No Better Time To FRET

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 4

Hayward et al.

Journal of Biology 2010, 9:12


http://jbiol.com/content/9/2/12

MINIREVIEW

No better time to FRET: shedding light on host


pathogen interactions
Richard D Hayward*1, Jon D Goguen2 and John M Leong2
See research article http://www.biomedcentral.com/1741-7007/7/81

transduction, cytoskeletal dynamics, intracellular


Abstract trafficking and cytokinesis. An exciting new epoch of
Understanding the spatio-temporal subversion of molecular, cellular and structural microbiology has
host cell signaling by bacterial virulence factors is therefore dawned.
key to combating infectious diseases. Following a In recent years, substantial research efforts have
recent study by Buntru and co-workers published in focused on the actions of such bacterial virulence
BMC Biology, we review how fluorescence (Forster) proteins and the identity of their host targets, clearly an
resonance energy transfer (FRET) has been applied to essential initial step. However, an appreciation of the
studying host-pathogen interactions and consider the spatio-temporal dynamics underlying the intricate
prospects for its future application. molecular cross-talk that triggers complex events such as
bacterial internalization into host cells remains a distant
goal. Most biochemical and genetic approaches entail
Pathogen-induced signaling in mammalian cells cell disruption or artificial protein localization and
The incidence of diseases caused by pathogenic bacteria expression, and observations arise from population
is once again increasing with the resurgence of rather than single cell analyses. In addition, many
tuberculosis, the rise of nosocomial infections, and signaling interactions are transient or of low-affinity and
escalating resistance to antibiotics. Over the last two thus difficult to detect.
decades it has become increasingly apparent that many
bacterial pathogens of substantial medical relevance Fluorescence (Forster) resonance energy transfer
engage in complex cross-talk with cells of their (FRET) to detect direct interactions between
mammalian hosts. It is thus critical to decipher the labeled molecules
molecular mechanisms of the underlying bacterial One powerful approach is fluorescence (Forster)
virulence weaponry, which includes not only the secreted resonance energy transfer (FRET) [1]. FRET utilizes two
exotoxins, like those that cause anthrax and cholera, but fluorophores, a donor and an acceptor, of which the
also the multiple ‘effectors’ injected directly into donor emission spectrum overlaps with the acceptor
eukaryotic target cells via specialized nanomachines by absorption spectrum. If the two fluorophores are spatially
pathogens like Escherichia coli O157:H7 and Yersinia. segregated, excitation of the donor results in donor
Such studies have already yielded new insights into the emission with high efficiency. In contrast, when in close
molecular basis of microbial pathogenesis, suggesting proximity (usually 1-10 nm), excitation of the donor
new avenues for the development of novel diagnostics, results in acceptor emission due to the overlap in spectra
therapeutics and vaccines. These toxins and effectors can that allows resonance energy transfer between the donor
also be exploited as reagents to probe the pathways and the acceptor probes (Figure 1). A modification of
controlling key cellular processes such as signal FRET termed acceptor photobleaching FRET involves
selective photochemical destruction of the acceptor
*Correspondence: richard.hayward@ucl.ac.uk fluorophore, which, if the two fluorophores had
1
Institute of Structural and Molecular Biology, University College London and previously been physically close enough for FRET, results
Birkbeck, University of London, Gower Street, London WC1E 6BT, UK
2
Department of Molecular Genetics and Microbiology, UMass Medical School,
in a release from donor quenching and an increase in
55 Lake Ave North, Worcester, MA 01655, USA donor emission. Technically, this is particularly
Full list of author information is available at the end of the article advantageous as it reduces the requirements for
compensation and calibration associated with standard
© 2010 BioMed Central Ltd © 2010 BioMed Central Ltd FRET.
Hayward et al. Journal of Biology 2010, 9:12 Page 2 of 4
http://jbiol.com/content/9/2/12

