Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
J Virol ; 98(6): e0050324, 2024 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-38780245

RESUMEN

The henipaviruses, including Nipah virus (NiV) and Hendra virus (HeV), are biosafety level 4 (BSL-4) zoonotic pathogens that cause severe neurological and respiratory disease in humans. To study the replication machinery of these viruses, we developed robust minigenome systems that can be safely used in BSL-2 conditions. The nucleocapsid (N), phosphoprotein (P), and large protein (L) of henipaviruses are critical elements of their replication machinery and thus essential support components of the minigenome systems. Here, we tested the effects of diverse combinations of the replication support proteins on the replication capacity of the NiV and HeV minigenomes by exchanging the helper plasmids coding for these proteins among the two viruses. We demonstrate that all combinations including one or more heterologous proteins were capable of replicating both the NiV and HeV minigenomes. Sequence alignment showed identities of 92% for the N protein, 67% for P, and 87% for L. Notably, variations in amino acid residues were not concentrated in the N-P and P-L interacting regions implying that dissimilarities in amino acid composition among NiV and HeV polymerase complex proteins may not impact their interactions. The observed indiscriminate activity of NiV and HeV polymerase complex proteins is different from related viruses, which can support the replication of heterologous genomes only when the whole polymerase complex belongs to the same virus. This newly observed promiscuous property of the henipavirus polymerase complex proteins likely attributed to their conserved interaction regions could potentially be harnessed to develop universal anti-henipavirus antivirals.IMPORTANCEGiven the severity of disease induced by Hendra and Nipah viruses in humans and the continuous emergence of new henipaviruses as well as henipa-like viruses, it is necessary to conduct a more comprehensive investigation of the biology of henipaviruses and their interaction with the host. The replication of henipaviruses and the development of antiviral agents can be studied in systems that allow experiments to be performed under biosafety level 2 conditions. Here, we developed robust minigenome systems for the Nipah virus (NiV) and Hendra virus (HeV) that provide a convenient alternative for studying NiV and HeV replication. Using these systems, we demonstrate that any combination of the three polymerase complex proteins of NiV and HeV could effectively initiate the replication of both viral minigenomes, which suggests that the interaction regions of the polymerase complex proteins could be effective targets for universal and effective anti-henipavirus interventions.


Asunto(s)
Genoma Viral , Virus Nipah , Replicación Viral , Virus Nipah/genética , Virus Nipah/fisiología , Humanos , Proteínas Virales/metabolismo , Proteínas Virales/genética , Virus Hendra/genética , Virus Hendra/metabolismo , Virus Hendra/fisiología , Animales , Henipavirus/genética , Henipavirus/metabolismo , Infecciones por Henipavirus/virología , Fosfoproteínas/metabolismo , Fosfoproteínas/genética , Proteínas de la Nucleocápside/metabolismo , Proteínas de la Nucleocápside/genética , Línea Celular
2.
J Virol ; 98(3): e0183823, 2024 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-38426726

RESUMEN

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.


Asunto(s)
Infecciones por Henipavirus , Henipavirus , Receptores Virales , Humanos , Aminoácidos/genética , Anticuerpos Monoclonales/metabolismo , Proteínas Portadoras/metabolismo , Efrina-B3/genética , Efrina-B3/química , Efrina-B3/metabolismo , Epítopos/genética , Epítopos/metabolismo , Ghana , Virus Hendra/metabolismo , Henipavirus/clasificación , Henipavirus/genética , Henipavirus/metabolismo , Mutagénesis , Virus Nipah/metabolismo , Proteínas del Envoltorio Viral/genética , Internalización del Virus , Receptores Virales/metabolismo
3.
J Virol ; 97(6): e0043323, 2023 06 29.
Artículo en Inglés | MEDLINE | ID: mdl-37278642

RESUMEN

Langya virus (LayV) is a paramyxovirus in the Henipavirus genus, closely related to the deadly Nipah (NiV) and Hendra (HeV) viruses, that was identified in August 2022 through disease surveillance following animal exposure in eastern China. Paramyxoviruses present two glycoproteins on their surface, known as attachment and fusion proteins, that mediate entry into cells and constitute the primary antigenic targets for immune response. Here, we determine cryo-electron microscopy (cryo-EM) structures of the uncleaved LayV fusion protein (F) ectodomain in pre- and postfusion conformations. The LayV-F protein exhibits pre- and postfusion architectures that, despite being highly conserved across paramyxoviruses, show differences in their surface properties, in particular at the apex of the prefusion trimer, that may contribute to antigenic variability. While dramatic conformational changes were visualized between the pre- and postfusion forms of the LayV-F protein, several domains remained invariant, held together by highly conserved disulfides. The LayV-F fusion peptide (FP) is buried within a highly conserved, hydrophobic interprotomer pocket in the prefusion state and is notably less flexible than the rest of the protein, highlighting its "spring-loaded" state and suggesting that the mechanism of pre-to-post transition must involve perturbations to the pocket and release of the fusion peptide. Together, these results offer a structural basis for how the Langya virus fusion protein compares to its Henipavirus relatives and propose a mechanism for the initial step of pre- to postfusion conversion that may apply more broadly to paramyxoviruses. IMPORTANCE The Henipavirus genus is quickly expanding into new animal hosts and geographic locations. This study compares the structure and antigenicity of the Langya virus fusion protein to other henipaviruses, which have important vaccine and therapeutic development implications. Furthermore, the study proposes a new mechanism to explain the early steps of the fusion initiation process that can be more broadly applied to the Paramyxoviridae family.


