ArticlePDF AvailableLiterature Review

Single molecule techniques for the study of membrane proteins

Authors:

Abstract and Figures

Single molecule techniques promise novel information about the properties and behavior of individual particles, thus enabling access to molecular heterogeneities in biological systems. Their recent developments to accommodate membrane studies have significantly deepened the understanding of membrane proteins. In this short review, we will describe the basics of the three most common single-molecule techniques used on membrane proteins: fluorescence correlation spectroscopy, single particle tracking, and atomic force microscopy. We will discuss the most relevant findings made during the recent years and their contribution to the membrane protein field.
Content may be subject to copyright.
MINI-REVIEW
Single molecule techniques for the study
of membrane proteins
Ana J. García-Sáez & Petra Schwille
Received: 2 February 2007 / Revised: 17 April 2007 / Accepted: 17 April 2007 / Published online: 12 May 2007
#
Springer-Verlag 2007
Abstract Single molecule techniques promise novel infor-
mation about the properties and behavior of individual
particles, thus enabling access to molecular heter ogeneities
in biological systems. Their recent developments to accom-
modate membrane studies have significantly deepened the
understanding of membrane proteins. In this short review, we
will describe the basics of the three most common single-
molecule techniques used on membrane proteins: fluores-
cence correlation spectroscopy, single particle tracking, and
atomic force microscopy. We will discuss the most relevant
findings made during the recent years and their contribution
to the membrane protein field.
Keywords Membrane protein
.
Single molecule technique
.
Atomic force microscopy
.
Fluorescence correlation spectroscopy
.
Single particle tracking
.
Single molecule microscopy
Introduction
Cell membranes are noncovalent, supramolecular assemblies
of lipids and proteins that form self-contained volumes. They
define cellular compartments by acting as permeability
barriers to polar molecules. But besides this structural
function, cell membranes are involved in the transport of
energy, matter, and information into and out of the cell.
Such a diversity of functions is reflected by the complex
protein and lipid composition of natural membranes. Lipids
organize into bilayers that contain integral and associated
membrane proteins. According to genomic data, half of a
cells genes code for membrane proteins, which are
responsible for around one third of the dry cell-weight
(Douglass and Vale 2005; Zimmerberg and Gawrisch 2006).
This makes cellular membranes crowded environments.
For the lipids, the combination of different backbones,
head groups, acyl chains, and degree of saturation can give
rise to hundreds of different species. This allows fine-
tuning of the physical and chemical properties of mem-
branes, including lateral fluidity, permeability, rigidity, and
curvature. Indeed, this diversity is fully exploited by the
cell: Mass -spectrometry analys es of lipid composition in
cells revealed high levels of temporal and spatial variability
of membrane composition (van Meer 2005).
Besides controlling the majority of membrane properties,
lipids can affect membrane protein function through
specific interactions (Lin et al. 2005). And on the other
hand, membrane proteins can alter lipid organization and
modify the physical properties of mem branes (Douglass
and Vale 2005).
Information about how membrane proteins work, their
interactions wi th lipids and other proteins, and their
dynamics and structure, is essential for the understanding
of biological membranes and cell functioning in general.
However, the difficulties in reconstituting functional mem-
brane proteins to controllable model bilayers have delaye d
their structural and functional characterization in compari-
son to their soluble counterparts.
Recently, a number of single-molecule techniques have
been extended for the study of membrane proteins. The
applications performed to date have contributed substan-
tially to the current knowledge of membrane proteins and
membrane organization.
Appl Microbiol Biotechnol (2007) 76:257266
DOI 10.1007/s00253-007-1007-8
A. J. García-Sáez
:
P. Schwille (*)
Biophysics Group,
Biotechnologisches Zentrum (BIOTEC) der TU Dresden,
Tatzberg 47-51, 01307 Dresden, Germany
e-mail: petra.schwille@biotec.tu-dresden.de
Biological systems are intrinsically heterogeneous, im-
plying distributions of dynamic coefficients, structural
conformations, interactions among molecules, and catalytic
rates. The main advantage of single-molecule techniques vs
averaging techniques is that individual inhomogeneities in
the system of study can be assessed (Haustein and Schwille
2004). The analysis of these distributions can provide a
deeper insight into the mechanism of action of biolo gical
molecules. For example, it can be distinguished if changes
in the measured parameter are observed for all mol ecules in
the system or only a subpopulation of them (Johnson et al.
2005). In addition, single-molecule techniques usually need
low concentrations similar to physiological conditions and
are normally applied to systems in equilibrium.
In this review, we will focus on the recent progress in the
membraneprotein field achieved with single-molecule tech-
niques. We will describe the principles of the three main single-
molecule techniques that have been applied to membrane
proteins: fluorescence correlation spectroscopy (FCS), single-
particle tracking (SPT), and atomic force microscopy/spectros-
copy (AFM), and we will discuss the main findings and their
contribution to the understanding of membrane proteins.
Fluorescence correlation spectroscopy
Fluorescence correlation spectroscopy characterizes the
fluctuations in fluorescence intensity of a system in
equilibrium (Haustein and Schwille 2004). A scheme of a
typical setup is shown in Fig. 1a. It usually contains a
microscope objective with high numerical aperture to focus
the laser and achieve efficient detection. The photons
emitted within the focal volume pass through a pinhole,
which reduces the detection volume in the z-direction. As a
result, the measurement volume is confined to dimensions in
the range of fractions of a femtoliter, which is a micrometer
cube. Then, the photons pass proper filters and are detected
by an avalanche photodiode. This configuration allows high
signal/noise ratio when using fluorophore concentrations
ranging from 10
10
to 10
6
M.
The diffusion of fluorophore molecules through the focal
volume or changes in their emission properties [due to
chemical reactions, association/dissociation events, photo-
dynamic processes, or conformational changes (Haustein
and Schwille 2004; Kahya 2006)] produce fluctuations in
fluorescence intensity that can be quantified and autocorre-
lated as a function of time (Fig. 1b). The autocorrel ation
function is a mathematical tool that provides a measure of
the self-similarity of the fluorescence signal over time. The
characteristic decay time of the autocorrelation function is
related to the time that the fluorophore spends within the
focal volume. It depends on the mobility of the particle:
The larger the diffusion coefficient, the faster the decay
(Fig. 1c).
FCS averages thousands of single-diffusion events,
allowing precise estimation of diffusion coefficients and
particle concentrations in relatively short acquisition times
(Schwille 2001). It thus can be used to inves tigate not only
molecule mobility, transport, and diffusion processes but
also rate constants of inter- or intramolecular reactions or
binding (Bacia et al. 2006).
As with other fluorescence techniques, the protein of
interest must be fluorescently labeled. Fusion to fluorescent
proteins and chemical labeling with organic dyes are the
most common strategies. Care must be taken that labeling
does not affect the protein behavior.
Fig. 1 Principles of fluorescence correlation spectroscopy. a The
sample is illuminated by focusing the laser beam with an objective.
The emitted photons are then spectrally filtered and detected with an
avalanche phot odiode. A pinhole before the detector limits the
detection volume in the z-direction, so that the light from upper and
lower planes is eliminated. b The emission of photons diffusing in and
out the focal volume induces fluctuations in the intensity signal. The
fluorescence trace recorded during a given peri od of time is
autocorrelated. The autocorrelation function measures the self-
similarity of the fluorescence signal with time. c The graph shows
the values obtained for the autocorrelated fluorescence signal in b
(white circles). The diffusion coefficient and the particle concentration
can be determined by fitting the experimental values to the
autocorrelation function in b (solid line) (courtesy of J. Ries)
258 Appl Microbiol Biotechnol (2007) 76:257266
In the case of membrane measurements, some additional
considerations are required (Kahya 2006). It is critical to
correctly position the laser focus on the membrane bilayer
to avoid artifacts during the collection of autocorrelation
curves. For this purpose, it is recommended to combine the
FCS setup with an imaging device (preferably a laser scanning
microscope) and a z-stepping motor that allows measurement
of the diffusion times at different z-positions of the bilayer
(Benda et al. 2003). The measured membrane should be
planar in the focal plane, and fluorescence fluctuations due
to undulations or movement of the whole membrane should
be avoided. The analysis of aggregation processes is
restricted by sensitivity, as for freely diffusing molecules in
the two-dimensional plane of the membrane, the mass of the
monomer/oligomer should differ at least by a factor of 1520
(Kahya 2006).