subunit embedded within an isopentamer of CTX-B


L L moieties. The secreted holotoxin binds to the host cell
surface via interactions between CTX-B and plasma
membrane GM1 ganglioside. After internalization and
processing, CTX-B and CTX-A dissociate, whereupon a
a liberated sub-fragment of CTX-A irreversibly ADP-
YFP ribosylates the α-subunits of heterotrimeric Gs proteins,
leading to aberrant persistent activation of host adenylate
436nm CFP CFP 535nm cyclase. Compartmentalization and separation of the
FRET
holotoxin were evaluated by measuring acceptor
photobleaching FRET between CTX-B labeled with the
480nm 480nm sulfoindocyanine dye Cy3 and an antibody against
CTX-A labeled with Cy5 in single cells, exploiting
confocal laser scanning microscopy. The data illuminated
a YFP that after holotoxin internalization, CTX-A is trafficked
back towards the plasma membrane by retrograde
Figure 1. Schematic representation of a hypothetical FRET
experiment. In the resting state (left), a transmembrane receptor is
transport, whereas the CTX-B multimer persists with the
fused to cyan fluorescent protein (CFP, donor). Adaptor protein (a) is Golgi apparatus. This study was one of the first to apply
fused to yellow fluorescent protein (YFP, acceptor). a-YFP is distal FRET to a pathogen system and contributed to
from receptor-CFP, so upon excitation at 436 nm, donor fluorescence understanding the unexpected complexities of
at 480 nm is recorded. Upon binding to a ligand (L, right), a-YFP binds intracellular CTX transport.
to receptor-CFP and the reduction in distance enables FRET. Upon
equivalent excitation at 436 nm, donor fluorescence (480 nm) is
A related study of Helicobacter pylori vacuolating
reduced, but acceptor fluorescence at 535 nm is now recorded due cytotoxin (VacA), which binds and enters mammalian
to FRET. FRET can similarly be performed with two transmembrane or cells to induce cellular vacuolation, also addressed toxin
two soluble factors. multimerization. In this case FRET was monitored within
cells co-expressing VacA and derivative proteins fused to
cyan or yellow fluorescent protein, where association
FRET is now widely applied in the context of cellular induces energy transfer between the VacA-CFP donor
signaling to detect interactions such as the formation of and VacA-YFP acceptor pairs [3]. These FRET data
protein complexes, and to detail molecular dynamics suggested that intermolecular interactions between
such as changes in protein conformation. When discrete monomers are critical for intracellular activity of
combined with fluorescence microscopy, it can the toxin.
simultaneously provide information about the location
within a single cell in which an interaction is occurring in Probing pathogen-host cell interplay that triggers
two and three dimensions. Given the power of this or prevents bacterial uptake
technique, its use in understanding host-pathogen These early FRET-based studies of bacterial exotoxins
interplay has, to date, been surprisingly limited. Here we focused on understanding the intermolecular inter­
review the principal studies that have utilized FRET to actions between toxin subunits rather than between
understand signaling events, in each case initiated at the toxins and their host targets. Another timely and
plasma membrane by a bacterium, and we consider the ambitious application of FRET has been to examine the
prospects for future applications. cellular signaling pathways underpinning the interaction
of bacterial pathogens with both phagocytic and non-
Probing the intermolecular interactions of phagocytic host cells.
bacterial exotoxins The Yersinia outer membrane protein invasin acts as a
Some pathogenic bacteria release exotoxins that poison high affinity ligand for cellular β1-family integrins,
mammalian target cells directly by disrupting their transmembrane receptors involved in the formation of
membrane integrity or indirectly using associated multi-protein structures termed focal adhesions that link
enzymatic activities that override cellular pathways. the extracellular matrix to the intracellular cytoskeleton.
These virulence factors alone are often sufficient to cause Invasin-mediated clustering of integrins triggers host
disease, and their activities normally require toxin signaling on the cytoplasmic face of the plasma
oligomerization and interaction with host targets. membrane. This requires the activation of the small
Bastiaens and colleagues investigated one example of GTPase Rac1, which subsequently binds downstream
this [2]. Cholera toxin (CTX) has an oligomeric AB5 adaptors that promote cytoskeletal rearrangements and
structure, comprising one enzymatically active CTX-A bacterial internalization. Ralph Isberg’s laboratory
Hayward et al. Journal of Biology 2010, 9:12 Page 3 of 4
http://jbiol.com/content/9/2/12

