You are on page 1of 7

Journal of Structural Biology 175 (2011) 120–126

Contents lists available at ScienceDirect

Journal of Structural Biology


journal homepage: www.elsevier.com/locate/yjsbi

Review

Cells under siege: Viral glycoprotein interactions at the cell surface


Thomas A. Bowden a,⇑, E. Yvonne Jones a, David I. Stuart a,b
a
Division of Structural Biology, University of Oxford, Wellcome Trust Centre for Human Genetics, Roosevelt Drive, Oxford OX3 7BN, United Kingdom
b
Science Division, Diamond Light Source Ltd., Diamond House, Harwell Science and Innovation Campus, Didcot, Oxfordshire 0X11 0DE, United Kingdom

a r t i c l e i n f o a b s t r a c t

Article history: As obligate parasites, viruses are required to enter and replicate within their host, a process which
Available online 31 March 2011 employs many of their proteins to hijack natural cellular processes. High resolution X-ray crystallo-
graphic analysis has proven to be an ideal method to visualize the mechanisms by which such virus-host
Keywords: interactions occur and has revealed the innovative capacity of viruses to adapt efficiently to their hosts. In
Glycoprotein structure this review, we draw upon recently elucidated paramyxovirus-, arenavirus-, and poxvirus-host protein
Virus entry complex crystal structures to reveal both the capacity of viruses to appropriate one component of a phys-
Cell signaling
iological protein–protein binding event (often modifying it to out-compete the host-protein), and the
X-ray crystallography
Cell surface receptors
ability to utilize novel binding sites on host cell surface receptors. The structures discussed shed light
on a number of biological processes ranging from viral entry to virulence and host antagonism. Drawn
together they reveal the common strategies which viruses have evolved to interact with their natural
host. The structures also support molecular level rationales for how viruses can be transmitted to unre-
lated organisms and thus pose severe health risks.
Ó 2011 Elsevier Inc. All rights reserved.

1. Introduction Structural investigations of viral attachment glycoproteins, for


example, have shown that enveloped viruses adopt a wide range
Viruses have tremendous genetic diversity (Edwards and Roh- of folds optimized for engagement of their cognate cellular recep-
wer, 2005), a property largely accounted for by rapid replication, tors. These folds vary from the compact and novel a/b fold of
frequent and unspecific mutations arising from error-prone poly- Arenaviridae (Fig. 1A) (Abraham et al., 2010; Bowden et al.,
merases, and an ability to recombine host genes into their own 2009a), to the trimeric GP1 ‘chalice’ of the Filoviridae (Fig. 1B)
genome. The resulting capability of a single virion to generate a (Lee et al., 2008), the globular six-bladed b-propeller of the
genetically diverse complement of progeny provides a simple Paramyxovirinae (Fig. 1C) (Bowden et al., 2010a), the large trimeric
mechanism by which virus-host cell interactions can rapidly be- hemagglutinin of the Orthomyxoviridae (Fig. 1D) (Weis et al., 1988;
come specialized for specific host ranges and tissues. Combined Wilson et al., 1981), and the highly glycosylated GP120 trimer of
with the ability to ‘steal’ host proteins, this provides a powerful Lentiviruses in the Retroviridae (Wyatt et al., 1998; Zhu et al.,
method by which viruses hijack natural host cell functions, facili- 2003). It is noteworthy that the associated fusion glycoproteins
tating processes such as viral attachment and antagonism of the from each of the above virus families, in contrast to the attachment
host’s innate immune response (Bahar et al., 2011). glycoproteins, are similar in architecture and have all been
Crystallographic studies of viral proteins alone and in complex grouped into the first of the three known structural classes of
with their functional ligands have led to a greater appreciation of fusion proteins (Eschli et al., 2006; Lamb and Jardetzky, 2007;
how the structurally dissimilar fold architectures resulting from Lee et al., 2008) (Fig. 1AD). It has been suggested that these
viral genomic diversity can achieve analogous biological processes. proteins are related (Kadlec et al., 2008).
Fusion and receptor-binding proteins are often synthesized on
the same polypeptide and fold together to form complexes on
Abbreviations: GAP, GTPase-activating protein; IPT, Ig-like plexins and tran-
the virus surface. As a result, these protein pairs have not evolved
scription factors; HeV, Hendra virus; HeV-G, Hendra virus attachment glycoprotein;
HNV, Henipavirus; HNV-G, Henipavirus attachment glycoprotein; NiV, Nipah virus; entirely independently. Nevertheless, receptor-binding attachment
NiV-G, Nipah virus attachment glycoprotein; MACV, Machupo virus; PDB, protein glycoproteins display much greater structural diversity than their
databank; PSI, plexin-semaphrorin-integrin domain; r.m.s.d., root mean square fusion glycoprotein counter-parts. This is likely to stem from
deviation; Tf, transferrin; TfR1, transferrin receptor 1; SLAM, Signaling Lymphocytic whether there is a functional requirement for a given viral protein
Activation Molecule; SPINE, Structural Proteomics In Europe.
⇑ Corresponding author. Fax: +44 (0)1865 287547. to adapt to and interact with its hosts. Viral nucleoproteins and fu-
E-mail address: tom@strubi.ox.ac.uk (T.A. Bowden). sion glycoproteins (with the exception of the immunosuppressive

1047-8477/$ - see front matter Ó 2011 Elsevier Inc. All rights reserved.
doi:10.1016/j.jsb.2011.03.016
T.A. Bowden et al. / Journal of Structural Biology 175 (2011) 120–126 121