Giant unilamellar vesicles (GUVs) are an excellent
model membrane system for FCS studies: They have
similar size to eukaryotic cells, so they can be visualized
with a microscope, and the membrane area focused in the
detection volume has virtually no curvature. In addition,
they form stable structures composed of freestanding bilayers
whose lipid composition can be controlled. In spite of some
technical difficulties, membrane proteins can be reconstituted
into them. FCS measurements in GUVs have been success-
fully used during the recent years for the study of the lateral
organization and dynamics of several membrane proteins, like
an ATP-binding cassette transport system from Lactobacillus
lactis, the lactose transporter LacS from Streptococcus
thermophilus, the mechanosensitive channel MscL from
Escherichia coli (Doeven et al. 2005), and bateriorhodopsin
(Kahya et al. 2002). The possibility to produce phase-
separated membranes in GUVs has extended the use of FCS
to characterize the partition and the dynamics of membrane
proteins in liquid-disordered (L
d
) and liquid-ordered (L
o
)
raft-like phases. The SNARE proteins syntaxin 1A and
synaptobrevin 2 preferentially localized to the L
d
phase when
functionally reconstituted into domain-exhibiting GUVs
(Bacia et al. 2004b). A significantly higher partition into
the L
o
phase, around 2030%, was found in the cases of
BACE and human placental alkaline phosphatase (Kahya et
al. 2005; Kalvodova et al. 2005). Due to the differential
diffusion properties of raft-associated cholera toxin and the
L
d
-located lipid dye DiI, FCS could be use to analyze the
relationship between domain-exhibiting GUVs and rafts in
living cells (Bacia et al. 2004a).
FCS has been employed several times to study mem-
brane receptors dynamics. By using fluorescently labeled
ligands, the ligand-binding constant to the γ-amino butyric
acid A (GABA
A
) and to the β
2
-adrenergic receptors could
be determined in living cells. Analysis of the mobility of
ligand-receptor complexes revealed two different diffusion
coefficients in both cases, which were assigned to fast and
hindered mobility (Hegener et al. 2004; Meissner and
Haberlein 2003). A similar approach was used to investigate
the interactions of T-cell receptor with CD8 during T-cell
activation (Gakamsky et al. 2005).
The interaction kinetics of fluor escently labeled IgG and
the FcγRII receptor was studied in supported planar
membranes with a combination of FCS and total internal
reflection f luorescence (TIR-FCS; Lieto et al. 2003).
Recently, the technique was extended to living cells and
applied to measure the lateral diffusion of membrane-
bound, farnesylated enhanced green fluorescent protein
(EGFP; Ohsugi et al. 2006).
The organization of the cell membrane has been investi-
gated by FCS. Diffusion measurements at different spatial
scales give rise to the so-called FCS diffusion law, which
provides information about the confinement parameters of
microdomains in cell membranes (Wawrezinieck et al.
2005). The area of measured membrane can be modified
by using laser beams of different size (Wawrezini eck et al.
2005), by performing a z-scan (Humpolickova et al. 2006),
or by means of nanometric apertures that allow to observe
areas below the diffraction limit (Wenger et al. 2007).
Interestingly, the latter approach can give an estimate of the
size of mem brane heterogeneities. The diffusion properties
of GFP proteins bound to the cell membrane through
different tags were affected by the disruption of lipid-based
or cytoskeleton-based microdomains, revealing the impor-
tance of these factors in membrane organization (Lenne et
al. 2006).
Dual-color fluorescence cross-correlation spectroscopy
(FCCS) is a powerful variation of FCS to probe interactions
between two species labeled with spectrally different dyes
(Bacia et al. 2006; Haustein and Schwille 2004). In this
mode, the two fluorophore s are excited within the same
focal volume using two lasers or two-photon excitation, and
the photons emitted are measured using spectral channels.
The analysis of the fluorescence fluctuations in the two
channels yields a cross-correlation function whose ampli-
tude is proportion al to the relative concentration of the
double-labeled particles (interacting molecules that move
together through the diffusion volume). Spectrally distinct
dyes should be used to avoid cross talk and false positive
cross-correlation.
The destiny of two cargoes along the endocytic pathway
was followed with FCCS (Bacia et al. 2002). The A and B
subunits of cholera toxin were labeled with Cy2 an d Cy5,
respectively. Positive cross-correlation could be measured
for the two subunits along the endocytic pathway, until they
were separated in the Golgi network. Larson et al. (2003)
also used a cross-correlation strategy to analyze the dynamic
interactions between labeled Lyn kinase and FcɛRI receptor
stimulated with fluorescently labeled IgE. In an elegant
approach, they were able to perform FCCS measurements in
Appl Microbiol Biotechnol (2007) 76:257266 259
real time in living cells that revealed a decrease in Lyn diffusion
associated to FcɛRI receptor binding upon stimulation.
Scanning FCS (SFCS) extends FCS to larger diffusion
times and has thus been successfully applied to analyze
membrane dynamics (Ries and Schwille 2006). The extension
to two-focus SFCS, realized by measuring simultaneously in
two detection volumes instead of one, eliminates the need to
calibrate the detection volume, and dual-color cross-correlation
analysis is easily implemented. Accurate diffusion coefficients
could be determined on GUVs, on planar supported bilayers,
and on the membrane of yeast cells.
Single-particle tracking
SPT uses video microscopy combined with digital computer
processing to monitor the motion of single proteins or lipids
on the plane of the membrane. The molecules of interest are
often labeled with submicrometer particles (latex beads of
200500 nm in diameter or colloidal gold of 40100 nm in
size) coated with specific antibodies or ligands. These
particles are smaller than the wavelength of light; thus, they
scatter light, giving rise to a diffraction pattern, which is an
airy disc, and allows determination of the centroid position
with around 10 nm precision (Kusumi et al. 2005a).
Molecules can also be fluorescently labeled. The most
common fluorophores are organic dyes, quantum dots, or
fluorescent proteins. Fluorescence labels are usual ly smaller
in size than the particles mentioned above and probably less
invasive. But unlike latex or gold beads, they undergo
photobleaching, thus reducing the achiev able acquisition
time. In both cases, proper controls have to be made to
assure that labeling does not alter the behavior of the
system under study. When noncovalent labeling is used,
there is risk of label exchange among target molecules,
which increases tracking difficulties. In addition, multiva-
lent labels can promote cross-linking, modifying molecule
mobility or inducing cellular responses (Lenne et al. 2006).
Particles are usually ima ged in bright field mode with
enhanced contrast and projected onto a video camera
(Fig. 2a). TIRF microscopy can also be used for SPT, as
only a thin plane of the sample is illuminated, such that the
background fluorescence is strongly reduced and single
particles can be detected in the membrane. The video
images are processed and recorded in real time. Analysis of
the movies is performed digitally. The tracking software
used must be able to accurately track a single molecule
without mistaking it with other ones, while being flexible
enough to allow changes in the particle characteristics due
to out-of-focus movement (Lenne et al. 2006).
Analysis of a single-particle trajectory is done by
determining the x, y positions of the particle in every image
of the movie an d calculating the displacements at Δt time
intervals (Fig. 2b). Then, the mean square displacement can
be related to the diffusion coefficient through different
models, depending on the type of motion. Slow diffusing
particles are experimentally favored, and statistical quality
strongly depends on the time interval. SPT is a powerful
technique to detect deviations from Brownian diffusion,
like confined diffusion, anomalous diffusion, or directed
flow of particles. However, classification of trajectories
must be done with a proper algorithm, which can statistically
distinguish among the different ways of motion (Schmidt
et al. 1996; Schutz et al. 1997).
SPT studies on fluorescent lipid analogs and membrane
proteins led Kusumi et al. to propose a new model of
membrane organization, known as the compartmentalized
fluid model (Kusumi et al. 2005a,b; Ritchie et al. 2005;
Suzuki et al. 2005). The analysis of single-particle mobility
at high frame rates in living cells revealed short-term
confined Brownian diffusion and long-term hop diffusion
between the compartments. According to this model, the
compartment boundaries are form ed by the membrane
skeleton and the membrane proteins associated to it. This
model explains why the diffusion coefficients of lipids and
membrane proteins are larger in artifici al model membranes
than in living cell s and why their mobility is reduced after
molecular complex formation.