demonstrated that Rac1 was activated at the site of They used FRET to investigate the activation of two host
internalization by normally non-phagocytic cells factors critical for the entry process, Rac1 and
(Figure 2), detecting the presence of activated CFP-Rac1 phosphoinositide (PI)-3-kinase, which generates the
by its ability to bind to a YFP-labeled domain of its bioactive signaling lipids phosphatidylinositol-3,4-
downstream adaptor PAK1 [4]. Although Yersinia is bisphosphate [PI(3,4)P2] and phosphatidylinositol-3,4,5-
capable of entry into non-phagocytic cells, the bacterium triphosphate [PI(3,4,5)P3]. Rac1 activation was
paradoxically also utilizes a specialized (type III) monitored similarly to that described above. To monitor
secretion system to translocate effector proteins that activation of PI-3-kinase, the investigators co-expressed
prevent its uptake by phagocytic immune cells. Isberg YFP and CFP derivatives of the pleckstrin-homology
and co-workers used FRET to show that the concerted (PH) domain of the serine/threonine kinase Akt, which
action of two effectors, YopE, which suppresses Rac1 specifically interacts with these phosphoinositides.
activation, and YopT, which alters Rac1 membrane Bacterial attachment resulted in a localized FRET signal, as
localization, generated two spatially distinct Rac1 PI(3,4)P2 and PI(3,4,5)P3 were sufficiently concentrated
populations, an active pool in the nucleus and an inactive at the plasma membrane to permit a FRET signal
pool in the cytoplasm, leading to cellular paralysis. Thus, between the bound Akt PH domains. Their kinetic analy­
FRET, being uniquely suited to investigating the location sis of living cells showed that activation of PI3-kinase and
and activation state of host molecules, was integral to concomitant generation of 3’-phosphoinositides at
uncovering this multifaceted manipulation of GTPase- bacterial entry sites occurs upstream of Rac1 activation,
dependent signaling [5]. which in turn is critical for F-actin assembly. Thus, FRET
FRET can be combined with live cell imaging to reveal enabled both the spatial and temporal mapping of lipid-
not only detailed interactions critical to manipulation of and protein-based signaling at the plasma membrane.
host cells by bacteria, as described above, but also the Most recently, in their study in BMC Biology [7], Hauck
kinetics of those interactions, thereby establishing a and co-workers combined live fluorescence microscopy
specific sequence of events. Such a study resulted from a and FRET techniques to study events triggered by the
collaboration between the laboratories of Pascale Cossart association of Neisseria gonorrhoeae surface (Opa)
and Joel Swanson, who combined FRET with live cell proteins with the mammalian transmembrane receptor
imaging to document the kinetics of signaling during CEACAM3. Uptake of CEACAM3-bound bacteria
Listeria cell entry [6]. Listeria monocytogenes employs depends on an immunoreceptor tyrosine-based activa­
outer membrane proteins termed internalins to hijack tion motif (ITAM)-like sequence within the cytoplasmic
Rac1-dependent receptor-mediated endocytosis. domain of the receptor, which is rapidly phosphorylated
Internalin B (InlB) binds the hepatocyte growth factor upon ligand binding. This is engaged by multiple host
receptor (HGFR/c-Met) to stimulate actin reorganization. signaling proteins that contain a Src-homology 2 (SH2)

PBD-mYFP mCFP-Rac1 phase FRET

Figure 2. Yersinia pseudotuberculosis binding to host cells leads to local activation of Rac1 GTPase. COS1 cells, expressing mCFP-Rac1 and
mYFP fused to the p21 binding domain of Pak1 (PBD) were incubated with an effector-deficient Y. pseudotuberculosis for 20 minutes, then fixed.
GTP-loaded Rac1 interaction with the mYFP-PBD brings fused mCFP and mYFP into close proximity, allowing energy transfer. This energy transfer
is recorded microscopically as a corrected FRET image. CFP, YFP, and FRET images were captured using appropriate filter cube sets and the FRET
image was corrected for bleed-through and cross-excitation. Scale bar (applicable to all images except insets) = 10 μm. Activation of Rac1 in
response to bacterial binding is depicted here. Images courtesy of Sima Mohammadi and Ralph Isberg.
Hayward et al. Journal of Biology 2010, 9:12 Page 4 of 4
http://jbiol.com/content/9/2/12

domain, including the Src-family kinase (SFK) Hck. agents, but also fundamental features of mammalian cell
Using acceptor photobleaching FRET, as well as other signaling. As illustrated here, FRET-based techniques
approaches, Hauck and colleagues demonstrated that that enable the description of the activation state of key
Hck and CEACAM3 transiently but directly interact signaling molecules, their cellular location, proximity to
specifically at sites of bacterial attachment [7]. Although each other, and the timing of their interactions, have
the interaction of these two mammalian signaling already proved to be powerful tools to decipher the
proteins was predicted from earlier biochemical studies, spatiotemporal features of critical signaling events. Given
this investigation confirmed the prediction and also the fundamental importance of microbial-host cell
revealed the dynamic nature of the association in living communication and the growing capacity for its
cells. application, FRET should be more widely employed by
investigators of infectious agents. Making FLIM-capable
FLIM-FRET: it’s easy to FRET instruments more widely available and accessible for use
Measuring FRET can be simplified considerably by with samples containing live infectious agents will be
determining the fluorescence lifetime rather than needed to realize this welcome development.
fluorescence intensities. Energy transfer from the donor
Acknowledgements
to the acceptor accelerates the decay of donor We thank Ralph Isberg for helpful discussion and Sima Mohammadi for Figure
fluorescence, allowing energy transfer efficiency to be 2. This was supported by NIH R01 AI46454 to JML. RDH is a Royal Society
determined directly from measurements of fluorescence University Research Fellow.

lifetime. Only a single measurement is required and, Author details


because many sources of artifact and noise that modulate 1
Institute of Structural and Molecular Biology, University College London and
fluorescence intensity such as sample absorption and Birkbeck, University of London, Gower Street, London WC1E 6BT, UK
2
Department of Molecular Genetics and Microbiology, UMass Medical School,
variation in laser intensity have no effect on fluorescence 55 Lake Ave North, Worcester, MA 01655, USA
lifetime, the requirements for calibration are minimal.
For these reasons, fluorescence lifetime imaging (FLIM) Published: 18 February 2010

is rapidly becoming the preferred method for making References


FRET measurements. This technique has been utilized to 1. Förster T: Intermolecular energy migration and fluorescence. Ann Phys
investigate plant cell invasion by the ascomycete powdery 1948, 2:55-75.
2. Bastiaens PHI, Majoul IV, Verveer PJ, Söling H-D, Jovin TM: Imaging the
mildew fungus [8], but has yet to be applied to any intracellular trafficking and state of the toxin AB5 quaternary structure of
human pathogen. cholera toxin. EMBO J 1996, 15:4246-4253.
In addition, FLIM-FRET facilitates the use of FRET as a 3. Willhite DC, Ye D, Blanke SR: Fluorescence resonance energy transfer
microscopy of the Helicobacter pylori vacuolating cytotoxin within
‘spectroscopic ruler’ to measure the physical distance mammalian cells. Infect Immun 2002, 70:3824-3832.
between donor and acceptor fluorophores, a technique 4. Wong KW, Isberg RR: Yersinia pseudotuberculosis spatially controls
utilized by Latz and colleagues [9] to decipher signaling activation and misregulation of host cell Rac1. PLoS Pathog 2005, 1:e16.
5. Wong KW, Mohammadi S, Isberg RR: The polybasic region of Rac1
associated with immune recognition of CpG DNA, a modulates bacterial uptake independently of self-association and
pathogen-associated molecular pattern (PAMP), by the membrane targeting. J Biol Chem 2008, 283:35954-35965.
Toll-like receptor TLR9. To determine how engagement 6. Seveau S, Tham TN, Payrastre B, Hoppe AD, Swanson JA, Cossart P: A FRET
analysis to unravel the role of cholesterol in Rac1 and PI 3-kinase
of the ectodomain of TLR9 might transmit an activation in the InlB/Met signalling pathway. Cell Microbiol 2007,
intracellular signal, they expressed both CFP-TLR9 and 9:790-803.
YFP-TLR9 in cultured cells and measured the distance 7. Buntru A, Zimmermann T, Hauck CR: Fluorescence resonance energy
transfer (FRET) based subcellular visualization of pathogen-induced host
between adjacent TLR9 cytoplasmic domains using receptor signalling. BMC Biol 2009, 7:81.
FLIM-FRET. They calculated that receptor engagement 8. Bhat RA, Miklis M, Schmelzer E, Schulze-Lefert P, Panstruga R: Recruitment
resulted in a decrease in the intermolecular distance and interaction dynamics of plant penetration resistance components in a
plasma membrane microdomain. Proc Natl Acad Sci USA 2005,
from 7.0 nm to less than 5.4 nm, a change apparently
102:3135-3140.
sufficient to trigger immune signaling by recruiting the 9. Latz E, Verma A, Visintin A, Gong M, Sirois CM, Klein DC, Monks BG, McKnight
downstream adaptor molecule MyD88. CJ, Lamphier MS, Duprex WP, Espevik T, Golenbock DT: Ligand-induced
conformational changes allosterically activate Toll-like receptor 9. Nat
Immunol 2007, 8:772-779.
Future perspectives
The investigation of pathogen-induced signal transduc­ doi:10.1186/jbiol225
tion in mammalian cells is a rich vein to tap for Cite this article as: Hayward RD et al. No better time to FRET: shedding light
on host pathogen interactions. Journal of Biology 2010, 9:12.
understanding not only the pathogenesis of infectious

You might also like