Fig. 1. Contrasts in fold conservation between attachment and fusion glycoproteins from negative-sense, single-stranded RNA viruses. Receptor-binding attachment
glycoproteins are shown above and the six-helix bundles from their cognate fusion glycoproteins below from (A) Machupo Arenavirus (PDB identification number 2WFO), (B)
Ebola Filovirus (PDB ID 3CSY and 1EBO), (C) Nipah (above) and Parainfluenza type-III (below) Paramyxoviruses (PDB ID 2VSM and 1ZTM, respectively), and (D) Flu
Orthomyxovirus (PDB ID 3LZG). Note for panel A, there are currently no known crystal structures of arenaviral fusion glycoproteins, in panel B, Ebola GP1 is shown with its
non-covalently associated GP2 subunit (gray cartoon), and in panel D, Flu virus haemagglutinin is shown with the HA1 domain colored as a rainbow and the HA2 domain
colored gray.

segments of some fusion glycoproteins) (Avota et al., 2010; Cianciolo well established viral glycoprotein systems including HIV and
et al., 1985; Kleinerman et al., 1987; Volchkov et al., 2001; Yang Measles virus, we illustrate that viruses not only subvert binding
et al., 2000), interact less-specifically with their host cell and have sites that are used in natural physiological signaling processes,
a relatively self-contained function (e.g. insertion into and merging but can also exploit novel sites on host proteins previously not
of the viral and host envelopes or packaging of genomic material) used as interaction surfaces.
which requires minimal adaption. Certain non-structural proteins
and attachment glycoproteins, on the other hand, are examples of
viral proteins which more often interact specifically with their host 2. Viral semaphorins and immune antagonism
cell and adapt rapidly to cellular host factors.
Using recently elucidated paramyxovirus-, arenavirus-, and Semaphorins comprise a family of cell surface signaling glyco-
poxvirus-host complex crystal structures resulting from the Spine proteins which, through binding to the family of plexin glycopro-
(Structural Proteomics In Europe) 2-Complexes initiative as exam- tein cell surface receptors, activate repulsive guidance pathways
ples (detailed in Table 1), we draw upon this second, adaptive which are fundamental to a number of physiological processes
group of viral proteins to reveal the varied strategies employed including axon guidance, immune regulation and activation, and
by viruses when interacting with their hosts. Setting these results vascular development (Kruger et al., 2005; Suzuki et al., 2007).
in a broader context, through comparison with other structurally There are eight known classes of semaphorins: two found in inver-
tebrates, five in vertebrates, and the eighth class in viruses which
are known as ‘viral semaphorins’ (Comeau et al., 1998; Ensser
Table 1 and Fleckenstein, 1995). Whilst the ectodomains of cellular sem-
Relevant structures solved under the European Spine initiatives. aphorins contain C-terminal domain elaborations such as PSI
Structure Reference PDB accession code
(plexin, semaphorin and integrin) domains, immunoglobulin (Ig)-
a
like domains, thrombospondin domains and PDZ-domain-binding
Semaphorin4DEcto Love et al. (2003) 1OLZ
PlexinA2D1-4b Janssen et al. (2010) 3OKT
sites which may or may not attach to the cell-surface, the
Semaphorin6AEcto Janssen et al. (2010) 3OKW N-terminal portion, comprising a plexin-binding sema-domain, is
Semaphorin4DEcto-PlexinB1D1-2 Janssen et al. (2010) 3OL2 well conserved. The sema-domain is the only component found
Semaphorin6AEcto-PlexinA2D1-4 Janssen et al. (2010) 3OKY in viruses. Crystallographic studies, by ourselves and others, have
EphA4LBDc Bowden et al. (2009b) 2WO1
shown that the human Sema3A and mouse Sema4D semadomains
EphA4LBD-ephrinB2RBDd Bowden et al. (2009b) 2WO2
EphA4LBD-ephrinA2RBD Bowden et al. (2009b) 2WO3 consist of a structurally conserved homodimer of seven-bladed
EphA2Ecto Seiradake et al. (2010) 2X10 b-propellers (1.7 Å root mean square deviation, r.m.s.d., for match-
EphA2Ecto-ephrinA5RBD Seiradake et al. (2010) 2X11 ing Ca atoms) (Antipenko et al., 2003; Janssen et al., 2010; Liu
NiV-G Bowden et al. (2008b) 2VWD et al., 2010; Love et al., 2003; Nogi et al., 2010).
HeV-G Bowden et al. (2010b) 2X9M
NiV-G-ephrinB2 Bowden et al. (2008a) 2VSM
The domain architecture is conserved amongst the four classes
HeV-G-ephrinB2 Bowden et al. (2008a) 2VSK (A–D) of vertebrate plexin type-I membrane glycoproteins and
MACV-GP1 Bowden et al. (2009a) 2WFO consists of an N-terminal, membrane distal sema-domain which
a is anchored to the membrane by PSI domains and IPT (Ig-like, plex-
Ecto, entire ectodomain.
b
D1–4, domains 1–4. ins and transcription factors) domains (Bork et al., 1999). A
c
LBD, ligand binding domain. GTPase-binding domain and a C-terminal segment GAP (GTPase-
d
RBD, receptor binding domain. activating protein) domain constitute the intracellular portion of
122 T.A. Bowden et al. / Journal of Structural Biology 175 (2011) 120–126