Asunto(s)
Henipavirus , Proteínas Virales de Fusión , Animales , Microscopía por Crioelectrón , Henipavirus/metabolismo , Péptidos , Conformación Proteica , Proteínas Virales de Fusión/metabolismo , Internalización del Virus
4.
J Virol ; 95(20): e0066621, 2021 09 27.
Artículo en Inglés | MEDLINE | ID: mdl-34288734

RESUMEN

Cedar virus (CedV) is a nonpathogenic member of the Henipavirus (HNV) genus of emerging viruses, which includes the deadly Nipah (NiV) and Hendra (HeV) viruses. CedV forms syncytia, a hallmark of henipaviral and paramyxoviral infections and pathogenicity. However, the intrinsic fusogenic capacity of CedV relative to NiV or HeV remains unquantified. HNV entry is mediated by concerted interactions between the attachment (G) and fusion (F) glycoproteins. Upon receptor binding by the HNV G head domain, a fusion-activating G stalk region is exposed and triggers F to undergo a conformational cascade that leads to viral entry or cell-cell fusion. Here, we demonstrate quantitatively that CedV is inherently significantly less fusogenic than NiV at equivalent G and F cell surface expression levels. We then generated and tested six headless CedV G mutants of distinct C-terminal stalk lengths, surprisingly revealing highly hyperfusogenic cell-cell fusion phenotypes 3- to 4-fold greater than wild-type CedV levels. Additionally, similarly to NiV, a headless HeV G mutant yielded a less pronounced hyperfusogenic phenotype compared to wild-type HeV. Further, coimmunoprecipitation and cell-cell fusion assays revealed heterotypic NiV/CedV functional G/F bidentate interactions, as well as evidence of HNV G head domain involvement beyond receptor binding or G stalk exposure. All evidence points to the G head/stalk junction being key to modulating HNV fusogenicity, supporting the notion that head domains play several distinct and central roles in modulating stalk domain fusion promotion. Further, this study exemplifies how CedV may help elucidate important mechanistic underpinnings of HNV entry and pathogenicity. IMPORTANCE The Henipavirus genus in the Paramyxoviridae family includes the zoonotic Nipah (NiV) and Hendra (HeV) viruses. NiV and HeV infections often cause fatal encephalitis and pneumonia, but no vaccines or therapeutics are currently approved for human use. Upon viral entry, Henipavirus infections yield the formation of multinucleated cells (syncytia). Viral entry and cell-cell fusion are mediated by the attachment (G) and fusion (F) glycoproteins. Cedar virus (CedV), a nonpathogenic henipavirus, may be a useful tool to gain knowledge on henipaviral pathogenicity. Here, using homotypic and heterotypic full-length and headless CedV, NiV, and HeV G/F combinations, we discovered that CedV G/F are significantly less fusogenic than NiV or HeV G/F, and that the G head/stalk junction is key to modulating cell-cell fusion, refining the mechanism of henipaviral membrane fusion events. Our study exemplifies how CedV may be a useful tool to elucidate broader mechanistic understanding for the important henipaviruses.


Asunto(s)
Henipavirus/metabolismo , Proteínas Virales de Fusión/genética , Proteínas Virales de Fusión/metabolismo , Células Gigantes/metabolismo , Glicoproteínas/genética , Células HEK293 , Henipavirus/genética , Infecciones por Henipavirus/metabolismo , Infecciones por Henipavirus/virología , Humanos , Fusión de Membrana/fisiología , Receptores Virales/metabolismo , Proteínas del Envoltorio Viral/genética , Proteínas Virales de Fusión/fisiología , Acoplamiento Viral , Internalización del Virus
5.
Int J Mol Sci ; 23(2)2022 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-35055108

