Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
J Virol ; 98(3): e0183823, 2024 Mar 19.
Article in English | MEDLINE | ID: mdl-38426726

ABSTRACT

Nipah virus (NiV) is a highly lethal, zoonotic Henipavirus (HNV) that causes respiratory and neurological signs and symptoms in humans. Similar to other paramyxoviruses, HNVs mediate entry into host cells through the concerted actions of two surface glycoproteins: a receptor-binding protein (RBP) that mediates attachment and a fusion glycoprotein (F) that triggers fusion in an RBP-dependent manner. NiV uses ephrin-B2 (EFNB2) and ephrin-B3 (EFNB3) as entry receptors. Ghana virus (GhV), a novel HNV identified in a Ghanaian bat, uses EFNB2 but not EFNB3. In this study, we employ a structure-informed approach to identify receptor-interfacing residues and systematically introduce GhV-RBP residues into a NiV-RBP backbone to uncover the molecular determinants of EFNB3 usage. We reveal two regions that severely impair EFNB3 binding by NiV-RBP and EFNB3-mediated entry by NiV pseudotyped viral particles. Further analyses uncovered two-point mutations (NiVN557SGhV and NiVY581TGhV) pivotal for this phenotype. Moreover, we identify NiV interaction with Y120 of EFNB3 as important for the usage of this receptor. Beyond these EFNB3-related findings, we reveal two domains that restrict GhV binding of EFNB2, confirm the HNV-head as an immunodominant target for polyclonal and monoclonal antibodies, and describe putative epitopes for GhV- and NiV-specific monoclonal antibodies. Cumulatively, the work presented here generates useful reagents and tools that shed insight to residues important for NiV usage of EFNB3, reveal regions critical for GhV binding of EFNB2, and describe putative HNV antibody-binding epitopes. IMPORTANCE: Hendra virus and Nipah virus (NiV) are lethal, zoonotic Henipaviruses (HNVs) that cause respiratory and neurological clinical features in humans. Since their initial outbreaks in the 1990s, several novel HNVs have been discovered worldwide, including Ghana virus. Additionally, there is serological evidence of zoonotic transmission, lending way to concerns about future outbreaks. HNV infection of cells is mediated by the receptor-binding protein (RBP) and the Fusion protein (F). The work presented here identifies NiV RBP amino acids important for the usage of ephrin-B3 (EFNB3), a receptor highly expressed in neurons and predicted to be important for neurological clinical features caused by NiV. This study also characterizes epitopes recognized by antibodies against divergent HNV RBPs. Together, this sheds insight to amino acids critical for HNV receptor usage and antibody binding, which is valuable for future studies investigating determinants of viral pathogenesis and developing antibody therapies.


Subject(s)
Henipavirus Infections , Henipavirus , Receptors, Virus , Humans , Amino Acids/genetics , Antibodies, Monoclonal/metabolism , Carrier Proteins/metabolism , Ephrin-B3/genetics , Ephrin-B3/chemistry , Ephrin-B3/metabolism , Epitopes/genetics , Epitopes/metabolism , Ghana , Hendra Virus/metabolism , Henipavirus/classification , Henipavirus/genetics , Henipavirus/metabolism , Mutagenesis , Nipah Virus/metabolism , Viral Envelope Proteins/genetics , Virus Internalization , Receptors, Virus/metabolism
2.
Infect Genet Evol ; 116: 105516, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37924857

ABSTRACT

Nipah virus (NiV) is a lethal bat-borne zoonotic virus that causes mild to acute respiratory distress and neurological manifestations in humans with a high mortality rate. NiV transmission to humans occurs via consumption of bat-contaminated fruit and date palm sap (DPS), or through direct contact with infected individuals and livestock. Since NiV outbreaks were first reported in pigs from Malaysia and Singapore, non-neutralizing antibodies against NiV attachment Glycoprotein (G) have also been detected in a few domestic mammals. NiV infection is initiated after NiV G binds to the host cell receptors Ephrin-B2 and Ephrin-B3. In this study, we assessed the degree of NiV host tropism in domestic and peridomestic mammals commonly found in Bangladesh that may be crucial in the transmission of NiV by serving as intermediate hosts. We carried out a protein-protein docking analysis of NiV G complexes (n = 52) with Ephrin-B2 and B3 of 13 domestic and peridomestic species using bioinformatics tools. Protein models were generated by homology modelling and the structures were validated for model quality. The different protein-protein complexes in this study were stable, and their binding affinity (ΔG) scores ranged between -8.0 to -19.1 kcal/mol. NiV Bangladesh (NiV-B) strain displayed stronger binding to Ephrin receptors, especially with Ephrin-B3 than the NiV Malaysia (NiV-M) strain, correlating with the observed higher pathogenicity of NiV-B strains. From the docking result, we found that Ephrin receptors of domestic rat (R. norvegicus) had a higher binding affinity for NiV G, suggesting greater susceptibility to NiV infections compared to other study species. Investigations for NiV exposure to domestic/peridomestic animals will help us knowing more the possible role of rats and other animals as intermediate hosts of NiV and would improve future NiV outbreak control and prevention in humans and domestic animals.


