain has been
adjusted to fill the entire intensity range.
MICROSCOPY, CONFOCAL 457

High end confocal microscope systems have several photo- utilized to provide monochromatic illumination for multi-
multipliers that enable simultaneous imaging of different spectral imaging, as well as to attenuate the beam for
fluorophores in multiply labeled specimens. Often, an addi- intensity control, and the filters are often mechanically
tional photomultiplier is included for imaging the specimen interchanged by a rotating turret mechanism. Interference
with transmitted light using differential interference or filter turrets and wheels have the disadvantages of limited
phase-contrast techniques. In general, confocal micro- wavelength selection, vibration, relatively slow switching
scopes contain three photomultipliers for the fluorescence speed, and potential image shift (70). They are also sus-
color channels (red, green, and blue; each with a separate ceptible to damage and deterioration caused by exposure to
pinhole aperture) utilized to discriminate between fluor- heat, humidity, and intense illumination, which changes
ophores, along with a fourth for transmitted or reflected their spectral characteristics over time. In addition, the
light imaging. Signals from each channel can be collected utilization of filter wheels for illumination wavelength
simultaneously and the images merged into a single profile selection has become progressively more complex and
that represents the real colors of the stained specimen. If expensive as the number of lasers being employed has
the specimen is also imaged with a brightfield contrast- increased with current applications.
enhancing technique, such as differential interference con- Rotation of filter wheels and optical block turrets intro-
trast (65), the fluorophore distribution in the fluorescence duces mechanical vibrations into the imaging and illumi-
image can be overlaid onto the brightfield image to deter- nation system, which consequently requires a time delay
mine the spatial location of fluorescence emission within for damping of perhaps 50 ms, even if the filter transition
the structural domains. itself can be accomplished more quickly. Typical filter
change times are considerably slower in practice, however,
and range on the order of 0.1–0.5 s. Mechanical imprecision
ACOUSTOOPTIC TUNABLE FILTERS IN CONFOCAL in the rotating mechanism can introduce registration
MICROSCOPY errors when sequentially acquired multicolor images are
processed. Furthermore, the fixed spectral characteristics
The integration of optoelectronic technology into confocal of interference filters do not allow optimization for different
microscopy has provided a significant enhancement in the fluorophore combinations, nor for adaptation to new fluor-
versatility of spectral control for a wide variety of fluores- escent dyes, limiting the versatility of both the excitation
cence investigations. The acoustooptic tunable filter and detection functions of the microscope. Introduction of
(AOTF) is an electrooptical device that functions as an the AOTF to confocal systems overcomes most of the filter
electronically tunable excitation filter to simultaneously wheel disadvantages by enabling rapid simultaneous elec-
modulate the intensity and wavelength of multiple laser tronic tuning and intensity control of multiple laser lines
lines from one or more sources (66). Devices of this type rely from several lasers.
on a specialized birefringent crystal whose optical proper- As applied in laser scanning confocal microscopy, one of
ties vary upon interaction with an acoustic wave. Changes the most significant benefits of the AOTF is its capability to
in the acoustic frequency alter the diffraction properties of replace much more complex and unwieldy filter mechan-
the crystal, enabling very rapid wavelength tuning, limited isms for controlling light transmission, and to apply inten-
only by the acoustic transit time across the crystal. sity modulation for wavelength discrimination purposes
An acoustooptic tunable filter designed for microscopy (67,70). The ability to perform extremely rapid adjust-
typically consists of a tellurium dioxide or quartz aniso- ments in the intensity and wavelength of the diffracted
tropic crystal to which a piezoelectric transducer is bonded beam gives the AOTF unique control capabilities. By vary-
(67–70). In response to the application of an oscillating ing the illumination intensity at different wavelengths, the
radio frequency (RF) electrical signal, the transducer gen- response of multiple fluorophores, for example, can be
erates a high frequency vibrational (acoustic) wave that balanced for optimum detection and recording (71). In
propagates into the crystal. The alternating ultrasonic addition, digital signal processors along with phase and
acoustic wave induces a periodic redistribution of the frequency lock-in techniques can be employed to discrimi-
refractive index through the crystal that acts as a trans- nate emission from multiple fluorophores or to extract low
mission diffraction grating or Bragg diffracter to deviate a level signals from background.
portion of incident laser light into a first-order beam, which A practical light source configuration scheme utilizing
is utilized in the microscope (or two first-order beams when an acoustooptic tunable filter for confocal microscopy is
the incident light is nonpolarized). Changing the frequency illustrated in Fig. 9. The output of three laser systems
of the transducer signal applied to the crystal alters the (violet diode, argon, and argon–krypton) are combined by
period of the refractive index variation, and therefore, the dichromatic mirrors and directed through the AOTF,
wavelength of light that is diffracted. The relative intensity where the first-order diffracted beam (green) is collinear
of the diffracted beam is determined by the amplitude and is launched into a single-mode fiber. The undiffracted
(power) of the signal applied to the crystal. laser beams (violet, green, yellow, and red) exit the AOTF
In the traditional fluorescence microscope configura- at varying angles and are absorbed by a beam stop (not
tion, including many confocal systems, spectral filtering illustrated). The major lines (wavelengths) produced by
of both excitation and emission light is accomplished uti- each laser are indicated (in nm) beneath the hot and cold
lizing thin-film interference filters (7). These filters are mirrors. The dichromatic mirror reflects wavelengths
limiting in several respects. Because each filter has a fixed < 525 nm and transmits longer wavelengths. Two longer
central wavelength and passband, several filters must be wavelength lines produced by the argon–krypton laser
458 MICROSCOPY, CONFOCAL

less restrictive limitations because of the lower acoustic


frequencies associated with diffraction of these longer
wavelengths.
The maximum size of an individual acoustic transducer
is constrained by performance and power requirements in
addition to the geometric limitations of the instrument
configuration, and AOTF designers may use an array of
transducers bonded to the crystal in order to increase the
effective lateral dimensions of the propagating acoustic
beam, and to enlarge the area of acoustooptic interaction
(66,67,70). The required drive power is one of the most
important variables in acoustooptic design, and generally
increases with optical aperture and for longer wave-
lengths. In contrast to acoustic attenuation, which is
reduced in the IR spectral range, the higher power required
to drive transducers for infrared AOTFs is one of the
Figure 9. Configuration scheme utilizing an AOTF for laser
intensity control and wavelength selection in confocal microscopy. greatest limitations in these devices. High drive power
levels result in heating of the crystal, which can cause
thermal drift and instability in the filter performance (66).
(568 and 648 nm) are reflected by the hot mirror, while the This is particularly a problem when acoustic power and
output of the argon laser (458, 476, 488, and 514 nm) is frequency are being varied rapidly over a large range, and
reflected by the dichromatic mirror and combined with the the crystal temperature does not have time to stabilize,
transmitted light from the argon–krypton laser. Output producing transient variations in refractive index. If an
from the violet diode laser (405 nm) is reflected by the cold application requires wavelength and intensity stability
mirror and combined with the longer wavelengths from and repeatability, the AOTF should be maintained at a
the other two lasers, which are transmitted through the constant temperature. One approach taken by equipment
mirror. manufacturers to minimize this problem is to heat the
Because of the rapid optical response from the AOTF crystal above ambient temperature, to a level at which it
crystal to the acoustic transducer, the acoustooptic inter- is relatively unaffected by the additional thermal input of
action is subject to abrupt transitions resembling a rec- the transducer drive power. An alternative solution is to
tangular rather than sinusoidal waveform (66). This house the AOTF in a thermoelectrically cooled housing
results in the occurrence of sidelobes in the AOTF that provides precise temperature regulation. Continuing
passband on either side of the central transmission peak. developmental efforts promise to lead to new materials
Under ideal acoustooptic conditions, these sidelobes should that can provide relatively large apertures combined with
be symmetrical about the central peak, with the first lobe effective separation of the filtered and unfiltered beams
having 4.7% of the central peak’s intensity. In practice, the without use of polarizers, while requiring a fraction of the
sidelobes are commonly asymmetrical and exhibit other typical device drive power.
deviations from predicted structure caused by variations in In a noncollinear AOTF, which spatially separates the
the acoustooptic interaction, among other factors. In order incident and diffracted light paths, the deflection angle (the
to reduce the sidelobes in the passband to insignificant angle separating diffracted and undiffracted light beams
levels, several types of amplitude apodization of the acoustic exiting the crystal) is an additional factor limiting the
wave are employed (66,67), including various window func- effective aperture of the device (67). As discussed pre-
tions, which have been found to suppress the highest side- viously, the deflection angle is greater for crystals having
lobe by 30–40 dB. One method that can be used in reduction greater birefringence, and determines in part the propaga-
of sidelobe level with noncollinear AOTFs is to apply spatial tion distance required for adequate separation of the dif-
apodization by means of weighted excitation of the trans- fracted and undiffracted beams to occur after exiting the
ducer. In the collinear AOTF, a different approach has been crystal. The required distance is increased for larger
employed, which introduces an acoustic pulse, apodized in entrance apertures, and this imposes a practical limit on
time, into the filter crystal. maximum aperture size because of constraints on the
The effective linear aperture of an AOTF is limited by physical dimensions of components that can be incorpo-
the acoustic beam height in one dimension (ID) and by the rated into a microscope system. The angular aperture is
acoustic attenuation across the optical aperture (the acous- related to the total light collecting power of the AOTF, an
tic transit distance) in the other dimension (67). The height important factor in imaging systems, although in order to
of the acoustic beam generated within the AOTF crystal is realize the full angular aperture without the use of polar-
determined by the performance and physical properties of izers in the noncollinear AOTF, its value must be smaller
the acoustic transducer. Acoustic attenuation in crystalline than the deflection angle. Because the acoustooptic tunable
materials, such as tellurium dioxide, is proportional to filter is not an image-forming component of the microscope
the square of acoustic frequency, and is therefore a more system (it is typically employed for source filtering), there
problematic limitation to linear aperture size in the shorter is no specific means of evaluating the spatial resolution
wavelength visible light range, which requires higher RF for this type of device (70). However, the AOTF may restrict
frequencies for tuning. Near-IR and IR radiation produces the attainable spatial resolution of the imaging system
MICROSCOPY, CONFOCAL 459

because of its limited linear aperture size and acceptance enhance the versatility of the latest generation of confocal
angle, in the same manner as other optical components. instruments, and these devices are becoming increasing
Based on the Rayleigh criterion and the angular and linear popular for control of excitation wavelength ranges and
apertures of the AOTF, the maximum number of resolvable intensity. The primary characteristic that facilitates
image elements may be calculated for a given wavelength, nearly every advantage of the AOTF is its capability to
utilizing different expressions for the polar and azimuthal allow the microscopist control of the intensity and/or illu-
planes. Although diffraction limited resolution can be mination wavelength on a pixel-by-pixel basis while main-
attained in the azimuthal plane, dispersion in the AOTF taining a high scan rate (7). This single feature translates
limits the resolution in the polar plane, and measures must into a wide variety of useful analytical microscopy tools,
be taken to suppress this factor for optimum performance. which are even further enhanced in flexibility when laser
The dependence of deflection angle on wavelength can illumination is employed.
produce one form of dispersion, which is typically negligi- One of the most useful AOTF functions allows the
ble when tuning is performed within a relatively narrow selection of small user-defined specimen areas (commonly
bandwidth, but significant in applications involving opera- termed regions of interest; ROI) that can be illuminated
tion over a broad spectral range. Changes in deflection with either greater or lesser intensity, and at different
angle with wavelength can result in image shifts during wavelengths, for precise control in photobleaching techni-
tuning, producing errors in techniques, such as ratio ima- ques, excitation ratio studies, resonance energy-transfer
ging of fluorophores excited at different wavelengths, and investigations, or spectroscopic measurements (see Fig. 10).
in other multispectral applications. When the image shift The illumination intensity can not only be increased in
obeys a known relationship to wavelength, corrections can selected regions for controlled photobleaching experiments
be applied through digital processing techniques (1,7). (71–73), but can be attenuated in desired areas in order to
Other effects of dispersion, including reduced angular minimize unnecessary photobleaching. When the illumina-
resolution, may result in image degradation, such as blur- tion area is under AOTF control, the laser exposure is
ring, that requires more elaborate measures to suppress. restricted to the scanned area by default, and the extremely
rapid response of the device can be utilized to provide beam
blanking during the flyback interval of the galvanometer
SUMMARY OF AOTF BENEFITS IN CONFOCAL scanning mirror cycle, further limiting unnecessary speci-
MICROSCOPY men exposure. In practice, the regions of excitation are
typically defined by freehand drawing or using tools to
Considering the underlying principles of operation and produce defined geometrical shapes in an overlay plane
performance factors that relate to the application of AOTFs on the computer monitor image. Some systems allow any
in imaging systems, a number of virtues from such devices number of specimen areas to be defined for laser exposure,
for light control in fluorescence confocal microscopy are and the laser intensity to be set to different levels for each
apparent. Several benefits of the AOTF combine to greatly area, in intensity increments as small as 0.1%. When the

Figure 10. AOTF selection of specific regions for excitation in


confocal microscopy. (a) Region of Interest (ROI) selected for
fluorescence recovery after photobleaching (FRAP) experi-
ments. (b) Freehand ROIs for selective excitation. (c) ROI for
fluorescence resonance energy-transfer (FRET) analysis. (d) ROI
for photoactivation and photoconversion of fluorescent proteins.
460 MICROSCOPY, CONFOCAL