In accordance with the above model, Douglass and Vale
(2005) used SPT in an elegant study to show that the adaptor
protein LAT and the tyrosine kinase Lck localized to
membrane microdomains depending on protein
protein
interactions but not on actin or lipid rafts. These protein
microdomains affected the diffusion of other membrane
proteins by partial exclusion or trapping. These clusters were
proposed to have a role in cell signaling, so that partition of
molecules in and out of the microdomains would decide the
phosphorylation/dephosporylation balance.
Relevant information about the G-protein coupled
receptors (GPCRs) mechanism of action has been obtained
with single-molecule detection and SPT studies. Single-
molecule analysis of ep idermal growth fac tor (EGF)
binding to cells showed that Ca
2+
-signaling was activated
in a sigmoidal fashion. A 50% probability of cell activation
was achieved after binding of 300 EGF molecules, a very
low number compared with the tens of thousands of
receptors expressed on the cell surface (Uyemura et al.
2005). Data analysis revealed that EGF bound preferential-
ly to pre-dimeric EGF receptor complexes, showing coop-
erativity for the binding of the second ligand. This would
allow sensitive signaling even at low EGF concentrations
(Teramura et al. 2006). In addit ion, a decrease in receptor
mobility upon ligand binding was measured for nerve
growth factor receptor, neurokinin-1 receptor, and odorant
receptor OR17-40 (Jacquier et al. 2006; Lill et al. 2005;
Shibata et al. 2006).
260 Appl Microbiol Biotechnol (2007) 76:257266
The effect of cholesterol concentration on the mobility of
proteins in the plasma membrane has been investigated by
SPT. Interestingly, the diffusion of EGFR, HER2, and the I-
Ek proteins of the major histocompatibility complex
(MCH) decreased after cholesterol depletion in a reversible
way (Nishimura et al. 2006; Orr et al. 2005). In addition,
the mobility of EGFR and HER2, but not I-Ek proteins,
was affected by the cytoskeletal organization in cholesterol-
depleted cells.
SPT experiments were used to monitor the diffusion
properties of the G protein Ras as a function of its activation
state (Lommerse et al. 2005; Murakoshi et al. 2004). Upon
EGF or insulin activation, Ras was greatly immobilized and
confined to 20 nm domains, suggesting the formation of
Ras-signaling complexes. A sim ilar strategy was used to
investigate the diffusion properties of the heterotrimeric G
protein Gq (Perez et al. 2006). Experiments in supported
cell-membrane sheets wi th immobilized GPCRs showed
that Gq in its heterotrimeric state had a lower mobility than
in the monomeric state, suggesting a role of oligomeric
state in differential partition into signaling complexes.
Atomic force spectroscopy
AFM is a scanning probe technique that allows samples to
be imaged with nanometer to atomic level resolution
(Morris et al. 1999). As schematically shown in Fig. 3a,
the AFM microscope consists of an extremely sharp tip
mounted on the end of a cantilever, which allows the tip to
move vertically as it tracks the sample. The scanning
mechanism works via a piezoelectric transducer, which
accurately positions the tip on the sample surface. For
detection, a laser beam is focused on the cantilever and
reflected onto a photodiode. During measurements, the tip
is brought into proximity with the sample surface, which is
scanned underneath the tip. The force experienced between
the tip and samp le causes a deflection of the canti lever,
which changes the position of the refl ected beam on the
photodiode. This, in turn, can be used to provide a
topographical map of the surface (Fig. 3b).
In addition to topographical imaging, AFM can also
probe the nanomechanical properties relating to the force
experienced between the tip and sample. In these measure-
Fig. 2 Principles of single-particle tracking. a Single particles are
detected and time lapse recorded in a stack file. The raw images are
deconvoluted, filtered, and spatially aligned. Then, fluorescence
peaks are accurately localized with a two-dimensional Gaussian fit.
The peak positions are connected in the successive frames, and
particle traces are obtained. b Analysis of single-particle trajectories.
On the left, the trajectories of free-diffusing (A) and confined (B)
particles are shown. The graph on the right depicts the corresponding
mean-square-displacement vs lag time for the same particles
Appl Microbiol Biotechnol (2007) 76:257266 261
ments, the AFM tip is brought into contact with the sample
and pulled away. Then, the deflection of the cantilever
during this approach and retraction is recorded. The plot of
cantilever deflection vs tip/sample distance is called force
curve (Fig. 3c). Small force differences can be obtained,
thanks to the low spring constant of the cantilever (Janovjak
et al. 2006; Zlatanova et al. 2000).
For sample preparation, the membrane protein of interest
is embedded in planar lipid bilayers (with a defined lipid
composition, coming from native membranes or in the form
of 2D protein-lipi d crystals) supported on a solid substrate.
Unlike with FCS and SPT, AFM does not need protein
labeling. Measurements are usually done under physiolog-
ical conditions in the presence of a bathing buffer solution
and at ambient temperature.
The AFM mode provides images of single-membrane
proteins at subnano meter resolution. The organization of
the photosynthetic machinery in several bacteria has been
visualized by AFM at high resolution, providing informa-
tion about the molecular architecture of the complexes in
native membranes (Fotiadis et al. 2002; Scheuring et al.
2001; Scheuring et al. 2003a,b; Scheuring et al. 2004a,b;
Scheuring et al. 2006). Interestingly, a comparative AFM
analysis of Rhodospirillum photometricum membranes
under low- and high-light conditions showed adaptation in
the photosynthetic complex assembly to optimize photo-
synthetic efficiency under different environmental condi-
tions (Scheuring and Sturgis 2005).
On the other hand, the single-molecule force spectros-
copy mode allows the characterization of inter- and
intramolecular interactions with high sensitivity. In unfold-
ing studies, the AFM tip is brought into contact with a
membrane protein and bound to it. Then, retraction of the
tip from the membrane stretches the polypeptide chain.
When the applied force overcomes the stability of the
protein, it unfolds and gives rise to peaks in the force curve
that correspond to elements of secondary structure in the
protein (Janovjak et al. 2006; Muller et al. 2006). The first
Fig. 3 Atomic force microscopy. a Scheme of an AFM setup. A very
sharp tip is mounted onto a cantilever and brought into proximity of the
sample, which is supported on a solid substrate. Relative positioning of
the sample and the AFM tip is performed with high accuracy , thanks to a
piezoelectric transducer . The detection module is composed of a laser
beam focused on the cantilever and reflected onto a photodiode detector
(courtesy of C. Bippes). b Three-dimensional topographical image of a
SM/DOPC/Chol supported lipid bilayer containing placental alkaline
phosphatase. The liquid-disordered and liquid-ordered phases are shown
with a height scale. Peaks of protein are depicted in white (kindly
provided by S. Chiantia). c Force curve obtained for the unfolding of a
single sodium/proton antiporter . Characteristic peaks are obtained that
correspond to the unfolding of secondary structure elements in the protein
(gently supplied by A. Kedrov)
262 Appl Microbiol Biotechnol (2007) 76:257266
unfolding experiments exploring the stability o f the
structural elements of membrane proteins were performed
with bacteriorhodopsin (Muller et al. 2002b; Oesterhelt
et al. 2000; Scheuring et al. 2006). The peaks obtained in
the forcedistance curves could be assigned to secondary
structure elements within the protein. Since then, severa l
publications have analyzed in depth how the molecular
interactions that stabilize the membrane structure of this
and other rhodopsins are affected by extra - or intracellular
unfolding, disulfide bonding, and oligomerization state
(Cisneros et al. 2005; Janovjak et al. 2004; Kessler et al.
2006; Kessler and Gaub 2006; Sapra et al. 2006; Tanuj
et al. 200 6 ). Single-molecule force spectroscopy was also
used in unfolding and refolding experiments with a
bacterial sodium/proton antiporter (Kedrov et al. 2004).
The effect of pH and of an inhibitory ligand on the molecular
interactions was investigated (Kedrov et al. 2005, 2006b).
Recently, Kedrov et al. (2006a ) reported an improved AFM
setup that enabled the study of the folding kinetics for this
antiporter with a time resolution of a few milliseconds.
Single-molecule recognition studies can be performed by
immobilizing one of the interacting partners to the AFM tip and
measuring the force necessary to break down the interacting
complex. This application allows recognition imaging, a
combination of topographical imaging and force measure-
ments.Ithasbeenusedtostudyligand/receptor interactions
(Kienberge r et al. 2006). Recently, it was also employed to
study the topology of the Na
+
/glucose co-transporter in living
cells (Puntheeranurak et al. 2006). The AFM tip was modified
with an antibody against the C-terminal loop 13 of the protein,
and single-mole cule recognition events probed that the
antigenic region was oriented towards the extracellular region.