Subject(s)
Chiroptera , Henipavirus Infections , Nipah Virus , Animals , Rats , Ephrin-B2/genetics , Ephrin-B2/chemistry , Ephrin-B2/metabolism , Ephrin-B3/chemistry , Ephrin-B3/metabolism , Glycoproteins/metabolism , Receptors, Cell Surface/metabolism , Receptors, Eph Family/metabolism , Swine , Virus Attachment
3.
J Orthop Surg Res ; 16(1): 499, 2021 Aug 14.
Article in English | MEDLINE | ID: mdl-34389038

ABSTRACT

BACKGROUND: Heterotopic ossification (HO) represents pathological lesions that refer to the development of heterotopic bone in extraskeletal tissues around joints. This study investigates the genetic characteristics of bone marrow mesenchymal stem cells (BMSCs) from HO tissues and explores the potential pathways involved in this ailment. METHODS: Gene expression profiles (GSE94683) were obtained from the Gene Expression Omnibus (GEO), including 9 normal specimens and 7 HO specimens, and differentially expressed genes (DEGs) were identified. Then, protein-protein interaction (PPI) networks and Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses were performed for further analysis. RESULTS: In total, 275 DEGs were differentially expressed, of which 153 were upregulated and 122 were downregulated. In the biological process (BP) category, the majority of DEGs, including EFNB3, UNC5C, TMEFF2, PTH2, KIT, FGF13, and WISP3, were intensively enriched in aspects of cell signal transmission, including axon guidance, negative regulation of cell migration, peptidyl-tyrosine phosphorylation, and cell-cell signaling. Moreover, KEGG analysis indicated that the majority of DEGs, including EFNB3, UNC5C, FGF13, MAPK10, DDIT3, KIT, COL4A4, and DKK2, were primarily involved in the mitogen-activated protein kinase (MAPK) signaling pathway, Ras signaling pathway, phosphatidylinositol-3-kinase/protein kinase B (PI3K/Akt) signaling pathway, and Wnt signaling pathway. Ten hub genes were identified, including CX3CL1, CXCL1, ADAMTS3, ADAMTS16, ADAMTSL2, ADAMTSL3, ADAMTSL5, PENK, GPR18, and CALB2. CONCLUSIONS: This study presented novel insight into the pathogenesis of HO. Ten hub genes and most of the DEGs intensively involved in enrichment analyses may be new candidate targets for the prevention and treatment of HO in the future.


Subject(s)
ADAMTS Proteins/genetics , Ephrin-B3/genetics , Extracellular Matrix Proteins/genetics , Membrane Proteins/genetics , Neoplasm Proteins/genetics , Ossification, Heterotopic , Phosphatidylinositol 3-Kinases/genetics , Transcriptome , ADAMTS Proteins/chemistry , Computational Biology , Ephrin-B3/chemistry , Extracellular Matrix Proteins/chemistry , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Gene Regulatory Networks , Humans , Membrane Proteins/chemistry , Neoplasm Proteins/chemistry , Ossification, Heterotopic/genetics , Phosphatidylinositol 3-Kinases/chemistry , Phosphatidylinositol 3-Kinases/metabolism , Protein Interaction Maps
4.
Proc Natl Acad Sci U S A ; 112(17): E2156-65, 2015 Apr 28.
Article in English | MEDLINE | ID: mdl-25825759