RESUMEN

Henipaviruses are severe human pathogens within the Paramyxoviridae family. Beyond the P protein, the Henipavirus P gene also encodes the V and W proteins which share with P their N-terminal, intrinsically disordered domain (NTD) and possess a unique C-terminal domain. Henipavirus W proteins antagonize interferon (IFN) signaling through NTD-mediated binding to STAT1 and STAT4, and prevent type I IFN expression and production of chemokines. Structural and molecular information on Henipavirus W proteins is lacking. By combining various bioinformatic approaches, we herein show that the Henipaviruses W proteins are predicted to be prevalently disordered and yet to contain short order-prone segments. Using limited proteolysis, differential scanning fluorimetry, analytical size exclusion chromatography, far-UV circular dichroism and small-angle X-ray scattering, we experimentally confirmed their overall disordered nature. In addition, using Congo red and Thioflavin T binding assays and negative-staining transmission electron microscopy, we show that the W proteins phase separate to form amyloid-like fibrils. The present study provides an additional example, among the few reported so far, of a viral protein forming amyloid-like fibrils, therefore significantly contributing to enlarge our currently limited knowledge of viral amyloids. In light of the critical role of the Henipavirus W proteins in evading the host innate immune response and of the functional role of phase separation in biology, these studies provide a conceptual asset to further investigate the functional impact of the phase separation abilities of the W proteins.


Asunto(s)
Amiloide/metabolismo , Henipavirus/metabolismo , Proteínas Virales/química , Proteínas Virales/metabolismo , Benzotiazoles/metabolismo , Dicroismo Circular , Simulación por Computador , Rojo Congo/metabolismo , Proteínas Intrínsecamente Desordenadas/química , Proteínas Intrínsecamente Desordenadas/metabolismo , Modelos Moleculares , Dominios Proteicos , Proteolisis , Dispersión del Ángulo Pequeño , Difracción de Rayos X
6.
Virol J ; 15(1): 56, 2018 03 27.
Artículo en Inglés | MEDLINE | ID: mdl-29587789

RESUMEN

BACKGROUND: Hendra virus and Nipah virus are zoonotic viruses that have caused severe to fatal disease in livestock and human populations. The isolation of Cedar virus, a non-pathogenic virus species in the genus Henipavirus, closely-related to the highly pathogenic Hendra virus and Nipah virus offers an opportunity to investigate differences in pathogenesis and receptor tropism among these viruses. METHODS: We constructed full-length cDNA clones of Cedar virus from synthetic oligonucleotides and rescued two replication-competent, recombinant Cedar virus variants: a recombinant wild-type Cedar virus and a recombinant Cedar virus that expresses a green fluorescent protein from an open reading frame inserted between the phosphoprotein and matrix genes. Replication kinetics of both viruses and stimulation of the interferon pathway were characterized in vitro. Cellular tropism for ephrin-B type ligands was qualitatively investigated by microscopy and quantitatively by a split-luciferase fusion assay. RESULTS: Successful rescue of recombinant Cedar virus expressing a green fluorescent protein did not significantly affect virus replication compared to the recombinant wild-type Cedar virus. We demonstrated that recombinant Cedar virus stimulated the interferon pathway and utilized the established Hendra virus and Nipah virus receptor, ephrin-B2, but not ephrin-B3 to mediate virus entry. We further characterized virus-mediated membrane fusion kinetics of Cedar virus with the known henipavirus receptors ephrin-B2 and ephrin-B3. CONCLUSIONS: The recombinant Cedar virus platform may be utilized to characterize the determinants of pathogenesis across the henipaviruses, investigate their receptor tropisms, and identify novel pan-henipavirus antivirals. Moreover, these experiments can be conducted safely under BSL-2 conditions.


Asunto(s)
Efrina-B2/metabolismo , Infecciones por Henipavirus/virología , Henipavirus/fisiología , Receptores Virales/metabolismo , Fusión Celular , Línea Celular , Efecto Citopatogénico Viral , Genes Reporteros , Proteínas Fluorescentes Verdes/genética , Henipavirus/genética , Henipavirus/metabolismo , Henipavirus/patogenicidad , Infecciones por Henipavirus/metabolismo , Interferón Tipo I/genética , Pruebas de Neutralización , Unión Proteica , Recombinación Genética , Genética Inversa , Proteínas del Envoltorio Viral/metabolismo , Tropismo Viral , Internalización del Virus , Replicación Viral
7.
J Gen Virol ; 97(5): 1066-1076, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-26813519

RESUMEN

The amino-terminal cytoplasmic domains of paramyxovirus attachment glycoproteins include trafficking signals that influence protein processing and cell surface expression. To characterize the role of the cytoplasmic domain in protein expression, fusion support and particle assembly in more detail, we constructed chimeric Nipah virus (NiV) glycoprotein (G) and canine distemper virus (CDV) haemagglutinin (H) proteins carrying the respective heterologous cytoplasmic domain, as well as a series of mutants with progressive deletions in this domain. CDV H retained fusion function and was normally expressed on the cell surface with a heterologous cytoplasmic domain, while the expression and fusion support of NiV G was dramatically decreased when its cytoplasmic domain was replaced with that of CDV H. The cell surface expression and fusion support functions of CDV H were relatively insensitive to cytoplasmic domain deletions, while short deletions in the corresponding region of NiV G dramatically decreased both. In addition, the first 10 residues of the CDV H cytoplasmic domain strongly influence its incorporation into virus-like particles formed by the CDV matrix (M) protein, while the co-expression of NiV M with NiV G had no significant effect on incorporation of G into particles. The cytoplasmic domains of both the CDV H and NiV G proteins thus contribute differently to the virus life cycle.