AOTF is combined with multiple lasers and software that cervical carcinoma) cell nuclei using selected illumination
allows time course control of sequential observations, time- (yellow box) with a 405 nanometer violet–blue diode laser.
lapse experiments can be designed to acquire data from The rapid intensity and wavelength switching capabil-
several different areas in a single experiment, which might, ities of the AOTF enable sequential line scanning of multi-
for example, be defined to correspond to different cellular ple laser lines to be performed in which each excitation
organelles. wavelength can be assigned a different intensity in order to
Figure 10 illustrates several examples of several user- balance the various signal levels for optimum imaging (77).
defined ROIs that were created for advanced fluorescence Sequential scanning of individual lines minimizes the time
applications in laser scanning confocal microscopy. In each differential between signal acquisitions from the various
image, the ROI is outlined with a yellow border. The rat fluorophores while reducing crossover, which can be a sig-
kangaroo kidney epithelial cell (PtK2 line) presented in nificant problem with simultaneous multiple-wavelength
Fig. 10a has a rectangular area in the central portion of the excitation (Fig. 11). The synchronized incorporation of
cytoplasm that has been designated for photobleaching multiple fluorescent probes into living cells has grown
experiments. Fluorophores residing in this region can be into an extremely valuable technique for study of protein–
selectively destroyed by high power laser intensity, and the protein interactions, and the dynamics of macromolecular
subsequent recovery of fluorescence back into the photo- complex assembly. The refinement of techniques for
bleached region monitored for determination of diffusion incorporating green fluorescent protein (GFP) and its
coefficients. Several freehand ROIs are illustrated in numerous derivatives into the protein-synthesizing
Fig. 10b, which can be targets for selective variation of mechanisms of the cell has revolutionized living cell
illumination intensities or photobleaching and photo- experimentation (78–80). A major challenge in multiple-
activation experiments. Fluorescence resonance energy- probe studies using living tissue is the necessity to acquire
transfer emission ratios can be readily determined using the complete multispectral data set quickly enough to
selected regions in confocal microscopy by observing the minimize specimen movement and molecular changes
effect of bleaching the acceptor fluorescence in these areas that might distort the true specimen geometry or dynamic
(Fig. 10c; African green monkey kidney epithelial cells sequence of events (32–34). The AOTF provides the speed
labeled with Cy3 and Cy5 conjugated to cholera toxin, and versatility to control the wavelength and intensity
which localizes in the plasma membrane). The AOTF con- illuminating multiple specimen regions, and to simulta-
trol of laser excitation in selected regions with confocal neously or sequentially scan each at sufficient speed to
microscopy is also useful for investigations of protein diffu- accurately monitor dynamic cellular processes.
sion in photoactivation studies (74–76) using fluorescent A comparison between the application of AOTFs and
proteins, as illustrated in Fig. 10d. This image frame neutral density filters (78) to control spectral separation of
presents the fluorescence emission peak of the Kaede fluorophore emission spectra in confocal microscopy is
protein as it shifts from green to red in HeLa (human presented in Fig. 11. The specimen is a monolayer culture

Figure 11. Fluorophore bleedthrough control with neutral


density filters and sequential scanning using AOTF laser
modulation. Adherent human lung fibroblast (MRC-5 line)
cells were stained with Texas Red conjugated to phalloidin
(actin; red) and counterstained with SYTOX green (nuclei;
green). (a) Typical cell imaged with neutral density filters. (b)
The same cell imaged using sequential line scanning controlled
by an AOTF laser combiner. (c) and (d) Colocalization scatterplots
derived from the images in (a) and (b), respectively.
MICROSCOPY, CONFOCAL 461

of adherent human lung fibroblast (MRC-5 line) cells The method of utilizing photoactivated fluorescence has
stained with Texas Red conjugated to phalloidin (targeting been very useful in studies, such as those examining the
the filamentous actin network) and SYTOX Green (stain- role of calcium ion concentration in cellular processes, but
ing DNA in the nucleus). A neutral density filter that has been limited in its sensitivity to localized regional
produces the high excitation signals necessary for both effects in small organelles or in close proximity to cell
fluorophores leads to a significant amount of bleedthrough membranes. Typically, fluorescent species that are inacti-
of the SYTOX Green emission into the Texas Red channel vated by being bound to a photosensitive species (referred
(Fig. 11a; note the yellow nuclei). The high degree of to as being caged) are activated by intense illumination
apparent colocalization between SYTOX Green and Texas that frees them from the caging compound and allows them
Red is clearly illustrated by the scatterplot in Fig. 11b. The to be tracked by the sudden appearance of fluorescence
two axes in the scatterplot represent the SYTOX Green (84). The use of the AOTF has facilitated the refinement of
(abscissa) and the Texas Red (ordinate) channels. In order such studies to assess highly localized processes such as
to balance the excitation power levels necessary to selec- calcium ion mobilization near membranes, made possible
tively illuminate each fluorophore with greater control of because of the precise and rapid control of the illumination
emission intensity, an AOTF was utilized to selectively triggering the activation (uncaging) of the fluorescent
reduce the SYTOX Green excitation power (Argon-ion laser molecule of interest.
line at 488 nm). Note the subsequent reduction in bleed- Because the AOTF functions, without use of moving
through as manifested by green color in the cellular nuclei mechanical components, to electronically control the wave-
in Fig. 11c. The corresponding scatterplot (Fig. 11d) indi- length and intensity of multiple lasers, great versatility is
cates a dramatically reduced level of bleed-through (and provided for external control and synchronization of laser
apparent colocalization) of SYTOX Green into the Texas illumination with other aspects of microscopy experiments.
Red channel. When the confocal instrument is equipped with a controller
The development of the AOTF has provided substantial module having input and output trigger terminals, laser
additional versatility to techniques, such as fluorescence intensity levels can be continuously monitored and
recovery after photobleaching (FRAP; 81,82), fluorescence recorded, and the operation of all laser functions can be
loss in photobleaching (FLIP; 83), as well as in localized controlled to coordinate with other experimental specimen
photoactivated fluorescence (uncaging; 84) studies measurements, automated microscope stage movements,
(Fig. 10). The FRAP technique (81,82) was originally con- sequential time-lapse recording, and any number of other
ceived to measure diffusion rates of fluorescently tagged operations.
proteins in organelles and cell membranes. In the conven-
tional FRAP procedure, a small spot on the specimen is
continuously illuminated at a low light flux level and the RESOLUTION AND CONTRAST IN CONFOCAL
emitted fluorescence is measured. The illumination level is MICROSCOPY
then increased to a very high level for a brief time to
destroy the fluorescent molecules in the illuminated region All optical microscopes, including conventional widefield,
by rapid bleaching. After the light intensity is returned to confocal, and two-photon instruments are limited in the
the original low level, the fluorescence is monitored to resolution that they can achieve by a series of fundamental
determine the rate at which new unbleached fluorescent physical factors (1,3,5–7,24,85–89). In a perfect optical
molecules diffuse into the depleted region. The technique, system, resolution is restricted by the numerical aperture
as typically employed, has been limited by the fixed geo- of optical components and by the wavelength of light, both
metry of the bleached region, which is often a diffraction- incident (excitation) and detected (emission). The concept
limited spot, and by having to mechanically adjust the of resolution is inseparable from contrast, and is defined as
illumination intensity (using shutters or galvanometer- the minimum separation between two points that results in
driven components). The AOTF not only allows near- a certain level of contrast between them (24). In a typical
instantaneous switching of light intensity, but also can be fluorescence microscope, contrast is determined by the num-
utilized to selectively bleach randomly specified regions of ber of photons collected from the specimen, the dynamic
irregular shape, lines, or specific cellular organelles, and to range of the signal, optical aberrations of the imaging
determine the dynamics of molecular transfer into the region. system, and the number of picture elements (pixels) per
By enabling precise control of illuminating beam geo- unit area in the final image (66,86–88).
metry and rapid switching of wavelength and intensity, The influence of noise on the image of two closely spaced
the AOTF is a significant enhancement to application of small objects is further interconnected with the related
the FLIP technique in measuring the diffusional mobility factors mentioned above, and can readily affect the quality
of certain cellular proteins (83). This technique monitors of resulting images (29). While the effects of many instru-
the loss of fluorescence from continuously illuminated mental and experimental variables on image contrast, and
localized regions and the redistribution of fluorophore consequently on resolution, are familiar and rather obvious,
from distant locations into the sites of depletion. The data the limitation on effective resolution resulting from the
obtained can aid in the determination of the dynamic division of the image into a finite number of picture elements
interrelationships between intracellular and intercellular (pixels) may be unfamiliar to those new to digital micro-
components in living tissue, and such fluorescence loss scopy. Because all digital confocal images employing laser
studies are greatly facilitated by the capabilities of the scanners and/or camera systems are recorded and processed
AOTF in controlling the microscope illumination. in terms of measurements made within discrete pixels (66),
462 MICROSCOPY, CONFOCAL

some discussion of the concepts of sampling theory is THE AIRY DISK AND LATERAL RESOLUTION
required. This is appropriate to the subject of contrast
and resolution because it has a direct bearing on the ability Imaging a point-like light source in the microscope pro-
to record two closely spaced objects as being distinct. duces an electromagnetic field in the image plane whose
In addition to the straightforward theoretical aspects of amplitude fluctuations can be regarded as a manifestation
resolution, regardless of how it is defined, the reciprocal of the response of the optical system to the specimen. This
relationship between contrast and resolution has practical field is commonly represented through the amplitude
significance because the matter of interest to most micro- point spread function, and allows evaluation of the
scopists is not resolution, but visibility. The ability to optical transfer properties of the combined system compo-
recognize two closely spaced features as being separate nents (29,86–88). Although variations in field amplitude
relies on advanced functions of the human visual system to are not directly observable, the visible image of the point
interpret intensity patterns, and is a much more subjective source formed in the microscope and recorded by its
concept than the calculation of resolution values based on imaging system is the intensity point spread function,
diffraction theory (24). Experimental limitations and the which describes the system response in real space. Actual
properties of the specimen itself, which vary widely, dictate specimens are not point sources, but can be regarded as a
that imaging cannot be performed at the theoretical max- superposition of an infinite number of objects having
imum resolution of the microscope. dimensions below the resolution of the system. The proper-
The relationship between contrast and resolution with ties of the intensity point spread function (PSF; see Fig. 12)
regard to the ability to distinguish two closely spaced in the image plane as well as in the axial direction are
specimen features implies that resolution cannot be major factors in determining the resolution of a microscope
defined without reference to contrast, and it is this inter- (1,24,29,40,85–89).
dependence that has led to considerable ambiguity invol- It is possible to experimentally measure the intensity
ving the term resolution and the factors that influence it in point spread function in the microscope by recording the
microscopy (29). As discussed above, recent advances in image of a subresolution spherical bead as it is scanned
fluorescent protein technology have led to an enormous through focus (a number of examples may be found in the
increase in studies of dynamic processes in living cells and literature). Because of the technical difficulty posed in
tissues (71–76,78–83). Such specimens are optically thick direct measurement of the intensity point spread function,
and inhomogeneous, resulting in a far-from-ideal imaging calculated point spread functions are commonly utilized to
situation in the microscope. Other factors, such as cell evaluate the resolution performance of different optical
viability and sensitivity to thermal damage and photo- systems, as well as the optical-sectioning capabilities of
bleaching, place limits on the light intensity and duration confocal, two-photon, and conventional widefield micro-
of exposure, consequently limiting the attainable resolu- scopes. Although the intensity point spread function
tion. Given that the available timescale may be dictated by extends in all three dimensions, with regard to the rela-
these factors and by the necessity to record rapid dynamic tionship between resolution and contrast, it is useful to
events in living cells, it must be accepted that the quality of consider only the lateral components of the intensity dis-
images will not be as high as those obtained from fixed and tribution, with reference to the familiar Airy disk (24).
stained specimens. The most reasonable resolution goal for The intensity distribution of the point-spread function
imaging in a given experimental situation is that the in the plane of focus is described by the rotationally sym-
microscope provides the best resolution possible within metric Airy pattern. Because of the cylindrical symmetry
the constraints imposed by the experiment. of the microscope lenses, the two lateral components

Figure 12. Schematic diagram of an Airy disk diffraction


pattern and the corresponding three-dimensional point spread
functions for image formation in confocal microscopy. Intensity
profiles of a single Airy disk, as well as the first and higher order
maxima are illustrated in the graphs.
MICROSCOPY, CONFOCAL 463

(x and y) of the Airy pattern are equivalent, and the objects. By specifying the depth of the dip in intensity
pattern represents the lateral intensity distribution as between two overlapping point spread functions, the ambi-
a function of distance from the optical axis (24). The guity in evaluating resolution can be removed, and a
lateral distance is normalized by the numerical aperture quantitative aspect introduced.
of the system and the wavelength of light, and therefore is In order to quantify resolution, the concept of contrast
dimensionless. Figure 12 (Airy disk and intensity func- is employed, which is defined for two objects of equal
tion) illustrates diagrammatically the formation and intensity as the difference between their maximum inten-
characteristics of the Airy disk, the related 3D point sity and the minimum intensity occurring in the space
spread function, and Airy patterns in the fluorescence between them (55,86,89). Because the maximum intensity
microscope. Following the excitation of fluorophores in a of the Airy disk is normalized to one, the highest achievable
point-like specimen region, fluorescence emission occurs contrast is also one, and occurs only when the spacing
in all directions, a small fraction of which is selected and between the two objects is relatively large, with sufficient
focused by the optical components into an image plane separation to allow the first zero crossing to occur in their
where it forms an Airy disk surrounded by concentric combined intensity distribution. At decreased distance, as
rings of successively decreasing maximum and minimum the two point spread functions begin to overlap, the dip in
intensity (the Airy pattern). The Airy pattern intensity intensity between the two maxima (and the contrast) is
distribution is the result of Fraunhofer diffraction of light increasingly reduced. The distance at which two peak
passing through a circular aperture, and in a perfect maxima are no longer discernible, and the contrast
optical system exhibits a central intensity maximum becomes zero, is referred to as the contrast cut-off distance
and higher order maxima separated by regions of zero (24,40). The variation of contrast with distance allows
intensity (85). The distance of the zero crossings from the resolution, in terms of the separation of two points, to be
optical axis, when the distance is normalized by the defined as a function of contrast.
numerical aperture and wavelength, occur periodically The relationship between contrast and separation dis-
(Fig. 12). When the intensity on the optical axis is normal- tance for two point-like objects is referred to as the con-
ized to one (100%), the proportional heights of the first trast/distance function or contrast transfer function
four higher order maxima are 1.7%, 0.4%, 0.2%, and (31,90). Resolution can be defined as the separation dis-
0.08%, respectively. tance at which two objects are imaged with a certain
A useful approach to the concept of resolution is based contrast value. It is obvious that when zero contrast exists,
on consideration of an image formed by two point-like the points are not resolved; the so-called Sparrow criter-
objects (specimen features), under the assumption that ion defines the resolution of an optical system as being
the image-forming process is incoherent, and that the equivalent to the contrast cut-off distance (24). It is com-
interaction of the separate object images can be described mon, however, to specify that greater contrast is necessary
using intensity point spread functions. The resulting image to adequately distinguish two closely spaced points
is then composed of the sum of two Airy disks, the char- visually, and the well-known Rayleigh criterion (24) for
acteristics of which depend on the separation distance resolution states that two points are resolved when the
between the two points (24,86). When sufficiently sepa- first minimum (zero crossing) of one Airy disk is aligned
rated, the intensity change in the area between the objects with the central maximum of the second Airy disk. Under
is the maximum possible, cycling from the peak intensity optimum imaging conditions, the Rayleigh criterion
(at the first point) to zero and returning to the maximum separation distance corresponds to a contrast value of
value at the center of the second point. At decreased 26.4%. Although any contrast value >0 can be specified
distance in object space, the intensity distribution func- in defining resolution, the 26% contrast of the Rayleigh
tions of the two points, in the image plane, begin to overlap criterion is considered reasonable in typical fluorescence
and the resulting image may appear to be that of a single microscopy applications, and is the basis for the common
larger or brighter object or feature rather than being expression defining lateral resolution according to the
recognizable as two objects. If resolution is defined, in following equation (24), in which the point separation (r)
general terms, as the minimum separation distance at in the image plane is the distance between the central
which the two objects can be sufficiently distinguished, maximum and the first minimum in the Airy disk:
it is obvious that this property is related to the width of the
intensity peaks (the point spread function). Microscope rlateral ¼ 1:22 l=ð2NAÞ ¼ 0:6 l=NA
resolution is directly related, therefore, to the full-width
at half maximum (fwhm) of the instrument’s intensity point where l is the emitted light wavelength and NA is the
spread function in the component directions (29,86,87). numerical aperture of the objective.
Some ambiguity in use of the term resolution results Resolution in the microscope is directly related to the
from the variability in defining the degree of separation fwhm dimensions of the microscope’s point spread function,
between features and their point spread functions that is and it is common to measure this value experimentally in
sufficient to allow them to be distinguished as two objects order to avoid the difficulty in attempting to identify inten-
rather than one. In general, minute features of interest in sity maxima in the Airy disk. Measurements of resolution
microscopy specimens produce point images that overlap to utilizing the fwhm values of the point spread function are
some extent, displaying two peaks separated by a gap somewhat smaller than those calculated employing the
(1,24,29,40,86). The greater the depth of the gap between Rayleigh criterion. Furthermore, in confocal fluorescence
the peaks, the easier it is to distinguish, or resolve, the two configurations, single-point illumination scanning and
464 MICROSCOPY, CONFOCAL