Moreover,
D-glucose modified tips were used to investigate
the effect of the presence/absence of Na
+
on ligand binding
and to test the transporter specificity in competition experi-
ments with
L-glucose and D-galactose. Similarly, th e
appearance of HSP60 on the cell membrane of living and
fixed endothelial cells upon heat stress was confirmed by
single-molecule recognition events using an AFM tip modi-
fied with an antibody against the protein (Pfister et al. 2005).
The organization of eukaryotic membrane complexes has
been investigated by AFM. Gap junctions are channels
between neighboring cells, which are important for the flux
of ions and metabolites as well as for cell-to-ce ll signaling.
Connexon complexes that form the gap junctions were
purified and reconstituted into supported planar bilayers.
The structure and mechanical properties of the cytoplasmic
and extracellular loops were studied by imaging AFM and
single-molecule force spectroscopy (Liu et al. 2006; Muller
et al. 2002a). Aquaporins have also been studied by AFM
(Fotiadis et al. 2002; Moller et al. 2003; Schenk et al.
2005), and recently, the structural organization of mem-
brane patches containing junctional microdomains com-
posed by connexons and aquaporins was visualized by
high-resolution AFM (Buzhynskyy et al. 2007).
Recently, Gonçalves et al. (2006) reported high-resolution
AFM imaging and force spectroscopy measurements on
unsupported membranes. This two-chamber AFM setup is a
significant step towards more physiological conditions. In
addition, it allows the use of different buffer solutions
across the membranes for the study of gradient-excitable
membrane proteins.
The combination of AFM with FCS is a very promising
application that has been developed during the last couple of
years. It was initially applied to study the dynamic properties
of supported lipid bilayers exhibiting phase separation (Burns
et al. 2005; Chiantia et al. 2006a). Now, the first studies
including membrane proteins start to appear. Chiantia et al.
(2006b) used a combination of AFM and SFCS to solve the
existing controversy about the lateral organization of the
placental alkaline phosphatase, a raft associated protein, in
domain-exhibiting membranes. The dynamics and lateral
distribution of other membrane proteins, like cholera toxin,
which partitions to the liquid-ordered phase, and synapto-
brevin, found in the liquid-disordered phase, have also been
investigated (Chiantia et al. 2006b and unpublished results).
Conclusions
The use of single-molecule techniques for the study of
membrane proteins has already shown its usefulness. Among
their major achievements, single-molecule studies have
revealed a more complex organization of the cell membrane,
allowing significant extension of the fluid mosaic model.
Besides, the molecular details of signaling processes, including
receptor activation, lateral organization, and dynamics or the
formation of signaling complexes, are now better understood.
However, technical difficulties to reconstitute membrane
proteins into model membranes still are the bottleneck for the
advancement of knowledge in the field. Therefore, the
development of general, reproducible methods is highly
desirable not only for single-molecule applications but also
for averaging biochemical and biophysical techniques. Cur-
rent approaches involve detergent-mediated reconstitution of
membrane proteins into proteoliposomes (Bacia et al. 2004b;
Girard et al. 2004; Kahya et al. 2002), which can be used for
GUV or supported bilayer production. Alternatively, mem-
brane proteins can be in vitro translated and simultaneously
incorporated into supported bilayers (Robelek et al. 2007).
Alternatively to in vitro systems, single-molecule tech-
niques also offer the possibility of studying membrane
proteins in their natural environment with minor alte rations
of the biological system. Living-cell applications open a
new way to investigate the structure, molecular mecha-
nisms, and dynamics of membrane proteins.
Appl Microbiol Biotechnol (2007) 76:257266 263
The maturation and easier accessibility of these method-
ologies for membrane research will continue to make single-
molecule analysis a major tool in the membrane protein field.
Aknowledgments We thank J. Suckale, J. Ries, and S. Chiantia for
careful reading. This work was supported by a Marie Curie Intra-
European Fellowship (FP6) and German DGF (SCHW716/4-1).
References
Bacia K, Majoul IV, Schwille P (2002) Probing the endocytic pathway
in live cells using du al-color fluorescence cross-correlatio n
analysis. Biophys J 83:11841193
Bacia K, Scherfeld D, Kahya N, Schwille P (2004a) Fluorescence
correlation spectroscopy relates rafts in model and native
membranes. Biophys J 87:10341043
Bacia K, Schuette CG, Kahya N, Jahn R, Schwille P (2004b)
SNAREs prefer liquid-disordered over raft (liquid-ordered)
domains when reconstituted into giant unilamellar vesicles. J Biol
Chem 279:3795137955
Bacia K, Kim SA, Schwille P (2006) Fluorescence cross-correlation
spectroscopy in living cells. Nat Methods 3:8389
Benda A, Benes M, Marecek V, Lhotsky A, Hermens WT, Hof M
(2003) How to determine diffusion coeff icients in planar
phospholipid systems by confo cal fluorescence correlation
spectroscopy. Langmuir 19:41204126
Burns AR, Frankel DJ, Buranda T (2005) Local mobility in lipid
domains of supported bilayers characterized by atomic force
microscopy and fluorescence correlation spectroscopy. Biophys J
89:10811093
Buzhynskyy N, Hite RK, Walz T, Scheuring S (2007) The
supramolecular architecture of junctional microdomains in native
lens membranes. EMBO Rep 8:5155
Chiantia S, Kahya N, Ries J, Schwille P (2006a) Effects of ceramide
on liquid-ordered domains investigated by simultaneous AFM
and FCS. Biophys J 90:45004508
Chiantia S, Ries J, Kahya N, Schwille P (2006b) Combined AFM and
two-focus SFCS study of Raft-exhibiting model membranes.
ChemPhysChem 7:24092418
Cisneros DA, Oesterhelt D, Muller DJ (2005) Probing origins of
molecular interactions stabilizing the membrane proteins halor-
hodopsin and bacteriorhodopsin. Structure 13:235242
Doeven MK, Folgering JH, Krasnikov V, Geertsma ER, van den BG,
Poolman B (2005) Distribution, lateral mobility and function of
membrane proteins incorporated into giant unilamellar vesicles.
Biophys J 88:11341142
Douglass AD, Vale RD (2005) Single-molecule microscopy reveals
plasma membrane microdomains created by protein-protein
networks that exclude or trap signaling molecules in T cells.
Cell 121:937950
Fotiadis D, Suda K, Tittmann P, Jeno P, Philippsen A, Muller DJ,
Gross H, Engel A (2002) Identification and structure of a putative
Ca2+-binding domain at the C terminus of AQP1. J Mol Biol
318:13811394
Gakamsky DM, Luescher IF, Pramanik A, Kopito RB, Lemonnier F,
Vogel H, Rigler R, Pecht I (2005) CD8 kinetically promotes ligand
binding to the T-cell antigen receptor. Biophys J 89:21212133
Girard P, Pecreaux J, Lenoir G, Falson P, Rigaud JL, Bassereau P
(2004) A new method for the reconstitution of membrane
proteins into giant unilamellar vesicles. Biophys J 87:419429
Gonçalves RP, Agnus G, Sens P, Houssin C, Bartenlian B, Scheuring S
(2006) Two-chamber AFM: probing membrane proteins separat-
ing two aqueous compartments. Nat Methods 3:10071012
Haustein E, Schwille P (2004) Single-molecule spectroscopic meth-
ods. Curr Opin Struct Biol 14:531540
Hegener O, Jordan R, Haberlein H (2004) Dye-labeled benzodiaze-
pines: development of small ligands for receptor binding studies
using fluorescence correlation spectroscopy. J Med Chem
47:36003605
Humpolickova J, Gielen E, Benda A, Fagulova V, Vercammen J,
Vandeven M, Hof M, Ameloot M, Engelborghs Y (2006)
Probing diffusion laws within cellular membranes by Z-scan
fluorescence correlation spectroscopy. Biophys J 91:L23L25
Jacquier V, Prummer M, Segura JM, Pick H, Vogel H (2006)
Visualizing odorant receptor trafficking in living cells down to
the single-molecul e level. Proc Natl Acad Sci USA 103:
1432514330
Janovjak H, Struckmeier J, Hubain M, Kedrov A, Kessler M, Muller
DJ (2004) Probing the energy landscape of the membrane protein
bacteriorhodopsin. Structure 12:871879
Janovjak H, Kedrov A, Cisneros DA, Sapra KT, Struckmeier J, Muller
DJ (2006) Imaging and detecting molecular interactions of single
transmembrane proteins. Neurobiol Aging 27:546561
Johnson CK, Osborn KD, Allen MW, Slaughter BD (2005) Single-
molec ule fluorescence spectroscop y: new probes of prote in
function and dynamics. Physiology (Bethesda) 20:1014
Kahya N (2006) Targeting membrane proteins to liquid-ordered
phases: molecular self-organization explored by fluorescence
correlation spectroscopy. Chem Phys Lipids 141:158168
Kahya N, Wiersma DA, Poolman B, Hoekstra D (2002) Spatial
organization of bacteriorhodopsin in model membranes. Light-
induced mobility changes. J Biol Chem 277:3930439311
Kahya N, Brown DA, Schwille P (2005) Raft partitioning and
dynamic behavior of human placental alkaline phosphatase in
giant unilamellar vesicles. Biochemistry 44:74797489
Kalvodova L, Kahya N, Schwille P, Ehehalt R, Verkade P, Drechsel
D, Simons K (2005) Lipids as modulators of proteolytic
activity of BACE: involvement of cholesterol, glycosphingoli-
pids, and anion ic phos pholipids i n vitro. J Biol Ch em
280:3681536823
Kedrov A, Ziegler C, Janovjak H, Kuhlbrandt W, Muller DJ (2004)
Controlled unfolding and refolding of a single sodium-proton
antiporter using atomic force microscopy. J Mol Biol 340:11431152
Kedrov A, Krieg M, Ziegler C, Kuhlbrandt W, Muller DJ (2005)
Locating ligand binding and activation of a single antiporter.