Asunto(s)
Regulación Viral de la Expresión Génica/fisiología , Henipavirus/metabolismo , Morbillivirus/metabolismo , Proteínas del Envoltorio Viral/metabolismo , Ensamble de Virus/fisiología , Internalización del Virus , Secuencia de Aminoácidos , Henipavirus/genética , Morbillivirus/genética , Proteínas del Envoltorio Viral/genética
8.
J Virol ; 88(20): 11973-80, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25100832

RESUMEN

Henipaviruses are associated with pteropodid reservoir hosts. The glycoproteins G and F of an African henipavirus (strain M74) have been reported to induce syncytium formation in kidney cells derived from a Hypsignathus monstrosus bat (HypNi/1.1) but not in nonchiropteran BHK-21 and Vero76 cells. Here, we show that syncytia are also induced in two other pteropodid cell lines from Hypsignathus monstrosus and Eidolon helvum bats upon coexpression of the M74 glycoproteins. The G protein was transported to the surface of transfected chiropteran cells, whereas surface expression in the nonchiropteran cells was detectable only in a fraction of cells. In contrast, the G protein of Nipah virus is transported efficiently to the surface of both chiropteran and nonchiropteran cells. Even in chiropteran cells, M74-G was predominantly expressed in the endoplasmic reticulum (ER), as indicated by colocalization with marker proteins. This result is consistent with the finding that all N-glycans of the M74-G proteins are of the mannose-rich type, as indicated by sensitivity to endo H treatment. These data indicate that the surface transport of M74-G is impaired in available cell culture systems, with larger amounts of viral glycoprotein present on chiropteran cells than on nonchiropteran cells. The restricted surface expression of M74-G explains the reduced fusion activity of the glycoproteins of the African henipavirus. Our results suggest strategies for the isolation of infectious viruses, which is necessary to assess the risk of zoonotic virus transmission. Importance: Henipaviruses are highly pathogenic zoonotic viruses associated with pteropodid bat hosts. Whether the recently described African bat henipaviruses have a zoonotic potential as high as that of their Asian and Australian relatives is unknown. We show that surface expression of the attachment protein G of an African henipavirus, M74, is restricted in comparison to the G protein expression of the highly pathogenic Nipah virus. Transport to the cell surface is more restricted in nonchiropteran cells than it is in chiropteran cells, explaining the differential fusion activity of the M74 surface proteins in these cells. Our results imply that surface expression of viral glycoproteins may serve as a major marker to assess the zoonotic risk of emerging henipaviruses.


Asunto(s)
Quirópteros/virología , Henipavirus/metabolismo , Proteínas del Envoltorio Viral/metabolismo , Animales , Compartimento Celular , Línea Celular , Cricetinae , Citometría de Flujo , Células Gigantes , Humanos , Especificidad de la Especie
9.
J Gen Virol ; 95(Pt 3): 539-548, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24296468

RESUMEN

In recent years, novel henipavirus-related sequences have been identified in bats in Africa. To evaluate the potential of African bat henipaviruses to spread in non-bat mammalian cells, we compared the biological functions of the surface glycoproteins G and F of the prototype African henipavirus GH-M74a with those of the glycoproteins of Nipah virus (NiV), a well-characterized pathogenic member of the henipavirus genus. Glycoproteins are central determinants for virus tropism, as efficient binding of henipavirus G proteins to cellular ephrin receptors and functional expression of fusion-competent F proteins are indispensable prerequisites for virus entry and cell-to-cell spread. In this study, we analysed the ability of the GH-M74a G and F proteins to cause cell-to-cell fusion in mammalian cell types readily permissive to NiV or Hendra virus infections. Except for limited syncytium formation in a bat cell line derived from Hypsignathus monstrosus, HypNi/1.1 cells, we did not observe any fusion. The highly restricted fusion activity was predominantly due to the F protein. Whilst GH-M74a G protein was found to interact with the main henipavirus receptor ephrin-B2 and induced syncytia upon co-expression with heterotypic NiV F protein, GH-M74a F protein did not cause evident fusion in the presence of heterotypic NiV G protein. Pulse-chase and surface biotinylation analyses revealed delayed F cleavage kinetics with a reduced expression of cleaved and fusion-active GH-M74a F protein on the cell surface. Thus, the F protein of GH-M74a showed a functional defect that is most likely caused by impaired trafficking leading to less efficient proteolytic activation and surface expression.