the glycoprotein and are responsible for activation of Rho family 3. Henipavirus entry: The ephrin gateway
GTPase signaling pathways within the plexin-expressing cell (Kru-
ger et al., 2005). Nipah virus and Hendra virus compose the genus Henipavirus
Immunoregulatory semaphorins including Sema3A, 4A, 4D, and within the Paramyxoviridae family and are emergent and highly
7A contribute to B cell mediated immunity (Sema4D), T cell activa- virulent bat-borne pathogens found in Africa, Australia, and South
tion and differentiation (Sema4A, Sema3A, and Sema4D), and East Asia (Eaton et al., 2006; Wild, 2009). Oligomeric complexes of
inflammation (Sema7A) (Suzuki et al., 2008). Genomic sequencing two glycoproteins extending outward from the viral envelope are
of eukaryotic viruses has revealed that Poxviruses (e.g. Smallpox required for efficient attachment (G glycoprotein) and fusion (F
virus and Vaccinia virus) and alcelaphine herpes virus encode viral glycoprotein) into their respective host cells. During henipaviral
semaphorins which have been shown to modulate these processes attachment, the F glycoprotein is activated in a pH independent
(Comeau et al., 1998; Ensser and Fleckenstein, 1995; Suzuki et al., mechanism (Aguilar et al., 2009) to undergo classical class I fusion
2008). These viral encoded proteins were presumably ‘stolen’ from rearrangements which merge the host and viral membranes (Lou
their host during the process of virus-host co-evolution (Suzuki et al., 2006).
et al., 2008). The glycoprotein A39R, encoded by Vaccinia virus, NiV-G and HeV-G (collectively referred to as HNV-G) are
for example, shares highest sequence homology with Sema7A type-II transmembrane glycoproteins consisting of an N-terminal
(30%) and undermines the host immune response by binding to cytoplasmic tail, a short transmembrane region, an ectodomain
plexinC1 (Comeau et al., 1998). stalk region, and a C-terminal receptor binding six-bladed b-pro-
Recent crystallographic studies of semaphorin-plexin signaling peller domain. HNV-G glycoproteins are important in determin-
complexes, in part within the activity of Spine2-Complexes, have ing the broad species and cellular tropism of these viruses as
enabled a detailed comparison of the mechanism by which they have been observed to bind specifically with ephrinB2
natural- and viral-semaphorins bind to plexins. Structures of and ephrinB3 cell surface receptors at nanomolar affinity (Bona-
Sema7A-plexinC1 (Liu et al., 2010), Sema6A-plexinA2 (Janssen parte et al., 2005; Negrete et al., 2005, 2006). The sequences of
et al., 2010; Nogi et al., 2010), and Sema4D-plexinB1(Janssen ephrinB2 and ephrinB3 are well conserved amongst many verte-
et al., 2010), have revealed structurally similar signaling com- brate species including humans, bats, horses, and pigs (>95% se-
plexes, all composed of a Sema-plexin heterotetramer where each quence identity) (Bossart et al., 2008), and they are ubiquitously
protomer of the semaphorin dimer binds to one plexin seven- expressed in most human tissues due to their importance in fun-
bladed b-propeller sema domain (Fig. 2A and B). Elucidation of damental bi-directional cell signaling processes such as osteo-
the crystal structure of vaccinia virus A39R in complex with plex- genesis, axon guidance, and vascular development (Hafner
inC1 demonstrates that poxviruses take advantage of an almost et al., 2004; Pasquale, 2005).
identical binding mechanism to that of physiological semaphorin- Crystal structures of ephrin ligands alone and in complex with
plexin signaling, where both the viral and the cell semaphorin their Eph receptors (determined by others and as part of Spine2-
b-propellers bind to their cognate plexin b-propellers in a side-on Complexes) have been invaluable for identifying the molecular
orientation (Fig. 2C) (Liu et al., 2010). Furthermore, although the specificity which underlies normal physiological signaling events.
binding interface of the viral complex is less extensive to that These studies reveal that the ephrin ectodomain forms a compact
observed in analogous physiological complexes (Sema7A-plex- greek-key fold containing a 10 amino acid (GH) binding loop,
inC1), the binding strength of the A39R-plexinC1 interaction is which is predominantly responsible for Eph receptor binding
significantly enhanced (Kd of 10 nM versus 300 nM) (Liu through its insertion into the receptor binding cleft of the mem-
et al., 2010). Whilst Sema-plexin binding affinities are delicately brane-distal Eph receptor b-sandwich domain (Bowden et al.,
balanced to contribute to the complex interplay of interactions 2009b; Chrencik et al., 2006a, 2006b; Himanen et al., 2001, 2009,
required for physiological functions, Vaccinia virus protein A39R 2010, 2004; Nikolov et al., 2007, 2005; Qin et al., 2010; Seiradake
can simply optimize a single interaction. These studies are an et al., 2010; Toth et al., 2001) (Fig. 3A).
example of how genetic variability can give rise to mechanisms Site-directed mutagenesis of ephrinB2 and ephrinB3 confirmed
which enhance virus virulence and replication. In addition to incor- that HNV-G subverts natural Eph receptor binding by also utilizing
porating genes from their host organism, these structures provide a this GH loop during viral attachment (Negrete et al., 2006). How-
molecular basis for how viruses can optimize their own proteins to ever, rather than completely imitating the exact binding mode ob-
override normal physiological interactions. served in physiological Eph-ephrin interactions, structural studies

Fig. 2. Poxviral appropriation of semaphorin-plexin interactions. (A) Crystal structure of Sema7a in complex with PlexinC1 (PDB ID 3NVQ). The PlexinC1 seven-bladed b-
propeller domains are rendered as gray surfaces and the Sema7A dimer is shown as a cartoon with each protomer colored as a rainbow with the N-terminus in blue and the C-
terminus in red. Close-up view of protein-protein interactions in the (B) Sema7a-PlexinC1 and (C) poxvirus A39R-PlexinC1 complex (PDB ID 3NVN) interfaces reveal nearly
identical binding modes (colored as in panel A).
T.A. Bowden et al. / Journal of Structural Biology 175 (2011) 120–126 123

Fig. 3. Differential utilization of the GHephrinB2 loop by Eph receptors and Henipaviruses. (A) Crystal structure of EphB2 in complex with ephrinB2 (PDB ID 1KGY). (B) Crystal
structure of NiV-G in complex with ephrinB2 (PDB ID 2VSM). For both panels, structures are shown as cartoons with EphB2 and NiV-G colored in gray and ephrinB2 colored as
a rainbow with the N-terminus in blue and the C-terminus in red. The primary ephrinB2 interaction loop is highlighted with a thicker radius and the side-chains of residues
important for both protein-protein interactions are labeled and shown in a ball and stick representation.