single-point detection are employed, so that only the most applicable to fluorescence emission are similar in
fluorophores in the shared volume of the illumination form to the expressions evaluating depth of field, and
and detection point spread functions are able to be demonstrate that axial resolution is proportional to the
detected. The intensity point spread function in the con- wavelength, and refractive index of the specimen medium,
focal case is, therefore, the product of the independent and inversely proportional to the square of the numerical
illumination intensity and detection intensity point aperture. Consequently, the NA of the microscope objective
spread functions. For confocal fluorescence, the lateral has a much greater effect on axial resolution than does the
(and axial) extent of the point spread function is reduced emission wavelength. One equation (89) commonly used to
by 30% compared to that in the wide-field microscope. describe axial resolution for the confocal configuration is
Because of the narrower intensity point spread function, given below, with h representing the index of refraction,
the separation of points required to produce acceptable and the other variables as specified previously:
contrast in the confocal microscope (29,31) is reduced to a
distance approximated by raxial ¼ 1:4 lh=NA2

rlateral ¼ 0:4l=NA Although the confocal microscope configuration exhibits


only a modest improvement in measured axial resolution
If the illumination and fluorescence emission wave- over that of the widefield microscope, the true advantage of
lengths are approximately the same, the confocal fluor- the confocal approach is in the optical sectioning capability
escence microscope Airy disk size is the square of the in thick specimens, which results in a dramatic improve-
wide-field microscope Airy disk. Consequently, the con- ment in effective axial resolution over conventional tech-
trast cut-off distance is reduced in the confocal arrange- niques. The optical sectioning properties of the confocal
ment, and equivalent contrast can be achieved at a shorter microscope result from the characteristics of the integrated
distance compared to the widefield illumination config- intensity point spread function, which has a maximum in
uration. Regardless of the instrument configuration, the the focal plane when evaluated as a function of depth. The
lateral resolution displays a proportional relationship to equivalent integral of intensity point spread function for the
wavelength, and is inversely proportional to the objective conventional widefield microscope is constant as a function
lens numerical aperture. of depth, producing no optical sectioning capabilities.
As noted previously, lateral resolution is of primary
interest in discussing resolution and contrast, although
the axial extent of the microscope intensity point spread FLUOROPHORES FOR CONFOCAL MICROSCOPY
function is similarly reduced in the confocal arrangement
as compared to the widefield fluorescence configuration Biological laser scanning confocal microscopy relies heavily
(86,89). Reasonable contrast between point-like objects on fluorescence as an imaging mode, primarily due to the
lying on the optical axis occurs when they are separated high degree of sensitivity afforded by the technique coupled
by the distance between the central maximum and the first with the ability to specifically target structural components
minimum of the axial point spread function component. and dynamic processes in chemically fixed as well as living
Figure 13 presents the axial intensity distributions (89) cells and tissues. Many fluorescent probes are constructed
for a typical widefield (Fig. 13a) and confocal (Fig. 13b) around synthetic aromatic organic chemicals designed
fluorescence microscope. Note the dramatic reduction in to bind with a biological macromolecule (e.g., a protein
intensity of the wings in the confocal distribution as a or nucleic acid) or to localize within a specific structural
function of distance from the central maximum. region, such as the cytoskeleton, mitochondria, Golgi appa-
A variety of equations are presented in the literature ratus, endoplasmic reticulum, and nucleus (90). Other
that pertains to different models for calculating axial probes are employed to monitor dynamic processes and
resolution for various microscope configurations. The ones localized environmental variables, including concentra-
tions of inorganic metallic ions, pH, reactive oxygen spe-
cies, and membrane potential (91). Fluorescent dyes are
also useful in monitoring cellular integrity (live versus
dead and apoptosis), endocytosis, exocytosis, membrane
fluidity, protein trafficking, signal transduction, and enzy-
matic activity (92). In addition, fluorescent probes have
been widely applied to genetic mapping and chromosome
analysis in the field of molecular genetics.
The history of synthetic fluorescent probes dates back
over a century to the late-1800s when many of the corner-
stone dyes for modern histology were developed. Among
these were pararosaniline, methyl violet, malachite green,
safranin O, methylene blue, and numerous azo (nitrogen)
dyes, such as Bismarck brown (93). Although these dyes
were highly colored and capable of absorbing selected
bands of visible light, most were only weakly fluorescent
Figure 13. Comparison of axial (x–z) point spread functions for and would not be useful for the fluorescence microscopes
widefield (left) and confocal (right) microscopy. that would be developed several decades later. However,
MICROSCOPY, CONFOCAL 465

several synthetic dye classes synthesized during this period,


based on the xanthene and acridine heterocyclic ring sys-
tems, proved to be highly fluorescent and provided a foun-
dation for the development of modern synthetic fluorescent
probes. Most notable among these early fluorescent dyes
were the substituted xanthenes, fluorescein and rhodamine
B, and the biaminated acridine derivative, acridine orange.
Fluorochromes were introduced to fluorescence micro-
scopy in the early twentieth century as vital stains for
bacteria, protozoa, and trypanosomes, but did not see
widespread use until the 1920s when fluorescence micro-
scopy was first used to study dye binding in fixed tissues Figure 14. Fluorescent spectral profiles, plotted as normalized
absorption or emission as a function of wavelength, for popular
and living cells (7,93). However, it was not until the early
synthetic fluorophores emitting in the blue, green, and red regions
1940s that Coons developed a technique for labeling anti-
of the visible spectrum. Each profile is identified with a colored
bodies with fluorescent dyes, thus giving birth to the field of bullet in (a), which illustrates excitation spectra. (b) The emission
immunofluorescence (94). Over the past 60 years, advances spectra for the fluorophores according to the legend in (a).
in immunology and molecular biology have produced a wide
spectrum of secondary antibodies and provided insight into
the molecular design of fluorescent probes targeted at spe- yield represents the probability that a given excited fluor-
cific regions within macromolecular complexes. ochrome will produce an emitted (fluorescence) photon.
Fluorescent probe technology and cell biology were Quantum yields typically range between a value of 0 and 1
dramatically altered by the discovery of the GFP from and fluorescent molecules commonly employed as probes
jellyfish and the development of mutant spectral variants, in microscopy have quantum yields ranging from very low
which have opened the door to noninvasive fluorescence (0.05 or less) to almost unity. In general, a high quantum
multicolor investigations of subcellular protein localiza- yield is desirable in most imaging applications. The quan-
tion, intermolecular interactions, and trafficking using tum yield of a given fluorophore varies, sometimes to large
living cell cultures (79,80,95). More recently, the develop- extremes, with environmental factors, such as metallic
ment of nanometer-sized fluorescent semiconductor quan- ion concentration, pH, and solvent polarity (92).
tum dots has provided a new avenue for research in In most cases, the molar extinction coefficient for photon
confocal and widefield fluorescence microscopy (96). absorption is quantitatively measured and expressed at a
Despite the numerous advances made in fluorescent dye specific wavelength, whereas the quantum efficiency is an
synthesis during the past few decades, there is very little assessment of the total integrated photon emission over the
solid evidence about molecular design rules for developing entire spectral band of the fluorophore (Fig. 14b). As
new fluorochromes, particularly with regard to matching opposed to traditional arc-discharge lamps used with the
absorption spectra to available confocal laser excitation shortest range (10–20 nm) bandpass interference filters in
wavelengths. As a result, the number of fluorophores that wide-field fluorescence microscopy, the laser systems used
have found widespread use in confocal microscopy is a for fluorophore excitation in scanning confocal microscopy
limited subset of the many thousands that have been restrict excitation to specific laser spectral lines that
discovered. encompass only a few nanometers (1,7). The fluorescence
emission spectrum for both techniques, however, is con-
trolled by similar bandpass or longpass filters that can
BASIC CHARACTERISTICS OF FLUOROPHORES cover tens to hundreds of nanometers (7). Below saturation
levels, fluorescence intensity is proportional to the product
Fluorophores are catalogued and described according to of the molar extinction coefficient and the quantum yield of
their absorption and fluorescence properties, including the the fluorophore, a relationship that can be utilized to judge
spectral profiles, wavelengths of maximum absorbance and the effectiveness of emission as a function of excitation
emission, and the fluorescence intensity of the emitted wavelength(s). These parameters display an approximate
light (92). One of the most useful quantitative parameters 20-fold range in variation for the popular fluorophores
for characterizing absorption spectra is the molar extinc- commonly employed for investigations in confocal micro-
tion coefficient (denoted with the Greek symbole, see scopy with quantum yields ranging from 0.05 to 1.0, and
Fig. 14a), which is a direct measure of the ability of a extinction coefficients ranging from 10,000 to 0.25 million
molecule to absorb light. The extinction coefficient is useful (L  mol1). In general, the absorption spectrum of a fluor-
for converting units of absorbance into units of molar ophore is far less dependent on environmental conditions
concentration, and is determined by measuring the absor- than the fluorescence emission characteristics (spectral
bance at a reference wavelength (usually the maximum, wavelength profile and quantum yield; 92).
characteristic of the absorbing species) for a molar concen- Fluorophores chosen for confocal applications must
tration in a defined optical path length. The quantum yield exhibit a brightness level and signal persistence sufficient
of a fluorochrome or fluorophore represents a quantitative for the instrument to obtain image data that does not suffer
measure of fluorescence emission efficiency, and is expressed from excessive photobleaching artifacts and low signal/
as the ratio of the number of photons emitted to the noise ratios. In widefield fluorescence microscopy, excita-
number of photons absorbed. In other words, the quantum tion illumination levels are easily controlled with neutral
466 MICROSCOPY, CONFOCAL

Table 1. Laser and Arc-Discharge Spectral Lines in Widefield and Confocal Microscopy
Laser Type Ultraviolet Violet Blue Green Yellow Orange Red

Argon-ion 351, 364 457, 477, 488 514


Blue diode 405, 440
Diode-pumped solid state 355 430, 442 457, 473 532 561
Helium–cadmium 322, 354 442
Krypton–argon 488 568 647
Green helium–neon 543
Yellow helium–neon 594
Orange helium–neon 612
Red helium-neon 633
Red diode 635, 650
Mercury arc 365 405, 436 546 579
Xenon arc 467

density filters (40), and the intensity can be reduced levels is, therefore, a critical condition for achieving the opti-
(coupled with longer emission signal collection periods) mal signal/noise ratio in confocal experiments (1,7,92,97). The
to avoid saturation and curtail irreversible loss of fluores- number of fluorescent probes currently available for con-
cence. Excitation conditions in confocal microscopy are focal microscopy runs in the hundreds (90,93), with many
several orders of magnitude more severe, however, and dyes having absorption maxima closely associated with
restrictions imposed by characteristics of the fluorophores common laser spectral lines (90). An exact match between
and efficiency of the microscope optical system become the a particular laser line and the absorption maximum of a
dominating factor in determining excitation rate and emis- specific probe is not always possible, but the excitation
sion collection strategies (1,7,92). efficiency of lines near the maximum is usually sufficient
Because of the narrow and wavelength-restricted laser to produce a level of fluorescence emission that can be
spectral lines employed to excite fluorophores in confocal readily detected.
microscopy (Table 1), fluorescence emission intensity can Instrumentally, fluorescence emission collection can be
be seriously restricted due to poor overlap of the excitation optimized by careful selection of objectives, detector aper-
wavelengths with the fluorophore absorption band. In ture dimensions, dichromatic and barrier filters, as well as
addition, the confocal pinhole aperture, which is critical maintaining the optical train in precise alignment (63). In
in obtaining thin optical sections at high signal/noise most cases, low magnification objectives with a high
ratios, is responsible for a 25–50% loss of emission inten- numerical aperture should be chosen for the most demand-
sity, regardless of how much effort has been expended on ing imaging conditions because light collection intensity
fine-tuning and alignment of the microscope optical system increases as the fourth power of the numerical aperture,
(7). Photomultiplier tubes are the most common detectors but only decreases as the square of the magnification.
in confocal microscopy, but suffer from a quantum effi- However, the most important limitations in light collection
ciency that varies as a function of wavelength (especially in efficiency in confocal microscopy arise from restrictions
the red and IR regions), further contributing to a wave- imposed by the physical properties of the fluorophores
length-dependent loss of signal across the emission spec- themselves. As previously discussed, fluorescent probe
trum (59–62). Collectively, the light losses in confocal development is limited by a lack of knowledge of the specific
microscopy can result in a reduction of intensity exceeding molecular properties responsible for producing optimum
50 times of the level typically observed in widefield fluor- fluorescence characteristics, and the design rules are insuf-
escence instruments. It should be clear from the preceding ficiently understood to be helpful as a guide to the devel-
argument that fluorophore selection is one of the most opment of more efficient fluorophores. The current success
critical aspects of confocal microscopy, and instrumental in development of new fluorescent probes capable of satis-
efficiency must be carefully considered, as well, in order to factory performance in confocal microscopy is a testament
produce high quality images. to the progress made through use of empirical data and
In confocal microscopy, irradiation of the fluorophores assumptions about molecular structure extrapolated from
with a focused laser beam at high power densities increases the properties of existing dyes, many of which were first
the emission intensity up to the point of dye saturation, a synthesized over a hundred years ago.
condition whose parameters are dictated by the excited
state lifetime (97). In the excited state, fluorophores are
unable to absorb another incident photon until they emit a TRADITIONAL FLUORESCENT DYES
lower energy photon through the fluorescence process.
When the rate of fluorophore excitation exceeds the rate The choice of fluorescent probes for confocal microscopy
of emission decay, the molecules become saturated and the must address the specific capabilities of the instrument to
ground state population decreases. As a result, a majority excite and detect fluorescence emission in the wavelength
of the laser energy passes through the specimen undimin- regions made available by the laser systems and detectors.
ished and does not contribute to fluorophore excitation. Although the current lasers used in confocal microscopy
Balancing fluorophore saturation with laser light intensity (Table 1) produce discrete lines in the UV, visible, and
MICROSCOPY, CONFOCAL 467

near-IR portions of the spectrum, the location of these acid (DNA), but has a longer wavelength fluorescence
spectral lines does not always coincide with absorption maximum ( 640 nm; red) when bound to these macromo-
maxima of popular fluorophores. In fact, it is not necessary lecules. In living cells, acridine orange diffuses across the
for the laser spectral line to correspond exactly with the cell membrane (by virtue of the association constant for
fluorophore wavelength of maximum absorption, but the protonation) and accumulates in the lysosomes and other
intensity of fluorescence emission is regulated by the fluor- acidic vesicles. Similar to most acridines and related poly-
ophore extinction coefficient at the excitation wavelength nuclear nitrogen heterocycles, acridine orange has a rela-
(as discussed above). The most popular lasers for confocal tively broad absorption spectrum, which enables the probe to
microscopy are air-cooled argon and krypton–argon ion be used with several wavelengths from the argon-ion laser.
lasers, the new blue diode lasers, and a variety of helium– Another popular traditional probe that is useful in
neon systems (7,40). Collectively, these lasers are capable of confocal microscopy is the phenanthridine derivative, pro-
providing excitation at 10–12 specific wavelengths between pidium iodide, first synthesized as an antitrypanosomal
400 and 650 nm. agent along with the closely related ethidium bromide).
Many of the classical fluorescent probes that have been Propidium iodide binds to DNA in a manner similar to the
successfully utilized for many years in widefield fluores- acridines (via intercalation) to produce orange-red fluor-
cence (92,93), including fluorescein isothiocyanate, Lissa- escence centered at 617 nm (101,102). The positively
mine rhodamine, and Texas red, are also useful in confocal charged fluorophore also has a high affinity for double-
microscopy. Fluorescein is one of the most popular fluor- stranded RNA. Propidium has an absorption maximum at
ochromes ever designed, and has enjoyed extensive appli- 536 nm, and can be excited by the 488 or 514-nm spectral
cation in immunofluorescence labeling. This xanthene dye lines of an argon-ion (or krypton–argon) laser, or the
has an absorption maximum at 495 nm, which coincides 543 nm line from a green helium–neon laser. The dye is
quite well with the 488 nm (blue) spectral line produced by often employed as a counterstain to highlight cell nuclei
argon-ion and krypton–argon lasers, as well as the 436 and during double or triple labeling of multiple intracellular
467 principal lines of the mercury and xenon arc-discharge structures. Environmental factors can affect the fluores-
lamps, respectively. In addition, the quantum yield of cence spectrum of propidium, especially when the dye is
fluorescein is very high and a significant amount of infor- used with mounting media containing glycerol. The struc-
mation has been gathered on the characteristics of this dye turally similar ethidium bromide, which also binds to DNA
with respect to the physical and chemical properties (98). by intercalation (102), produces more background staining,
On the negative side, the fluorescence emission intensity of and is therefore not as effective as propidium.
fluorescein is heavily influenced by environmental factors The DNA and chromatin can also be stained with dyes
(e.g., pH), and the relatively broad emission spectrum often that bind externally to the double helix. The most popular
overlaps with those of other fluorophores in dual and triple fluorochromes in this category are 40 ,6-diamidino-2-pheny-
labeling experiments (92,98,99). lindole (DAPI) and the bis (benzimide) Hoechst dyes that
Tetramethyl rhodamine (TMR) and the isothiocyanate are designated by the numbers 33258, 33342, and 34580
derivative (TRITC) are frequently employed in multiple (103–106). These probes are quite water soluble and bind
labeling investigations in widefield microscopy due to their externally to AT-rich base pair clusters in the minor groove
efficient excitation by the 546 nm spectral line from mer- of double-stranded DNA with a dramatic increase in fluor-
cury arc-discharge lamps. The fluorochromes, which have escence intensity. Both dye classes can be stimulated by
significant emission spectral overlap with fluorescein, the 351 nm spectral line of high power argon-ion lasers or
can be excited very effectively by the 543 nm line from the 354 nm line from a helium–cadmium laser. Similar to
helium–neon lasers, but not by the 514 or 568 nanometer the acridines and phenanthridines, these fluorescent
lines from argon-ion and krypton–argon lasers (99). When probes are popular choices as a nuclear counterstain for
using krypton-based laser systems, Lissamine rhodamine use in multicolor fluorescent labeling protocols. The vivid
is a far better choice in this fluorochrome class due to the blue fluorescence emission produces dramatic contrast
absorption maximum at 575 nm and its spectral separation when coupled to green, yellow, and red probes in adjacent
from fluorescein. Also, the fluorescence emission intensity cellular structures.
of rhodamine derivatives is not as dependent upon strict
environmental conditions as that of fluorescein.
Several of the acridine dyes, first isolated in the nine- ALEXA FLUOR DYES
teenth century, are useful as fluorescent probes in confocal
microscopy (93). The most widely utilized, acridine orange, The dramatic advances in modern fluorophore technology
consists of the basic acridine nucleus with dimethylamino are exemplified by the Alexa Fluor dyes (90,107,108) intro-
substituents located at the 3 and 6 positions of the tri- duced by Molecular Probes (Alexa Fluor is a registered
nuclear ring system. In physiological pH ranges, the mole- trademark of Molecular Probes). These sulfonated rhoda-
cule is protonated at the heterocyclic nitrogen and exists mine derivatives exhibit higher quantum yields for more
predominantly as a cationic species in solution. Acridine intense fluorescence emission than spectrally similar
orange binds strongly to DNA by intercalation of the probes, and have several additional improved features,
acridine nucleus between successive base pairs, and exhi- including enhanced photostability, absorption spectra
bits green fluorescence with a maximum wavelength of matched to common laser lines, pH insensitivity, and a
530 nm (92,93,100). The probe also binds strongly to ribo- high degree of water solubility. In fact, the resistance to
nucleic acid (RNA) or single-stranded deoxyribonucleic photobleaching of Alexa Fluor dyes is so dramatic (108)
468 MICROSCOPY, CONFOCAL

that even when subjected to irradiation by high intensity indole nitrogen heterocyclic nucleus with two aromatic
laser sources, fluorescence intensity remains stable for units being connected via a polyalkene bridge of varying
relatively long periods of time in the absence of antifade carbon number (92,109). These probes exhibit fluorescence
reagents. This feature enables the water soluble Alexa excitation and emission profiles that are similar to many of
Fluor probes to be readily utilized for both live-cell and the traditional dyes, such as fluorescein and tetramethylr-
tissue section investigations, as well as in traditional fixed hodamine, but with enhanced water solubility, photostabil-
preparations. ity, and higher quantum yields. Most of the cyanine dyes
Alexa Fluor dyes are available in a broad range of are more environmentally stable than their traditional
fluorescence excitation and emission wavelength maxima, counterparts, rendering their fluorescence emission inten-
ranging from the UV and deep blue to the near-IR regions sity less sensitive to pH and organic mounting media.
(90). Alphanumeric names of the individual dyes are asso- In a manner similar to the Alexa Fluors, the excitation
ciated with the specific excitation laser or arc-discharge wavelengths of the Cy series of synthetic dyes are tuned
lamp spectral lines for which the probes are intended. For specifically for use with common laser and arc-discharge
example, Alexa Fluor 488 is designed for excitation by the sources, and the fluorescence emission can be detected with
blue 488 nm line of the argon or krypton–argon ion lasers, traditional filter combinations.
while Alexa Fluor 568 is matched to the 568 nm spectral Marketed by a number of distributors, the cyanine dyes
line of the krypton–argon laser. Several of the Alexa Fluor are readily available as reactive dyes or fluorophores
dyes are specifically designed for excitation by either the coupled to a wide variety of secondary antibodies, dextrin,
blue diode laser (405 nm), the orange/yellow helium–neon streptavidin, and eggwhite avidin (110). The cyanine dyes
laser (594 nm), or the red helium–neon laser (633 nm). generally have broader absorption spectral regions than
Other Alexa Fluor dyes are intended for excitation with members of the Alexa Fluor family, making them some-
traditional mercury arc-discharge lamps in the visible what more versatile in the choice of laser excitation sources
(Alexa Fluor 546) or UV (Alexa Fluor 350, also useful with for confocal microscopy (7). For example, using the 547 nm
high power argon-ion lasers), and solid-state red diode spectral line from an argon-ion laser, Cy2 is about twice as
lasers (Alexa Fluor 680). Because of the large number of efficient in fluorescence emission as Alexa Fluor 488. In an
available excitation and emission wavelengths in the Alexa analogous manner, the 514 nm argon-ion laser line excites
Fluor series, multiple labeling experiments can often be Cy3 with a much higher efficiency than Alexa Fluor 546, a
conducted exclusively with these dyes. spectrally similar probe. Emission profiles of the cyanine
Alexa Fluor dyes are commercially available as reactive dyes are comparable in spectral width to the Alexa Fluor
intermediates in the form of maleimides, succinimidyl series.
esters, and hydrazides, as well as prepared cytoskeletal Included in the cyanine dye series are the long-wave-
probes (conjugated to phalloidin, G-actin, and rabbit ske- length Cy5 derivatives, which are excited in the red region
letal muscle actin) and conjugates to lectin, dextrin, strep- (650 nm) and emit in the far-red (680 nm) wavelengths. The
tavidin, avidin, biocytin, and a wide variety of secondary Cy5 fluorophore is very efficiently excited by the 647 nm
antibodies (90). In the latter forms, the Alexa Fluor fluor- spectral line of the krypton–argon laser, the 633 nm line of
ophores provide a broad palette of tools for investigations in the red helium–neon laser, or the 650 nm line of the red
immunocytochemistry, neuroscience, and cellular biology. diode laser, providing versatility in laser choice. Because
The family of probes has also been extended into a series of the emission spectral profile is significantly removed from
dyes having overlapping fluorescence emission maxima traditional fluorophores excited by UV and blue illumina-
targeted at sophisticated confocal microscopy detection tion, Cy5 is often utilized as a third fluorophore in triple
systems with spectral imaging and linear unmixing cap- labeling experiments. However, similar to other probes
abilities. For example, Alexa Fluor 488, Alexa Fluor 500, with fluorescence emission in the far-red spectral region,
and Alexa Fluor 514 are visually similar in color with Cy5 is not visible to the human eye and can only be
bright green fluorescence, but have spectrally distinct detected electronically (using a specialized CCD camera
emission profiles. In addition, the three fluorochromes system or photomultiplier). Therefore, the probe is seldom
can be excited with the 488 or 514 nm spectral line from used in conventional widefield fluorescence experiments.
an argon-ion laser and are easily detected with traditional
fluorescein filter combinations. In multispectral (x–y–l;
referred to as a lambda stack) confocal imaging experi- FLUORESCENT ENVIRONMENTAL PROBES
ments, optical separation software can be employed to
differentiate between the similar signals (32–35). The over- Fluorophores designed to probe the internal environment
lapping emission spectra of Alexa Fluor 488, 500, and 514 of living cells have been widely examined by a number of
are segregated into separate channels and differentiated investigators, and many hundreds have been developed to
using pseudocolor techniques when the three fluorophores monitor such effects as localized concentrations of alkali
are simultaneously combined in a triple label investigation. and alkaline earth metals, heavy metals (employed bio-
chemically as enzyme cofactors), inorganic ions, thiols, and
sulfides, nitrite, as well as pH, solvent polarity, and mem-
CYANINE DYES brane potential (7,90–93,111,112). Originally, the experi-
ments in this arena were focused on changes in the
The popular family of cyanine dyes, Cy2, Cy3, Cy5, Cy7, wavelength and/or intensity of absorption and emission
and their derivatives, are based on the partially saturated spectra exhibited by fluorophores upon binding calcium
MICROSCOPY, CONFOCAL 469