Asunto(s)
Quirópteros/virología , Glicoproteínas/metabolismo , Infecciones por Henipavirus/veterinaria , Henipavirus/aislamiento & purificación , Henipavirus/metabolismo , Proteínas Virales/metabolismo , África , Animales , Quirópteros/metabolismo , Glicoproteínas/genética , Henipavirus/clasificación , Henipavirus/genética , Infecciones por Henipavirus/metabolismo , Infecciones por Henipavirus/virología , Virus Nipah/genética , Virus Nipah/metabolismo , Receptores Virales/metabolismo , Proteínas del Envoltorio Viral/genética , Proteínas del Envoltorio Viral/metabolismo , Proteínas Virales/genética
10.
J Virol ; 87(24): 13889-91, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24067951

RESUMEN

Serological screening and detection of genomic RNA indicates that members of the genus Henipavirus are present not only in Southeast Asia but also in African fruit bats. We demonstrate that the surface glycoproteins F and G of an African henipavirus (M74) induce syncytium formation in a kidney cell line derived from an African fruit bat, Hypsignathus monstrosus. Despite a less broad cell tropism, the M74 glycoproteins show functional similarities to glycoproteins of Nipah virus.


Asunto(s)
Quirópteros/virología , Células Gigantes/virología , Infecciones por Henipavirus/veterinaria , Henipavirus/aislamiento & purificación , Henipavirus/metabolismo , Proteínas del Envoltorio Viral/metabolismo , África , Animales , Asia Sudoriental , Línea Celular , Henipavirus/genética , Infecciones por Henipavirus/virología , Proteínas del Envoltorio Viral/genética
11.
J Virol ; 82(13): 6259-71, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18417573

RESUMEN

Emerging viruses in the paramyxovirus genus Henipavirus evade host antiviral responses via protein interactions between the viral V and W proteins and cellular STAT1 and STAT2 and the cytosolic RNA sensor MDA5. Polo-like kinase (PLK1) is identified as being an additional cellular partner that can bind to Nipah virus P, V, and W proteins. For both Nipah virus and Hendra virus, contact between the V protein and the PLK1 polo box domain is required for V protein phosphorylation. Results indicate that PLK1 is engaged by Nipah virus V protein amino acids 100 to 160, previously identified as being the STAT1 binding domain responsible for host interferon (IFN) signaling evasion, via a Thr-Ser-Ser-Pro motif surrounding residue 130. A distinct Ser-Thr-Pro motif surrounding residue 199 mediates the PLK1 interaction with Hendra virus V protein. Select mutations in the motif surrounding residue 130 also influenced STAT1 binding and innate immune interference, and data indicate that the V:PLK1 and V:STAT complexes are V mediated yet independent of one another. The effects of STAT1/PLK1 binding motif mutations on the function the Nipah virus P protein in directing RNA synthesis were tested. Remarkably, mutations that selectively disrupt the STAT or PLK1 interaction site have no effects on Nipah virus P protein-mediated viral RNA synthesis. Therefore, mutations targeting V protein-mediated IFN evasion will not alter the RNA synthetic capacity of the virus, supporting an attenuation strategy based on disrupting host protein interactions.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Henipavirus/metabolismo , Interferón Tipo I/inmunología , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Factor de Transcripción STAT1/metabolismo , Proteínas Estructurales Virales/metabolismo , Línea Celular , Electroforesis en Gel de Poliacrilamida , Ensayo de Inmunoadsorción Enzimática , Técnica del Anticuerpo Fluorescente Indirecta , Henipavirus/inmunología , Humanos , Immunoblotting , Inmunoprecipitación , Luciferasas , Mutación/genética , Proteínas Estructurales Virales/genética , Quinasa Tipo Polo 1
12.
Viruses ; 11(9)2019 08 29.
Artículo en Inglés | MEDLINE | ID: mdl-31470664

RESUMEN

The Ghana virus (GhV) is phylogenetically related to the zoonotic henipaviruses Nipah (NiV) and Hendra virus. Although GhV uses the highly conserved receptor ephrin-B2, the fusogenicity is restricted to cell lines of bat origin. Furthermore, the surface expression of the GhV attachment glycoprotein (G) is reduced compared to NiV and most of this protein is retained in the endoplasmic reticulum (ER). Here, we generated truncated as well as chimeric GhV G proteins and investigated the influence of the structural domains (cytoplasmic tail, transmembrane domain, ectodomain) of this protein on the intracellular transport and the fusogenicity following coexpression with the GhV fusion protein (F). We demonstrate that neither the cytoplasmic tail nor the transmembrane domain is responsible for the intracellular retention of GhV G. Furthermore, the cytoplasmic tail of GhV G modulates the fusogenicity of GhV F and therefore controls the species-restricted fusogenicity of the GhV surface glycoproteins.