of HNV-G alone and in complex with ephrinB2 and ephrinB3 show 4. Convergent viral attachment through transferrin receptor
that the GH loop is plastic and undergoes a unique rearrangement targeting
that allows it to bind to the top center portion of the HNV-G b-pro-
peller (Fig. 3B) (Bowden et al., 2008a; Xu et al., 2008). As the GH The transferrin receptor (TfR1) is a type-2 membrane glycopro-
loop is well conserved between many vertebrate species including tein which regulates the cellular uptake of iron through binding to
bats and humans, this observation provides a molecular level its ligand, transferrin (Tf) and is almost ubiquitously expressed in
rationale for Henipaviral zoonosis (Bowden et al., 2008a). The different human tissues (Ponka and Lok, 1999). Upon binding to
conformational changes observed in the ephrins, in addition to mono-ferric or di-ferric Tf, the TfR1-Tf complex is internalized
those occurring to the HNV-G b-propeller upon binding (Bowden through clathrin-dependent endocytosis and later is freed from
et al., 2010b, 2008a, 2008b; Xu et al., 2008), result in a protein- TfR1 in acidic compartments (Ponka and Lok, 1999). TfR1 exists
protein interface which is similarly tight (nanomolar affinity) but as a disulfide-linked dimer which consists of an N-terminal cyto-
more extensive than physiological Eph-ephrin interactions (an plasmic domain, a transmembrane region and a 650 amino acid
HNV-G-ephrinB2 interface of approximately 2700 Å2 compared to ectodomain. A major portion of the TfR1 ectodomain has been
an average of 2200 Å2 buried surface area for an Eph-ephrin com- crystallized and shown to consist of a protease-like domain, a heli-
plex). Despite differences in the extent of these interactions, the cal domain and an apical domain (Fig. 4A) (Bennett et al., 2000;
protein-protein interfaces in both sets of structures are dominated Lawrence et al., 1999). Structures of TfR1 in complex with Tf and
by hydrophobic contacts between aromatic sidechains of ephrinB2 HFE, a membrane glycoprotein associated with hereditary haemo-
and ephrinB3 (e.g. Phe120ephrinB2 and Trp 125ephrinB2) with binding chromatosis (Bomford, 2002; Lebron et al., 1999), have been eluci-
pockets on the physiological Eph and viral HNV-G glycoproteins. dated by cryo-electron microscopy (Cheng et al., 2004) and
Such binding surfaces are reminiscent to the hydrophobic contacts crystallography (Bennett et al., 2000), respectively. HFE can com-
observed in structures of CD4 in complex with MHC class II and pete with Tf for binding and both complex structures revealed a
HIV GP120 glycoproteins. The interfaces in both CD4 complexes 2:2 stoichiometry (Fig. 4B). In these structures, the Tf and HFE
are dominated by the insertion of Phe43CD4 into hydrophobic binding sites overlap at the membrane proximal TfR1 helical do-
cavities present on the MHC and HIV GP120 glycoproteins (Kwong main (Fig. 4B) and are extensive; the crystal structure of the
et al., 1998; Wang et al., 2001). TfR1–HFE complex revealed the occlusion of approximately
The structural properties observed in henipavirus-ephrin inter- 2000 Å2 of solvent accessible surface.
actions, in addition to those observed in the attachment glycopro- In addition to its importance in iron delivery into cells, TfR1 has
teins from other paramyxoviruses (e.g. Measles hemagglutinin in emerged as an entry receptor for a number of important pathogens
complex with cell surface SLAM (Hashiguchi et al., 2011) and including mouse mammary tumor virus (Ross et al., 2002), canine
CD46 (Santiago et al., 2010)), underscore the adaptability of the and panleukopenia feline parvoviruses (Parker et al., 2001), and
viral six-bladed b-propeller scaffold (Bowden et al., 2010a; Stehle New World hemorrhagic fever arenaviruses (Radoshitzky et al.,
and Casasnovas, 2009), and in a broader context, the innovative 2007). These viruses differ markedly in properties: canine and pan-
ability of viruses to alter their glycoprotein repertoire to target leukopenia feline parvoviruses are small (26 nm in diameter),
new receptors and hosts. icosohedral, single-stranded DNA viruses that do not contain a
124 T.A. Bowden et al. / Journal of Structural Biology 175 (2011) 120–126

Fig. 4. Contrasting modes of TfR1-host and-virus interactions. (A) Crystal structure of the unbound TfR1ectodomain (PDB ID 1CX8). One TfR1 protomer of the dimer is colored
with the helical domain in blue, the protease-like domain orange, and the apical domain green. (B) Crystal structure of hereditary haemochromatosis protein HFE in complex
with human TfR1 (PDB ID 1DE4). HFE molecules are rendered as gray surfaces and bind to helical TfR1 domains with a 2:2 stoichiometry. TfR1 is rotated by 90° along the
vertical axis with respect to panel A. (C) Crystal structure of Machupo virus attachment glycoprotein GP1 (MACV GP1) in complex with human TfR1 (PDB ID 3KAS). MACV GP1
molecules are rendered as gray surfaces and bind to apical TfR1 domains in a 2:2 stoichiometry. TfR1 is shown in the same orientation as in panel A.