ions in order to measure intracellular flux densities. These decreases, an isosbestic point is obtained when the dye
probes bind to the target ion with a high degree of speci- concentrations are constant within the localized area
ficity to produce the measured response and are often being investigated. Another benefit of using these probes
referred to as spectrally sensitive indicators. Ionic concen- together is the ability to measure fluorescence intensity
tration changes are determined by the application of fluctuations with a standard FITC/Texas red interference
optical ratio signal analysis to monitor the association filter combination.
equilibrium between the ion and its host. The concen- Quantitative measurements of ions other than calcium,
tration values derived from this technique are largely such as magnesium, sodium, potassium, and zinc, are
independent of instrumental variations and probe concen- conducted in an analogous manner using similar fluoro-
tration fluctuations due to photobleaching, loading para- phores (7,90,92). One of the most popular probes for mag-
meters, and cell retention. In the past few years, a number nesium, mag-fura-2 (structurally similar to fura red), is
of new agents have been developed that bind specific ions or also excited in the ultraviolet range and presents the same
respond with measurable features to other environmental problems in confocal microscopy as fura-2 and indo-1.
conditions (7,90). Fluorophores excited in the visible light region are becom-
Calcium is a metabolically important ion that plays a ing available for the analysis of many monovalent and
vital role in cellular response to many forms of external divalent cations that exist at varying concentrations in
stimuli (113). Because transient fluctuations in calcium ion the cellular matrix. Several synthetic organic probes have
concentration are typically involved when cells undergo a also been developed for monitoring the concentration of
response, fluorophores must be designed to measure not simple and complex anions.
only localized concentrations within segregated compart- Important fluorescence monitors for intracellular pH
ments, but should also produce quantitative changes when include a pyrene derivative known as HPTS or pyranine,
flux density waves progress throughout the entire cyto- the fluorescein derivative, BCECF, and another substi-
plasm. Many of the synthetic molecules designed to mea- tuted xanthene termed carboxy SNARF-1 (90,119–122).
sure calcium levels are based on the nonfluorescent Because many common fluorophores are sensitive to pH
chelation agents EGTA and BAPTA, which have been used in the surrounding medium, changes in fluorescence inten-
for years to sequester calcium ions in buffer solutions sity that are often attributed to biological interactions may
(7,114,115). Two of the most common calcium probes are actually occur as a result of protonation. In the physiological
the ratiometric indicators fura-2 and indo-1, but these pH range (pH 6.8–7.4), the probes mentioned above are
fluorophores are not particularly useful in confocal micro- useful for dual-wavelength ratiometric measurements and
scopy (7,116). The dyes are excited by UV light and exhibit differ only in dye loading parameters. Simultaneous mea-
a shift in the excitation or emission spectrum with the surements of calcium ion concentration and pH can often be
formation of isosbestic points when binding calcium. How- accomplished by combining a pH indicator, such as SNARF-
ever, the optical aberrations associated with UV imaging, 1, with a calcium ion indicator (e.g., fura-2). Other probes
limited specimen penetration depths, and the expense of have been developed for pH measurements in subcellular
ultraviolet lasers have limited the utility of these probes in compartments, such as the lysosomes, as described below.
confocal microscopy.
Fluorophores that respond in the visible range to cal-
cium ion fluxes are, unfortunately, not ratiometric indica- ORGANELLE PROBES
tors and do not exhibit a wavelength shift (typical of fura-2
and indo-1) upon binding, although they do undergo an Fluorophores targeted at specific intracellular organelles,
increase or decrease in fluorescence intensity. The best such as the mitochondria, lysosomes, Golgi apparatus, and
example is fluo-3, a complex xanthene derivative, which endoplasmic reticulum, are useful for monitoring a variety
undergoes a dramatic increase in fluorescence emission at of biological processes in living cells using confocal micro-
525 nm (green) when excited by the 488 nm spectral line of scopy (7,90,92). In general, organelle probes consist of a
an argon-ion or krypton–argon laser (7,117). Because iso- fluorochrome nucleus attached to a target-specific moiety
sbestic points are not present to assure the absence of con- that assists in localizing the fluorophore through covalent,
centration fluctuations, it is impossible to determine whether electrostatic, hydrophobic, or similar types of bonds. Many
spectral changes are due to complex formation or a variation of the fluorescent probes designed for selecting organelles
in concentration with fluo-3 and similar fluorophores. are able to permeate or sequester within the cell membrane
To overcome the problems associated with using visible (and therefore, are useful in living cells), while others must
light probes lacking wavelength shifts (and isosbestic be installed using monoclonal antibodies with traditional
points), several of these dyes are often utilized in combina- immunocytochemistry techniques. In living cells, organelle
tion for calcium measurements in confocal microscopy probes are useful for investigating transport, respiration,
(118). Fura red, a multinuclear imidazole and benzofuran mitosis, apoptosis, protein degradation, acidic compart-
heterocycle, exhibits a decrease in fluorescence at 650 nm ments, and membrane phenomena. Cell impermeant fluor-
when binding calcium. A ratiometric response to calcium ophore applications include nuclear functions, cytoskeletal
ion fluxes can be obtained when a mixture of fluo-3 and fura structure, organelle detection, and probes for membrane
red is excited at 488 nm and fluorescence is measured at the integrity. In many cases, living cells that have been labeled
emission maxima (525 and 650 nm, respectively) of the two with permeant probes can subsequently be fixed and coun-
probes. Because the emission intensity of fluo-3 increases terstained with additional fluorophores in multicolor label-
monotonically while that of fura red simultaneously ing experiments.
470 MICROSCOPY, CONFOCAL

Mitochondrial probes are among the most useful fluor- live cells. Several of the most useful fluorophores for Golgi
ophores for investigating cellular respiration and are often apparatus contain the complex heterocyclic BODIPY
employed along with other dyes in multiple labeling inves- nucleus developed by Molecular Probes (90,92,131). When
tigations. The traditional probes, rhodamine 123 and tet- coupled to sphingolipids, the BODIPY fluorophore is highly
ramethylrosamine, are rapidly lost when cells are fixed selective and exhibits a tolerance for photobleaching that is
and have largely been supplanted by newer, more specific, far superior to many other dyes. In addition, the emission
fluorophores developed by Molecular Probes (90,123,124). spectrum is dependent upon concentration (shifting from
These include the popular MitoTracker and MitoFluor green to red at higher concentrations), making the probes
series of structurally diverse xanthene, benzoxazole, useful for locating and identifying intracellular structures
indole, and benzimidazole heterocycles that are available that accumulate large quantities of lipids. During live-cell
in a variety of excitation and emission spectral profiles. The experiments, fluorescent lipid probes can undergo meta-
mechanism of action varies for each of the probes in this bolism to derivatives that may bind to other subcellular
series, ranging from covalent attachment to oxidation features, a factor that can often complicate the analysis of
within respiring mitochondrial membranes. experimental data.
MitoTracker dyes are retained quite well after cell The most popular traditional probes for endoplasmic
fixation in formaldehyde and can often withstand lipophilic reticulum fluorescence analysis are the carbocyanine and
permeabilizing agents (123). In contrast, the MitoFluor xanthene dyes, DiOC (6) and several rhodamine deriva-
probes are designed specifically for actively respiring cells tives, respectively (90,92). These dyes must be used with
and are not suitable for fixation and counterstaining pro- caution, however, because they can also accumulate in the
cedures (90). Another popular mitochondrial probe, mitochondria, Golgi apparatus, and other intracellular
entitled JC-1, is useful as an indicator of membrane poten- lipophilic regions. Newer, more photostable, probes have
tial and in multiple staining experiments with fixed cells been developed for selective staining of the endoplasmic
(125). This carbocyanine dye exhibits green fluorescence at reticulum by several manufacturers. In particular, oxazole
low concentrations, but can undergo intramolecular asso- members of the Dapoxyl family produced by Molecular
ciation within active mitochondria to produce a shift in Probes are excellent agents for selective labeling of the
emission to longer (red) wavelengths. The change in emis- endoplasmic reticulum in living cells, either alone or in
sion wavelength is useful in determining the ratio of active combination with other dyes (90). These probes are
to nonactive mitochondria in living cells. retained after fixation with formaldehyde, but can be lost
In general, weakly basic amines that are able to with permeabilizing detergents. Another useful probe is
pass through membranes are the ideal candidates for Brefeldin A (131), a stereochemically complex fungal meta-
investigating biosynthesis and pathogenesis in lysosomes bolite that serves as an inhibitor of protein trafficking out
(90–92,112). Traditional lysosomal probes include the non- of the endoplasmic reticulum. Finally, similar to other
specific phenazine and acridine derivatives neutral red and organelles, monoclonal antibodies (127–129) have been
acridine orange, which are accumulated in the acidic vesi- developed that target the endoplasmic reticulum in fixed
cles upon being protonated (92,93). Fluorescently labeled cells for immunocytochemistry investigations.
latex beads and macromolecules, such as dextran, can also
be accumulated in lysosomes by endocytosis for a variety of
experiments. However, the most useful tools for investi- QUANTUM DOTS
gating lysosomal properties with confocal microscopy are
the LysoTracker and LysoSensor dyes developed by Mole- Nanometer-sized crystals of purified semiconductors
cular Probes (90,92,126). These structurally diverse agents known as quantum dots are emerging as a potentially
contain heterocyclic and aliphatic nitrogen moieties that useful fluorescent labeling agent for living and fixed cells
modulate transport of the dyes into the lysosomes of living in both traditional widefield and laser scanning confocal
cells for both short- and long-term studies. The LysoTracker fluorescence microscopy (132–136). Recently introduced
probes, which are available in a variety of excitation and techniques enable the purified tiny semiconductor crystals
emission wavelengths (91), have high selectivity for acidic to be coated with a hydrophilic polymer shell and conju-
organelles and are capable of labeling cells at nanomolar gated to antibodies or other biologically active peptides and
concentrations. Several of the dyes are retained quite well carbohydrates for application in many of the classical
after fixing and permeabilization of cells. In contrast, the immunocytochemistry protocols (Fig. 15). These probes
LysoSensor fluorophores are designed for studying dynamic have significant benefits over organic dyes and fluorescent
aspects of lysosome function in living cells. Fluorescence proteins, including long-term photostability, high fluores-
intensity dramatically increases in the LysoSensor series cence intensity levels, and multiple colors with single-
upon protonation, making these dyes useful as pH indica- wavelength excitation for all emission profiles (136).
tors (91). A variety of Golgi apparatus specific monoclonal Quantum dots produce illumination in a manner similar
antibodies have also been developed for use in immunocy- to the well-known semiconductor light emitting diodes, but
tochemistry assays (90,127–129). are activated by absorption of a photon rather than an
Proteins and lipids are sorted and processed in the Golgi electrical stimulus. The absorbed photon creates an elec-
apparatus, which is typically stained with fluorescent tron-hole pair that quickly recombines with the concurrent
derivatives of ceramides and sphingolipids (130). These emission of a photon having lower energy. The most useful
agents are highly lipophilic, and are therefore useful as semiconductor discovered thus far for producing biological
markers for the study of lipid transport and metabolism in quantum dots is cadmium selenide (CdSe), a material in
MICROSCOPY, CONFOCAL 471

argon-ion, helium–cadmium, krypton–argon, and the


green helium–neon. Particularly effective at exciting quan-
tum dots in the UV and violet regions are the new blue
diode and diode-pumped solid-state lasers that have pro-
minent spectral lines at 442 nm and below (135,136). The
405 nm blue diode laser is an economical excitation source
that is very effective for use with quantum dots due to their
high extinction coefficient at this wavelength. Another
advantage of using these fluorophores in confocal micro-
scopy is the ability to stimulate multiple quantum dot sizes
(and spectral colors) in the same specimen with a single
Figure 15. Anatomy and spectral profiles of quantum dot excitation wavelength, making these probes excellent can-
conjugates. The cadmium selenide core is encapsulated with
didates for multiple labeling experiments (137).
zinc sulfide, and then a polymer coating is applied followed by a
The exceptional photostability of quantum dot conjugates
hydrophilic exterior to which the biological conjugate is attached
(left). The absorption profile displays a shoulder at 400 nm, while is of great advantage in confocal microscopy when optical
the emission spectra all feature similar symmetrical profiles. sections are being collected. Unlike the case of organic
fluorophores, labeled structures situated away from the
focal plane do not suffer from excessive photobleaching
which the energy of the emitted photons is a function of the during repeated raster scanning of the specimen and yield
physical size of the nanocrystal particles. Thus, quantum more accurate 3D volume models. In widefield fluorescence
dots having sizes that differ only by tenths of a nanometer microscopy, quantum dot conjugates are available for use
emit different wavelengths of light, with the smaller sizes with conventional dye-optimized filter combinations that
emitting shorter wavelengths, and vice versa. are standard equipment on many microscopes. Excitation
Unlike typical organic fluorophores or fluorescent pro- can be further enhanced by substituting a shortpass filter
teins, which display highly defined spectral profiles, quan- for the bandpass filter that accompanies most filter sets,
tum dots have an absorption spectrum that increases thus optimizing the amount of lamp energy that can be
steadily with decreasing wavelength (Fig. 15). Also, in utilized to excite the quantum dots. Several of the custom
contrast, the fluorescence emission intensity is confined fluorescence filter manufacturers offer combinations speci-
to a symmetrical peak with a maximum wavelength that is fically designed to be used with quantum dot conjugates.
dependent on the dot size, but independent of the excita-
tion wavelength (135). As a result, the same emission FLUORESCENT PROTEINS
profile is observed regardless of whether the quantum
dot is excited at 300, 400, 500, or 600 nm, but the fluores- Over the past few years, the discovery and development of
cence intensity increases dramatically at shorter excitation naturally occurring fluorescent proteins and mutated deri-
wavelengths. For example, the extinction coefficient for a vatives have rapidly advanced to center stage in the inves-
typical quantum dot conjugate that emits in the orange tigation of a wide spectrum of intracellular processes in
region (605 nm) is approximately five-fold higher when the living organisms (75,78,80). These biological probes have
semiconductor is excited at 400 versus 600 nm. The fwhm provided scientists with the ability to visualize, monitor,
value for a typical quantum dot conjugate is  30 nm (135), and track individual molecules with high spatial and tem-
and the spectral profile is not skewed towards the longer poral resolution in both steady-state and kinetic experi-
wavelengths (having higher intensity tails), such is the case ments. A variety of marine organisms have been the source
with most organic fluorochromes. The narrow emission of >100 fluorescent proteins and their analogs, which arm
profile enables several quantum dot conjugates to be simul- the investigator with a balanced palette of noninvasive
taneously observed with a minimal level of bleed through. biological probes for single, dual, and multispectral fluor-
For biological applications, a relatively uniform popula- escence analysis (75). Among the advantages of fluorescent
tion of cadmium selenide crystals is covered with a sur- proteins over the traditional organic and new semiconduc-
rounding semiconductor shell composed of zinc sulfide to tor probes described above is their response to a wider
improve the optical properties. Next, the core material is variety of biological events and signals. Coupled with the
coated with a polymeric film and other ligands to decrease ability to specifically target fluorescent probes in subcel-
hydrophobicity and to improve the attachment efficiency of lular compartments, the extremely low or absent photo-
conjugated macromolecules. The final product is a biologi- dynamic toxicity, and the widespread compatibility with
cally active particle that ranges in size from 10 to 15 nm, tissues and intact organisms, these biological macromole-
somewhere in the vicinity of a large protein (133). Quan- cules offer an exciting new frontier in live-cell imaging.
tum dot conjugates are solubilized as a colloidal suspension The first member of this series to be discovered, GFP,
in common biological buffers and may be incorporated into was isolated from the North Atlantic jellyfish, Aequorea
existing labeling protocols in place of classical staining Victoria, and found to exhibit a high degree of fluorescence
reagents (such as organic fluorochrome-labeled secondary without the aid of additional substrates or coenzymes (138–
antibodies). 142). In native green fluorescent protein, the fluorescent
In confocal microscopy, quantum dots are excited with moiety is a tripeptide derivative of serine, tyrosine, and
varying degrees of efficiency by most of the spectral lines glycine that requires molecular oxygen for activation, but
produced by the common laser systems, including the no additional cofactors or enzymes (143). Subsequent
472 MICROSCOPY, CONFOCAL