Asunto(s)
Fusión Celular , Henipavirus/metabolismo , Proteínas del Envoltorio Viral/química , Proteínas del Envoltorio Viral/metabolismo , Proteínas Virales de Fusión/metabolismo , Animales , Línea Celular , Quirópteros , Chlorocebus aethiops , Células HEK293 , Henipavirus/genética , Especificidad del Huésped , Humanos , Dominios Proteicos , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Eliminación de Secuencia , Células Vero , Proteínas del Envoltorio Viral/genética , Proteínas Virales de Fusión/genética
13.
Vet Microbiol ; 218: 90-97, 2018 May.
Artículo en Inglés | MEDLINE | ID: mdl-29685227

RESUMEN

Hendra virus (HeV) and Nipah virus (NiV) are highly pathogenic henipaviruses originating from fruit bats in Australia and Asia that can cause severe infections in livestock and humans. In recent years, also African bat henipaviruses were identified at the nucleic acid level. To assess their potential to replicate in non-bat species, several studies were performed to characterize the two surface glycoproteins required for virus entry and spread by cell-cell fusion. It has been shown that surface expression and fusion-helper function of the receptor-binding G protein of Kumasi virus (KV), the prototypic Ghanaian bat henipavirus, is reduced compared to other non-African henipavirus G proteins. Immunostainings and pulse-chase analysis revealed a delayed export of KV G from the ER. As defects in oligomerization of viral glycoproteins can be responsible for limited surface transport thereby restricting the bioactivity, we analyzed the oligomerization pattern of KV G. In contrast to HeV and NiV whose G proteins are known to be expressed at a dimer-tetramer ratio of 1:1, KV G almost exclusively formed stable tetramers or higher oligomers. KV G also showed less stringent requirements for defined stalk cysteines to form dimers and tetramers. Interestingly, any changes in the oligomeric forms negatively affected the fusion-helper activity although surface expression and receptor binding was unchanged. This clearly indicates that the formation of mostly higher oligomeric KV G forms is not a deficiency responsible for ER retention, but is rather a basic structural feature essential for the bioactivity of this African bat henipavirus glycoprotein.


Asunto(s)
Quirópteros/virología , Proteínas de Unión al GTP/química , Henipavirus/metabolismo , Glicoproteínas de Membrana/química , Proteínas del Envoltorio Viral , Animales , Retículo Endoplásmico/virología , Proteínas de Unión al GTP/metabolismo , Ghana/epidemiología , Henipavirus/química , Henipavirus/genética , Infecciones por Henipavirus/epidemiología , Infecciones por Henipavirus/virología , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Proteínas del Envoltorio Viral/química , Proteínas del Envoltorio Viral/metabolismo , Internalización del Virus
14.
J Virol Methods ; 142(1-2): 29-40, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17292974

RESUMEN

Hendra virus (HeV) and Nipah virus (NiV) are related emerging paramyxoviruses classified in the genus Henipavirus. Both cause fatal disease in animals and humans and are classified as biosafety level 4 pathogens. Here we detail two new multiplexed microsphere assays, one for antibody detection and differentiation and another designed as a surrogate for virus neutralization. Both assays utilize recombinant soluble attachment glycoproteins (sG) whereas the latter incorporates the cellular receptor, recombinant ephrin-B2. Spectrally distinct sG(HeV)- and sG(NiV)-coupled microspheres preferentially bound antibodies from HeV- and NiV-seropositive animals, demonstrating a simple procedure to differentiate antibodies to these closely related viruses. Soluble ephrin-B2 bound sG-coupled microspheres in a dose-dependent fashion. Specificity of binding was further evaluated with henipavirus G-specific sera and MAbs. Sera from henipavirus-seropositive animals differentially blocked ephrin-B2 binding, suggesting that detection and differentiation of HeV and NiV neutralizing antibodies can be done simultaneously in the absence of live virus.


Asunto(s)
Anticuerpos Antivirales/sangre , Virus Hendra/inmunología , Pruebas de Neutralización , Virus Nipah/inmunología , Análisis por Matrices de Proteínas , Juego de Reactivos para Diagnóstico , Animales , Anticuerpos Monoclonales/inmunología , Anticuerpos Antivirales/inmunología , Gatos , Efrina-B2/genética , Efrina-B2/inmunología , Efrina-B2/metabolismo , Virus Hendra/genética , Virus Hendra/metabolismo , Henipavirus/genética , Henipavirus/inmunología , Henipavirus/metabolismo , Infecciones por Henipavirus/epidemiología , Infecciones por Henipavirus/virología , Humanos , Ratones , Microesferas , Virus Nipah/genética , Virus Nipah/metabolismo , Análisis por Matrices de Proteínas/instrumentación , Análisis por Matrices de Proteínas/métodos , Conejos , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología , Proteínas Recombinantes/metabolismo , Sensibilidad y Especificidad , Pruebas Serológicas , Proteínas del Envoltorio Viral/genética , Proteínas del Envoltorio Viral/inmunología , Proteínas del Envoltorio Viral/metabolismo
15.
Mol Biosyst ; 13(11): 2254-2267, 2017 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-28972216

