You are on page 1of 18

Basic Confocal Microscopy UNIT 2.

Carolyn L. Smith1
1
National Institute of Neurological Disorders and Stroke, Bethesda, Maryland

ABSTRACT
This unit introduces the reader to the basic principles of confocal microscopy and the design
and capabilities of current confocal microscopes. The advantages and disadvantages of confocal
microscopy compared to other techniques for fluorescence imaging are described. There are
also practical guidelines for sample preparation and optimization of imaging parameters, as
well as examples of some of the applications of confocal microscopy. Curr. Protoc. Neurosci.
56:2.2.1-2.2.18. C 2011 by John Wiley & Sons, Inc.

Keywords: confocal microscopy r fluorescence r imaging r resolution r


three-dimensional reconstruction

INTRODUCTION some dynamics (Bystricky et al., 2005), and


Confocal microscopy is a powerful tool for molecular binding interactions (Sprague and
visualizing fluorescent specimens. The prin- McNally, 2005). Confocal microscopy also is
cipal advantage of confocal microscopy over useful for visualizing cells in situ in fixed
conventional wide-field microscopy is that it (Fig. 2.2.1A) and living specimens. The maxi-
can reveal the three-dimensional structure of mum depth into the specimen at which images
the specimen. Fluorescent specimens viewed can be captured depends on the transparency
with a conventional wide-field fluorescent mi- of the specimen, characteristics of the objec-
croscope appear blurry and lack contrast be- tive, and excitation wavelength. Single-photon
cause fluorophores throughout the entire depth excitation with visible wavelengths (450 to
of the specimen are illuminated, and fluores- 650 nm) can typically penetrate up to 50 to
cence signals are collected not only from the 200 μm. Multiphoton excitation with infrared
plane of focus but also from areas above and wavelengths (>700 nm) can penetrate deeper,
below. A confocal microscope selectively col- up to 1 mm in some circumstances.
lects light from a thin (<1 μm) optical sec- Several types of confocal microscopes are
tion at the plane of focus in the specimen available. The most common type is the laser
(Fig. 2.2.1). Structures within the focal plane scanning confocal microscope (LSCM), which
appear more sharply defined than with a con- captures images by scanning the specimen
ventional microscope because there is essen- with a focused beam of light from a laser
tially no flare of light from out-of-focus areas. and collecting the emitted fluorescence sig-
A three-dimensional view of the specimen can nals with a photodetector. LSCMs sometimes
be reconstructed from a series of optical sec- are referred to as “spot-scanning” confocal
tions at different depths (Fig. 2.2.2). microscopes, to distinguish them from mi-
The capability for optical sectioning makes croscopes that scan the specimen with a slit
confocal microscopy well suited for study- of light (slit-scanning) or multiple spots of
ing the structure and function of cells using light (spinning-disk or Nipkow disk). Spot-
immunofluorescence reagents (Fig. 2.2.1C; scanning LSCMs typically have slower im-
Goldstein and Watkins, 2008), organic dyes age acquisition rates than slit-scanning or
(Fig. 2.2.1C), fluorescent fusion proteins spinning-disk microscopes (<1 frame/sec ver-
(Fig. 2.2.1D; Fig. 2.2.2; Kahana and Silver, sus 30 frames/sec or higher). However, they
1996), quantum dots (Michalet et al., 2005), are more versatile in a number of ways. They
and fluorescence in situ hybridization (FISH; can accommodate lasers with a wide range
Knoll et al., 2007). Confocal imaging in living of wavelengths (from the UV to the infrared)
specimens is feasible, making it possible to and can be configured to image multiple fluo-
study dynamic processes such as vesicle traf- rophores either simultaneously or sequentially.
ficking (Fernandez-Alfonso and Ryan, 2004), Some include spectral detectors that can cap-
spine motility (Bonhoeffer and Yuste, 2002), ture the entire spectrum of the fluorescence
cytoskeletal assembly and turnover, chromo- emitted at each pixel in the image. The most
Imaging

Current Protocols in Neuroscience 2.2.1-2.2.18, July 2011 2.2.1


Published online July 2011 in Wiley Online Library (wileyonlinelibrary.com).
DOI: 10.1002/0471142301.ns0202s56 Supplement 56
Copyright C 2011 John Wiley & Sons, Inc.
Figure 2.2.1 Applications of laser scanning microscopy. (A,B) Imaging in thick specimens. Neu-
rons in a Drosophila embryo were immunolabeled with antibodies against three different transcrip-
tion factors (images provided by Dr. Ward Odenwald of the National Institutes of Health, Bethesda,
Md.; reproduced from Kamabadur et al., 1998, by permission of Cold Spring Harbor Laboratory
Press). (A) A single optical section (∼2.5 μm) captured with a 25×, 0.8-NA objective. Labeled
neurons in the plane of focus appear sharply defined, while those outside it are not visualized.
(B) A maximum projection of 65 optical sections collected at 2-μm intervals in the z axis. (C)
Imaging intracellular structures. Dissociated rat fibroblasts were immunolabeled with anti-tubulin
antibodies to visualize microtubules (green), and stained with fluorescent probes for mitochondria
(Mitotracker, red) and DNA (DAPI, blue). The image is a projection of 20 optical sections (0.3-μm
intervals) captured with a 100×, 1.4-NA objective. (D) Measuring molecular mobility in living cells.
In a living fibroblast expressing a Golgi membrane protein (galactosyltransferase) fused to GFP
(S65T), GFP fluorescence (green) is localized in the Golgi complex, shown superimposed on a DIC
image of the cell. After the first image was collected, the boxed region (yellow) was scanned with
full laser power to photobleach the GFP in the boxed area. The second image was collected 2 sec
later. Subsequent images (not illustrated) showed that the GFP-galactosyltransferase rapidly dif-
fused back into the photobleached area. Images were captured with an LSM410 (Carl Zeiss, Inc.).
For the color version of this figure go to http://www.currentprotocols.com/protocol/ns0202.

sophisticated LSCMs allow the user to con- Lippincott-Schwartz et al., 2003; Sprague and
trol the illumination wavelength and inten- McNally, 2005). Photosensitive molecules in-
sity on a microsecond time scale. This feature clude certain fluorescent proteins (e.g., see
makes it possible to perform experiments that Patterson and Lippincott-Schwartz, 2002, for
require selectively illuminating fluorophores the fluorescent protein PaGFP, and Ando et al.,
in a defined region of interest in order to 2002, for the photosensitive protein Kaede),
photobleach (Fig. 2.2.1D) or photoactivate “caged” molecules such as caged Ca2+ chela-
them. Measurement of fluorescence recovery tors, neurotransmitters, and second messen-
after photobleach (FRAP) or fluorescence loss gers (Nerbonne, 1996). Confocal microscopy
in photobleach (FLIP) can provide informa- also can be used to measure fluorescence res-
Basic Confocal tion about molecular mobility and binding onance energy transfer (FRET; Wouters and
Microscopy (Cole et al., 1996; McNally and Smith, 2002; Bastiaens, 2000).

2.2.2
Supplement 56 Current Protocols in Neuroscience
D

xy

0 1 2 3 4 5 μm
H

xy

0 1 2 3 4 5 μm

Figure 2.2.2 Three-dimensional imaging in living specimens. Comparison of water- and oil-
immersion objectives. Living yeast cells expressing a GFP construct that targets the mitochondrial
matrix were embedded in an aqueous solution with 0.2% agarose and visualized with (A,B) a
C-APO 63×, 1.2-NA water-immersion objective or (E,F) a Plan Apochromat 100×, 1.4-NA oil-
immersion objective. The images show xy (A,E) and yz (B,F) projections of stacks of 40 images
collected at 0.2-μm intervals along the optical axis. The xy projections appear sharper than the
yz projections because the resolution is higher in the focal plane of the objective than along the
optical axis. (C,G) yz projections of images of 0.19-μm fluorescent beads embedded in an aqueous
solution with 2% agarose and captured with a 63× water-immersion (C) or 100× oil-immersion (G)
objective. (D,H) Intensity profiles along the horizontal and vertical axes of the beads. A 63×, 1.2-NA
water-immersion objective (D) provides better axial resolution than a 100×, 1.4-NA oil-immersion
objective (H) in specimens in an aqueous solution. Scale bars = 5 μm (A,B,E,F); 0.5 μm (C,G).
Images were captured with an LSM510 laser scanning confocal microscope (Carl Zeiss, Inc.).

The purpose of this unit is to provide through the pinhole to reach the detector. The
background information and practical tips for physical basis of optical sectioning is illus-
optimizing confocal imaging. The first sec- trated in Figure 2.2.3. The microscope objec-
tion (Basis of Optical Sectioning) explains tive focuses light from a point source (a laser)
the basic principle of confocal imaging as to a diffraction-limited spot in the specimen.
implemented in LSCM. The second section The irradiation is most intense at the focal spot,
(Configuration of an LSCM) describes the but areas of the specimen above and below
components and light path in a typical LSCM the focal spot are also illuminated. Fluores-
and compares this with the light paths of spot- cent molecules excited by the incident light
scanning microscopes and a new type of slit- emit fluorescence in all directions. The objec-
scanning confocal microscope. The third sec- tive captures a portion of the emitted light. The
tion (Practical Guidelines) provides guidelines objective projects light from the focal spot in
for preparing specimens and configuring the the specimen to a conjugate spot in an “image”
critical parameters for confocal imaging. The plane. The pinhole aperture is positioned in
Commentary provides references to sources of the image plane so as to be centered on this
additional information. spot. The light that passes through the aperture
is detected by a photomultiplier tube (PMT).
Light from out-of-focus areas of the specimen
BASIS OF OPTICAL SECTIONING is spread out at the image plane and is largely
Confocal microscopes accomplish optical blocked by the pinhole aperture.
sectioning by scanning the specimen with a The diameter of the pinhole determines how
focused beam of light and collecting the fluo- much of the fluorescence emitted by the illu-
rescence signals emitted by the specimen via a minated cone in the specimen is detected, as
pinhole aperture. The pinhole aperture blocks well as the thickness of the optical section.
signals from out-of-focus areas of the speci- From wave optics, it is known that a point light
Imaging
men, whereas light from the focal plane passes source in the plane of focus of an objective
2.2.3
Current Protocols in Neuroscience Supplement 56
photodetector

confocal pinhole

dichroic mirror

point
light
source

objective lens

specimen
focal plane

Figure 2.2.3 The basis of optical sectioning. Illumination from a point light source is reflected
by a dichroic mirror into the back aperture of a microscope objective. The objective lens focuses
the light to a diffraction-limited spot within the specimen. Fluorophores at the focal spot and within
the cones of illumination above and below it are excited, emitting fluorescence in all directions.
The fluorescence captured by the objective passes through the dichroic mirror because the fluo-
rescence is at a longer wavelength than the excitation. The confocal pinhole allows fluorescence
from the focal spot to reach the photodetector and blocks fluorescence from out-of-focus areas.
Redrawn from Shotton (1993).

produces a three-dimensional diffraction pat- of-focus background achieved by a properly


tern in the image plane. The cross-section at adjusted pinhole is the principal advantage of
the image plane is an Airy disk, a circular confocal microscopy for examination of thick
diffraction pattern with a bright central region. specimens.
The radius of the bright central region of the Point illumination and the presence of a
Airy disk in the reference frame of the spec- pinhole in the detection light path also produce
imen is given by RAiry = 0.61λ/NA, where λ improved lateral and axial resolution relative
is the emission wavelength and NA is the nu- to conventional microscopy (Table 2.2.1). The
merical aperture of the objective (Inoué and actual extent of improvement depends on the
Spring, 1997). At the image plane, the ra- size of the pinhole. Near-maximal axial res-
dius of the central region is RAiry multiplied olution is obtained with a pinhole radius of
by the magnification at that plane (Wilson, ∼0.7 × RAiry , whereas optimal lateral resolu-
1995). tion is obtained with a pinhole smaller than
Adjustment of the pinhole to a diameter 0.3 × RAiry (Wilson, 1995). However, a pin-
slightly less than the diameter of the central hole smaller than ∼0.7 × RAiry significantly
region of the Airy disk allows most of the reduces the total signal, a sacrifice that may
light from the focal point to reach the detec- not be worth the gain in resolution, espe-
tor and reduces the background from out-of- cially when imaging dim samples. In fluores-
focus areas by ∼1000-fold relative to wide- cence imaging, resolution is also influenced
field microscopy (Sandison et al., 1995). The by the emission and excitation wavelengths
Basic Confocal
Microscopy separation of the in-focus signal from the out- (Table 2.2.1).

2.2.4
Supplement 56 Current Protocols in Neuroscience
Table 2.2.1 Theoretical Resolutions of Confocal and Conventional Microscopesa

Objective
10×, 0.4 NA, air 40×, 0.85 NA, air 60×, 1.4 NA, oil
λex /λem b
Lateral Axial Lateral Axial Lateral Axial

Confocal fluorescence microscope


488/518 0.55 4.50 0.26 0.99 0.16 0.56
568/590 0.64 5.17 0.30 1.09 0.18 0.64
647/677 0.72 5.88 0.34 1.28 0.21 0.72
Conventional fluorescence microscope
518 0.79 6.48 0.37 1.43 0.24 0.93
590 0.90 7.38 0.42 1.63 0.28 1.06
680 1.04 8.50 0.49 1.88 0.32 1.22
a Data reprinted from Brelje et al. (1993) by permission of Academic Press.
bλ and λem , excitation and emission wavelengths (nm).
ex

CONFIGURATION OF AN LSCM The light path in a simple confocal micro-


Confocal microscopes use lasers for illu- scope is illustrated in Figure 2.2.4. The output
mination because they provide intense exci- of the laser (or the combined output of multi-
tation within a narrow range of wavelengths. ple lasers) is reflected into the optical axis of
Mixed krypton-argon gas lasers are popular the microscope by the primary dichroic beam
for multicolor confocal microscopy because splitter (splitter 1 in Fig. 2.2.4). Wavelength-
they emit at three wavelengths (488, 568, and selection filters are inserted into the light
647 nm) that excite many commonly used path to block specific laser lines, and neutral-
fluorophores—e.g., fluorescein (FITC), rho- density filters may be inserted to attenuate
damine, Cy3, Cy5, Alexa 488/555/568/647, the illumination. In current, high-end confocal
green fluorescent protein (GFP), and red flu- systems, the line selection and neutral-density
orescent protein (mRFP or DsRed). The dis- filters have been replaced with an electroni-
advantage of krypton-argon lasers is that their cally controlled acousto-optical tunable filter
life spans are short (∼2000 hr). Another way to (AOTF). An AOTF can alter its transmission
achieve multiwavelength excitation is to com- characteristics to allow selected wavelengths
bine the outputs of multiple lasers. Many of the to pass, while completely blocking others. An
confocal microscopes currently on the market AOTF also provides precise control over the
combine an argon laser (488 nm) with a green attenuation of the individual laser beams.
helium-neon (HeNe) laser (543 or 594 nm) and The scanner deflects the laser beam into
a red HeNe laser (633 nm). The argon laser also the objective at varying angles in order to scan
may provide 458- and 514-nm lines, which can the laser beam across the specimen. Several
be used to excite the cyan and yellow variants different technologies for scanning have been
of GFP (CFP and YFP). Some confocal mi- devised. The most common method employs
croscopes can accommodate a 405-nm diode a pair of galvanometer mirrors. One mirror os-
laser. The 405-nm laser is more optimal for ex- cillates rapidly to excite sequential spots along
citation of CFP than the 458-nm line of the ar- the x-axis of the specimen, and the second mir-
gon laser, and also excites photosensitive GFP ror oscillates more slowly to move the illumi-
(PaGFP). It can even be used to visualize some nation from line to line in the y-axis.
UV fluorophores such as DAPI and Hoechst The fluorescence emissions that are col-
DNA dyes, although 405 nm is not the opti- lected by the objective follow the reverse path
mal excitation wavelength for these dyes. UV through the scanner to the primary dichroic
argon lasers (351/364 nm) also are available. beam splitter, and are thereby “descanned”
Inclusion of a 405-nm or UV argon laser adds (Fig. 2.2.4). The fluorescence signals (which
considerably to the cost of the confocal mi- are at a longer wavelength than the excitation
croscope system due to the requirement for due to the Stokes shift) are transmitted through
additional optical components to handle these the beam splitter. To simultaneously image flu-
wavelengths. orescence from multiple fluorophores requires Imaging

2.2.5
Current Protocols in Neuroscience Supplement 56
krypton-argon
laser
variable emission
pinhole filter
specimen

line-selection filter
objective
photomultiplier
neutral-density filter
tube 2

x
mirror
y
dichroic dichroic photomultiplier
microscope scanner
beam beam tube 1
splitter 1 splitter 2

488-nm laser beam lissamine rhodamine


FITC 568-nm laser beam

Figure 2.2.4 The light path of a laser-scanning confocal microscope (LSCM) set up for simulta-
neous imaging of FITC and lissamine rhodamine. The 488- and 568-nm lines of a krypton-argon
laser are reflected by dichroic beam splitter 1 into the optical axis of the microscope. The beam is
reflected by a mirror into the microscope objective, which focuses the beam to a diffraction-limited
spot in the specimen. The scanner consists of a pair of galvanometer mirrors that deflect the laser
beams so as to scan the spot across the specimen in a raster pattern. Fluorescence emitted as
each point is illuminated travels the reverse path through the scanning system. The FITC fluores-
cence (peak at 520 nm) and lissamine rhodamine fluorescence (peak at 590 nm) pass through
dichroic beam splitter 1 to dichroic beam splitter 2, which transmits the lissamine rhodamine fluo-
rescence to photomultiplier tube 1 and reflects the FITC fluorescence to photomultiplier tube 2. A
variable pinhole in front of each photodetector blocks light from out-of-focus areas of the specimen
while allowing light from the focal plane to reach the detector.

selection of a primary dichroic beam split- the diameter of the pinhole to a value of 0.7
ter that reflects each of the required excita- to 1.0 Airy unit allows most of the in-focus
tion wavelengths and transmits the emissions light to reach the detector and blocks most of
of all of the fluorophores. Secondary dichroic the out-of-focus light. In systems with a sep-
mirrors split the fluorescence emissions from arate pinhole aperture for each detector, the
different fluorophores for detection by sepa- pinhole apertures are located immediately in
rate detectors. Emission filters are inserted in front of the detectors. Incorporation of a sep-
the light path to the detectors (Fig. 2.2.4) to arate pinhole for each detector allows the user
block back-scattered excitation light and to re- to optimize the pinhole settings for different
duce bleed-through of signals between chan- wavelengths.
nels. Current high-end confocal microscopes The photodetectors in LSCMs are photo-
use more sophisticated technology for emis- multiplier tubes (PMTs), which generate elec-
sion discrimination; descriptions of the de- trons at a rate proportional to the intensity of
signs of specific systems are available from the incoming fluorescence signal (Art, 2006).
the vendors. The PMT output is converted to a digital image
The fluorescence captured by the objective that can be displayed on a computer monitor
focuses to a stationary spot (Airy disk) in the and stored as a digital file for later analysis.
image plane (Fig. 2.2.3). The pinhole aperture Digitization may be at 8-bit (256 gray lev-
is positioned in the image plane so as to be els), 10-bit (1024 gray levels), or 12-bit (4096
centered on the Airy disk. The diameter of the gray levels) resolution . Conofcal microscopes
pinhole aperture can be adjusted to allow op- typically have two to four PMTs for reflected
timization for different Airy-disk sizes, which light/epifluorescence imaging and may have,
vary with the objective’s numerical aperture in addition, a photodetector for transmitted
Basic Confocal and the emission wavelength. Adjustment of light.
Microscopy

2.2.6
Supplement 56 Current Protocols in Neuroscience
In spinning-disk confocal microscopes, the aperture in front of the detector blocks out-of-
illumination from a laser or white light source focus light, analogous to the pinhole aperture
passes through pinholes in the Nipkow disk in an LSCM. The LSM 5 Live has somewhat
so as to excite fluorescence at multiple sites poorer resolution than a spot-scanning LSCM,
(∼1000) within the specimen. The disk re- but can capture images much more rapidly.
volves rapidly (1000 to 5000 rpm), causing the
illuminating spots to sweep across the spec-
imen as uniformly spaced scan lines (Inoué PRACTICAL GUIDELINES
and Inoué, 2002; Toomre and Pawley, 2006). Sample Preparation
Fluorescence emitted by the specimen that The pre-eminent goal in preparing samples
is collected by the objective returns through for imaging with a confocal microscope is to
the same pinholes in the Nipkow disk that maximize the fluorescence signals while pre-
provided the excitation light before it is de- serving the three-dimensional structure of the
tected by a full-field CCD camera. In this specimen. Ideally, the sample should be less
way, point light sources and detector pinholes than ∼50 μm in thickness, although thicker
to block out-of-focus fluorescence are pro- samples can be visualized. Guidelines for
vided, with the advantage of higher collec- preparing fixed and living samples are de-
tion speeds than a spot scanner. Drawbacks scribed below.
of this approach include decreased illumina-
tion to the specimen from light loss through
Fixation
the pinholes, and the inability to change the
A standard fixative for fluorescence mi-
pinhole diameter. This means that, unlike the
croscopy is 2% to 4% formaldehyde in
case with a spot scanner, optimal confocal-
PBS. Formaldehyde penetrates cells rapidly
ity is achieved only for one objective magni-
and preserves the antigen-recognition sites
fication, and the thickness of the optical sec-
for many antibodies. However, formaldehyde
tion cannot be changed. Although the reduced
cross-links proteins slowly and may cause
specimen illumination generates a smaller flu-
vesiculation of membranes. Some commercial
orescent signal, scientific-grade CCD cameras
preparations of formaldehyde (formalin) con-
have significantly higher quantum efficiencies
tain methanol, which shrinks cells. Techniques
than the PMTs used for fluorescence detection
for optimizing formaldehyde fixation are de-
in LSCMs and are able to detect these levels
scribed by Bacallao et al. (2006). Fixatives
of fluorescence more than adequately. Fluores-
containing a small amount of glutaraldehyde
cent specimens have been reported to undergo
(0.125% to 0.25%) in addition to formalde-
less photobleaching during examination with a
hyde preserve cellular morphology better, but
spinning-disk confocal microscope than with
glutaraldehyde destroys the epitopes for some
an LSCM. The lower rate of photobleaching
antibodies. Glutaraldehyde fixation induces
is thought to be due to the lower illumination
autofluorescence but autofluorescence can be
levels (Inoué and Inoué, 2002).
reduced by treating the sample after fixation
A new type of slit-scanning confocal mi-
with NaBH4 (1 mg/ml in PBS, pH 8.0, using
croscope (LSM 5 Live; Carl Zeiss, Inc.) has
two treatments of 5 min each for dissociated
recently been introduced that allows images to
cells, longer for thicker samples). An alter-
be acquired at rates as fast as or faster than
native procedure for preparing specimens for
can be achieved with a spinning-disk confo-
immunocytochemistry is to immerse them in
cal microscope, and with as low or lower rates
cold (−20◦ C) methanol or acetone but fixation
of photobleaching. The system adopts princi-
by this method causes severe shrinkage.
ples from both the spot scanner and the spin-
ning disk in that it uses a single scanning gal-
vanometer to move an illumination line that is Choices of fluorophores
combined with a sensitive single-line CCD de- The choice of fluorophores should take into
tector. The point source of light from the laser account the available laser lines and the detec-
is optically converted to a narrow line, which tor channels of the confocal microscope. Ex-
is reflected onto the specimen by a novel beam citation is most efficient at wavelengths near
splitter consisting of a mirrored line on trans- the peak of the excitation spectrum of the flu-
parent glass. The line illumination is scanned orophore, but a precise match is not required.
across the specimen. The emitted fluorescence For experiments that require imaging mul-
from the specimen that is collected by the ob- tiple fluorophores with standard photodetec-
jective passes through the beam splitter and tors (PMTs), it is best to select fluorophores
Imaging
is detected by a linear CCD detector. A slit that are excited by different laser lines, in
2.2.7
Current Protocols in Neuroscience Supplement 56
Marina Alexa Alexa Alexa
Blue 488 555 647
100

90

80

Relative excitation efficiency


70

60

50

40

30

20

10

0
300 350 400 450 500 550 600 650 700 750
Wavelength (nm)

krypton-argon
488 568 647

argon
458 488 514

UV argon
488 514

helium-neon
546 561 594 633

diode
405

Figure 2.2.5 Excitation spectra of representative fluorophores and emission wavelengths of


lasers for confocal microscopy. The graph at the top shows the excitation spectra of Marina Blue,
Alexa 488, Alexa 555, and Alexa 647 (Molecular Probes). The emission wavelengths of lasers
commonly used for confocal microscopy are shown below. Data for the excitation spectra are from
Molecular Probes.

order to minimize spectral crossover (bleed- imaging with a spectral detector and spectral
through) between the channels (Fig. 2.2.5). unmixing, it is important to select fluorophores
Excitation and emission spectra for many flu- that have distinct emission spectra, but there is
orophores are available via the Internet (see no advantage in using fluorophores that have
Internet Resources). A recommended combi- differing excitation spectra. Indeed, it is best
nation of fluorophores for excitation at 405 nm, to use fluorophores that have similar excita-
488 nm, 543/561 nm, and 633 nm is Pacific tion maxima, so that they can be excited with
Blue or Alexa 405, Alexa 488, Alexa 555, a single laser line reflected into the micro-
and Alexa 647 (all available from Molecu- scope with a single-wavelength dichroic mir-
lar Probes/Invitrogen). The nucleic acid stain ror. Other important criteria to consider in se-
DAPI can be excited by illumination at lecting fluorophores for confocal microscopy
405 nm, although ultraviolet excitation (350 are the quantum efficiencies and rates of pho-
nm) is more optimal. The cyanine dyes Cy2, tobleaching. In addition, the staining protocol
Cy3, and Cy5 (available from Jackson Im- should be designed to produce similar signal
munoResearch Laboratories) are also suitable intensities in each channel. For more informa-
Basic Confocal
Microscopy for confocal microscopy. For multiwavelength tion on fluorophores for confocal imaging, see

2.2.8
Supplement 56 Current Protocols in Neuroscience
Tsien et al. (2006), Giepmans et al. (2006), the mounting medium should have the same
and the Molecular Expressions Web site (see refractive index (RI) as the objective immer-
Internet Resources). sion medium. Mismatches in the refractive in-
dices produce spherical aberration leading to
loss of light at the detector, as well as de-
Control samples
creased z-axis resolution and incorrect depth
Confocal microscopes rely on electronic
discrimination. Image deterioration caused by
image enhancement techniques that can make
spherical aberration increases with depth into
even dim autofluorescence signals or nonspe-
the specimen; therefore, matching the immer-
cific background staining look bright. In or-
sion and mounting medium refractive indices
der to distinguish a real signal from back-
is particularly important for thick specimens.
ground, it is essential to prepare and examine
The refractive indices of some commonly used
appropriate control samples. For immunoflu-
mounting media are listed in Table 2.2.2.
orescence experiments with one primary an-
Mounting media that have refractive in-
tibody, the appropriate control samples are
dices close to that of immersion oil (n = 1.51)
unstained specimens and specimens treated
include DPX (n = 1.5; ProSciTech) and Per-
with the secondary antibody but no primary
mount (n = 1.52; ProSciTech). However, spec-
antibody. Other control experiments may be
imens must be dehydrated prior to mounting in
required to verify the specificity of label-
these media, and dehydration causes shrinkage
ing. Experiments with primary and secondary
and distortion. Moreover, some fluorophores
antibodies require additional controls to test
cannot withstand dehydration. Cells retain
whether the secondary antibodies cross-react
their three dimensional shapes when they are
with the “wrong” primary antibody. Singly
kept in physiological saline (PBS) or a mixture
stained samples also should be prepared and
of PBS and glycerol (Bacallao et al., 2006). A
imaged to determine the extent of spectral
water-soluble mounting medium with a refrac-
cross-over between the channels.
tive index matching that of immersion oil can
be made with 97% 2,2 -thiodiethanol (TDE;
Mounting the specimen Sigma-Aldrich, cat. no. 88559) and 3% PBS
Selection of a mounting medium should (Staudt et al., 2007). The specimen should be
take into account the type of microscope ob- successively immersed in increasing concen-
jective that will be used to observe the spec- trations of TDE in PBS (10:90, 25:75, 50:50
imen (see section on Microscope objectives). each for 5 to 10 min, followed by three changes
In order for an objective to perform optimally, of 97:3 TDE:PBS). If the specimen is to be

Table 2.2.2 Refractive Indexes of Common Immersion and Mounting Media

Medium RI

Immersion media
Air 1.00
Water 1.338
Glycerol 1.47
Immersion oil 1.518
Mounting media
50% glycerol/PBS/DABCO 1.416a
5% n-propyl gallate/0.0025% p-phenylene diamine (PPD) in 1.474a
glycerol
0.25% PPD/0.0025% DABCO/5% n-propyl gallate in glycerol 1.473a
VectaShield (Vector Labs) 1.458a
Slow Fade (Molecular Probes) 1.415b
ProLong Gold (Molecular Probes) 1.46b,c
a Data from Bacallao et al. (2006).
b Data from Molecular Probes.
c After curing. Imaging

2.2.9
Current Protocols in Neuroscience Supplement 56
mounted under a coverglass, it may be neces- medium, and then the coverglass with cells at-
sary to support the coverglass to avoid damag- tached is sealed onto the well. More elaborate
ing the specimen. The coverslip edges should chambers, some having built-in heaters and/or
be sealed with transparent nail polish. ports for perfusion, are available from com-
Addition of an antioxidant (antifade agent) mercial sources (see Internet Resources for a
to the mounting medium helps to alleviate pho- list of suppliers).
tobleaching of synthetic fluorophores such as Specimens that need to be kept warm dur-
those used for immunocytochemistry. One of ing observation pose a particular challenge be-
the best antifade agents is 100 mg/ml 1,4- cause temperature fluctuations can make it dif-
diazabicyclo[2,2,2]octane (DABCO; Sigma; ficult to maintain focus. Probably the best way
Bacallao et al., 2006). n-Propyl gallate (Giloh to keep specimens warm is to place the entire
and Sedat, 1982) and p-phenylenediamine microscope in a temperature-controlled en-
(PPD; Johnson et al., 1982) are also effective closure. Alternative strategies include warm-
antifade agents, but the former may cause dim- ing the microscope stage with heated air (us-
ming of the fluorescence while the latter may ing an air stream incubator or hair dryer)
damage the specimen (Bacallao et al., 2006). or infrared lamps, or using a temperature-
A wide variety of mounting media is available controlled specimen chamber (Terasaki and
from commercial sources (Biomeda, Electron Dailey, 1995; Dailey et al., 2006). If an oil-
Microscopy Sciences, ProSciTech, Invitrogen, or water-immersion objective is used, heating
Vector Laboratories), and many of these con- the objective helps to maintain the specimen
tain antifade agents. It is wise to check with at the desired temperature. Microscope enclo-
the fluorophore provider for recommendations sures, stage warmers, temperature-controlled
about which mounting media and antifade chambers, and objective heaters are available
agents to use. Antioxidants do not reduce pho- from suppliers of microscopes and microscope
tobleaching of fluorescent proteins. accessories.
Living specimens should be kept in a
Living specimens medium that is buffered to maintain the correct
Microscopy on living specimens grown pH. Many commonly used culture media are
in vitro is most conveniently performed with buffered with bicarbonate and require an atmo-
an inverted microscope, because the speci- sphere with 5% to 10% CO2 to maintain the
mens can be viewed through the bottom of the correct pH. For microscopy, it is more conve-
culture chamber and the top can be opened for nient to use a buffer that maintains the correct
access. To allow imaging with an oil- or water- pH in air. Many types of cells can be main-
immersion objective, the culture chamber sub- tained for several hours in a balanced saline so-
strate should be a coverglass. The coverglass lution or culture medium that is buffered with
should be 0.17 mm thick (no. 1.5 thickness) HEPES (10 to 20 mM). Use of a medium that
for optimal performance of standard micro- contains phenol red should be avoided because
scope objectives. The coverglass can be coated phenol red adds background fluorescence and
with poly-L-lysine (using a 1 mg/ml solution; can produce oxygen radicals when exposed
Sigma) to promote adhesion of the specimens. to intense illumination. Addition of 0.3 U/ml
Nonadherent specimens can be immobilized Oxyrase (Oxyrase, Inc.) to the medium can
by embedding them in a thin layer of low- help to alleviate photobleaching of synthetic
melting-point agar (0.2%). Culture chambers fluorophores (Waterman-Storer et al., 1993).
with coverglass substrates can be made from
standard plastic petri dishes by boring holes
in their bottoms and affixing coverglasses to
Optimizing Imaging Parameters
the holes with Silgard (Dow-Corning). Culture Microscope objectives
chambers with coverglass substrates are also High-NA objectives are optimal for flu-
available from commercial sources (Labtek orescence microscopy because they col-
coverglass chamber, Fisher Scientific; MatTek lect more light than low-NA objectives
glass-bottom culture dish, MakTek Corp). Al- (brightness is proportional to NA4 ). Oil-
ternatively, cells may be grown on a coverglass immersion objectives have the highest nu-
that can be mounted in a chamber for observa- merical apertures (NA = 1.4 to 1.46). How-
tion on a microscope. A simple chamber can be ever, oil-immersion objectives have short
constructed from a gasket cut from a sheet of working distances (100 to 200 μm). More-
silicon rubber or a soft plastic ruler and affixed over, they work optimally only with speci-
Basic Confocal to a glass microscope slide with silicon grease. mens mounted in a medium with a refractive
Microscopy
The well formed by the gasket is filled with index the same as that of immersion oil
2.2.10
Supplement 56 Current Protocols in Neuroscience
(n = 1.51). Mismatch of the refractive indices Pawley, 1995, 2006), although not as good as
leads to a deterioration of image quality that can be achieved with a smaller pinhole. Lat-
becomes increasingly severe with depth into eral resolution continues to improve as pin-
the specimen (Fig. 2.2.2). When a high-NA oil hole radius is decreased down to a pinhole
objective is used to image a specimen mounted size of ∼0.2 × RAiry , but a pinhole this small
in an aqueous medium, image quality and sig- excludes ∼95% of the signal (Wilson, 1995).
nal brightness decline noticeably at distances Axial resolution improves as pinhole size de-
of 5 to 10 μm from the coverglass. Mismatch creases, down to ∼0.7× RAiry , then levels off.
of the refractive indices also leads to spatial The best trade-off between signal intensity and
distortion in the z-axis. The actual movement resolution will depend on the characteristics of
of the focal plane in the specimen (ds ) pro- the sample and the required resolution.
duced by a movement of the objective (dobj )
depends on the ratio of the refractive indices:
Scan zoom
ds /dobj = ns /nobj (Majlof and Forsgren, 2002).
The scan zoom determines the dimensions
A water-immersion objective is useful for
of the area in the specimen that is scanned.
imaging living specimens that are more than
Increasing the zoom reduces the dimensions
a few microns thick (Fig. 2.2.2). Water-
of the scan area. The pixel number remains
immersion objectives with numerical aper-
the same; consequently, individual pixels rep-
tures of 1.2 are available. These objectives are
resent a smaller area. For example, the scan
designed for viewing specimens mounted un-
area at zoom 2 is one quarter the scan area at
der a coverglass (0.17 μm; no. 1.5) and have
zoom 1, and the pixel dimensions are half as
fairly short working distances (130 to 220 μm).
large in each dimension. That is, if the pixel
“Dipping” objectives, which are intended for
dimensions represent 0.25 μm × 0.25 μm
use without a coverglass, have lower numeri-
at zoom 1, then dimensions are 0.125 ×
cal apertures (NA = 0.9) and longer working
0.125 μm at zoom 2.
distances (1 to 2 mm).
For each objective, there is an optimal
Objectives differ in their transmission ef-
zoom setting that yields pixel dimensions
ficiency and degree of correction for spheri-
small enough to take advantage of the full
cal and chromatic aberration and flatness of
resolution of the objective, but large enough
field. Plan Apochromat objectives provide the
to avoid oversampling. In order for the min-
flattest fields of view and color correction for
imum resolvable entity to be visible on the
three wavelengths. Plan Apochromat objec-
display monitor, the pixel dimension needs to
tives generally transmit efficiently throughout
be smaller than (less than one-half) the optical
the visible spectrum (400 nm to 700 nm), but
resolution. However, if the pixel size is made
may transmit poorly in the UV (<400 nm)
too small by using a higher-than-optimal zoom
or infrared (>700 nm; Keller, 1995, 2006).
factor, the specimen is subjected to more irra-
Some objectives that are less highly corrected
diation than necessary, with an increased risk
(Fluar, Plan NeoFluar, Plan Fluor) provide
of photobleaching. The rate of photobleach-
higher transmission at visible, UV, and in-
ing increases proportionally to the square of
frared wavelengths. For additional informa-
the zoom factor (Centonze and Pawley, 1995,
tion about objectives for confocal microscopy,
2006). A guideline for selecting an appropriate
see Salmon and Canman (1998), Keller (1995,
zoom factor derived from information theory
2006), Benham (2002), and the Molecular
(the Nyquist Sampling Theorem) states that
Expressions Web site (see Internet Resources).
the pixel dimensions should be equal to the
optical resolution divided by 2.3 (see Webb
Pinhole size and Dorey, 1995; Pawley, 2006). The opti-
As explained in the section on the Basis of cal resolution in confocal imaging depends
Optical Sectioning, the size of the detector pin- on the numerical aperture of the objective,
hole has a critical influence on image quality. the refractive index of the immersion medium,
A pinhole with a diameter slightly less than the excitation and emission wavelengths, and
or equal to the diameter of the bright central the diameter of the pinhole aperture. Values
region of the Airy disk will let most of the calculated for different objectives and wave-
light from the plane of focus reach the detec- lengths using the point-spread functions (PSF)
tor, while blocking most of the out-of-focus for wide-field and confocal microscopy are
flare. The lateral resolution will be ∼10% bet- given in Table 2.2.1. The lateral resolution
ter than that obtainable by conventional mi- for confocal microscopy can be approximated
croscopy with the same optics (Centonze and by: Reselx,y confocal = 0.4λ/NA (see Webb and Imaging

2.2.11
Current Protocols in Neuroscience Supplement 56
Dorey, 1995). This equation assumes the use of titative imaging experiments. Confocal imag-
an infinitesimal pinhole; Reselx,y will be larger ing software typically includes a pseudocolor
with a pinhole of 0.7 to 1 Airy unit. image display mode (“range indicator”) that
facilitates selection of appropriate offset and
z-axis sectioning interval gain settings by highlighting pixels with in-
In order to study the three-dimensional tensity values near 0 or 255.
structure of a specimen, a series of images are
captured at fixed intervals throughout the en- Reducing noise
tire depth of the specimen. The interval needed Confocal images are inherently noisy due
between focal planes to achieve optimal res- to the statistics of photon emission and limited
olution in the z-axis is not as small as the quantum efficiency of the detectors (Pawley,
x,y pixel dimensions because the axial res- 2006). Improved signal-to-noise ratios can be
olution is poorer than the lateral resolution attained by scanning the specimen at a slower
(see Table 2.2.1). The optimal interval (ac- rate or by scanning multiple times and av-
cording to the Nyquist Sampling Theorem) eraging the signals. Current LSCMs allow
is equal to the axial resolution divided by individual lines in the image to be scanned
2.3. The axial resolution for an objective in repeatedly and averaged. Line averaging gen-
confocal imaging can be approximated by: erally produces sharper images than frame av-
Reselz confocal = 1.4λn/NA2 , where n is the re- eraging (which averages full frames) because
fractive index (see Webb and Dorey, 1995). there is less risk of blurring due to movements
Collecting images at shorter intervals results or changes in the specimen. √Averaging reduces
in oversampling, with an increased risk of noise by a factor of (1/ n, where n is the
photobleaching. number of frames or lines averaged), but more
scans will result in more bleaching. Residual
noise can be reduced by image processing (see
Illumination intensity
below).
Fluorescence emission increases linearly
with illumination intensity up to a level at
which emission saturates. Optimal signal-to- Imaging multiple fluorophores
Confocal microscopes can typically be con-
background and signal-to-noise ratios are ob-
figured to capture images of two or more fluo-
tained with illumination levels well below sat-
rophores simultaneously or sequentially. Each
uration (Tsien et al., 2006). The illumination
approach has advantages and disadvantages.
intensity on a laser-scanning microscope can
For simultaneous imaging, the specimen is
be adjusted by operating the laser at submax-
scanned with all of the required excitation
imal power and by inserting neutral-density
wavelengths, and the emissions of the differ-
filters into the light path or varying the trans-
ent fluorophores are split for detection by sep-
mission through the AOTF. In general, the best
arate photodetectors (Fig. 2.2.4). The draw-
images are obtained with illumination levels
back of this approach is that spectral crossover
that are as high as possible without producing
between channels may occur if the emission
unacceptable rates of photobleaching.
spectra of the fluorophores overlap. If each flu-
orophore is excited by only one laser line, then
PMT black level and gain exciting them sequentially will avoid spectral
The contrast and background of confocal crossover. The disadvantage of sequential ex-
images are determined by the gain and offset citation is that there may be misalignment of
settings of the PMT amplifiers. To obtain max- the signals in different channels, particularly if
imal information, the offset and gain should be the specimen is alive and moving. A third way
adjusted to take advantage of the full dynamic of imaging multiple fluorophores is available
range of the PMTs. The appropriate offset set- in confocal systems in which the laser excita-
ting can be found by scanning while the light tion is controlled with an AOTF; such systems
path to the PMT is blocked. The image that can scan each line of the specimen sequentially
appears on the display monitor should be just with different excitation wavelengths with a
barely brighter than the background, which is time delay between scan lines of less than a
black (gray level = 0). To set the gain, scan the millisecond. Line-by-line wavelength switch-
specimen and adjust the gain so that the bright- ing provides rapid acquisition of fluorescence
est pixel in the image is slightly below white signals from each spot in the specimen while
(gray level = 255, for 8-bit images). Ensuring avoiding the spectral crossover between chan-
Basic Confocal that all signals fall within the dynamic range nels that may occur when the fluorophores are
Microscopy
of the PMT is especially important for quan- excited simultaneously.
2.2.12
Supplement 56 Current Protocols in Neuroscience
Image display trast and resolution and reduces noise. Several
Confocal images are typically displayed as algorithms for deconvolution have been de-
8-bit grayscale or 24-bit RGB (red/green/blue) vised that differ in computational intensity and
color images. Each channel of an RGB im- the extent to which they are quantitative (i.e.,
age can represent a different fluorophore retain intensity information). The importance
(Fig. 2.2.1A to C). Color mixtures indicate of eliminating noise for quantitative imaging
colocalization of fluorophores within a pixel. with confocal microscopy is discussed in de-
A RGB fluorescence image can be merged tail by Pawley (2006). Although deconvolution
with a grayscale transmitted-light image by is the most robust method for reducing noise,
adding the transmitted-light image to each it is computationally intensive. An alternative
channel of the RGB image (Fig. 2.2.1D; and quicker method for reducing noise is to
fluorescence in green channel merged with perform a linear deblurring operation (Russ,
DIC image). 2002).
The three-dimensional data set obtained by
capturing a series of optical sections through
COMMENTARY
the specimen can be used to compute views
Effective use of a confocal microscope re-
of the specimen from different viewing an-
quires understanding of the principles of im-
gles. Commercial confocal microscopes typ-
age formation and knowledge of how to set up
ically include the capability to generate or-
and use a microscope. Salmon and Canman
thogonal views of the specimen (xy, xz, and
(1998) describe the components of a light mi-
yz) and may permit views from arbitrary an-
croscope and provide protocols for setting up
gles. An xy projection or “z-series projec-
a microscope for transmitted light and epiflu-
tion” is a two-dimensional display formed by
orescence imaging. This unit also lists refer-
merging multiple image planes (Fig. 2.2.1B,C;
ences to literature on light microscopy. The
Fig. 2.2.2A,E). The most common type of pro-
Handbook of Biological Confocal Microscopy
jection is a “maximum” projection in which
(Pawley, 1995, 2006) is a comprehensive ref-
each pixel represents the intensity of the
erence book on confocal microscopy. It in-
brightest pixel in the z-axis. Another type of
cludes chapters on fundamental principles, in-
projection, referred to as “surface render,” dis-
strumentation, image acquisition and display,
plays the most superficial pixels with intensi-
sample preparation, and much more. Confocal
ties above a defined threshold. Projections also
Microscopy (Wilson, 1990) provides a thor-
can be created for different viewing angles
ough discussion of the principles behind con-
(Fig. 2.2.2B,F). Projections of the specimen
focal imaging. Cell Biological Applications
from different viewing angles can be combined
of Confocal Microscopy (Matsumoto, 2002)
to create an animation in which the specimen
discusses the performance of different types
appears to rotate in space. Such animations
of confocal microscopes and contains prac-
give the viewer a striking impression of the
tical information about common applications
three-dimensional geometry of the specimen.
such as imaging immunofluorescence and cal-
Generating two projections at azimuths differ-
cium ion indicators. Additional applications
ing by 4◦ to 10◦ creates stereo pairs that can
are described in Confocal and Two Photon Mi-
be visualized with a stereo viewer or color-
croscopy: Foundations, Applications and Ad-
coded and merged to form a stereo anaglyph.
vances (Diaspro, 2002). The Molecular Ex-
Volume-rendering imaging software is avail-
pressions Web site also is an excellent source
able that provides additional options for three-
of information about light microscopy, in-
dimensional visualization and measurements
cluding confocal microscopy (see Internet Re-
(see Internet Resources).
sources).
Confocal microscopy is only one of sev-
Image processing eral available techniques for capturing optical
Confocal images can be enhanced by sections in fluorescent specimens. An alterna-
an image processing technique known as tive to confocal microscopy is computational
“deconvolution” or “restoration” (Canell et al., “deconvolution” of images captured by wide-
2006; Holmes et al., 2006). The purpose of field epifluorescence microscopy (McNally
deconvolution is to remove the blur caused et al., 1999; Boccacci and Bertero, 2002).
by diffraction. By measuring or approximat- Computational deconvolution makes use of
ing the point spread function of the optics, one all of the fluorescence captured by the objec-
can mathematically “work backwards” to ac- tive, in contrast to confocal microscopy, which
count for the distorting effects of the optics on discards fluorescence from out-of-focus ar- Imaging
the true image. Deconvolution improves con- eas. In addition, wide-field microscopy can
2.2.13
Current Protocols in Neuroscience Supplement 56
employ CCD cameras that have higher quan- tained depends on the objective and the opti-
tum efficiencies than the photodetectors used cal properties of the specimen. With a high-
for confocal microscopy. For these reasons, NA immersion objective, it may be possible
wide-field microscopy and deconvolution can to capture images at depths of >100 μm in a
be superior to confocal microscopy for imag- specimen that is transparent and not heavily
ing dim specimens or specimens that are sus- stained (Centonze and Pawley, 1995). How-
ceptible to photobleaching or photodamage. ever, if the specimen scatters light, both the il-
However, computational deconvolution of im- lumination intensity and the proportion of the
ages is time-consuming and does not work emitted fluorescence that is captured by the
well in specimens with high levels of dispersed objective decline with increasing focal depth.
fluorescence. Confocal microscopy allows di- Mismatch of the refractive indices of the im-
rect visualization of optical sections and is ap- mersion medium and specimen will further re-
plicable to a wider range of specimens. duce the depth at which adequate signal can
Another technique for confocal imaging be obtained. A low-NA objective can capture
takes advantage of the optical phenomenon images at greater depths but provides much
known as multiphoton excitation. Multiphoton poorer axial resolution.
microscopy allows deeper penetration into tis- A three-dimensional reconstruction of the
sue than either wide-field microscopy or con- specimen can be generated from a series of
ventional (single-photon) microscopy, and is optical sections at appropriately spaced inter-
particularly useful for imaging in thick speci- vals along the optical axis. The reconstruction
mens such as tissue slices or multicellular or- can be viewed from any angle, but the view
ganisms. However, this method suffers from along the optical axis of the objective will
loss of resolution due to the longer illumi- appear sharper than off-axial views, because
nation wavelength and absence of a detector the lateral resolution of the objective is better
pinhole. than the axial resolution (Fig. 2.2.2). The axial
distortion can be corrected by computational
Troubleshooting deconvolution (Wouterlood, 2005).
Test samples are useful for monitoring the Confocal imaging in living specimens is
performance of a confocal microscope. A mi- feasible although care must be taken to avoid
crometer slide should be used to check the spa- phototoxicity and photodamage. Robust flu-
tial calibration of each objective. Fluorescent orophores such as the fluorescent proteins
microspheres with mixtures of fluorophores EGFP and EYFP can be imaged hundreds of
(FluoSpheres; Molecular Probes) are useful times with minimal photobleaching and no
for checking the x,y and axial alignment of apparent phototoxicity, provided that the il-
images acquired at different excitation wave- lumination is kept at a low level. Synthetic
lengths. Misalignment of the images in the xy fluorophores, such as organelle-specific dyes
plane may indicate that the pinholes are not (Molecular Probes), are generally more pho-
centered or that the lasers need to be aligned; tosensitive, although in some applications the
misalignment in the z axis may be due to in- rate of bleaching can be reduced by the addi-
correct setting of a collimating lens, misalign- tion of Oxyrase. The maximum rate at which
ment of pinholes, or chromatic aberration in images can be collected will depend on the
the objective. The optical resolution of the mi- scan speed, resolution, and area. Typical scan
croscope can be measured by capturing im- times for a 512 × 512 image with a spot-
ages of submicroscopic (<0.2 nm) fluorescent scanning LSCM are 1 to 4 sec/frame.
microspheres (Fig. 2.2.2C,G). The images of A common application of confocal mi-
the microspheres should be radially symmet- croscopy is to determine the relative dis-
rical in the xy plane and elliptical in the z-axis tributions and extent of co-localization of
(Fig. 2.2.2C). Horizontal and axial resolutions molecules tagged with different fluorophores
are defined by the full width at half-maximal (Brelje et al., 2002). As many as four dif-
intensity (FWHM) of intensity profiles along ferent fluorophores can be discriminated on
the horizontal and vertical axes of the beads a confocal microscope with laser excitation
(Fig. 2.2.2D,H). at 350/405, 488, 546/568, and 633/647 nm
and standard photodetectors, provided that
Anticipated Results the excitation spectra of the fluorophores are
Confocal microscopy provides sharp im- well separated and matched to the laser lines.
ages of fluorescent structures in thick speci- Confocal microscopes with spectral detectors
Basic Confocal mens (Figs. 2.2.1 and 2.2.2). The maximum can discriminate larger combinations of fluo-
Microscopy
depth at which adequate images can be ob- rophores on the basis of their emission spectra.
2.2.14
Supplement 56 Current Protocols in Neuroscience
Spectral detection and linear unmixing allows molecules into the bleached area may be gov-
discrimination of fluorophores with highly erned by diffusion, binding interactions with
overlapping emission spectra, such as GFP and other molecules, or a combination of both, and
YFP (Dickinson et al., 2001). appropriate mathematical models have been
Confocal microscopy also is well suited for developed to analyze these responses (Sprague
visualizing fluorescent proteins such as GFP, and McNally, 2005). In FLIP, a region of the
cyan FP (CFP), yellow FP (YFP), and DsRed. specimen is photobleached several times with
CFP and YFP can be visualized with minimal a delay between the bleach scans, and images
cross-talk between channels on a confocal mi- are collected during this process to monitor the
croscope with 405-nm and 514-nm excitation. distributions of bleached and nonbleached flu-
The overlap of the excitation and emission orescent molecules. Observation of FLIP can
spectra of CFP and GFP or GFP and DsRed show whether there is exchange of fluorescent
may result in cross-talk between channels in molecules between two compartments of a cell
experiments with these combinations of fluo- or whether a fluorescent structure is a single
rophores. New fluorescent proteins have been organelle or a network of contiguous but inde-
developed that are less prone to dimerization pendent organelles (Cole et al., 1996).
and provide more optimal combinations for The spatial precision by which two fluo-
multi-color imaging (Shaner et al., 2004). Flu- rophores can be said to co-localize on the
orescent proteins have also been incorporated basis of light microscopy is limited by the
into biochemical reporters for measuring in- optical resolution (∼0.2 μm in the xy plane
tracellular calcium, kinase activity, and other and 0.6 μm in the z-axis). The phenomenon
signaling molecules (Zhang et al., 2002). of fluorescence resonance energy transfer
Photosensitive fluorescent proteins are (FRET) can potentially reveal whether two
available that undergo a change in spectral fluorophores are within <10 nm proximity.
properties upon photoactivation. Photactivat- FRET (Wouters and Bastiaens, 2000) is the
able GFP (PaGFP; Patterson and Lippincott- nonradiative transfer of energy from a fluores-
Schwartz, 2002) exhibits little fluorescence cent donor molecule to an acceptor molecule.
under 488-nm illumination prior to activation, Energy transfer occurs only if the molecules
but undergoes a 100-fold increase in fluores- are within a distance of less than ∼10 nm,
cence after photoactivation at 400 to 430 nm. and only if the emission spectrum of the donor
An LSCM with a 405- or 413-nm laser can overlaps the excitation spectrum of the accep-
be used to photoactivate PaGFP within a user- tor. One application of FRET is to determine
defined region of interest within the specimen, whether two populations of molecules undergo
and thereby selectively “highlight” GFP flu- binding interactions. One population is labeled
orescence within that region. The activated with donor fluorophores (e.g., CFP) and the
GFP retains its fluorescence indefinitely and, second is labeled with acceptor fluorophores
importantly, manifests these properties even (e.g., YFP). Several techniques for measuring
when fused to another protein. PaGFP fusion FRET have been devised (Jares-Erijman and
proteins provide a useful tool for studying Jovin, 2003) and many of these can be carried
the intracellular dynamics of proteins and or- out with current LSCMs.
ganelles (Karbowski et al., 2004). Current LSCMs are much superior to their
LSCMs that incorporate an AOTF to con- predecessors in sensitivity, speed of image ac-
trol the illumination wavelength and intensity quisition, and versatility. Although they are
can be configured to perform various types expensive ($200,000 to $600,000) and require
of photobleaching experiments. Measurement costly service contracts to ensure optimal per-
of fluorescence recovery after photobleach formance, their many benefits justify these
(FRAP) or fluorescence loss in photobleach costs.
(FLIP) can provide information about molec-
ular mobility and binding (Cole et al., 1996; ACKNOWLEDGEMENTS
McNally and Smith, 2002; Lippincott- The author wishes to thank Dr. James
Schwartz et al., 2003). In FRAP, fluorescence Galbraith (National Institutes of Health,
in a small region of the specimen is pho- Bethesda, Md.) for helpful comments on the
tobleached by scanning with high-intensity manuscript. This research was supported by
illumination, and recovery of fluorescence the Intramural Research Program of the Na-
into the bleached area is then monitored tional Institute of Neurological Diseases and
by scanning with low-intensity illumination Stroke at the National Institutes of Health,
(Fig. 2.2.1D). The rate of return of fluorescent Bethesda, Md.
Imaging

2.2.15
Current Protocols in Neuroscience Supplement 56
LITERATURE CITED Cole, N., Smith, C., Sciaky, N., Terasaki, M.,
Ando, R., Hama, H., Yamamoto-Hino, M., Mizuno, Edidin, M., and Lippincott-Schwartz, J. 1996.
H., and Miyawaki, A. 2002. An optical marker Diffusional mobility of Golgi proteins in mem-
based on the UV-induced green-to-red photo- branes of living cells. Science 237:797-801.
conversion of a fluorescent protein. Proc. Natl. Dailey, M.E., Manders, E., Soll, D.R., and Terasaki,
Acad. Sci. U.S.A. 99:12651-12656. M. 2006. Confocal microscopy of living cells.
Art, J. 2006. Photon detectors for confocal mi- In Handbook of Biological Confocal Mi-
croscopy. In Handbook of Biological Confocal croscopy, 3rd ed. (J. Pawley, ed.) pp. 381-403.
Microscopy, 3rd ed. (J. Pawley, ed.) pp. 251-264. Springer, New York.
Springer, New York. Diaspro, A. (ed.) 2002. Confocal and Two Photon
Bacallao, R., Sohrab, S., and Phillips, C. 2006. Microscopy: Foundations, Applications and
Guiding principles of specimen preservation for Advances. Wiley-Liss, New York.
confocal fluorescence microscopy. In Handbook Dickinson, M.E., Bearman, G., Tille, S., Lansford,
of Biological Confocal Microscopy, 3rd ed. (J. R., and Fraser, S.E. 2001. Multi-spectral
Pawley, ed.) pp. 368-380. Springer, New York. imaging and llinear unmixing add a whole
Benham, G. 2002. Practical aspects of objective new dimension to laser scanning fluorescence
lens selection for confocal and multiphoton microscopy. Biotechniques 31:1274-1278.
digital imaging techniques. In Cell Biological Fernandez-Alfonso, T. and Ryan, T.A. 2004. The
Applications of Confocal Microscopy, 2nd ed. kinetics of synaptic vesicle pool depletion at
(B. Matsumoto, ed.) pp. 247-300. Academic CNS synaptic terminals. Neuron 41:943-953.
Press, San Diego, Calif.
Giepmans, B.N.G., Adams, S.R., Ellisman, M.H.,
Boccacci, P. and Bertero, M. 2002. Image- and Tsien, R.Y. 2006. The fluorescent tool-
restoration methods: Basics and algorithms. In box for assessing protein location and function.
Confocal and Two-Photon Microscopy: Founda- Science 312:217-224.
tions, Applications, and Advances (A. Diaspro, Giloh, H. and Sedat, J.W. 1982. Fluorescence
A., ed.), pp. 253-269. John Wiley & Sons, microscopy: Reduced photobleaching of rho-
Hoboken, N.J. damine and fluorescein protein conjugates by
Bonhoeffer, T. and Yuste, R. 2002. Spine motil- n-propyl gallate. Science 217:1252-1255.
ity: Phenomenology, mechanisms, and function. Goldstein, M. and Watkins, S. 2008. Immunohis-
Neuron 35:1019-1027. tochemistry. Curr. Protoc. Mol. Biol. 81:14.6.1-
Brelje, T.C., Wessendorf, M.W., and Sorenson, 14.6.23.
R.L. 1993. Multicolor laser scanning confocal Holmes, T.J., Biggs, D., and Abu-Tarif, A. 2006.
immunofluorescence microscopy: Practical ap- Blind deconvolution. In Handbook of Biological
plications and limitations. In Cell Biological Confocal Microscopy, 3rd ed. (J. Pawley, ed.)
Applications of Confocal Microscopy (B. pp. 468-487. Springer, New York.
Matsumoto, ed.) pp. 98-182. Academic Press,
San Diego, Calif. Inoué, S. and Inoué, T. 2002. Direct-View High-
Speed Confocal Scanner: The CSU-10. In
Brelje, T.C., Wessendorf, M.W., and Sorenson, Cell Biological Applications of Confocal Mi-
R.L. 2002. Multicolor laser scanning confo- croscopy, 2nd ed. (B. Matsumoto, ed.) pp. 88-
cal immunofluorescence microscopy: Practical 128. Academic Press, San Diego, Calif.
applications and limitations. In Cell Biological
Applications of Confocal Microscopy, 2nd ed. Inoué, S. and Spring, K.R. 1997. Video Microscopy:
(B. Matsumoto, ed.), pp. 166-244. Academic The Fundamentals, 2nd ed. Plenum, New York.
Press, San Diego, Calif. Jares-Erijman, E.A. and Jovin, T.M. 2003. FRET
Bystricky, K., Laroche, T., van Houwe, G., imaging. Nat. Biotechnol. 21:1387-1395.
Blaszczyk, M., and Gasser, S.M. 2005. Chro- Johnson, G.D., Davidson, R.S., McNamee, K.C.,
mosome looping in yeast: Telomere pairing and Russell, G., Goodwin, D., and Holborow, E.J.
coordinated movement reflect anchoring effi- 1982. Fading of immunofluorescence during mi-
ciency and territorial organization. J. Cell Biol. croscopy: A study of the phenomenon and its
168:375-387. remedy. J. Immunol. Methods 55:231-242.
Canell, M.B., McMorland, A., and Soeller, C. Kahana, J.A. and Silver, P.A. 1996. Use of the A.
2006. Image enhancement by deconvolution. In victoria green fluorescent protein to study pro-
Handbook of Biological Confocal Microscopy, tein dynamics in vivo. Curr. Protoc. Mol. Biol.
3rd ed. (J. Pawley, ed.) pp. 488-500. Springer, 34:9.7.22-9.7.28.
New York.
Kamabadur, R., Koizumi, K., Stivers, C., Nagle, J.,
Centonze, V. and Pawley, J. 1995. Tutorial on prac- Poole, S., and Odenwald, W. 1998. Regulation of
tical confocal microscopy and use of the con- POU genes by castor and hunchback establishes
focal test specimen. In Handbook of Biological layered compartments in the Drosophila CNS.
Confocal Microscopy, 2nd ed. (J. Pawley, ed.) Genes Dev. 12:246-260.
pp. 549-570. Plenum, New York.
Karbowski, M., Arnoult, D., Chen, H., Chan, D.C.,
Centonze, V. and Pawley, J. 2006. Tutorial on prac- Smith, C.L., and Youle, R.J. 2004. Quantitation
tical confocal microscopy and use of the con- of mitochondrial dynamics by photolabeling of
focal test specimen. In Handbook of Biological individual organelles shows that mitochondrial
Basic Confocal
Microscopy Confocal Microscopy, 3rd ed. (J. Pawley, ed.) fusion is blocked during the Bax activation phase
pp. 627-649. Springer, New York. of apoptosis. J. Cell Biol. 164:493-499.
2.2.16
Supplement 56 Current Protocols in Neuroscience
Keller, E. 1995. Objective lenses for confocal mi- 2004. Improved monomeric red, orange and yel-
croscopy. In Handbook of Biological Confocal low fluorescent proteins derived from Disco-
Microscopy, 2nd ed. (J. Pawley, ed.) pp. 111- soma sp. Red fluorescent protein. Nat. Biotech-
126. Plenum, New York. nol. 22:1567-1572.
Keller, E. 2006. Objective lenses for confocal mi- Shotton, D.M. 1993. Electronic acquisition of light
croscopy. In Handbook of Biological Confocal microscope images. In Electronic Light Mi-
Microscopy, 3rd ed. (J. Pawley, ed.) pp. 145-161. croscopy (D.M. Shotton, ed.) pp. 1-38. Wiley-
Springer, New York. Liss, New York.
Knoll, J.H., Lichter, P., Bakdounes, K., and Eltoum, Sprague, B.L. and McNally, J.G. 2005. FRAP anal-
I.-E.A. 2007. In situ hybridization and detection ysis of binding: Proper and fitting. Trends Cell
using nonisotopic probes. Curr. Protoc. Mol. Biol. 15:84-91.
Biol. 79:14.7.1-14.7.17. Staudt, T., Lang, M.C., Medda, R., Engelhardt, J.,
Lippincott-Schwartz, J., Altan-Bonnet, N., and and Hell, S.W. 2007. 2,2 -Thiodiethanol: A new
Patterson, G.H. 2003. Photobleaching and pho- water-soluble mounting medium for high reso-
toactivation: Following protein dynamics in lution optical microscopy. Microsc. Res. Tech.
living cells. Nat. Cell Biol. Suppl:S7-S14. 70:1-9.
Majlof, L. and Forsgren, P. 2002. Confocal mi- Terasaki, M. and Dailey, M.E. 1995. Confocal mi-
croscopy: Important considerations for accu- croscopy of living cells. In Handbook of Biolog-
rate imaging. In Cell Biological Applications of ical Confocal Microscopy, 2nd ed. (J. Pawley,
Confocal Microscopy, 2nd ed. (B. Matsumoto, ed.) pp. 327-346. Plenum, New York.
ed.) pp. 149-164. Academic Press, San Diego, Toomre, D. and Pawley, J.B. 2006. Disk-scanning
Calif. confocal microscopy. In Handbook of Biolog-
Matsumoto, B. (ed.) 2002. Cell Biological Appli- ical Confocal Microscopy, 3rd ed. (J. Pawley,
cations of Confocal Microscopy. 2nd ed. Aca- ed.) pp. 221-238. Springer, New York.
demic Press, London.
Tsien, R.Y., Ernst, L., and Waggoner, A. 2006. Fluo-
McNally, J.G. and Smith, C.L. 2002. Photobleach- rophores for confocal microscopy: Photophysics
ing by confocal microscopy. In Confocal and and photochemistry. In Handbook of Biological
Two Photon Microscopy: Foundations, Appli- Confocal Microscopy, 3rd ed. (J. Pawley, ed.)
cations and Advances (A. Diaspro, ed.) pp. 525- pp. 338-352. Springer, New York.
538. Wiley-Liss, New York.
Waterman-Storer, C.M., Sanger, J.W., and Sanger,
McNally, J., Karpova, T., Cooper, J., and Conchello, J.M. 1993. Dynamics of organelles in the mi-
J. 1999. Three-dimensional imaging by decon- totic spindles of living cells: Membrane and mi-
volution microscopy. Methods 19:373-385. crotubule interactions. Cell Motil. Cytoskeleton
Michalet, X., Pinaud, F.F., Bentolila, L.A., Tsay, 26:19-39.
J.M., Doose, S., Li, J.J., Sundaresan, G., Wu, Webb, R.H. and Dorey, C.K. 1995. The pixilated
A.M., Gambhir, S.S., and Weiss, S. 2005. Quan- image. In Handbook of Biological Confocal Mi-
tum dots for live cells, in vivo imaging, and di- croscopy, 2nd ed. (J. Pawley, ed.) pp. 55-68.
agnostics. Science 307:538-544. Plenum, New York.
Nerbonne, J.M. 1996. Caged compounds: Tools Wilson, T. (ed.) 1990. Confocal Microscopy. Aca-
for illuminating neuronal responses and connec- demic Press, London.
tions. Curr. Opin. Neurobiol. 6:379-386.
Wilson, T. 1995. The role of the pinhole in confo-
Patterson, G.H. and Lippincott-Schwartz, J. 2002. cal imaging system. In Handbook of Biological
A photoactivatable GFP for selective photola- Confocal Microscopy, 2nd ed. (J. Pawley, ed.)
beling of proteins and cells. Science 297:1873- pp. 167-182. Plenum, New York.
1877.
Wouterlood, F.G. 2005. 3-D reconstruction of neu-
Pawley, J.B. (ed.) 1995. Handbook of Biological rons from multichannel confocal laser scanning
Confocal Microscopy, 2nd ed. Plenum, New image series. Curr. Protoc. Neurosci. 32:2.8.1-
York. 2.2.8.16.
Pawley, J.B. (ed.) 2006. Handbook of Biological Wouters, F.S. and Bastiaens, P.I.H. 2000. Imag-
Confocal Microscopy, 3rd ed. Springer, New ing protein-protein interactions by fluorescence
York. resonance energy transfer (FRET) microscopy.
Russ, J. 2002. The Image Processing Handbook, Curr. Protoc. Cell Biol. 7:17.1.1-17.1.15.
4th ed. CRC Press, Boca Raton, Fla.
Zhang, J., Campbell, R.E., Ting, A., and Tsien, R.Y.
Salmon, E.D. and Canman, J.C. 1998. Proper align- 2002. Creating new fluorescent probes for cell
ment and adjustment of the light microscope. biology. Nature Rev. Mol. Cell Biol. 3:906-918
Curr. Protoc. Cell. Biol. 0:4.1.1-4.1.26.
Sandison, D.R., Williams, R.M., Wells, K.S.,
Strickler, J., and Webb, W.W. 1995. Quantita- KEY REFERENCES
tive fluorescence confocal laser scanning mi- Inoué and Spring, 1997. See above.
croscopy. In Handbook of Biological Confocal Covers the basics of light microscopy, video mi-
Microscopy, 2nd ed. (J. Pawley, ed.) pp. 39-54. croscopy, and much more.
Plenum, New York. Matsumoto, 2002. See above.
Shaner, N.C., Campbell, R.E., Steinbach, P.A., Good source of practical information about confo- Imaging
Giepmans, B.N., Palmer, A.E., and Tsien, R.Y. cal imaging.
2.2.17
Current Protocols in Neuroscience Supplement 56
Pawley, 1995, 2006. See above. Vendors of Confocal Microscopes
Two editions of comprehensive reference book on These provide product descriptions, manuals, tuto-
confocal microscopy. rials and literature.
Russ, 2002. See above. http://www.zeiss.com
Guide to digital image processing. Carl Zeiss, Inc.
http://www.leica-microsystems.com/
company
INTERNET RESOURCES Leica Microsystems.
http://rsb.info.nih.gov/ij
ImageJ is a public domain image analysis pro- http://www.nikonusa.com
gram developed by W. Rasband (Research Services Nikon, Inc.
Branch, National Institute of Mental Health, NIH) http://www.olympusconfocal.com
for operating systems running Java (including Win- Olympus, Inc.
dows/PC and OSX/Macintosh). ImageJ has many
useful tools for analysis of confocal images. http://www.perkinelmer.com
PerkinElmer, Inc.
http://www.uhnres.utoronto.ca/facilities/wcif/
imagej/ http://www.solameretech.com
A manual written by Tony Collins that describes Solamere Technology.
the use of ImageJ to visualize and analyze confocal
images.
http://www.molecularexpressions.com Spectra of Fluorophores
The Molecular Expressions Web site is a rich source http://fluorescence.nexus-solutions.net/
of information about all aspects of light microscopy, frames6.htm
including confocal microscopy. It includes sections Biorad Microsciences fluorochrome database and
on the basic principles of confocal imaging, instru- charting application.
mentation, sample preparation, and choices of flu- http://home.earthlink.net/∼fluorescentdyes
orophores. An interactive tutorial “Choosing flu- George McNamara Multiprobe Microscopy.
orophore combinations for confocal microscopy”
allows the user to determine the extent of spec- http://www.invitrogen.com/site/us/en/home/
tral crossover that will occur when imaging differ- support/Research-Tools/Fluorescence-
ent combinations of fluorophores with specific laser SpectraViewer.html
lines and filter sets. Invitrogen Spectral Viewer.
http://www.olympusfluoview.com/resources/ http://www.bdbiosciences.com/research/
specimenchambers.html multicolor/spectrum viewer/index.jsp
Sources of chambers for maintaining living speci- BD Fluorescence Spectral Viewer.
mens during observation by microscopy.
http://listserv.buffalo.edu/user/sub.html
Many topics of interest to confocal microscopists
are discussed on the confocal listserver operated by
the listserver at the University at Buffalo. To sub-
scribe to the list, go to the URL and type “confocal”
in the box that asks which list one wishes to join.

Basic Confocal
Microscopy

2.2.18
Supplement 56 Current Protocols in Neuroscience

You might also like