investigations revealed that the GFP gene could be 405 nm blue diode, the 442 nm helium–cadmium spectral
expressed in other organisms, including mammals, to yield line, and the 457 nm line from the standard argon-ion laser.
fully functional analogs that display no adverse biological Another popular fluorescent protein derivative, the yel-
effects (144). In fact, fluorescent proteins can be fused to low fluorescent protein (YFP), was designed on the basis of
virtually any protein in living cells using recombinant the GFP crystalline structural analysis to red-shift the
complementary DNA cloning technology, and the resulting absorption and emission spectra (148). Yellow fluorescent
fusion protein gene product expressed in cell lines adapted protein is optimally excited by the 514 nm spectral line of
to standard tissue culture methodology. Lack of a need for the argon-ion laser, and provides more intense emission
cell-specific activation cofactors renders the fluorescent than enhanced green fluorescent protein, but is more
proteins much more useful as generalized probes than sensitive to low pH and high halogen ion concentrations.
other biological macromolecules, such as the phycobilipro- The enhanced yellow fluorescent protein derivative
teins, which require insertion of accessory pigments in (EYFP) is useful with the 514 argon-ion laser line, but
order to produce fluorescence. can also be excited with relatively high efficiency by the
Mutagenesis experiments with green fluorescent pro- 488 nm line from argon and krypton–argon lasers. Both of
tein have produced a large number of variants with these fluorescent protein derivatives have been widely
improved folding and expression characteristics, which applied to protein–protein FRET investigations in combi-
have eliminated wild-type dimerization artifacts and fine nation with CFP, and in addition, have proven useful in
tuned the absorption and fluorescence properties. One of studies involving multiprotein trafficking.
the earliest variants, known as enhanced green fluores- Attempts to shift the absorption and emission spectra of
cence protein (EGFP), contains codon substitutions (com- Aequorea Victoria fluorescent proteins to wavelengths in
monly referred to as the S65T mutation) that alleviates the the orange and red regions of the spectrum have met with
temperature sensitivity and increases the efficiency of GFP little success. However, fluorescent proteins from other
expression in mammalian cells (145). Proteins fused with marine species have enabled investigators to extend the
EGFP can be observed at low light intensities for long time available spectral regions to well within the red wave-
periods with minimal photobleaching. Enhanced green length range. The DsRed fluorescent protein and its
fluorescent protein fusion products are optimally excited derivatives, originally isolated from the sea anemone
by the 488 nm spectral line from argon and krypton–argon Discosoma striata, are currently the most popular analogs
ion lasers in confocal microscopy. This provides an excel- for fluorescence analysis in the 575–650 nm region (149).
lent biological probe and instrument combination for exam- Another protein, HcRed from the Heteractis crispa purple
ining intracellular protein pathways along with the anemone, is also a promising candidate for investigations
structural dynamics of organelles and the cytoskeleton. in the longer wavelengths of the visible spectrum (150).
Additional mutation studies have uncovered GFP var- Newly developed photoactivation fluorescent proteins,
iants that exhibit a variety of absorption and emission including photoactivatable green fluorescent protein
characteristics across the entire visible spectral region, (PA-GFP;74), Kaede (76), and kindling fluorescent protein
which have enabled researchers to develop probe combina- 1 (KFP1; 151), exhibit dramatic improvements over GFP
tions for simultaneous observation of two or more distinct (up to several 1000-fold) in fluorescence intensity when
fluorescent proteins in a single organism (see the spectral stimulated by violet laser illumination. These probes
profiles in Fig. 16). Early investigations yielded the blue should prove useful in fluorescence confocal studies invol-
fluorescent protein (BFP) and cyan fluorescent protein ving selective irradiation of specific target regions and the
(CFP) mutants from simple amino acid substitutions that subsequent kinetic analysis of diffusional mobility and
shifted the absorption and emission spectral profiles of compartmental residency time of fusion proteins.
wild-type GFP to lower wavelength regions (146–148).
Used in combination with GFP, these derivatives are use-
ful in resonance energy transfer (FRET) experiments and QUENCHING AND PHOTOBLEACHING
other investigations that rely on multicolor fluorescence
imaging (73). Blue fluorescent protein can be efficiently The consequences of quenching and photobleaching are
excited with the 354 nm line from a high power argon laser, suffered in practically all forms of fluorescence micro-
while the more useful cyan derivative is excited by a scopy, and result in an effective reduction in the levels
number of violet and blue laser lines, including the of emission (152,153). These artifacts should be of primary

Figure 16. Fluorescent spectral profiles, plotted as normalized


absorption or emission as a function of wavelength, for fluores-
cent proteins emitting in the blue to orange-red regions of the
visible spectrum. Each profile is identified with a colored bullet in
(a), which illustrates excitation spectra. (b) The emission spectra
for the proteins according to the legend in (a).
MICROSCOPY, CONFOCAL 473

consideration when designing and executing fluorescence Some fluorophores bleach quickly after emitting only a few
investigations. The two phenomena are distinct in that photons, while others that are more robust can undergo
quenching is often reversible whereas photobleaching is thousands or even millions of cycles before bleaching.
not (154). Quenching arises from a variety of competing Figure 17 presents a typical example of photobleaching
processes that induce nonradiative relaxation (without (fading) observed in a series of digital images captured at
photon emission) of excited-state electrons to the ground different time points for a multiply stained culture of
state, which may be either intramolecular or intermole- normal Tahr ovary (HJ1.Ov line) fibroblast cells. The
cular in nature. Because nonradiative transition path- nuclei were stained with DAPI (blue fluorescence), while
ways compete with the fluorescence relaxation, they the mitochondria and actin cytoskeleton were stained with
usually dramatically lower or, in some cases, completely MitoTracker Red CMXRos (red fluorescence) and an Alexa
eliminate emission. Most quenching processes act to Fluor phalloidin derivative (Alexa Fluor 488; green fluor-
reduce the excited state lifetime and the quantum yield escence), respectively. Time points were taken in 2 min
of the affected fluorophore. intervals using a fluorescence filter combination with
A common example of quenching is observed with the bandwidths tuned to excite the three fluorophores simul-
collision of an excited state fluorophore and another (non- taneously while also recording the combined emission
fluorescent) molecule in solution, resulting in deactivation of signals. Note that all three fluorophores have a relatively
the fluorophore and return to the ground state. In most high intensity in Fig. 17a, but the DAPI (blue) intensity
cases, neither of the molecules is chemically altered in the starts to drop rapidly at two min and is almost completely
collisional quenching process. A wide variety of simple gone at six min (Fig. 17f). The mitochondrial and actin
elements and compounds behave as collisional quenching stains are more resistant to photobleaching, but the inten-
agents, including oxygen, halogens, amines, and many elec- sity of both drops dramatically over the course of the timed
tron-deficient organic molecules (154). Collisional quench- sequence (10 min).
ing can reveal the presence of localized quencher molecules An important class of photobleaching events is repre-
or moieties, which via diffusion or conformational change, sented by events that are photodynamic, meaning they
may collide with the fluorophore during the excited state involve the interaction of the fluorophore with a combina-
lifetime. The mechanisms for collisional quenching include tion of light and oxygen (157–161). Reactions between
electron transfer, spin–orbit coupling, and intersystem fluorophores and molecular oxygen permanently destroy
crossing to the excited triplet state (154,155). Other terms fluorescence and yield a free-radical singlet oxygen species
that are often utilized interchangeably with collisional that can chemically modify other molecules in living cells.
quenching are internal conversion and dynamic quenching. The amount of photobleaching due to photodynamic events
A second type of quenching mechanism, termed static is a function of the molecular oxygen concentration and
or complex quenching, arises from nonfluorescent com- the proximal distance between the fluorophore, oxygen
plexes formed between the quencher and fluorophore that molecules, and other cellular components. Photobleaching
serve to limit absorption by reducing the population of can be reduced by limiting the exposure time of
active, excitable molecules (154,156). This effect occurs
when the fluorescent species forms a reversible complex
with the quencher molecule in the ground state, and does
not rely on diffusion or molecular collisions. In static
quenching, fluorescence emission is reduced without alter-
ing the excited state lifetime. A fluorophore in the excited
state can also be quenched by a dipolar resonance energy
transfer mechanism when in close proximity with an
acceptor molecule to which the excited-state energy can
be transferred nonradiatively. In some cases, quenching
can occur through non molecular mechanisms, such as
attenuation of incident light by an absorbing species
(including the chromophore itself).
In contrast to quenching, photobleaching (also
termed fading) occurs when a fluorophore permanently
loses the ability to fluoresce due to photon-induced chemi-
cal damage and covalent modification (153–156). Upon
transition from an excited singlet state to the excited
triplet state, fluorophores may interact with another mole- Figure 17. Photobleaching in multiply stained specimens. Normal
cule to produce irreversible covalent modifications. The Tahr ovary fibroblast cells were stained with MitoTracker Red
triplet state is relatively long lived with respect to the CMXRos (mitochondria; red fluorescence), Alexa Fluor 488
conjugated to phalloidin (actin; green fluorescence), and
singlet state, thus allowing excited molecules a much
subsequently counterstained with DAPI (nuclei; blue fluore-
longer timeframe to undergo chemical reactions with com-
scence). Time points were taken in two-minute intervals over a
ponents in the environment (155). The average number of 10 min period using a fluorescence filter combination with
excitation and emission cycles that occur for a particular bandwidths tuned to excite the three fluorophores simultaneously
fluorophore before photobleaching is dependent on the while also recording the combined emission signals. (a–f) Time ¼0, 2,
molecular structure and the local environment (154,156). 4, 6, 8, 10 min, respectively.
474 MICROSCOPY, CONFOCAL

fluorophores to illumination or by lowering the excitation 3. Diaspro A, editor. Confocal and Two-Photon Microscopy:
energy. However, these techniques also reduce the mea- Foundations, Applications, and Advances. New York:
surable fluorescence signal. In many cases, solutions of Wiley-Liss; 2002.
fluorophores or cell suspensions can be deoxygenated, but 4. Matsumoto B, editor. Cell Biological Applications of Confocal
Microscopy. Methods in Cell Biology, Vol. 70. New York:
this is not feasible for living cells and tissues. Perhaps the
Academic Press; 2002.
best protection against photobleaching is to limit exposure 5. Sheppard CJR, Shotton DM. Confocal Laser Scanning Micro-
of the fluorochrome to intense illumination (using neutral scopy. Oxford (UK): BIOS Scientific Publishers; 1997.
density filters) coupled with the judicious use of commer- 6. Müller M. Introduction to Confocal Fluorescence Microscopy.
cially available antifade reagents that can be added to the Maastricht, The Netherlands: Shaker; 2002.
mounting solution or cell culture medium (153). 7. Hibbs AR. Confocal Microscopy for Biologists. New York:
Under certain circumstances, the photobleaching effect Kluwer Academic; 2004.
can also be utilized to obtain specific information that 8. Conn PM. Confocal Microscopy. Methods in Enzymology, Vol.
would not otherwise be available. For example, in FRAP 307. New York: Academic Press; 1999.
experiments, fluorophores within a target region are inten- 9. Corle TR, Kino GS. Confocal Scanning Optical Microscopy and
Related Imaging Systems. New York: Academic Press; 1996.
tionally bleached with excessive levels of irradiation (82).
10. Wilson T, editor. Confocal Microscopy. New York: Academic
As new fluorophore molecules diffuse into the bleached Press; 1990.
region of the specimen (recovery), the fluorescence emis- 11. Gu M. Principles of Three-Dimensional Imaging in Confocal
sion intensity is monitored to determine the lateral diffu- Microscopes. New Jersey: World Scientific; 1996.
sion rates of the target fluorophore. In this manner, the 12. Masters BR, editor. Selected Papers on Confocal Microscopy.
translational mobility of fluorescently labeled molecules SPIE Milestone Series, Vol. MS 131. Bellingham (WA): SPIE
can be ascertained within a very small (2–5 mm) region of a Optical Engineering Press; 1996.
single cell or section of living tissue. 13. Mason WT. Fluorescent and Luminescent Probes for Biolo-
Although the subset of fluorophores that are advanta- gical Activity. New York: Academic Press; 1999.
geous in confocal microscopy is rapidly growing, many of 14. Peterman EJG, Sosa H, Moerner WE. Single-Molecule Fluor-
escence Spectroscopy and Microscopy of Biomolecular
the traditional probes that have been useful for years in
Motors. Ann Rev Phys Chem 2004;55:79–96.
widefield applications are still of little utility when con- 15. Goldman RD, Spector DL. Live Cell Imaging: A Laboratory
strained by fixed-wavelength laser spectral lines. Many of Manual. New York: Cold Spring Harbor Press; 2005.
the limitations surrounding the use of fluorophores excited 16. Minsky M. Microscopy Apparatus. US Patent 3,013,467. 1961.
in the ultraviolet region will be eliminated with the intro- 17. Minsky M. Memoir on Inventing the Confocal Scanning
duction of advanced objectives designed to reduce aberra- Microscopy. Scanning 1988;10:128–138.
tion coupled to the gradual introduction of low cost, high 18. Egger MD, Petran M. New Reflected-Light Microscope for
power diode laser systems with spectral lines in these Viewing Unstained Brain and Ganglion Cells. Science
shorter wavelengths. The 405 nm blue diode laser is a 1967;157:305–307.
rather cheap alternative to more expensive ion and Noble 19. Davidovits P, Egger MD. Photomicrography of Corneal
Endothelial Cells in vivo. Nature (London) 1973;244:366–
gas based ultraviolet lasers, and is rapidly becoming
367.
available for most confocal microscope systems. Helium– 20. Amos WB, White JG. How the Confocal Laser Scanning Micro-
neon lasers with spectral lines in the yellow and orange scope entered Biological Research. Biol Cell 2003;95:335–342.
region have rendered some fluorophores useful that were 21. Brakenhoff GJ, Blom P, Barends P. Confocal Scanning Light
previously limited to widefield applications. In addition, Microscopy with High Aperture Immersion Lenses. J Micros
new diode-pumped solid-state lasers are being introduced 1979;117:219–232.
with emission wavelengths in the UV, violet, and blue 22. Sheppard CJR, Wilson T. Effect of Spherical Aberration on
regions. the Imaging Properties of Scanning Optical Microscopes.
Continued advances in fluorophore design, dual-laser Appl Opt 1979;18:1058.
scanning, multispectral imaging, endoscopic instruments, 23. Hamilton DK, Wilson T. Scanning Optical Microscopy by
Objective Lens Scanning. J Phys E: Sci Instr 1986;19:52–54.
and spinning disk applications will also be important in the
24. Spring KR, Inoué S. Video Microscopy: The Fundamentals.
coming years. The persistent problem of emission crossover New York: Plenum Press; 1997.
due to spectral overlap, which occurs with many synthetic 25. Wilson T. Optical Sectioning in Confocal Fluorescence Micro-
probes and fluorescent proteins in multicolor investiga- scopes. J Micros 1989;154:143–156.
tions, benefits significantly from spectral analysis and 26. Lichtmann JW. Confocal Microscopy. Sci Am Aug. 1994; 40–45.
deconvolution of lambda stacks. Combined, these advances 27. White JG, Amos WB, Fordham M. An Evaluation of Confocal
and will dramatically improve the collection and analysis of versus Conventional Imaging of Biological Structures by
data obtained from complex fluorescence experiments in Fluorescence Light Microscopy. J Cell Biol 1987;105:41–48.
live-cell imaging. 28. Swedlow JR, et al. Measuring Tubulin Content in Toxo-
plasma gondii: A Comparison of Laser-Scanning Confocal
and Wide-Field Fluorescence Microscopy. Proc Natl Acad
Sci USA 2002;99:2014–2019.
BIBLIOGRAPHY 29. Stelzer EHK. Practical Limits to Resolution in Fluorescence
Light Microscopy. In: Yuste R, Lanni F, Konnerth A, editors.
1. Pawley JB, editor. Handbook of Biological Confocal Micro- Imaging Neurons: A Laboratory Manual. New York: Cold
scopy. New York: Plenum Press; 1995. Spring Harbor Press; 2000. pp 12.1–12.9.
2. Paddock SW, editor. Confocal Microscopy: Methods and Pro- 30. Rost FWD. Fluorescence Microscopy. Vol. 1. New York: Cam-
tocols. Totowa (NJ): Humana Press; 1999. bridge University Press; 1992.
MICROSCOPY, CONFOCAL 475