ABSTRACT

The discovery of African henipaviruses (HNVs) related to pathogenic Hendra virus (HeV) and Nipah virus (NiV) from Southeast Asia and Australia presents an open-ended health risk. Cell receptor use by emerging African HNVs at the stage of host-cell entry is a key parameter when considering the potential for spillover and infection of human populations. The attachment glycoprotein from a Ghanaian bat isolate (GhV-G) exhibits <30% sequence identity with Asiatic NiV-G/HeV-G. Here, through functional and structural analysis of GhV-G, we show how this African HNV targets the same human cell-surface receptor (ephrinB2) as the Asiatic HNVs. We first characterized this virus-receptor interaction crystallographically. Compared with extant HNV-G-ephrinB2 structures, there was significant structural variation in the six-bladed ß-propeller scaffold of the GhV-G receptor-binding domain, but not the Greek key fold of the bound ephrinB2. Analysis revealed a surprisingly conserved mode of ephrinB2 interaction that reflects an ongoing evolutionary constraint among geographically distal and phylogenetically divergent HNVs to maintain the functionality of ephrinB2 recognition during virus-host entry. Interestingly, unlike NiV-G/HeV-G, we could not detect binding of GhV-G to ephrinB3. Comparative structure-function analysis further revealed several distinguishing features of HNV-G function: a secondary ephrinB2 interaction site that contributes to more efficient ephrinB2-mediated entry in NiV-G relative to GhV-G and cognate residues at the very C terminus of GhV-G (absent in Asiatic HNV-Gs) that are vital for efficient receptor-induced fusion, but not receptor binding per se. These data provide molecular-level details for evaluating the likelihood of African HNVs to spill over into human populations.


Subject(s)
Ephrin-B2 , Henipavirus Infections/metabolism , Henipavirus , Viral Proteins , Virus Internalization , Ephrin-B2/chemistry , Ephrin-B2/genetics , Ephrin-B2/metabolism , Ephrin-B3/chemistry , Ephrin-B3/genetics , Ephrin-B3/metabolism , HEK293 Cells , Henipavirus/chemistry , Henipavirus/physiology , Henipavirus Infections/genetics , Humans , Protein Structure, Quaternary , Protein Structure, Secondary , Protein Structure, Tertiary , Structure-Activity Relationship , Viral Proteins/chemistry , Viral Proteins/genetics , Viral Proteins/metabolism
5.
Nucleic Acids Res ; 42(Web Server issue): W227-33, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24848013

ABSTRACT

Structure validation has become a major issue in the structural biology community, and an essential step is checking the ligand structure. This paper introduces MotiveValidator, a web-based application for the validation of ligands and residues in PDB or PDBx/mmCIF format files provided by the user. Specifically, MotiveValidator is able to evaluate in a straightforward manner whether the ligand or residue being studied has a correct annotation (3-letter code), i.e. if it has the same topology and stereochemistry as the model ligand or residue with this annotation. If not, MotiveValidator explicitly describes the differences. MotiveValidator offers a user-friendly, interactive and platform-independent environment for validating structures obtained by any type of experiment. The results of the validation are presented in both tabular and graphical form, facilitating their interpretation. MotiveValidator can process thousands of ligands or residues in a single validation run that takes no more than a few minutes. MotiveValidator can be used for testing single structures, or the analysis of large sets of ligands or fragments prepared for binding site analysis, docking or virtual screening. MotiveValidator is freely available via the Internet at http://ncbr.muni.cz/MotiveValidator.


Subject(s)
Macromolecular Substances/chemistry , Software , Acetylglucosamine/chemistry , Binding Sites , Cholic Acid/chemistry , Ephrin-B3/chemistry , Glycoproteins/chemistry , Internet , Ligands , Proteins/chemistry
6.
Proteins ; 82(12): 3241-54, 2014 Dec.
Article in English | MEDLINE | ID: mdl-24615845

ABSTRACT

The fusion of Nipah with host cells is facilitated by two of their glycoproteins, the G and the F proteins. The binding of cellular ephrins to the G head domain causes the G stalk domain to interact differently with F, which activates F to mediate virus-host fusion. To gain insight into how the ephrin-binding signal transduces from the head to the stalk domain of G, we examine quantitatively the differences between the conformational ensembles of the G head domain in its ephrin-bound and unbound states. We consider the human ephrins B2 and B3, and a double mutant of B2, all of which trigger fusion. The ensembles are generated using molecular dynamics, and the differences between them are quantified using a new machine learning method. We find that the portion of the G head domain whose conformational density is altered equivalently by the three ephrins is large, and comprises ∼25% of the residues in the G head domain. This subspace also includes the residues that are known to be important to F activation, which suggests that it contains at least one common signaling pathway. The spatial distribution of the residues constituting this subspace supports the model of signal transduction in which the signal transduces via the G head dimer interface. This study also adds to the growing list of examples where signaling does not depend solely on backbone deviations. In general, this study provides an approach to filter out conserved patterns in protein dynamics.


Subject(s)
Ephrin-B2/metabolism , Ephrin-B3/metabolism , Models, Biological , Nipah Virus/physiology , Signal Transduction , Viral Envelope Proteins/metabolism , Allosteric Regulation , Amino Acid Substitution , Artificial Intelligence , Databases, Protein , Ephrin-B2/chemistry , Ephrin-B2/genetics , Ephrin-B3/chemistry , Host-Pathogen Interactions , Humans , Ligands , Molecular Dynamics Simulation , Mutation , Protein Conformation , Protein Interaction Domains and Motifs , Support Vector Machine , Viral Envelope Proteins/agonists , Viral Envelope Proteins/chemistry , Virus Activation , Virus Attachment , Virus Integration
7.
Curr Top Microbiol Immunol ; 359: 59-78, 2012.
Article in English | MEDLINE | ID: mdl-22695915

ABSTRACT

Nipah (NiV) and Hendra (HeV) viruses are the deadliest human pathogens within the Paramyxoviridae family, which include human and animal pathogens of global biomedical importance. NiV and HeV infections cause respiratory and encephalitic illness with high mortality rates in humans. Henipaviruses (HNV) are the only Paramyxoviruses classified as biosafety level 4 (BSL4) pathogens due to their extreme pathogenicity, potential for bioterrorism, and lack of licensed vaccines and therapeutics. HNV use ephrin-B2 and ephrin-B3, highly conserved proteins, as viral entry receptors. This likely accounts for their unusually broad species tropism, and also provides opportunities to study how receptor usage, cellular tropism, and end-organ pathology relates to the pathobiology of HNV infections. The clinical and pathologic manifestations of NiV and HeV virus infections are reviewed in the chapters by Wong et al. and Geisbert et al. in this issue. Here, we will review the biology of the HNV receptors, and how receptor usage relates to HNV cell tropism in vitro and in vivo.


Subject(s)
Ephrin-B2/metabolism , Ephrin-B3/metabolism , Hendra Virus/physiology , Nipah Virus/physiology , Receptors, Virus/metabolism , Viral Fusion Proteins/metabolism , Viral Tropism , Animals , Blood Vessels/pathology , Blood Vessels/virology , Brain/pathology , Brain/virology , Endothelial Cells/pathology , Endothelial Cells/virology , Ephrin-B2/chemistry , Ephrin-B3/chemistry , Hendra Virus/pathogenicity , Henipavirus Infections/pathology , Henipavirus Infections/virology , Humans , Models, Molecular , Nipah Virus/pathogenicity , Receptors, Virus/chemistry , Viral Fusion Proteins/chemistry , Virus Internalization
8.
Curr Top Microbiol Immunol ; 359: 79-94, 2012.
Article in English | MEDLINE | ID: mdl-22427111

ABSTRACT

Nipah (NiV) and Hendra (HeV) viruses cause cell-cell fusion (syncytia) in brain, lung, heart, and kidney tissues, leading to encephalitis, pneumonia, and often death. Membrane fusion is essential to both viral entry and virus-induced cell-cell fusion, a hallmark of henipavirus infections. Elucidiation of the mechanism(s) of membrane fusion is critical to understanding henipavirus pathobiology and has the potential to identify novel strategies for the development of antiviral therapeutic agents. Henipavirus membrane fusion requires the coordinated actions of the viral attachment (G) and fusion (F) glycoproteins. Current henipavirus fusion models posit that attachment of NiV or HeV G to its cell surface receptors releases F from its metastable pre-fusion conformation to mediate membrane fusion. The identification of ephrinB2 and ephrinB3 as henipavirus receptors has paved the way for recent advances in our understanding of henipavirus membrane fusion. These advances highlight mechanistic similarities and differences between membrane fusion for the henipavirus and other genera within the Paramyxoviridae family. Here, we review these mechanisms and the current gaps in our knowledge in the field.


Subject(s)
Ephrin-B2/metabolism , Ephrin-B3/metabolism , Hendra Virus/physiology , Nipah Virus/physiology , Receptors, Virus/metabolism , Viral Fusion Proteins/metabolism , Animals , Brain/pathology , Brain/virology , Ephrin-B2/chemistry , Ephrin-B3/chemistry , Hendra Virus/pathogenicity , Henipavirus Infections/pathology , Henipavirus Infections/virology , Humans , Kidney/pathology , Kidney/virology , Lung/pathology , Lung/virology , Membrane Fusion , Nipah Virus/pathogenicity , Receptors, Virus/chemistry , Viral Fusion Proteins/chemistry , Virus Internalization
9.
Semin Cell Dev Biol ; 23(1): 116-23, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22227101

ABSTRACT

Members of the ephrin cell-surface protein family interact with the Eph receptors, the largest family of receptor tyrosine kinases, mediating bi-directional signaling during tumorogenesis and various developmental events. Surprisingly, ephrin-B2 and -B3 were recently identified as entry receptors for henipaviruses, emerging zoonotic paramyxoviruses responsible for repeated outbreaks in humans and animals in Australia, Southeast Asia, India and Bangladesh. Nipah virus (NiV) and Hendra virus (HeV) are the only two identified members in the henipavirus genus. While the initial human infection cases came from contact with infected pigs (NiV) or horses (HeV), in the more recent outbreaks of NiV both food-borne and human-to-human transmission were reported. These characteristics, together with high mortality and morbidity rates and lack of effective anti-viral therapies, make the henipaviruses a potential biological-agent threat. Viral entry is an important target for the development of anti-viral drugs. The entry of henipavirus is initiated by the attachment of the viral G envelope glycoprotein to the host cell receptors ephrin-B2 and/or -B3, followed by activation of the F fusion protein, which triggers fusion between the viral envelop and the host membrane. We review recent progress in the study of henipavirus entry, particularly the identification of ephrins as their entry receptors, and the structural characterization of the ephrin/Henipa-G interactions.


Subject(s)
Ephrin-B2/metabolism , Ephrin-B3/metabolism , Henipavirus/physiology , Receptors, Virus/metabolism , Virus Attachment , Animals , Antibodies, Neutralizing/chemistry , Ephrin-B2/chemistry , Ephrin-B3/chemistry , Henipavirus/pathogenicity , Host-Pathogen Interactions , Humans , Models, Molecular , Protein Binding , Protein Conformation , Signal Transduction , Viral Proteins/chemistry , Viral Proteins/metabolism
10.
ACS Nano ; 5(7): 5444-56, 2011 Jul 26.
Article in English | MEDLINE | ID: mdl-21702475

ABSTRACT

Controlling and understanding the changes in metastatic cancer cell adhesion, shape, and motility are of paramount importance in cancer research, diagnosis, and treatment. Here, we used gold nanoparticles (AuNPs) as nanotopological structures and protein nanocluster forming substrates. Cell adhesion controlling proteins [in this case, fibronection (Fn) and ephrinB3] were modified to AuNPs, and these particles were then modified to the layer-by-layer (LbL) polymer surface that offers a handle for tuning surface charge and mechanical property of a cell-interfacing substrate. We found that metastatic cancer cell adhesion is affected by nanoparticle density on a surface, and ∼140 particles per 400 µm(2) (∼1.7 µm spacing between AuNPs) is optimal for effective metastatic cell adhesion. It was also shown that the AuNP surface density and protein nanoclustering on a spherical AuNP are controlling factors for the efficient interfacing and signaling of metastatic cancer cells. Importantly, the existence of nanotopological features (AuNPs in this case) is much more critical in inducing more dramatic changes in metastatic cell adhesion, protrusion, polarity, and motility than the presence of a cell adhesion protein, Fn, on the surface. Moreover, cell focal adhesion and motility-related paxillin clusters were heavily formed in cell lamellipodia and filopodia and high expression of phospho-paxillins were observed when the cells were cultured on either an AuNP or Fn-modified AuNP polymer surface. The ephrin signaling that results in the decreased expression of paxillin was found to be more effective when ephrins were modified to the AuNP surface than when ephrinB3 was directly attached to the polymer film. The overall trend for cell motility change is such that a nanoparticle-modified LbL surface induces higher cell motility and the AuNP modification to the LbL surface results in more pronounced change in cell motility than Fn or ephrin modification to the LbL surface.


Subject(s)
Breast Neoplasms/pathology , Cell Movement/drug effects , Cell Shape/drug effects , Gold/chemistry , Metal Nanoparticles/chemistry , Nanotechnology/methods , Polymers/pharmacology , Cell Adhesion/drug effects , Cell Line, Tumor , Ephrin-B3/chemistry , Ephrin-B3/metabolism , Fibronectins/chemistry , Fibronectins/metabolism , Humans , Neoplasm Metastasis , Polymers/chemistry
11.
Proc Natl Acad Sci U S A ; 105(29): 9953-8, 2008 Jul 22.
Article in English | MEDLINE | ID: mdl-18632560

ABSTRACT

Nipah virus (NiV) and Hendra virus are the type species of the highly pathogenic paramyxovirus genus Henipavirus, which can cause severe respiratory disease and fatal encephalitis infections in humans, with case fatality rates approaching 75%. NiV contains two envelope glycoproteins, the receptor-binding G glycoprotein (NiV-G) that facilitates attachment to host cells and the fusion (F) glycoprotein that mediates membrane merger. The henipavirus G glycoproteins lack both hemagglutinating and neuraminidase activities and, instead, engage the highly conserved ephrin-B2 and ephrin-B3 cell surface proteins as their entry receptors. Here, we report the crystal structures of the NiV-G both in its receptor-unbound state and in complex with ephrin-B3, providing, to our knowledge, the first view of a paramyxovirus attachment complex in which a cellular protein is used as the virus receptor. Complex formation generates an extensive protein-protein interface around a protruding ephrin loop, which is inserted in the central cavity of the NiV-G beta-propeller. Analysis of the structural data reveals the molecular basis for the highly specific interactions of the henipavirus G glycoproteins with only two members (ephrin-B2 and ephrin-B3) of the very large ephrin family and suggests how they mediate in a unique fashion both cell attachment and the initiation of membrane fusion during the virus infection processes. The structures further suggest that the NiV-G/ephrin interactions can be effectively targeted to disrupt viral entry and provide the foundation for structure-based antiviral drug design.


Subject(s)
Ephrin-B3/chemistry , Nipah Virus/chemistry , Viral Envelope Proteins/chemistry , Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Binding Sites , Crystallography, X-Ray , Drug Design , Host-Pathogen Interactions , Humans , Ligands , Membrane Fusion/physiology , Models, Molecular , Multiprotein Complexes , Nipah Virus/pathogenicity , Nipah Virus/physiology , Protein Conformation , Receptors, Virus/chemistry , Viral Envelope Proteins/physiology , Virulence/physiology
12.
Virology ; 372(2): 357-71, 2008 Mar 15.
Article in English | MEDLINE | ID: mdl-18054977

ABSTRACT

Hendra virus (HeV) and Nipah virus (NiV) are closely related paramyxoviruses that infect and cause disease in a wide range of mammalian hosts. To determine whether host receptor molecules play a role in species-specific and/or virus-specific infection we have cloned and characterized ephrin-B2 and ephrin-B3 ligands from a range of species, including human, horse, pig, cat, dog, bats (Pteropus alecto and Pteropus vampyrus) and mouse. HeV and NiV were both able to infect cells expressing any of the ephrin-B2 and ephrin-B3 molecules. There did not appear to be significant differences in receptor function from different species or receptor usage by HeV and NiV. Soluble ephrin ligands, their receptors and G-specific human monoclonal antibodies differentially blocked henipavirus infections suggesting different receptor affinities, overlapping receptor binding domains of the henipavirus attachment glycoprotein (G) and that the functional domains of the ephrin ligands may be important for henipavirus binding.


Subject(s)
Ephrin-B2/metabolism , Ephrin-B3/metabolism , Henipavirus/metabolism , Receptors, Virus/metabolism , Amino Acid Sequence , Animals , Antibodies, Viral , Binding Sites , Cell Line , Cells, Cultured , Cloning, Molecular , DNA , Ephrin-B2/chemistry , Ephrin-B2/genetics , Ephrin-B3/chemistry , Ephrin-B3/genetics , Humans , Ligands , Molecular Sequence Data , Species Specificity
13.
PLoS Pathog ; 2(2): e7, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16477309

ABSTRACT

EphrinB2 was recently discovered as a functional receptor for Nipah virus (NiV), a lethal emerging paramyxovirus. Ephrins constitute a class of homologous ligands for the Eph class of receptor tyrosine kinases and exhibit overlapping expression patterns. Thus, we examined whether other ephrins might serve as alternative receptors for NiV. Here, we show that of all known ephrins (ephrinA1-A5 and ephrinB1-B3), only the soluble Fc-fusion proteins of ephrinB3, in addition to ephrinB2, bound to soluble NiV attachment protein G (NiV-G). Soluble NiV-G bound to cell surface ephrinB3 and B2 with subnanomolar affinities (Kd = 0.58 nM and 0.06 nM for ephrinB3 and B2, respectively). Surface plasmon resonance analysis indicated that the relatively lower affinity of NiV-G for ephrinB3 was largely due to a faster off-rate (K(off) = 1.94 x 10(-3) s(-1) versus 1.06 x 10(-4) s(-1) for ephrinB3 and B2, respectively). EphrinB3 was sufficient to allow for viral entry of both pseudotype and live NiV. Soluble ephrinB2 and B3 were able to compete for NiV-envelope-mediated viral entry on both ephrinB2- and B3-expressing cells, suggesting that NiV-G interacts with both ephrinB2 and B3 via an overlapping site. Mutational analysis indicated that the Leu-Trp residues in the solvent exposed G-H loop of ephrinB2 and B3 were critical determinants of NiV binding and entry. Indeed, replacement of the Tyr-Met residues in the homologous positions in ephrinB1 with Leu-Trp conferred NiV receptor activity to ephrinB1. Thus, ephrinB3 is a bona fide alternate receptor for NiV entry, and two residues in the G-H loop of the ephrin B-class ligands are critical determinants of NiV receptor activity.


Subject(s)
Ephrin-B3/chemistry , Ephrin-B3/metabolism , Nipah Virus/metabolism , Receptors, Virus/metabolism , Animals , Binding Sites , CHO Cells , Cricetinae , Cricetulus , Ephrin-B2/genetics , Ephrin-B2/metabolism , Ephrin-B3/genetics , Henipavirus Infections/virology , Humans , Leucine , Nipah Virus/physiology , Tryptophan
14.
Assay Drug Dev Technol ; 1(4): 555-63, 2003 Aug.
Article in English | MEDLINE | ID: mdl-15090252

ABSTRACT

We have characterized a recombinantly expressed N-terminally tagged GST fusion of the tyrosine kinase domain of human EphB3. The EphB3 kinase domain was shown to phosphorylate a group of synthetic tyrosine-containing peptides derived from a proprietary biotinylated kinase-biased peptide substrate library. In addition, the enzyme activity was stimulated by the divalent cation, manganese, and inhibited by addition of magnesium. The most active tyrosine-containing peptide, a biotinylated 49-mer, displayed saturation kinetics with an apparent K(m) of approximately 0.4 microM. The apparent K(m) for ATP was determined to be approximately 3 microM. The kinetics of the reaction was linear from concentrations of enzyme of 0.5 to 2 nM, and at or below the K(m) concentrations of the two substrates for at least 2 h at room temperature. Moreover, the tryrosine kinase inhibitor, PP2, produced an IC(50) of roughly 0.8 microM. In addition, the enzyme tolerated the solvent DMSO and was stable to multiple freeze/thaw cycles. Stability of the enzyme at 4 degrees C storage was seen out to 6 h with an approximately 50% reduction of activity by 24 h. Formatting the assay in a 384-well microtiter plate produced good uniformity of signal at 100% inhibition, 50% inhibition, and no inhibition. The coefficient of variance was at or below 10% with a signal-to-background ratio of approximately 24 and a z value of 0.72. Collectively, these results showed the ability to configure a robust HTS for a truncated recombinantly expressed family member of the Ephrin tyrosine kinases.


Subject(s)
Protein-Tyrosine Kinases/chemistry , Receptor, EphB3/chemistry , Scintillation Counting/methods , Ephrin-B3/chemistry , Humans , Receptor, EphB3/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...