lipid bilayer envelope, whilst mouse mammary tumor virus and In this review, recently elucidated virus-host crystal structures,
New World hemorrhagic fever arenaviruses are large, plieomor- many of which have emerged from the Spine2-Complexes project,
phic, enveloped viruses (100 and 120 nm in diameter, respec- have shown how viruses both appropriate existing cellular interac-
tively) which contain single-stranded RNA genomes. Structural tions (e.g. A39R binding to PlexinC1 and henipavirus attachment to
and functional studies of these viruses have shown that they attach ephrins) as well utilize novel modes for host-interaction (e.g. are-
to sites on TfR1 which do not overlap with the physiological Tf and naviral attachment to TfR1). These structural insights when drawn
HFE binding sites (Radoshitzky et al., 2007). The structure of the together reveal common molecular-level strategies which viruses
Machupo virus attachment glycoprotein, GP1, determined as a part have evolved to interact with their natural host and result in a dan-
of the Spine2-Complexes project, revealed a novel protein fold ger to human and animal health.
(Bowden et al., 2009a). A subsequent crystal structure of GP1 in
complex with TfR1 has revealed an extensive binding site occlud- Acknowledgments
ing over 1900 Å2 of solvent accessible surface at the tip of the
membrane distal TfR1 apical domain (Fig. 4C) (Abraham et al., We would like to thank Dr. Richard Elliott, Dr. Max Crispin, and
2010). Similarly, functional and electron microscopic data suggest Dr. Xiaoyun Ji for helpful discussions. David I. Stuart is an Medical
mouse mammary tumor virus and canine and feline parvoviruses Research Council Professor of Structural Biology, E. Yvonne Jones is
also utilize the TfR1 apical domain for attachment (Goodman a Cancer Research UK Principal Research Fellow, Thomas A. Bow-
et al., 2010; Hafenstein et al., 2007; Palermo et al., 2003; Wang den is a Sir Henry Wellcome Postdoctoral Fellow and Junior Re-
et al., 2006). search Fellow at University College, Oxford. This work was
It has been suggested that this TfR1 ‘viral binding patch’ is used funded by the Wellcome Trust [Grant Number 089026/Z/09/Z],
as it is remote from the known physiological Tf and HFE cellular Medical Research Council, Cancer Research UK, and Spine2-
binding sites and unlikely to disturb TfR1 endocytosis (Goodman Complexes [Grant Number FP6-RTD-031220].
et al., 2010). Given this hypothesis and the innate ability of viruses
to evolve rapidly, it is not surprising that these diverse viruses have References
independently evolved similar molecular mechanisms to rely on
the TfR1 cell surface receptor for virus entry. Abraham, J., Corbett, K.D., Farzan, M., Choe, H., Harrison, S.C., 2010. Structural basis
for receptor recognition by New World hemorrhagic fever arenaviruses. Nat.
Struct. Mol. Biol. 17, 438–444.
Aguilar, H.C., Ataman, Z.A., Aspericueta, V., Fang, A.Q., Stroud, M., Negrete, O.A.,
5. Concluding remarks Kammerer, R.A., Lee, B., 2009. A novel receptor-induced activation site in the
Nipah virus attachment glycoprotein (G) involved in triggering the fusion
The rapidity of sequence changes in viral genomes is fundamen- glycoprotein (F). J. Biol. Chem. 284, 1628–1635.
Antipenko, A., Himanen, J.P., van Leyen, K., Nardi-Dei, V., Lesniak, J., Barton, W.A.,
tal for the survival of many viruses. It enables co-evolution with Rajashankar, K.R., Lu, M., Hoemme, C., Puschel, A.W., Nikolov, D.B., 2003.
natural host reservoirs as well as opportunities to adapt to and in- Structure of the semaphorin-3A receptor binding module. Neuron 39, 589–598.
fect new hosts. Such genetic variability is thus extremely problem- Aricescu, A.R., Lu, W., Jones, E.Y., 2006. A time- and cost-efficient system for high-
level protein production in mammalian cells. Acta Crystallogr. D Biol.
atic from a biomedical perspective. For example, poxviruses such Crystallogr. 62, 1243–1250.
as smallpox virus have used these properties to ‘steal’ and optimize Avota, E., Gassert, E., Schneider-Schaulies, S., 2010. Measles virus-induced
host cell genes such as the plexinC1 interacting semaphorin, A39R, immunosuppression: from effectors to mechanisms. Med. Microbiol.
Immunol. 199, 227–237.
for antagonism of the host immune system. Emergent RNA viruses Bahar, M.W., Graham, S.C., Chen, R., Cooray, S., Smith, G.L., Stuart, D.I., Grimes,
such as henipa- and arena-viruses, on the other hand, have relied J.M., 2011. How vaccinia virus has evolved to subvert the host immune
on genetic diversity to develop very different attachment glycopro- response. J. Struct. Biol. 175, 127–134.
Bennett, M.J., Lebron, J.A., Bjorkman, P.J., 2000. Crystal structure of the hereditary
tein folds which can be used to bind to a variety of host cell surface
haemochromatosis protein HFE complexed with transferrin receptor. Nature
receptors. The methodological advances in eukaryotic cell expres- 403, 46–53.
sion for macromolecular crystallography developed in Spine Bomford, A., 2002. Genetics of haemochromatosis. Lancet 360, 1673–1681.
(Aricescu et al., 2006; Chang et al., 2007) have been invaluable Bonaparte, M.I., Dimitrov, A.S., Bossart, K.N., Crameri, G., Mungall, B.A., Bishop, K.A.,
Choudhry, V., Dimitrov, D.S., Wang, L.F., Eaton, B.T., Broder, C.C., 2005. Ephrin-
for understanding the molecular basis of these virus-host cell B2 ligand is a functional receptor for Hendra virus and Nipah virus. Proc. Natl.
interactions. Acad. Sci. USA 102, 10652–10657.
T.A. Bowden et al. / Journal of Structural Biology 175 (2011) 120–126 125