RESUMEN

Henipaviruses are severe human pathogens within the Paramyxoviridae family. Beyond the P protein, the Henipavirus P gene also encodes the V protein which shares with P its N-terminal, intrinsically disordered region (PNT) and possesses a unique C-terminal domain predicted to be folded and to bind zinc (ZnFD). Henipavirus V proteins antagonize IFN signaling through PNT-mediated binding to STAT1, and several paramyxoviral V proteins promote STAT1 degradation through binding to DDB1. Structural and molecular information on Henipavirus V proteins is lacking, and their ability to interact with DDB1 has not been documented yet. We cloned the V genes from Nipah and Hendra viruses and purified the V proteins from E. coli and DDB1 from insect cells. Using analytical size-exclusion chromatography, CD and SAXS we characterized the V proteins and their domains. Using pull-down and MST we assessed their binding abilities towards DDB1. We show that PNT remains disordered also in the context of the V protein, while the ZnFD adopts a predominant ß conformation. We also show that the V proteins interact with DDB1 predominantly via their ZnFD. This is the first experimental characterization of the Henipavirus V proteins and the first experimental evidence of their interaction with DDB1. The DDB1-ZnFD interaction constitutes a promising target for antiviral strategies. These studies provide a conceptual asset to design new antiviral strategies expected to reduce or abrogate the ability of these viruses to escape the innate immune response. They also contribute to illuminating the conformational behaviour of proteins encompassing large intrinsically disordered domains.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Henipavirus/metabolismo , Dominios y Motivos de Interacción de Proteínas , Desplegamiento Proteico , Proteínas Virales/química , Proteínas Virales/metabolismo , Dedos de Zinc , Secuencia de Aminoácidos , Dicroismo Circular , Interacciones Hidrofóbicas e Hidrofílicas , Espectrometría de Masas , Unión Proteica , Proteínas Recombinantes , Espectrofotometría Ultravioleta , Proteínas Virales/genética , Proteínas Virales/aislamiento & purificación , Difracción de Rayos X
16.
Antiviral Res ; 124: 69-76, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26526590

RESUMEN

Immune evasion by the lethal henipaviruses, Hendra (HeV) and Nipah virus, is mediated by its interferon (IFN) antagonist P gene products, phosphoprotein (P), and the related V and W proteins, which can target the signal transducer and activator of transcription 1 (STAT1) and STAT2 proteins to inhibit IFN/STAT signaling. However, it is not clear if the recently identified non-pathogenic Henipavirus, Cedar paramyxovirus (CedPV), is also able to antagonize the STAT proteins. We performed comparative studies between the HeV P gene products (P/V/W) and CedPV-P (CedPV does not encode V or W) and demonstrate that differences exist in their ability to engage the STAT proteins using immunoprecipitation and quantitative confocal microscopic analysis. In contrast to HeV-P gene encoded proteins, the ability of CedPV-P to interact with and relocalize STAT1 or STAT2 is compromised, correlating with a reduced capacity to inhibit the mRNA synthesis of IFN-inducible gene MxA. Furthermore, infection studies with HeV and CedPV demonstrate that HeV is more potent than CedPV in inhibiting the IFN-α-mediated nuclear accumulation of STAT1. These results strongly suggest that the ability of CedPV to counteract the IFN/STAT response is compromised compared to HeV.


Asunto(s)
Henipavirus/metabolismo , Fosfoproteínas/metabolismo , Factor de Transcripción STAT1/metabolismo , Factor de Transcripción STAT2/metabolismo , Proteínas Virales/metabolismo , Secuencia de Aminoácidos , Células HEK293 , Células HeLa , Henipavirus/genética , Henipavirus/inmunología , Infecciones por Henipavirus/metabolismo , Infecciones por Henipavirus/virología , Humanos , Interferón-alfa/inmunología , Interferón-alfa/metabolismo , Datos de Secuencia Molecular , Fosfoproteínas/genética , Fosfoproteínas/inmunología , ARN Mensajero/biosíntesis , ARN Mensajero/efectos de los fármacos , Reacción en Cadena en Tiempo Real de la Polimerasa , Factor de Transcripción STAT1/antagonistas & inhibidores , Factor de Transcripción STAT2/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Proteínas Virales/genética , Proteínas Virales/inmunología
17.
Trends Microbiol ; 19(8): 389-99, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21511478

RESUMEN

Henipavirus is a new genus of Paramyxoviridae that uses protein-based receptors (ephrinB2 and ephrinB3) for virus entry. Paramyxovirus entry requires the coordinated action of the fusion (F) and attachment viral envelope glycoproteins. Receptor binding to the attachment protein triggers F to undergo a conformational cascade that results in membrane fusion. The accumulation of structural and functional studies on many paramyxoviral fusion and attachment proteins, including the recent elucidation of structures of Nipah virus (NiV) and Hendra virus (HeV) G glycoproteins bound and unbound to cognate ephrinB receptors, indicate that henipavirus entry and fusion could differ mechanistically from paramyxoviruses that use glycan-based receptors.