31. Murray J. Confocal Microscopy, Deconvolution, and Struc- 51. Verveer PJ, Gemkow MJ, Jovin TM. A Comparison of Image
tured Illumination Methods. In: Goldman RD, Spector DL, Restoration Approaches Applied to Three-Dimensional Con-
editors. Live Cell Imaging: A Laboratory Manual. New York: focal and Wide-Field Fluorescence Microscopy. J Micros
Cold Spring Harbor Press; 2005. pp 239–280. 1998;193:50–61.
32. Dickinson ME, et al. Multi-Spectral Imaging and Linear 52. Conchello JA, Hansen EW. Enhanced 3-D Reconstruction
Unmixing Add a Whole New Dimension to Laser Scanning from Confocal Scanning Microscope Images. 1: Deterministic
Fluorescence Microscopy. Biotechniques 2001;31:1272–1278. and Maximum Likelihood Reconstructions. Appl Op 1990;29:
33. Zimmermann T, Rietdorf J, Pepperkok R. Spectral Imaging 3795–3804.
and its Applications in Live Cell Microscopy. FEBS Lett 53. Al-Kofahi O, et al. Algorithms for Accurate 3D Registration of
2003;546:87–92. Neuronal Images Acquired by Confocal Scanning Laser
34. Lansford R, Bearman G, Fraser SE. Resolution of Multiple Microscopy. J Micros 2003;211:8–18.
Green Fluorescent Protein Variants and Dyes using Two- 54. Conchello JA, et al., editors. Three-Dimensional and Multi-
Photon Microscopy and Imaging Spectroscopy. J Biomed Opt dimensional Microscopy: Image Acquisition and Processing.
2001;6:311–318. Vol. I–XII, Bellingham (WA): SPIE International Society for
35. Hiraoka Y, Shimi T, Haraguchi T. Multispectral Imaging Optical Engineering; 1994–2005.
Fluorescence Microscopy for Living Cells. Cell Struct Funct 55. Centonze V, Pawley J. Tutorial on Practical Confocal Micro-
2002;27:367–374. scopy and use of the Confocal Test Specimen. In: Pawley JB,
36. Gu Y, Di WL, Kelsell DP, Zicha D. Quantitative Fluorescence editor. Handbook of Biological Confocal Microscopy. New
Energy Transfer (FRET) Measurement with Acceptor Photo- York: Plenum Press; 1995. p 549–570.
bleaching and Spectral Unmixing. J Microsc 2004;215:162– 56. Gratton E, vandeVen MJ. Laser Sources for Confocal
173. Microscopy. In: Pawley JB, editor. Handbook of Biological
37. Ford BK, et al. Computed Tomography-Based Spectral Ima- Confocal Microscopy. New York: Plenum Press; 1995. p 69–98.
ging for Fluorescence Microscopy. Biophys J 2001;80:986– 57. Ashkin A, Dziedzic JM, Yamane T. Optical Trapping and
993. Manipulation of Single Cells using Infrared Laser Beams.
38. Bach H, Renn A, Wild UP. Spectral Imaging of Single Mole- London 1987;330:769–771.
cules. Single Mol 2000;1:73–77. 58. DeMaggio S. Running and Setting Up a Confocal Microscope
39. Wilson T, Carlini AR. Three-Dimensional Imaging in Con- Core Facility. In: Matsumoto B, editor. Cell Biological Appli-
focal Imaging Systems with Finite Sized Detectors. J Microsc cations of Confocal Microscopy. Methods in Cell Biology, Vol.
1988;149:51–66. 70. New York: Academic Press; 2002. p 475–486.
40. Murphy DB. Fundamentals of Light Microscopy and Electro- 59. Spring KR. Detectors for Fluorescence Microscopy. In: Peri-
nic Imaging. New York: Wiley-Liss; 2001. asamy A, editor. Methods in Cellular Imaging. New York:
41. Wright SJ, Wright DJ. Introduction to Confocal Microscopy. Oxford University Press; 2001. p 40–52.
In: Matsumoto B, editor. Cell Biological Applications of Con- 60. Art J. Photon Detectors for Confocal Microscopy. In: Pawley
focal Microscopy. Methods in Cell Biology, Vol. 70. New York: JB, editor. Handbook of Biological Confocal Microscopy. New
Academic Press; 2002. pp 1–85. York: Plenum Press; 1995. p 183–196.
42. Webb RH. Confocal Optical Microscopy. Rep Prog Phys 61. Amos WB. Instruments for Fluorescence Imaging. In: Allan
1996;59:427–471. VJ, editor. Protein Localization by Fluorescence Microscopy: A
43. Wilhelm S, Gröbler B, Gluch M, Hartmut H. Confocal Laser Practical Approach. New York: Oxford University Press; 2000.
Scanning Microscopy: Optical Image Formation and Electro- pp 67–108.
nic Signal Processing. Jena, Germany: Carl Zeiss Advanced 62. Hergert E. Detectors: Guideposts on the Road to Selection.
Imaging Microscopy; 2003. Photonics Design and Applications Handbook. 2001. pp
44. Ichihara A, et al. High-Speed Confocal Fluorescence Micro- H110–H113.
scopy using a Nipkow Scanner with Microlenses for 3-D 63. Piston DW, Patterson GH, Knobel SM. Quantitative Imaging
Imaging of Fluorescent Molecules in Real-Time. Bioimages of the Green Fluorescent Protein (GFP). In: Sullivan KF, Kay
1996;4:57–62. SA, editors. Green Fluorescent Proteins, Methods in Cell
45. Inoué S, Inoué T. Direct-View High-Speed Confocal Scan- Biology, Vol. 58. New York: Academic Press; 1999. pp 31–47.
ner—The CSU-10. In: Matsumoto B, editor. Cell Biological 64. Carter DR. Photomultiplier Handbook: Theory, Design,
Applications of Confocal Microscopy. Methods in Cell Biology, Application. Lancaster (PA): Burle Industries, Inc.; 1980.
Vol. 70. New York: Academic Press; 2002. p 88–128. 65. Cody SH, et al. A Simple Method Allowing DIC Imaging in
46. Nakano A. Spinning-Disk Confocal Microscopy —A Cutting- Conjunction with Confocal Microscopy. J Microsc 2005;217:
Edge Tool for Imaging of Membrane Traffic. Cell Struct Funct 265–274.
2002;27:349–355. 66. Chang IC. Acousto-Optic Devices and Applications. In: Bass
47. Chong FK, et al. Optimization of Spinning Disk Confocal M, Van Stryland EW, Williams DR, Wolfe WL, editors. Optics
Microscopy: Synchronization with the Ultra-Sensitive II: Fundamentals, Techniques, and Design. New York:
EMCCD. In: Conchello JA, Cogswell CJ, Wilson T, editors. McGraw-Hill; 1995. pp 12.1–12.54.
Three-Dimensional and Multidimensional Microscopy: Image 67. Wachman ES. Acousto-Optic Tunable Filters for Microscopy.
Acquisition and Processing XI. Proc. SPIE. 2004; 5324: 65–76. In: Yuste R, Lanni F, Konnerth A, editors. Imaging Neurons:
48. Sandison D, Webb W. Background Rejection and Signal-to- A Laboratory Manual. New York: Cold Spring Harbor Press;
Noise Optimization in the Confocal and Alternative Fluor- 2000. p 4.1–4.8.
escence Microscopes. Appl Op 1994;33:603–610. 68. Shonat RD, et al. Near-Simultaneous Hemoglobin Saturation
49. Centonze VE, White JG. Multiphoton Excitation Provides and Oxygen Tension Maps in Mouse Brain using an AOTF
Optical Sections from Deeper within Scattering Specimens Microscope. Biophy J 1997;73:1223–1231.
than Confocal Imaging. Biophys J 1998;75:2015. 69. Wachman ES, Niu W, Farkas DL. Imaging Acousto-Optic
50. Boccacci P, Bertero M. Image-Restoration Methods: Basics Tunable Filter with 0.35-Micrometer Spatial Resolution. App
and Algorithms. In: Diaspro A, editor. Confocal and Two- Opt 1996;35:5220–5226.
Photon Microscopy: Foundations, Applications, and Advances. 70. Wachman ES. AOTF Microscope for Imaging with Increased
New York: Wiley-Liss; 2002. pp 253–269. Speed and Spectral Versatility. Biophys J 1997;73:1215–1222.
476 MICROSCOPY, CONFOCAL

71. Chen Y, Mills JD, Periasamy A. Protein Localization in 95. Tsien RY. Building and Breeding Molecules to Spy on Cells
Living Cells and tissues using FRET and FLIM. Differentia- and Tumors. FEBS Lett 2005;579:927–932.
tion 2003;71:528–541. 96. Bruchez Jr M, et al. Semiconductor Nanocrystals as fluor-
72. Wallrabe H, Periasamy A. Imaging Protein Molecules using escent Biological Labels. Science 1998;218:2013–2016.
FRET and FLIM Microscopy. Curr Opin Biotech 2005;16: 19–27. 97. Tsien RY, Waggoner A. Fluorophores for Confocal Micro-
73. Day RN, Periasamy A, Schaufele F. Fluorescence Resonance scopy. In: Pawley JB, editor. Handbook of Biological Confocal
Energy Transfer Microscopy of Localized Protein Interac- Microscopy. New York: Plenum Press; 1995. p 267–280.
tions in the Living Cell Nucleus. Methods 2001;25:4–18. 98. Wessendorf MW, Brelje TC. Which Fluorophore is Brightest?
74. Patterson GH, Lippincott-Schwartz J. A Photoactivatable A Comparison of the Staining Obtained Using Fluorescein,
GFP for Selective Photolabeling of Proteins and Cells. Tetramethylrhodamine, Lissamine Rhodamine, Texas Red
Science 2002;297:1873–1877. and Cyanine 3.18. Histochemistry 1992;98:81–85.
75. Verkhusha VV, Lukyanov KA. The Molecular Properties and 99. Entwistle A, Noble M. The use of Lucifer Yellow, BODIPY,
Applications of Anthozoa Fluorescent Proteins and Chromo- FITC, TRITC, RITC and Texas Red for Dual Immunofluor-
proteins. Nature Biotechnol 2004;22:289–296. escence Visualized with a Confocal Scanning Laser Micro-
76. Ando R, et al. An Optical Marker Based on the UV-Induced scope. J Microsc 1992;168:219–238.
Green-to-Red Photoconversion of a Fluorescent Protein. Proc
100. Darzynkiewicz Z. Differential Staining of DNA and RNA in
Natl Acad Sci USA 2002;99:12651–12656.
Intact Cells and Isolated Cell Nuclei with Acridine Orange.
77. Sharma D. The Use of an AOTF to Achieve High Quality
Methods Cell Biol 1990;33:285–298.
Simultaneous Multiple Label Imaging. Bio-Rad Technical
101. Waring MJ. Complex Formation Between Ethidium Bromide
Notes. San Francisco: Bio-Rad, Note 4; 2001.
78. Miyawaki A, Sawano A, Kogure T. Lighting up Cells: Labeling and Nucleic Acids. J Mol Biol 1965;13:269–282.
Proteins with Fluorophores. Nature Cell Biol 2003;5:S1–S7. 102. Arndt-Jovin DJ, Jovin TM. Fluorescence Labeling and Micro-
79. Zhang J, Campbell RE, Ting AY, Tsien RY. Creating New scopy of DNA. Fluores Microsc Living Cells Culture Part B
Fluorescent Probes for Cell Biology. Nature Rev Mol Cell Bio Methods Cell Biol 1989;30:417–448.
2002;3:906–918. 103. Kubista M, Akerman B, Norden B. Characterization of Inter-
80. Lippincott-Schwartz J, Patterson G. Development and Use of action between DNA and 40 ,6-Diamidino-2-phenylindole by
Fluorescent Protein Markers in Living Cells. Science Optical Spectroscopy. Biochemistry 1987;26:4545–4553.
2003;300:87–91. 104. Loewe H, Urbanietz J. Basic Substituted 2,6-Bisbenzimida-
81. Klonis N, et al. Fluorescence Photobleaching Analysis for the zole Derivatives: A Novel Series of Substances with Che-
Study of Cellular Dynamics. Eur Biophys J 2002;31:36–51. motherapeutic Activity. Arzneim-Forsch 1974;24:1927–1933.
82. Lippincott-Schwartz J, Altan-Bonnet N, Patterson GH. 105. Arndt-Jovin DJ, Jovin TM. Analysis and Sorting of Living
Photobleaching and Photoactivation: Following Protein Cells According to Deoxyribonucleic Acid Content. J Histo-
Dynamics in Living Cells. Nature Cell Biol 2003;5:S7–S14. chem Cytochem 1977;25:585–589.
83. Phair RD, Misteli T. Kinetic Modelling Approaches to in vivo 106. Durand RE, Olive PL. Cytotoxicity, Mutagenicity and DNA
Imaging. Nature Rev Mol Cell Bio 2002;2:898–907. Damage by Hoechst 33342. J Histochem Cytochem 1982; 30:111–116.
84. Politz JC. Use of Caged Fluorophores to Track Macromolecular
107. Panchuk-Voloshina N, et al. Alexa Dyes, A Series of New
Movement in Living Cells. Trends Cell Biol 1999;9:284–287.
Fluorescent Dyes that Yield Exceptionally bright, Photostable
85. Born M, Wolf E. Principles of Optics. New York: Cambridge
Conjugates. J Histochem Cytochem 1999;47:1179–1188.
University Press; 1999.
86. Stelzer EHK. Contrast, Resolution, Pixelation, Dynamic range, 108. Berlier JE, et al. Quantitative Comparison of Long-Wavelength
and Signal-to-Noise Ratio: Fundamental Limits to Resolution in Alexa Fluor Dyes to Cy Dyes: Fluorescence of the Dyes and
Fluorescence Light Microscopy. J Microsc 1997;189:15–24. their Conjugates. J Histochem Cytochem 2003;51:1699–1712.
87. Pawley J. Fundamental Limits in Confocal Microscopy. In: 109. Mujumdar RB, et al. Cyanine Dye Labeling Reagents: Sulfoindo-
Pawley JB, editor. Handbook of Biological Confocal Micro- cyanine Succinimidyl Esters. Bioconjugate Chem 1993;4:105–111.
scopy. New York: Plenum Press; 1995. p 19–37. 110. Ballou B, et al. Tumor Labeling in vivo using Cyanine-
88. Webb RH, Dorey CK. The Pixelated Image. In: Pawley JB, Conjugated Monoclonal Antibodies. Cancer Immunol Immun-
editor. Handbook of Biological Confocal Microscopy. New other 1995;41:257–263.
York: Plenum Press; 1995. p 55–67. 111. Zorov DB, Kobrinsky E, Juhaszova M, Sollott SJ. Examining
89. Jonkman JEN, Stelzer EHK. Resolution and Contrast in Intracellular Organelle Function Using Fluorescent Probes.
Confocal and Two-Photon Microscopy. In: Diaspro A, editor. Circul Res 2004;95:239–252.
Confocal and Two-Photon Microscopy: Foundations, Applica- 112. Stephens DG, Pepperkok R. The Many Ways to Cross the Plasma
tions, and Advances. New York: Wiley-Liss; 2002. p 101–125. Membrane. Proc Natl Acad Sci USA 2001;98:4295–4298.
90. Haugland RP. The Handbook: A Guide to Fluorescent Probes 113. Rudolf R, Mongillo M, Rizzuto R, Pozzan T. Looking Forward to
and Labeling Technologies. Chicago: Invitrogen Molecular
Seeing Calcium. Nature Rev Mol Cell Biol 2003;4:579–586.
Probes; 2005.
114. Martin H, Bell MG, Ellis-Davies GC, Barsotti RJ. Activation
91. Lemasters JJ, et al. Confocal Imaging of Ca2þ, pH, Electrical
Kinetics of Skinned Cardiac Muscle by Laser Photolysis of
Potential and Membrane Permeability in Single Living Cells.
Nitrophenyl-EGTA. Biophys J 2004;86:978–990.
Methods Enzymol 1999;302:341–358.
92. Johnson I. Fluorescent Probes for Living Cells. Histochem J 115. White C, McGeown G. Imaging of Changes in Sarcoplasmic Reti-
1998;30:123–140. culum [Ca2þ] using Oregon Green BAPTA 5N and Confocal Laser
93. Kasten FH. Introduction to Fluorescent Probes: Properties, Scanning Microscopy. Cell Calcium 2002;31:151–159.
History, and Applications. In: Mason WT, editor. Fluorescent 116. Helm PJ, Patwardhan A, Manders EM. A Study of the Pre-
and Luminescent Probes for Biological Activity. New York: cision of Confocal, Ratiometric, Fura-2-Based [Ca2þ] Mea-
Academic Press; 1999. p 17–39. surements. Cell Calcium 1997;22:287–298.
94. Coons AH, Creech HJ, Jones RN, Berliner E. Demonstration 117. Rijkers GT, Justement LB, Griffioen AW, Cambier JC.
of Pneumococcal Antigen in Tissues by use of Fluorescent Improved Method for Measuring Intracellular Caþþ with
Antibody. J Immunol 1942;45:159–170. Fluo-3. Cytometry 1990;11:923–927.
477

118. Schild D, Jung A, Schultens HA. Localization of Calcium Entry 141. Hicks BW, editor. Green Fluorescent Protein, Methods in
through Calcium Channels in Olfactory Receptor Neurons Molecular Biology. Vol. 183. Totowa (NJ): Humana Press; 2002.
using a Laser Scanning Microscope and the Calcium Indicator 142. Chalfie M, Kain S, editors. Green Fluorescent Protein: Proper-
Dyes Fluo-3 and Fura-Red. Cell Calcium 1994;15: 341–348. ties, Applications, and Protocols. New York: Wiley-Liss; 1998.
119. Willoughby D, Thomas RC, Schwiening CJ. Comparison of 143. Zimmer M. Green Fluorescent Protein: Applications, Struc-
Simultaneous pH Measurements made with 8-Hydroxypyr- ture, and Related Photophysical Behavior. Chem Rev
ene-1,3,6-trisulphonic acid (HPTS) and pH-Sensitive Micro- 2002;102:759–781.
electrodes in Snail Neurons. Pflugers Arch 1998;436:615–622. 144. Chalfie M, et al. Green Fluorescent Protein as a Marker for
120. Ozkan P, Mutharasan R. A Rapid Method for Measuring Gene Expression. Science 1994;263:802–805.
Intracellular pH using BCECF-AM. Biochim Biophys Acta 145. Heim R, Cubitt AB, Tsien RY. Improved Green Fluores-
2002;1572:143. cence. Nature (London) 1995;373:664–665.
121. Cody SH, et al. Intracellular pH Mapping with SNARF-1 146. Heim R, Prasher DC, Tsien RY. Wavelength Mutations and
and Confocal Microscopy. I: A Quantitative Technique for Posttranslational Autoxidation of Green Fluorescent Pro-
Living Tissue and Isolated Cells. Micron 1993;24:573–580. tein. Proc Natl Acad Sci USA 1994;91:12501–12504.
122. Dubbin PN, Cody SH, Williams DA. Intracellular pH Mapping 147. Heim R, Tsien RY. Engineering Green Fluorescent Protein
with SNARF-1 and Confocal Microscopy. II: pH Gradients for Improved Brightness, Longer Wavelengths, and Fluor-
within Single Cultured Cells. Micron 1993;24:581–586. escence Resonance Energy Transfer. Curr Biol 1996;6:
123. Poot M, et al. Analysis of Mitochondrial Morphology and 178–182.
Function with Novel Fixable Fluorescent Stains. J Histo- 148. Wachter RM, et al. Structural Basis of Spectral Shifts in the
chem Cytochem 1996;44:1363–1372. Yellow-Emission Variants of Green Fluorescent Protein.
124. Keij JF, Bell-Prince C, Steinkamp JA. Staining of Mitochondrial Structure 1998;6:1267–1277.
Membranes with 10-Nonyl Acridine Orange, MitoFluor Green, 149. Matz MV, et al. Fluorescent Proteins from Nonbiolumines-
and MitoTracker Green is Affected by Mitochondrial Membrane cent Anthozoa Species. Nature Biotechnol 1999;17:969–973.
Potential Altering Drugs. Cytometry 2000;39: 203–210. 150. Matz MV, Lukyanov KA, Lukyanov SA. Family of the Green
125. Reers M. et al. Mitochondrial Membrane Potential Moni- Fluorescent Protein: Journey to the End of the Rainbow.
tored by JC-1 Dye. Methods Enzymol 1995;260:406–417. BioEssays 2002;24:953–959.
126. Price OT, Lau C, Zucker RM. Quantitative Fluorescence of 5- 151. Natural Animal Coloration can be Determined by a Non-
FU-Treated Fetal Rat Limbs using Confocal Laser Scanning fluorescent Green Fluorescent Protein Homolog. J Biol
Microscopy and LysoTracker Red. Cytometry 2003;53A:9–21. Chem 2000;275:25879–25882.
127. Kumar RK, Chapple CC, Hunter N. Improved Double 152. Song L, Hennink EJ, Young IT, Tanke HJ. Photobleaching
Immunofluorescence for Confocal Laser Scanning Micro- Kinetics of Fluorescein in Quantitative Fluorescence Micro-
scopy. J Histochem Cytochem 1999;47:1213–1217. scopy. Biophys J 1995;68:2588–2600.
128. Suzuki T, Fujikura K, Higashiyama T, Takata K. DNA 153. Berrios M, Conlon KA, Colflesh DE. Antifading Agents for
Staining for Fluorescence and Laser Confocal Microscopy. Confocal Fluorescence Microscopy. Methods Enzymol 1999;
J Histochem Cytochem 1997;45:49–53. 307:55–79.
129. Haugland RP. Coupling of Monoclonal Antibodies with 154. Lakowicz JR. Principles of Fluorescence Spectroscopy. New
Fluorophores. In: Davis WC, editor. Monoclonal Anibody York: Kluwer Academic/Plenum Publishers; 1999.
Protocols, Methods in Molecular Biology. Vol. 45. Totowa, 155. Song L, Varma CA, Verhoeven JW, Tanke HJ. Influence of
(NJ): Humana Press; 1995. p 205–221. the Triplet Excited State on the Photobleaching Kinetics of
130. Pagano RE, Martin OC. Use of Fluorescent Analogs of Fluorescein in Microscopy. Biophys J 1996;70:2959–2968.
Ceramide to Study the Golgi Apparatus of Animal Cells. 156. Herman B. Fluorescence Microscopy. New York: BIOS
In: Celis JE, editor. Cell Biology: A Laboratory Handbook. Scientific Publishers; 1998.
Vol. 2, 1998. p 507–512. 157. Bunting JR. A Test of the Singlet Oxygen Mechanism of
131. Cole L, Davies D, Hyde GJ, Ashford AE. ER-Tracker Dye and Cationic Dye Photosensitization of Mitochondrial Damage.
BODIPY-Brefeldin A Differentiate the Endoplasmic Reticulum Photochem Photobiol 1992;55:81–87.
and Golgi Bodies from the Tubular-Vacuole System in Living 158. Byers GW, Gross S, Henrichs PM. Direct and Sensitized
Hyphae of Pisolithus tinctorius. J Microsc 2000;197:239–249. Photooxidation of Cyanine Dyes. Photochem Photobiol 1976;
132. Jaiswal JK, Mattoussi H, Mauro JM, Simon SM. Long-Term 23:37–43.
Multiple Color Imaging of Live Cells using Quantum Dot 159. Dittrich PS, Schwille P. Photobleaching and Stabilization of
Bioconjugates. Nature Biotechnol 2003;21:47–52. Fluorophores used for Single Molecule Analysis with One-
133. Larson DR, et al. Water Soluble Quantum Dots for Multiphoton and Two-Photon Excitation. Appl Phys B 2001;73:829–837.
Fluorescence Imaging in vivo. Science 2003;300: 1434–1436. 160. Gandin E, Lion Y, Van de Vorst A. Quantum Yield of Singlet
134. Watson A, Wu X, Bruchez M. Lighting up Cells with Quan- Oxygen Production by Xanthene Derivatives. Photochem
tum Dots. Biotechniques 2003;34:296–303. Photobiol 1983;37:271–278.
135. Michalet X, et al. Quantum Dots for Live Cells, in vivo 161. Kanofsky JR, Sima PD. Structural and Environmental
Imaging, and Diagnostics. Science 2005;307:538–544. Requirements for Quenching of Singlet Oxygen by Cyanine
136. Gao X, et al. In vivo Molecular and Cellular Imaging with Dyes. Photochem Photobiol 2000;71:361–368.
Quantum Dots. Curr Opin Biotech 2005;16:63–72.
137. Lacoste TD, et al. Ultrahigh-Resolution Multicolor Coloca- Further Reading
lization of Single Fluorescent Probes. Proc Natl Acad Sci
USA 2000;97:9461–9466. Willison JR. Signal Detection and Analysis, In: Bass, M. Van
138. Tsien RY. The Green Fluorescent Protein. Ann Rev Biochem Stryland, E. W. Williams DR, Wolfe WL. Optics I: Fundamentals,
1998;67:509–544. Techniques, and Design. New York: McGraw-Hill; pp 1995. 18.1–
139. Sullivan KF, Kay SA, editors. Green Fluorescent Proteins, Meth- 18.16.
ods in Cell Biology. Vol. 58. New York: Academic Press; 1999.
140. Conn PM, editor. Green Fluorescent Protein, Methods in See also C ELLULAR IMAGING ; FLUORESCENCE MEASUREMENTS ;
ION-SENSITIVE FIELD EFFECT TRANSISTORS.
Enzymology. Vol. 302. New York: Academic Press; 1999.

You might also like