Bork, P., Doerks, T., Springer, T.A., Snel, B., 1999. Domains in plexins: links to Janssen, B.J., Robinson, R.A., Perez-Branguli, F., Bell, C.H., Mitchell, K.J., Siebold, C.,
integrins and transcription factors. Trends Biochem. Sci. 24, 261–263. Jones, E.Y., 2010. Structural basis of semaphorin-plexin signalling. Nature 467,
Bossart, K.N., Tachedjian, M., McEachern, J.A., Crameri, G., Zhu, Z., Dimitrov, D.S., 1118–1122.
Broder, C.C., Wang, L.F., 2008. Functional studies of host-specific ephrin-B Kadlec, J., Loureiro, S., Abrescia, N.G., Stuart, D.I., Jones, I.M., 2008. The postfusion
ligands as Henipavirus receptors. Virology 372, 357–371. structure of baculovirus gp64 supports a unified view of viral fusion machines.
Bowden, T.A., Crispin, M., Jones, E.Y., Stuart, D.I., 2010a. Shared paramyxoviral Nat. Struct. Mol. Biol. 15, 1024–1030.
glycoprotein architecture is adapted for diverse attachment strategies. Kleinerman, E.S., Lachman, L.B., Knowles, R.D., Snyderman, R., Cianciolo, G.J., 1987. A
Biochem. Soc. Trans. 38, 1349–1355. synthetic peptide homologous to the envelope proteins of retroviruses inhibits
Bowden, T.A., Crispin, M., Harvey, D.J., Jones, E.Y., Stuart, D.I., 2010b. Dimeric monocyte-mediated killing by inactivating interleukin 1. J. Immunol. 139,
architecture of the Hendra virus attachment glycoprotein: evidence for a 2329–2337.
conserved mode of assembly. J. Virol. 84, 6208–6217. Kruger, R.P., Aurandt, J., Guan, K.L., 2005. Semaphorins command cells to move. Nat.
Bowden, T.A., Aricescu, A.R., Gilbert, R.J., Grimes, J.M., Jones, E.Y., Stuart, D.I., 2008a. Rev. Mol. Cell Biol. 6, 789–800.
Structural basis of Nipah and Hendra virus attachment to their cell-surface Kwong, P.D., Wyatt, R., Robinson, J., Sweet, R.W., Sodroski, J., Hendrickson, W.A.,
receptor ephrin-B2. Nat. Struct. Mol. Biol. 15, 567–572. 1998. Structure of an HIV gp120 envelope glycoprotein in complex with the
Bowden, T.A., Crispin, M., Harvey, D.J., Aricescu, A.R., Grimes, J.M., Jones, E.Y., Stuart, CD4 receptor and a neutralizing human antibody. Nature 393, 648–659.
D.I., 2008b. Crystal structure and carbohydrate analysis of Nipah virus Lamb, R.A., Jardetzky, T.S., 2007. Structural basis of viral invasion: lessons from
attachment glycoprotein: a template for antiviral and vaccine design. J. Virol. paramyxovirus F. Curr. Opin. Struct. Biol. 17, 427–436.
82, 11628–11636. Lawrence, C.M., Ray, S., Babyonyshev, M., Galluser, R., Borhani, D.W., Harrison, S.C.,
Bowden, T.A., Crispin, M., Graham, S.C., Harvey, D.J., Grimes, J.M., Jones, E.Y., Stuart, 1999. Crystal structure of the ectodomain of human transferrin receptor.
D.I., 2009a. Unusual molecular architecture of the machupo virus attachment Science 286, 779–782.
glycoprotein. J. Virol. 83, 8259–8265. Lebron, J.A., West Jr., A.P., Bjorkman, P.J., 1999. The hemochromatosis protein HFE
Bowden, T.A., Aricescu, A.R., Nettleship, J.E., Siebold, C., Rahman-Huq, N., Owens, competes with transferrin for binding to the transferrin receptor. J. Mol. Biol.
R.J., Stuart, D.I., Jones, E.Y., 2009b. Structural plasticity of eph receptor A4 294, 239–245.
facilitates cross-class ephrin signaling. Structure 17, 1386–1397. Lee, J.E., Fusco, M.L., Hessell, A.J., Oswald, W.B., Burton, D.R., Saphire, E.O., 2008.
Chang, V.T., Crispin, M., Aricescu, A.R., Harvey, D.J., Nettleship, J.E., Fennelly, J.A., Yu, Structure of the Ebola virus glycoprotein bound to an antibody from a human
C., Boles, K.S., Evans, E.J., Stuart, D.I., Dwek, R.A., Jones, E.Y., Owens, R.J., Davis, survivor. Nature 454, 177–182.
S.J., 2007. Glycoprotein structural genomics: solving the glycosylation problem. Liu, H., Juo, Z.S., Shim, A.H., Focia, P.J., Chen, X., Garcia, K.C., He, X., 2010. Structural
Structure 15, 267–273. basis of semaphorin-plexin recognition and viral mimicry from Sema7A and
Cheng, Y., Zak, O., Aisen, P., Harrison, S.C., Walz, T., 2004. Structure of the human A39R complexes with PlexinC1. Cell 142, 749–761.
transferrin receptor-transferrin complex. Cell 116, 565–576. Lou, Z., Xu, Y., Xiang, K., Su, N., Qin, L., Li, X., Gao, G.F., Bartlam, M., Rao, Z., 2006.
Chrencik, J.E., Brooun, A., Kraus, M.L., Recht, M.I., Kolatkar, A.R., Han, G.W., Seifert, Crystal structures of Nipah and Hendra virus fusion core proteins. FEBS J. 273,
J.M., Widmer, H., Auer, M., Kuhn, P., 2006a. Structural and biophysical 4538–4547.
characterization of the EphB4-ephrinB2 protein–protein interaction and Love, C.A., Harlos, K., Mavaddat, N., Davis, S.J., Stuart, D.I., Jones, E.Y., Esnouf, R.M.,
receptor specificity. J. Biol. Chem. 281, 28185–28192. 2003. The ligand-binding face of the semaphorins revealed by the high-
Chrencik, J.E., Brooun, A., Recht, M.I., Kraus, M.L., Koolpe, M., Kolatkar, A.R., Bruce, resolution crystal structure of SEMA4D. Nat. Struct. Biol. 10, 843–848.
R.H., Martiny-Baron, G., Widmer, H., Pasquale, E.B., Kuhn, P., 2006b. Structure Negrete, O.A., Levroney, E.L., Aguilar, H.C., Bertolotti-Ciarlet, A., Nazarian, R., Tajyar,
and thermodynamic characterization of the EphB4-Ephrin-B2 antagonist S., Lee, B., 2005. EphrinB2 is the entry receptor for Nipah virus, an emergent
peptide complex reveals the determinants for receptor specificity. Structure deadly paramyxovirus. Nature 436, 401–405.
14, 321–330. Negrete, O.A., Wolf, M.C., Aguilar, H.C., Enterlein, S., Wang, W., Muhlberger, E., Su, S.V.,
Cianciolo, G.J., Copeland, T.D., Oroszlan, S., Snyderman, R., 1985. Inhibition of Bertolotti-Ciarlet, A., Flick, R., Lee, B., 2006. Two key residues in EphrinB3 are
lymphocyte proliferation by a synthetic peptide homologous to retroviral critical for its use as an alternative receptor for nipah virus. PLos Pathog. 2, e7.
envelope proteins. Science 230, 453–455. Nikolov, D., Li, C., Lackmann, M., Jeffrey, P., Himanen, J., 2007. Crystal structure of
Comeau, M.R., Johnson, R., DuBose, R.F., Petersen, M., Gearing, P., VandenBos, T., the human ephrin-A5 ectodomain. Protein Sci. 16, 996–1000.
Park, L., Farrah, T., Buller, R.M., Cohen, J.I., Strockbine, L.D., Rauch, C., Spriggs, Nikolov, D.B., Li, C., Barton, W.A., Himanen, J.P., 2005. Crystal structure of the
M.K., 1998. A poxvirus-encoded semaphorin induces cytokine production from ephrin-B1 ectodomain: implications for receptor recognition and signaling.
monocytes and binds to a novel cellular semaphorin receptor, VESPR. Immunity Biochemistry 44, 10947–10953.
8, 473–482. Nogi, T., Yasui, N., Mihara, E., Matsunaga, Y., Noda, M., Yamashita, N., Toyofuku, T.,
Eaton, B.T., Broder, C.C., Middleton, D., Wang, L.F., 2006. Hendra and Nipah viruses: Uchiyama, S., Goshima, Y., Kumanogoh, A., Takagi, J., 2010. Structural basis for
different and dangerous. Nat. Rev. Microbiol. 4, 23–35. semaphorin signalling through the plexin receptor. Nature 467, 1123–1127.
Edwards, R.A., Rohwer, F., 2005. Viral metagenomics. Nat. Rev. Microbiol. 3, 504– Palermo, L.M., Hueffer, K., Parrish, C.R., 2003. Residues in the apical domain of the
510. feline and canine transferrin receptors control host-specific binding and cell
Ensser, A., Fleckenstein, B., 1995. Alcelaphine herpesvirus type 1 has a semaphorin- infection of canine and feline parvoviruses. J. Virol. 77, 8915–8923.
like gene. J. Gen. Virol. 76 (Pt 4), 1063–1067. Parker, J.S., Murphy, W.J., Wang, D., O’Brien, S.J., Parrish, C.R., 2001. Canine and
Eschli, B., Quirin, K., Wepf, A., Weber, J., Zinkernagel, R., Hengartner, H., 2006. feline parvoviruses can use human or feline transferrin receptors to bind, enter,
Identification of an N-terminal trimeric coiled-coil core within arenavirus and infect cells. J. Virol. 75, 3896–3902.
glycoprotein 2 permits assignment to class I viral fusion proteins. J. Virol. 80, Pasquale, E.B., 2005. Eph receptor signalling casts a wide net on cell behaviour. Nat.
5897–5907. Rev. Mol. Cell Biol. 6, 462–475.
Goodman, L.B., Lyi, S.M., Johnson, N.C., Cifuente, J.O., Hafenstein, S.L., Parrish, C.R., Ponka, P., Lok, C.N., 1999. The transferrin receptor: role in health and disease. Int. J.
2010. Binding site on the transferrin receptor for the parvovirus capsid and Biochem. Cell Biol. 31, 1111–1137.
effects of altered affinity on cell uptake and infection. J. Virol. 84, 4969–4978. Qin, H., Noberini, R., Huan, X., Shi, J., Pasquale, E.B., Song, J., 2010. Structural
Hafenstein, S., Palermo, L.M., Kostyuchenko, V.A., Xiao, C., Morais, M.C., Nelson, C.D., characterization of the EphA4-Ephrin-B2 complex reveals new features
Bowman, V.D., Battisti, A.J., Chipman, P.R., Parrish, C.R., Rossmann, M.G., 2007. enabling Eph-ephrin binding promiscuity. J. Biol. Chem. 285, 644–654.
Asymmetric binding of transferrin receptor to parvovirus capsids. Proc. Natl. Radoshitzky, S.R., Abraham, J., Spiropoulou, C.F., Kuhn, J.H., Nguyen, D., Li, W., Nagel,
Acad. Sci. USA 104, 6585–6589. J., Schmidt, P.J., Nunberg, J.H., Andrews, N.C., Farzan, M., Choe, H., 2007.
Hafner, C., Schmitz, G., Meyer, S., Bataille, F., Hau, P., Langmann, T., Dietmaier, W., Transferrin receptor 1 is a cellular receptor for New World haemorrhagic fever
Landthaler, M., Vogt, T., 2004. Differential gene expression of Eph receptors and arenaviruses. Nature 446, 92–96.
ephrins in benign human tissues and cancers. Clin. Chem. 50, 490–499. Ross, S.R., Schofield, J.J., Farr, C.J., Bucan, M., 2002. Mouse transferrin receptor 1 is
Hashiguchi, T., Ose, T., Kubota, M., Maita, N., Kamishikiryo, J., Maenaka, K., Yanagi, the cell entry receptor for mouse mammary tumor virus. Proc. Natl. Acad. Sci.
Y., 2011. Structure of the measles virus hemagglutinin bound to its cellular USA 99, 12386–12390.
receptor SLAM. Nat. Struct. Mol. Biol. 18, 135–141. Santiago, C., Celma, M.L., Stehle, T., Casasnovas, J.M., 2010. Structure of the measles
Himanen, J.P., Rajashankar, K.R., Lackmann, M., Cowan, C.A., Henkemeyer, M., virus hemagglutinin bound to the CD46 receptor. Nat. Struct. Mol. Biol. 17, 124–
Nikolov, D.B., 2001. Crystal structure of an Eph receptor-ephrin complex. Nature 129.
414, 933–938. Seiradake, E., Harlos, K., Sutton, G., Aricescu, A.R., Jones, E.Y., 2010. An extracellular
Himanen, J.P., Goldgur, Y., Miao, H., Myshkin, E., Guo, H., Buck, M., Nguyen, M., steric seeding mechanism for Eph-ephrin signaling platform assembly. Nat.
Rajashankar, K.R., Wang, B., Nikolov, D.B., 2009. Ligand recognition by A-class Struct. Mol. Biol. 17, 398–402.
Eph receptors: crystal structures of the EphA2 ligand-binding domain and the Stehle, T., Casasnovas, J.M., 2009. Specificity switching in virus-receptor complexes.
EphA2/ephrin-A1 complex. EMBO Rep. 10, 722–728. Curr. Opin. Struct. Biol. 19, 181–188.
Himanen, J.P., Yermekbayeva, L., Janes, P.W., Walker, J.R., Xu, K., Atapattu, L., Suzuki, K., Kumanogoh, A., Kikutani, H., 2008. Semaphorins and their receptors in
Rajashankar, K.R., Mensinga, A., Lackmann, M., Nikolov, D.B., Dhe-Paganon, S., immune cell interactions. Nat. Immunol. 9, 17–23.
2010. Architecture of Eph receptor clusters. Proc. Natl. Acad. Sci. USA 107, Suzuki, K., Okuno, T., Yamamoto, M., Pasterkamp, R.J., Takegahara, N., Takamatsu, H.,
10860–10865. Kitao, T., Takagi, J., Rennert, P.D., Kolodkin, A.L., Kumanogoh, A., Kikutani, H.,
Himanen, J.P., Chumley, M.J., Lackmann, M., Li, C., Barton, W.A., Jeffrey, P.D., Vearing, 2007. Semaphorin 7A initiates T-cell-mediated inflammatory responses
C., Geleick, D., Feldheim, D.A., Boyd, A.W., Henkemeyer, M., Nikolov, D.B., 2004. through alpha1beta1 integrin. Nature 446, 680–684.
Repelling class discrimination: ephrin-A5 binds to and activates EphB2 receptor Toth, J., Cutforth, T., Gelinas, A.D., Bethoney, K.A., Bard, J., Harrison, C.J., 2001. Crystal
signaling. Nat. Neurosci. 7, 501–509. structure of an ephrin ectodomain. Dev. Cell 1, 83–92.
126 T.A. Bowden et al. / Journal of Structural Biology 175 (2011) 120–126

Volchkov, V.E., Volchkova, V.A., Muhlberger, E., Kolesnikova, L.V., Weik, M., Dolnik, Wilson, I.A., Skehel, J.J., Wiley, D.C., 1981. Structure of the haemagglutinin membrane
O., Klenk, H.D., 2001. Recovery of infectious Ebola virus from complementary glycoprotein of influenza virus at 3 A resolution. Nature 289, 366–373.
DNA: RNA editing of the GP gene and viral cytotoxicity. Science 291, 1965– Wyatt, R., Kwong, P.D., Desjardins, E., Sweet, R.W., Robinson, J., Hendrickson, W.A.,
1969. Sodroski, J.G., 1998. The antigenic structure of the HIV gp120 envelope
Wang, E., Albritton, L., Ross, S.R., 2006. Identification of the segments of the mouse glycoprotein. Nature 393, 705–711.
transferrin receptor 1 required for mouse mammary tumor virus infection. J. Xu, K., Rajashankar, K.R., Chan, Y.P., Himanen, J.P., Broder, C.C., Nikolov, D.B., 2008.
Biol. Chem. 281, 10243–10249. Host cell recognition by the henipaviruses: crystal structures of the Nipah G
Wang, J.H., Meijers, R., Xiong, Y., Liu, J.H., Sakihama, T., Zhang, R., Joachimiak, A., attachment glycoprotein and its complex with ephrin-B3. Proc. Natl. Acad. Sci.
Reinherz, E.L., 2001. Crystal structure of the human CD4 N-terminal two- USA 105, 9953–9958.
domain fragment complexed to a class II MHC molecule. Proc. Natl. Acad. Sci. Yang, Z.Y., Duckers, H.J., Sullivan, N.J., Sanchez, A., Nabel, E.G., Nabel, G.J., 2000.
USA 98, 10799–10804. Identification of the Ebola virus glycoprotein as the main viral determinant of
Weis, W., Brown, J.H., Cusack, S., Paulson, J.C., Skehel, J.J., Wiley, D.C., 1988. Structure vascular cell cytotoxicity and injury. Nat. Med.‘ 6, 886–889.
of the influenza virus haemagglutinin complexed with its receptor, sialic acid. Zhu, P., Chertova, E., Bess Jr., J., Lifson, J.D., Arthur, L.O., Liu, J., Taylor, K.A., Roux, K.H.,
Nature 333, 426–431. 2003. Electron tomography analysis of envelope glycoprotein trimers on HIV
Wild, T.F., 2009. Henipaviruses: a new family of emerging Paramyxoviruses. Pathol. and simian immunodeficiency virus virions. Proc. Natl. Acad. Sci. USA 100,
Biol. (Paris) 57, 188–196. 15812–15817.

You might also like