EMBO Rep 6:668674
Kedrov A, Janovjak H, Ziegler C, Kuhlbrandt W, Muller DJ (2006a)
Observing folding pathways and kinetics of a single sodium-
proton antiporter from Escherichia coli . J Mol Biol 355:28
Kedrov A, Ziegler C, Muller DJ (2006b) Differentiating ligand and
inhibitor interactions of a single antiporter. J Mol Biol 362:
925932
Kessler M, Gaub HE (2006) Unfolding barriers in bacteriorhodopsin
probed from the cytoplasmic and the extracellular side by AFM.
Structure 14:521527
Kessler M, Gottschalk KE, Janovjak H, Muller DJ, Gaub HE (2006)
Bacteriorhodopsin folds into the membrane against an external
force. J Mol Biol 357:644654
Kienberger F, Ebner A, Gruber HJ, Hinterdorfer P (2006) Molecular
recognition imaging and force spectroscopy of single biomole-
cules. Acc Chem Res 39:2936
Kusumi A, Ike H, Nakada C, Murase K, Fujiwara T (2005a) Single-
molecule tracking of membrane molecules: plasma membrane
compartmentalization and dynamic assembly of raft-philic
signaling molecules. Semin Immunol 17:321
Kusumi A, Nakada C, Ritchie K, Murase K, Suzuki K, Murakoshi H,
Kasai RS, Kondo J, Fujiwara T (2005b) Paradigm shift of the
plasma membrane concept from the two-dimensional continuum
fluid to the partitioned fluid: high-speed single-molecule tracking
264 Appl Microbiol Biotechnol (2007) 76:257266
of membrane molecules. Annu Rev Biophys Biomol Struct
34:351378
Larson DR, Ma YM, Vogt VM, Webb WW (2003) Direct measure-
ment of Gag-Gag interaction during retrovirus assembly with
FRET and fluorescence correlation spectroscopy. J Cell Biol
162:12331244
Lenne PF, Wawrezinieck L, Conchonaud F, Wurtz O, Boned A, Guo
XJ, Rigneault H, He HT, Marguet D (2006) Dynamic molecular
confinement in the plasma membrane by microdomains and the
cytoskeleton meshwork. EMBO J 25:32453256
Lieto AM, Cush RC, Thompson NL (2003) Ligand-receptor kinetics
measured by total internal reflection with fluorescence correlation
spectroscopy. Biophys J 85:32943302
Lill Y, Martinez KL, Lill MA, Meyer BH, Vogel H, Hecht B (2005)
Kinetics of the initial steps of g protein-coupled receptor-
mediated cellular signaling revealed by single-molecule imaging.
ChemPhysChem 6:16331640
Lin CW, Yan F, Shimamura S, Barg S, Shyng SL (2005)
Membrane phosphoinositides control insulin secretion through
their effects on ATP-sensitive K+ channel activity. Diabetes
54:28522858
Liu F, Arce FT, Ramachandran S, Lal R (2006) Nanomechanics of
hemichannel conformations: connexin flexibility underlying
channel opening and closing. J Biol Chem 281:2320723217
Lommerse PH, Snaar-Jagalska BE, Spaink HP, Schmidt T (2005)
Single-molecule diffusion measurements of H-Ras at the plasma
membrane of live cells reveal microdomain localization upon
activation. J Cell Sci 118:17991809
Meissner O, Haberlein H (2003) Lateral mobility and specific binding
to GABA(A) receptors on hippocampal neurons monitored by
fluorescence correlation spectroscopy. Biochemistry 42:
16671672
Moller C, Fotiadis D, Suda K, Engel A, Kessler M, Muller DJ (2003)
Determining molecular forces that stabilize human aquaporin-1. J
Struct Biol 142:369378
Morris VJ, Kirby AR, Gunning AP (1999) Atomic force microscopy
for biologists. Imperial College Press, London
Muller DJ, Hand GM, Engel A, Sosinsky GE (2002a) Conformational
changes in surface structures of isolated connexin 26 gap
junctions. EMBO J 21:35983607
Muller DJ, Kessler M, Oesterhelt F, Moller C, Oesterhelt D, Gaub H
(2002b) Stability of bacteriorhodopsin alpha-helices and loops
analyzed by single-molecule force spectroscopy. Biophys J
83:35783588
Muller DJ, Sapra KT, Scheuring S, Kedrov A, Frederix PL, Fotiadis
D, Engel A (2006) Single-molecule studies of membrane
proteins. Curr Opin Struct Biol 16:489495
Murakoshi H, Iino R, Kobayashi T, Fujiwara T, Ohshima C,
Yoshimura A, Kusumi A (2004) Single-molecule imaging
analysis of Ras activation in living cells. Proc Natl Acad Sci
USA 101:73177322
Nishimura SY, Vrljic M, Klein LO, McConnell HM, Moerner WE
(2006) Cholesterol depletion induces solid-like regions in the
plasma membrane. Biophys J 90:927938
Oesterhelt F, Oesterhelt D, Pfeiffer M, Engel A, Gaub HE, Muller DJ
(2000) Unfolding pathways of individual bacteriorhodopsins.
Science 288:143146
Ohsugi Y, Saito K, Tamura M, Kinjo M (2006) Lateral mobility of
membrane-binding proteins in living cells measured by total
internal reflection fluorescence correlation spectroscopy. Biophys
J 91:34563464
Orr G, Hu D, Ozcelik S, Opresko LK, Wiley HS, Colson SD (2005)
Cholesterol dictates the freedom of EGF receptors and HER2 in
the plane of the membrane. Biophys J 89:13621373
Perez JB, Segura JM, Abankwa D, Piguet J, Martinez KL, Vogel H
(2006) Mo nitoring the diffusion of single heterotrimeric G
proteins in supported cell-membrane sheets reveals their parti-
tioning into microdomains. J Mol Biol 363:918930
Pfister G, Stroh CM, Perschinka H, Kind M, Knoflach M, Hinterdorfer
P, Wick G (2005) Detection of HSP60 on the membrane surface of
stressed human endothelial cells by atomic force and confocal
microscopy. J Cell Sci 118:15871594
Puntheeranurak T, Wildling L, Gruber HJ, Kinne RK, Hinterdorfer P
(2006) Ligands on the string: single-molecule AFM studies on
the interaction of antibodies and substrates with the Na+-
glucose co-transporter SGLT1 in living cells. J Cell Sci
119:29602967
Ries J, Schwille P (2006) Studying slow membrane dynamics with
continuous wave scanning fluorescence correlation spectroscopy.
Biophys J 91:1915 1924
Ritchie K, Shan XY, Kondo J, Iwasawa K, Fujiwara T, Kusumi A
(2005) Detection of non-Brownian diffusion in the cell mem-
brane in single molecule tracking. Biophys J 88:22662277
Robelek R, Lemker ES, Wiltschi B, Kirste V, Naumann R, Oesterhelt
D, Sinner EK (2007) Incorporation of in vitro synthesized GPCR
into a tethered artificial lipid membrane system. Angew Chem Int
Ed Engl 46:605608
Sapra KT, Besir H, Oesterhelt D, Muller DJ (2006) Characterizing
molecular interactions in different bacteriorhodopsin assemblies
by single-molecule force spectroscopy. J Mol Biol 355:640
650
Schenk AD, Werten PJ, Scheuring S, de Groot BL, Muller SA,
Stahlberg H, Philippsen A, Engel A (2005) The 4.5 A structure
of human AQP2. J Mol Biol 350:278289
Scheuring S, Sturgis JN (2005) Chromatic adaptation of photosyn-
thetic membranes. Science 309:484487
Scheuring S, Reiss-Husson F, Engel A, Rigaud JL, Ranck JL (2001)
High-resolution AFM topographs of Rubrivivax gelatinosus
light-harvesting complex LH2. EMBO J 20:30293035
Scheuring S, Seguin J, Marco S, Levy D, Breyton C, Robert B,
Rigaud JL (2003a) AFM characterization of tilt and intrinsic
flexibility of Rhodobacter sphaeroides light harvesting complex
2 (LH2). J Mol Biol 325:569580
Scheuring S, Seguin J, Marco S, Levy D, Robert B, Rigaud JL
(2003b) Nanodissection and high-resolution imaging of the
Rhodopseudomonas viridis photosyn theti c core complex in
native membranes by AFM. Atomic force microscopy. Proc Natl
Acad Sci USA 100:16901693
Scheuring S, Francia F, Busselez J, Melandri BA, Rigaud JL, Levy D
(2004a) Structural role of PufX in the dimerization of the
photosynthetic core complex of Rhodobacter sphaeroides. J Biol
Chem 279:36203626
Scheuring S, Rigaud JL, Sturgis JN (2004b) Variable LH2 stoichiom-
etry and core clustering in native membranes of Rhodospirillum
photometricum. EMBO J 23:41274133
Scheuring S, Goncalves RP, Prima V, Sturgis JN (2006) The
photosynthetic apparatus of Rhodopseudomonas palustris: struc-
tures and organization. J Mol Biol 358:8396
Schmidt T, Schutz GJ, Baumgartner W, Gruber HJ, Schindler H
(1996) Imaging of single molecule diffusion. Proc Natl Acad Sci
USA 93:29262929
Schutz GJ, Schindler H, Schmidt T (1997) Single-molecule micros-
copy on model membranes reveals anomalous diffusion. Biophys
J 73:10731080
Schwille P (2001) Fluorescence correlation spectroscopy and its
potential for intracellular applications. Cell Biochem Biophys
34:383408
Shibata SC, Hibino K, Mashimo T , Yanagida T, Sako Y (2006) Formation
of signal transduction complexes during immobile phase of NGFR
movements. Biochem Biophys Res Commun 342:316322
Suzuki K, Ritchie K, Kajikawa E, Fujiwara T, Kusumi A (2005)
Rapid hop diffusion of a G-protein-coupled receptor in the
Appl Microbiol Biotechnol (2007) 76:257266 265
plasma membrane as revealed by single-molecule techniques.
Biophys J 88:3659 3680
Tanuj SK, Park PS, Filipek S, Engel A, Muller DJ, Palczewski K (2006)
Detecting molecular interactions that stabilize native bovine
rhodopsin. J Mol Biol 358:255269
Teramura Y, Ichinose J, Takagi H, Nishida K, Yanagida T, Sako Y (2006)
Single-molecule analysis of epidermal growth factor binding on the
surface of living cells. EMBO J 25:42154222
Uyemura T, Takagi H, Yanagida T, Sako Y (2005) Single-molecule
analysis of epidermal growth factor signaling that leads to
ultrasensitive calcium response. Biophys J 88:37203730
van Meer G (2005) Cellular lipidomics. EMBO J 24:31593165
Wawrezinieck L, Rigneault H, Marguet D, Lenne PF (2005) Fluores-
cence correlation spectroscopy diffusion laws to probe the submi-
cron cell membrane organization. Biophys J 89:40294042
Wenger J, Conchonaud F, Dintinger J, Wawrezinieck L, Ebbesen TW,
Rigneault H, Marguet D, Lenne PF (2007) Diffusion analysis
within single nanometric apertures reveals the ultrafine cell
membrane organization. Biophys J 92:913919
Zimmerberg J, Gawr isch K (2006) The physical chemistry of
biological membranes. Nat Chem Biol 2:564567
Zlatanova J, Lindsay SM, Leuba SH (2000) Single molecule force
spectroscopy in biology using the atomic force microscope. Prog
Biophys Mol Biol 74:3761
266 Appl Microbiol Biotechnol (2007) 76:257266
... Biological systems are complex and intrinsically heterogeneous with variable distributions of structural conformations. Analysis of these distributions can be useful in deciphering the mechanism of action of biological systems Garcia-Saez and Schwille, 2007). The individual inhomogeneities in the biological molecule can be studied in detail utilizing singlemolecule techniques compared to ensemble measurements (Haustein and Schwille, 2004). ...
... The FCCS technique has been successfully employed to monitor dynamics of protein interactions (Larson et al., 2003) and to track the endocytic pathway of cholera toxin in live cells (Bacia et al., 2002). Combination of FCS with atomic force microscopy (AFM), which is a scanning probe high-resolution imaging technique, for the study of membranes and membraneassociated proteins has attracted a lot of attention (Chiantia et al., 2006a,b;Garcia-Saez and Schwille, 2007). Two-photon excitation is advantageous for FCS studies (Berland et al., 1995;Schwille et al., 1999a,b). ...
Article
Full-text available
Membrane proteins mediate a number of cellular functions and are associated with several diseases and also play a crucial role in pathogenicity. Due to their importance in cellular structure and function, they are important drug targets for ~60% of drugs available in the market. Despite the technological advancement and recent successful outcomes in determining the high-resolution structural snapshot of membrane proteins, the mechanistic details underlining the complex functionalities of membrane proteins is least understood. This is largely due to lack of structural dynamics information pertaining to different functional states of membrane proteins in a membrane environment. Fluorescence spectroscopy is a widely used technique in the analysis of functionally-relevant structure and dynamics of membrane protein. This review is focused on various site-directed fluorescence (SDFL) approaches and their applications to explore structural information, conformational changes, hydration dynamics, and lipid-protein interactions of important classes of membrane proteins that include the pore-forming peptides/proteins, ion channels/transporters and G-protein coupled receptors.
... Techniques for measuring lipid diffusion, membrane dynamics and interactions include fluorescence correlation spectroscopy (FSC) (single molecule or several molecules), Förster Resonance Energy Transfer (FRET) (can be ensemble or single-molecule-based) and single particle tracking (SPT), a single-molecule method. Single-molecule based techniques have been used for assessing molecular heterogeneities in biological membranes through the incorporation of single particles, such as labelled proteins and tracking their behaviour [217]. They are often fluorescence based and are extensively used for the study of membrane proteins and their interactions in live cells. ...
Article
The lipid bilayer is a functional component of cells, forming a stable platform for the initiation of key biological processes, including cell signalling. There are distinct changes in the lipid composition of cell membranes during oncogenic transformation resulting in aberrant activation and inactivation of signalling transduction pathways. Studying the role of the cell membrane in cell signalling is challenging, since techniques are often limited to by timescale, resolution, sensitivity, and averaging. To overcome these limitations, combining ‘computational’, ‘wet-lab’ and ‘semi-dry’ approaches offers the best opportunity to resolving complex biological processes involved in membrane organisation. In this review, we highlight analytical tools that have been applied for the study of cell signalling initiation from the cancer cell membranes through computational microscopy, biological assays, and membrane biophysics. The cancer therapeutic potential of extracellular membrane-modulating agents, such as cholesterol-reducing agents is also discussed, as is the need for future collaborative inter-disciplinary research for studying the role of the cell membrane and its components in cancer therapy.
... Techniques for membrane protein structure investigations include X-ray crystallography (McPherson, 2004), cryoelectron microscopy (cryo-EM) (Thonghin et al., 2018), solution or solid-state nuclear magnetic resonance (NMR) (Liang & Tamm, 2016), and single-molecule tracking with fluorescence spectroscopy (García-Sáez & Schwille, 2007;Alcor, Gouzer, & Triller, 2009;Christie, Shi, & Smith, 2020). However, despite these techniques, the number of membrane protein with known structures is 2488 or 1.5% of the 165,422 structures in Protein Data Bank. ...
Article
Full-text available
Membrane proteins are incredibly important biomolecules because they mediate interactions between a cell's external and internal environment. Obtaining information about membrane protein structure and interactions is thus important for understanding these essential biomolecules. Compared with the analyses of water-soluble proteins, the structural analysis of membrane proteins is more challenging owing to their unique chemical properties and the presence of lipid components that are necessary to solubilize them. The combination of covalent labeling (CL) and mass spectrometry (MS) has recently been applied with great success to study membrane protein structure and interactions. These studies have demonstrated the many advantages that CL-MS methods have over other traditional biophysical techniques. In this review, we discuss both amino acid-specific and non-specific labeling approaches and the special considerations needed to address the unique challenges associated with interrogating membrane proteins. This review highlights the aspects of this approach that require special care to be applied correctly and provides a comprehensive review of the membrane protein systems that have been studied by CL-MS. © 2020 John Wiley & Sons Ltd. Mass Spec Rev.
... Enzyme encapsulation into liposomes has been shown to enable enzyme immobilization while preventing any negative impact due the surface-enzyme or protein-protein interactions 6,7 . In addition, liposomes offer a unique possibility to study single membrane proteins in their natural lipid bilayer environment 8,9,10 . A class of membrane proteins, transporters, exercises a directional translocation of substances across the cell membrane, a behavior that can only be studied when proteins are reconstituted into the lipid bilayers (e.g., liposomes) 11,12,13 . ...
Article
Proton-pumping enzymes of electron transfer chains couple redox reactions to proton translocation across the membrane, creating a proton-motive force used for ATP production. The amphiphilic nature of membrane proteins requires particular attention to their handling and reconstitution into the natural lipid environment is indispensable when studying membrane transport processes like proton translocation. Here, we detail a method that has been used for the investigation of proton-pumping mechanism of membrane redox enzymes, taking cytochrome bo3 from Escherichia coli as an example. A combination of electrochemistry and fluorescence microscopy is used to control the redox state of the quinone pool and monitor pH changes in the lumen. Due to the spatial resolution of fluorescent microscopy, hundreds of liposomes can be measured simultaneously while enzyme content can be scaled down to a single enzyme or transporter per liposome. The respective single enzyme analysis can reveal patterns in the enzyme functional dynamics that might be otherwise hidden by the behavior of whole population. We include a description of a script for automated image analysis.
... Most prominent are fluorescence imaging techniques, such as epifluorescence and confocal fluorescence microscopy. It is very common to employ lasers as an illumination or excitation source -especially in the case of nonlinear optical techniques including two photon microscopy (TPM), second and third generation imaging (SHIM, THIM) or fluorescence lifetime, fluorescence correlation and fluorescence resonance energy transfer microscopy (FLIM, FCM, FRET) [1][2][3][4][5]. ...
Chapter
Full-text available
Raman microscopic imaging of eukaryotic cells has become a well-established technique to address various biochemical questions related to cell biology. The combination of Raman spectroscopy with optical microscopy offers the possibility to study the composition of cells and alterations associated with metabolistic activity induced by internal or external factors. Because of the associated spectral information obtained, Raman microscopy can be utilized without the need for fluorescent dyes or laborious sample preparation. In order to contrast the cellular compartments within the cytosol, imaging algorithms based on multivariate data analysis are commonly employed. Apart from imaging the intracellular organelles and structures of eukaryotes based on their intrinsic biochemical composition, it is possible to follow the uptake of various substances. Externally administered molecules can be labeled with stable isotopes, such as deuterium. The unique spectroscopic features of isotopically labeled compounds provide high specificity and sensitivity without changing the chemical properties of the molecules of interest. The concept of isotopic labeling is illustrated for the uptake of lipids into macrophages, as well as for liposomal drug delivery systems.
Article
Membrane-associated proteins are important because they mediate interactions between a cell's external and internal environment and they are often targets of therapeutics. Characterizing their structures and binding interactions, however, is challenging because they typically must be solubilized using artificial membrane systems that can make measurements difficult. Mass spectrometry (MS) is emerging as a valuable tool for studying membrane-associated proteins, and covalent labeling MS has unique potential to provide higher order structure and binding information for these proteins in complicated membrane systems. Here, we demonstrate that diethylpyrocarbonate (DEPC) can be effectively used as a labeling reagent to characterize the binding interactions between a membrane-associated protein and its binding partners in an artificial membrane system. Using chemotaxis histidine kinase (CheA) as a model system, we demonstrate that DEPC-based covalent labeling MS can provide structural and binding information about the ternary complex of CheA with two other proteins that is consistent with structural models of this membrane-associated chemoreceptor system. Despite the moderate hydrophobicity of DEPC, we find that its reactivity with proteins is not substantially influenced by the presence of the artificial membranes. However, correct structural information for this multiprotein chemoreceptor system requires measurements of DEPC labeling at multiple reagent concentrations to enable an accurate comparison between CheA and its ternary complex in the chemoreceptor system. In addition to providing structural information that is consistent with the model of this complex system, the labeling data supplements structural information that is not sufficiently refined in the chemoreceptor model.
Article
Micro-chamber arrays enable highly sensitive and quantitative bioassays at the single-molecule level. Accordingly, they are widely used for ultra-sensitive biomedical applications, e.g., digital PCR and digital ELISA. However, the versatility of micro-chambers is generally limited to reactions in aqueous solutions, although various functions of membrane proteins are extremely important. To address this issue, microsystems using arrayed micro-sized chambers sealed with lipid bilayers, referred to here as a "biomembrane microsystems", have been developed by many research groups for the analysis of membrane proteins. In this review, I would like to introduce recent progress on the single molecule analysis of membrane transport proteins using a biomembrane microsystem, and discuss the future prospects for its use in analytical and pharmacological applications.
Article
Lipid droplets are dynamic subcellular organelles that participate in a range of physiological processes including metabolism, regulation and lipid storage. Their role in disease, such as cancer, where they are...
Article
The cell membrane is a complex milieu of lipids and proteins. In order to understand the behaviour of individual molecules is it often desirable to examine them as purified components in in vitro systems. Here, we detail the creation and use of droplet interface bilayers (DIBs) which, when coupled to TIRF microscopy, can reveal spatiotemporal and kinetic information for individual membrane proteins. A number of steps are required including modification of the protein sequence to enable the incorporation of appropriate fluorescent labels, expression and purification of the membrane protein and subsequent labelling. Following creation of DIBs, proteins are spontaneously incorporated into the membrane where they can be imaged via conventional single molecule TIRF approaches. Using this strategy, in conjunction with step-wise photobleaching, FRET and / or single particle tracking, a host of parameters can be determined such as oligomerisation state and dynamic information. We discuss advantages and limitations of this system and offer guidance for successful implementation of these approaches.
Chapter
In the past decades, single-molecule fluorescence (SMF) techniques have been booming as they provide researchers with valuable molecular information that are unobtainable by using ensemble techniques. Applications of SMF techniques to live cell membranes have revolutionized our views on many cellular processes. In this chapter, we describe the basics of SMF techniques such as SMF imaging, single-molecule fluorescence resonance energy transfer (sm-FRET), and fluorescence correlation spectroscopy (FCS) and then discuss their contributions to the understanding of live cell membranes. We finally provide the practical protocols of applying SMF techniques to live cell membranes, including the choice of fluorescent probes, labeling strategies, cell sample preparation, instrumentation setup, and data analysis.
Article
Full-text available
The mechanism of CD8 cooperation with the TCR in antigen recognition was studied on live T cells. Fluorescence correlation measurements yielded evidence of the presence of two TCR and CD8 subpopulations with different lateral diffusion rate constants. Independently, evidence for two subpopulations was derived from the experimentally observed two distinct association phases of cognate peptide bound to class I MHC (pMHC) tetramers and the T cells. The fast phase rate constant ((1.7 6 0.2) 3 10 5 M ÿ1 s ÿ1) was independent of examined cell type or MHC-bound peptides' structure. Its value was much faster than that of the association of soluble pMHC and TCR ((7.0 6 0.3) 3 10 3 M ÿ1 s ÿ1), and close to that of the association of soluble pMHC with CD8 ((1–2) 3 10 5 M ÿ1 s ÿ1). The fast binding phase disappeared when CD8-pMHC interaction was blocked by a CD8-specific mAb. The latter rate constant was slowed down ;10-fold after cells treatment with methyl-b-cyclodextrin. These results suggest that the most efficient pMHC-cell association route corresponds to a fast tetramer binding to a colocalized CD8-TCR subpopulation, which apparently resides within membrane rafts: the reaction starts by pMHC association with the CD8. This markedly faster step significantly increases the probability of pMHC-TCR encounters and thereby promotes pMHC association with CD8-proximal TCR. The slow binding phase is assigned to pMHC association with a noncolocali-zed CD8-TCR subpopulation. Taken together with results of cytotoxicity assays, our data suggest that the colocalized, raft-associated CD8-TCR subpopulation is the one capable of inducing T-cell activation.
Article
Full-text available
In photosynthesis, highly organized multiprotein assemblies convert sunlight into biochemical energy with high efficiency. A challenge in structural biology is to analyze such supramolecular complexes in native membranes. Atomic force microscopy (AFM) with high lateral resolution, high signal-to-noise ratio, and the possibility to nanodissect biological samples is a unique tool to investigate multiprotein complexes at molecular resolution in situ. Here we present high-resolution AFM of the photosynthetic core complex in native Rhodopseudomonas viridis membranes. Topographs at 10-Å lateral and 1-Å vertical resolution reveal a single reaction center (RC) surrounded by a closed ellipsoid of 16 light-harvesting (LH1) subunits. Nanodissection of the tetraheme cytochrome (4Hcyt) subunit from the RC allows demonstration that the L and M subunits exhibit an asymmetric topography intimately associated to the LH1 subunits located at the short ellipsis axis. This architecture implies a distance distribution between the antenna and the RC compared with a centered location of the RC within a circular LH1, which may influence the energy transfer within the core complex. The LH1 subunits rearrange into a circle after removal of the RC from the core complex.
Article
Full-text available
Despite the importance of trafficking for regulating G protein-coupled receptor signaling, for many members of the seven transmembrane helix protein family, such as odorant receptors, little is known about this process in live cells. Here, the complete life cycle of the human odorant receptor OR17-40 was directly monitored in living cells by ensemble and single-molecule imaging, using a double-labeling strategy. While the overall, intracellular trafficking of the receptor was visualized continuously by using a GFP tag, selective imaging of cell surface receptors was achieved by pulse-labeling an acyl carrier protein tag. We found that OR17-40 efficiently translocated to the plasma membrane only at low expression, whereas at higher biosynthesis the receptor accumulated in intracellular compartments. Receptors in the plasma membrane showed high turnover resulting from constitutive internalization along the clathrin pathway, even in the absence of ligand. Single-molecule microscopy allowed monitoring of the early, dynamic processes in odorant receptor signaling. Although mobile receptors initially diffused either freely or within domains of various sizes, binding of an agonist or an antagonist increased partitioning of receptors into small domains of ≈190 nm, which likely are precursors of clathrin-coated pits. The binding of a ligand, therefore, resulted in modulation of the continuous, constitutive internalization. After endocytosis, receptors were directed to early endosomes for recycling. This unique mechanism of continuous internalization and recycling of OR17-40 might be instrumental in allowing rapid recovery of odor perception. • cell signaling • G protein-coupled receptors • single-particle tracking • in vivo protein labeling • fluorescence imaging
Book
Atomic force microscopy (AFM) is part of a range of emerging microscopic methods for biologists which offer the magnification range of both the light and electron microscope, but allow imaging under the "natural" conditions usually associated with the light microscope. To biologists, AFM offers the prospect of high resolution images of biological material, images of molecules and their interactions even under physiological conditions, and the study of molecular processes in living systems. This book provides a realistic appreciation of the advantages and limitations of the technique and the present and future potential for improving the understanding of biological systems. The second edition of this bestseller has been updated to describe the latest developments in this exciting field, including a brand new chapter on force spectroscopy. The dramatic developments of AFM over the past ten years from a simple imaging tool to the multi-faceted, nano-manipulating technique that it is today are conveyed in a lively and informative narrative, which provides essential reading for students and experienced researchers alike.
Article
Atomic force microscopy and single-molecule force spectroscopy were combined to image and manipulate purple membrane patches from Halobacterium salinarum. Individual bacteriorhodopsin molecules were first localized and then extracted from the membrane; the remaining vacancies were imaged again. Anchoring forces between 100 and 200 piconewtons for the different helices were found. Upon extraction, the helices were found to unfold. The force spectra revealed the individuality of the unfolding pathways. Helices G and F as well as helices E and D always unfolded pairwise, whereas helices B and C occasionally unfolded one after the other. Experiments with cleaved loops revealed the origin of the individuality: stabilization of helix B by neighboring helices.
Article
Confocal fluorescence correlation spectroscopy (FCS) allows for the determination of lateral diffusion coefficients and surface densities in planar phospholipid systems. The determination of the vertical (z-) position of the laser focus relative to the phospholipid surface plane is of crucial importance for the accuracy of the confocal FCS experiment. In this work we determine for the first time this vertical (z-) position of the laser focus by a so-called "Z-scan", which is based on the determination of diffusion times and particle numbers in 0.2 µm steps along the vertical (z-) axis. Experiments on supported phospholipid bilayers composed of dioleoylphosphatidylcholine (DOPC) and small amounts of Rhodamine Red-X 1,2-dihexa-decanoyl-sn-glycero-3-phosphoethanolamine, triethylammonium salt (Rhodamine Red-X DHPE) adsorbed onto atomically flat mica and borosilicate glass demonstrate that results obtained by the Z-scan approach are significantly more precise than those results obtained when the fluorescence intensity maximum is used as an indicator in the determination of the vertical (z-) position of the sample. In addition to this basic contribution for the investigation of planar bilayer systems by confocal FCS, the lateral diffusion coefficients of Rhodamine Red-X DHPE in supported phospholipid bilayers composed of DOPC and cholesterol as well as in DOPC or dipalmitoylphosphatidylcholine (DPPC) monolayers adsorbed at a liquid-liquid interface were determined.
Article
We report on an in vivo single-molecule study of the signaling kinetics of G protein-coupled receptors (GPCR) performed using the neurokinin 1 receptor (NK1R) as a representative member. The NK1R signaling cascade is triggered by the specific binding of a fluorescently labeled agonist, substance P (SP). The diffusion of single receptor–ligand complexes in plasma membrane of living HEK 293 cells is imaged using fast single-molecule wide-field fluorescence microscopy at 100 ms time resolution. Diffusion trajectories are obtained which show intra- and intertrace heterogeneity in the diffusion mode. To investigate universal patterns in the diffusion trajectories we take the ligand-binding event as the common starting point. This synchronization allows us to observe changes in the character of the ligand–receptor-complex diffusion. Specifically, we find that the diffusion of ligand–receptor complexes is slowed down significantly and becomes more constrained as a function of time during the first 1000 ms. The decelerated and more constrained diffusion is attributed to an increasing interaction of the GPCR with cellular structures after the ligand–receptor complex is formed.
Article
Located in the principal cells of the collecting duct, aquaporin-2 (AQP2) is responsible for the regulated water reabsorbtion in the kidney and is indispensable for the maintenance of body water balance. Disregulation or malfunctioning of AQP2 can lead to severe diseases such as nephrogenic diabetes insipidus, congestive heart failure, liver cirrhosis and pre-eclampsia. Here we present the crystallization of recombinantly expressed human AQP2 into two-dimensional protein-lipid arrays and their structural characterization by atomic force microscopy and electron crystallography. These crystals are double-layered sheets that have a diameter of up to 30 μm, diffract to 3 Å−1 and are stacked by contacts between their cytosolic surfaces. The structure determined to 4.5 Å resolution in the plane of the membrane reveals the typical aquaporin fold but also a particular structure between the stacked layers that is likely to be related to the cytosolic N and C termini.
Article
um in translocation of AQP1 in cholangiocytes from intracellular vesicles to the plasma membrane and in triggering its fusion is discussed. Functional studies are now required to identify the physiological role of the Ca2+-binding site.