Asunto(s)
Henipavirus/fisiología , Fusión de Membrana , Proteínas del Envoltorio Viral/metabolismo , Acoplamiento Viral , Animales , Sitios de Unión , Glicosilación , Henipavirus/metabolismo , Interacciones Huésped-Patógeno , Humanos , Filogenia , Conformación Proteica , Receptores de Superficie Celular/metabolismo , Receptores de Superficie Celular/fisiología , Receptores de la Familia Eph/metabolismo , Receptores Virales/metabolismo , Receptores Virales/fisiología , Internalización del Virus
18.
PLoS One ; 5(7): e11684, 2010 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-20657787

RESUMEN

Henipaviruses are newly emerged viruses within the Paramyxoviridae family. Their negative-strand RNA genome is packaged by the nucleoprotein (N) within alpha-helical nucleocapsid that recruits the polymerase complex made of the L protein and the phosphoprotein (P). To date structural data on Henipaviruses are scarce, and their N and P proteins have never been characterized so far. Using both computational and experimental approaches we herein show that Henipaviruses N and P proteins possess large intrinsically disordered regions. By combining several disorder prediction methods, we show that the N-terminal domain of P (PNT) and the C-terminal domain of N (NTAIL) are both mostly disordered, although they contain short order-prone segments. We then report the cloning, the bacterial expression, purification and characterization of Henipavirus PNT and NTAIL domains. By combining gel filtration, dynamic light scattering, circular dichroism and nuclear magnetic resonance, we show that both NTAIL and PNT belong to the premolten globule sub-family within the class of intrinsically disordered proteins. This study is the first reported experimental characterization of Henipavirus P and N proteins. The evidence that their respective N-terminal and C-terminal domains are highly disordered under native conditions is expected to be invaluable for future structural studies by helping to delineate N and P protein domains amenable to crystallization. In addition, following previous hints establishing a relationship between structural disorder and protein interactivity, the present results suggest that Henipavirus PNT and NTAIL domains could be involved in manifold protein-protein interactions.


Asunto(s)
Henipavirus/metabolismo , Proteínas de la Nucleocápside/química , Proteínas de la Nucleocápside/metabolismo , Fosfoproteínas/química , Fosfoproteínas/metabolismo , Proteínas Virales/química , Proteínas Virales/metabolismo , Cromatografía en Gel , Dicroismo Circular , Henipavirus/genética , Espectroscopía de Resonancia Magnética , Proteínas de la Nucleocápside/genética , Fosfoproteínas/genética , Proteínas Virales/genética
19.
PLoS One ; 3(7): e2739, 2008 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-18648649

RESUMEN

Henipaviruses are emerging RNA viruses of fruit bat origin that can cause fatal encephalitis in man. Ghanaian fruit bats (megachiroptera) were tested for antibodies to henipaviruses. Using a Luminex multiplexed microsphere assay, antibodies were detected in sera of Eidolon helvum to both Nipah (39%, 95% confidence interval: 27-51%) and Hendra (22%, 95% CI: 11-33%) viruses. Virus neutralization tests further confirmed seropositivity for 30% (7/23) of Luminex positive serum samples. Our results indicate that henipavirus is present within West Africa.


Asunto(s)
Anticuerpos Antivirales/sangre , Quirópteros/virología , Infecciones por Henipavirus/diagnóstico , Henipavirus/metabolismo , Animales , Microesferas , Modelos Teóricos , Pruebas de Neutralización , Proteínas Recombinantes/química , Especificidad de la Especie
20.
Virology ; 372(2): 357-71, 2008 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-18054977

RESUMEN

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.


Asunto(s)
Efrina-B2/metabolismo , Efrina-B3/metabolismo , Henipavirus/metabolismo , Receptores Virales/metabolismo , Secuencia de Aminoácidos , Animales , Anticuerpos Antivirales , Sitios de Unión , Línea Celular , Células Cultivadas , Clonación Molecular , ADN , Efrina-B2/química , Efrina-B2/genética , Efrina-B3/química , Efrina-B3/genética , Humanos , Ligandos , Datos de Secuencia Molecular , Especificidad de la Especie
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA