Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Cell Rep ; 42(10): 113254, 2023 10 31.
Artigo em Inglês | MEDLINE | ID: mdl-37858466

RESUMO

Ebola virus (EBOV) and Bundibugyo virus (BDBV) belong to the family Filoviridae and cause a severe disease in humans. We previously isolated a large panel of monoclonal antibodies from B cells of human survivors from the 2007 Uganda BDBV outbreak, 16 survivors from the 2014 EBOV outbreak in the Democratic Republic of the Congo, and one survivor from the West African 2013-2016 EBOV epidemic. Here, we demonstrate that EBOV and BDBV are capable of spreading to neighboring cells through intercellular connections in a process that depends upon actin and T cell immunoglobulin and mucin 1 protein. We quantify spread through intercellular connections by immunofluorescence microscopy and flow cytometry. One of the antibodies, BDBV223, specific to the membrane-proximal external region, induces virus accumulation at the plasma membrane. The inhibiting activity of BDBV223 depends on BST2/tetherin.


Assuntos
Anticorpos Monoclonais , Anticorpos Antivirais , Antígeno 2 do Estroma da Médula Óssea , Ebolavirus , Doença pelo Vírus Ebola , Humanos , Antígenos CD , Antígeno 2 do Estroma da Médula Óssea/imunologia , Ebolavirus/imunologia , Proteínas Ligadas por GPI , Doença pelo Vírus Ebola/virologia
2.
Viruses ; 15(2)2023 01 30.
Artigo em Inglês | MEDLINE | ID: mdl-36851607

RESUMO

Canine influenza virus (CIV) significantly threatens the canine population and public health. Tetherin, an innate immune factor, plays an important role in the defense against pathogen invasion and has been discovered to restrict the release of various enveloped viruses. Two isoforms of canine tetherin (tetherin-X1 and tetherin-X2) were identified in peripheral blood leukocytes of mixed-breed dogs using reverse transcription polymerase chain reaction (RT-PCR). Amino acid alignment revealed that relative to full-length tetherin (tetherin-X1) and truncated canine tetherin (tetherin-X2) exhibited deletion of 34 amino acids. The deletion occurred at the C-terminus of the coiled-coiled ectodomain and the N-terminus of the glycosylphosphatidylinositol (GPI)-anchor domain. Tetherin-X2 was localized subcellularly at the cell membrane, which was consistent with the localization of tetherin-X1. In addition, canine tetherin-X1 and tetherin-X2 restricted the release of H3N2 CIV. However, canine tetherin-X1 had higher antiviral activity than canine tetherin-X2, indicating that the C-terminus of the coiled-coiled ectodomain and the N-terminus of the GPI-anchor domain of canine tetherin (containing the amino acids deleted in tetherin-X2) are critical for its ability to restrict H3N2 CIV release. This study provides insights for understanding the key functional domains of tetherin that restrict CIV release.


Assuntos
Antivirais , Antígeno 2 do Estroma da Médula Óssea , Doenças do Cão , Vírus da Influenza A Subtipo H3N2 , Infecções por Orthomyxoviridae , Animais , Cães , Aminoácidos , Antivirais/imunologia , Antivirais/uso terapêutico , Antígeno 2 do Estroma da Médula Óssea/imunologia , Antígeno 2 do Estroma da Médula Óssea/uso terapêutico , Glicosilfosfatidilinositóis , Vírus da Influenza A Subtipo H3N2/imunologia , Isoformas de Proteínas/genética , Doenças do Cão/prevenção & controle , Infecções por Orthomyxoviridae/prevenção & controle , Infecções por Orthomyxoviridae/veterinária
3.
Int J Mol Sci ; 23(19)2022 Sep 27.
Artigo em Inglês | MEDLINE | ID: mdl-36232695

RESUMO

Bone Marrow Stromal Cell Antigen 2 (BST2) is a type II transmembrane protein expressed on various cell types that tethers the release of viruses. Natural killer (NK) cells express low levels of BST2 under normal conditions but exhibit increased expression of BST2 upon activation. In this study, we show for the first time that murine BST2 can control the cytotoxicity of NK cells. The cytoplasmic tail of murine BST2 contains an immunoreceptor tyrosine-based inhibitory motif (ITIM). The absence of BST2 on NK cells can enhance their cytotoxicity against tumor cells compared to wild type NK cells. NK cells isolated from NZW mice, which express ITIM-deficient BST2, also showed higher cytotoxicity than wild type NK cells. In addition, we found that galectin-8 and galectin-9 were ligands of BST2, since blocking galectin-8 or -9 with monoclonal antibodies enhanced the cytotoxicity of NK cells. These results suggested that BST2 might be a novel NK cell inhibitory receptor as it was involved in regulating NK cell cytotoxicity through its interaction with galectins.


Assuntos
Antígeno 2 do Estroma da Médula Óssea , Citotoxicidade Imunológica , Células Matadoras Naturais , Animais , Anticorpos Monoclonais/imunologia , Antígenos CD/genética , Antígenos CD/imunologia , Antígeno 2 do Estroma da Médula Óssea/genética , Antígeno 2 do Estroma da Médula Óssea/imunologia , Proteínas de Transporte/imunologia , Citotoxicidade Imunológica/genética , Citotoxicidade Imunológica/imunologia , Galectinas/imunologia , Células Matadoras Naturais/imunologia , Ligantes , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/imunologia , Camundongos , Tirosina/metabolismo
4.
J Virol ; 94(12)2020 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-32238588

RESUMO

Tetherin/BST-2 is an antiviral protein that blocks the release of enveloped viral particles by linking them to the membrane of producing cells. At first, BST-2 genes were described only in humans and other mammals. Recent work identified BST-2 orthologs in nonmammalian vertebrates, including birds. Here, we identify the BST-2 sequence in domestic chicken (Gallus gallus) for the first time and demonstrate its activity against avian sarcoma and leukosis virus (ASLV). We generated a BST-2 knockout in chicken cells and showed that BST-2 is a major determinant of an interferon-induced block of ASLV release. Ectopic expression of chicken BST-2 blocks the release of ASLV in chicken cells and of human immunodeficiency virus type 1 (HIV-1) in human cells. Using metabolic labeling and pulse-chase analysis of HIV-1 Gag proteins, we verified that chicken BST-2 blocks the virus at the release stage. Furthermore, we describe BST-2 orthologs in multiple avian species from 12 avian orders. Previously, some of these species were reported to lack BST-2, highlighting the difficulty of identifying sequences of this extremely variable gene. We analyzed BST-2 genes in the avian orders Galliformes and Passeriformes and showed that they evolve under positive selection. This indicates that avian BST-2 is involved in host-virus evolutionary arms races and suggests that BST-2 antagonists exist in some avian viruses. In summary, we show that chicken BST-2 has the potential to act as a restriction factor against ASLV. Characterizing the interaction of avian BST-2 with avian viruses is important in understanding innate antiviral defenses in birds.IMPORTANCE Birds are important hosts of viruses that have the potential to cause zoonotic infections in humans. However, only a few antiviral genes (called viral restriction factors) have been described in birds, mostly because birds lack counterparts of highly studied mammalian restriction factors. Tetherin/BST-2 is a restriction factor, originally described in humans, that blocks the release of newly formed virus particles from infected cells. Recent work identified BST-2 in nonmammalian vertebrate species, including birds. Here, we report the BST-2 sequence in domestic chicken and describe its antiviral activity against a prototypical avian retrovirus, avian sarcoma and leukosis virus (ASLV). We also identify BST-2 genes in multiple avian species and show that they evolve rapidly in birds, which is an important indication of their relevance for antiviral defense. Analysis of avian BST-2 genes will shed light on defense mechanisms against avian viral pathogens.


Assuntos
Proteínas Aviárias/imunologia , Vírus do Sarcoma Aviário/imunologia , Antígeno 2 do Estroma da Médula Óssea/imunologia , Evolução Molecular , Galliformes/imunologia , Sarcoma Aviário/imunologia , Sequência de Aminoácidos , Animais , Proteínas Aviárias/genética , Vírus do Sarcoma Aviário/genética , Vírus do Sarcoma Aviário/patogenicidade , Antígeno 2 do Estroma da Médula Óssea/genética , Linhagem Celular , Fibroblastos/imunologia , Fibroblastos/virologia , Galliformes/genética , Galliformes/virologia , Regulação da Expressão Gênica , Células HEK293 , HIV-1/genética , HIV-1/imunologia , Interações Hospedeiro-Patógeno/genética , Interações Hospedeiro-Patógeno/imunologia , Humanos , Passeriformes/genética , Passeriformes/imunologia , Passeriformes/virologia , Sarcoma Aviário/genética , Sarcoma Aviário/virologia , Seleção Genética , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Transdução de Sinais , Liberação de Vírus , Replicação Viral , Produtos do Gene gag do Vírus da Imunodeficiência Humana/genética , Produtos do Gene gag do Vírus da Imunodeficiência Humana/imunologia
5.
Viruses ; 10(10)2018 10 16.
Artigo em Inglês | MEDLINE | ID: mdl-30332780

RESUMO

Tetherin (BST2/CD317/HM1.24) has emerged as a key host-cell ·defence molecule that acts by inhibiting the release and spread of diverse enveloped virions from infected cells. We analysed the biological features of canine tetherin and found it to be an unstable hydrophilic type I transmembrane protein with one transmembrane domain, no signal peptide, and multiple glycosylation and phosphorylation sites. Furthermore, the tissue expression profile of canine tetherin revealed that it was particularly abundant in immune organs. The canine tetherin gene contains an interferon response element sequence that can be regulated and expressed by canine IFN-α. A CCK-8 assay showed that canine tetherin was effective in helping mitigate cellular damage caused by canine influenza virus (CIV) infection. Additionally, we found that the overexpression of canine tetherin inhibited replication of the CIV and that interference with the canine tetherin gene enhanced CIV replication in cells. The impact of canine tetherin on CIV replication was mild. However, these results elucidate the role of the innate immune factor, canine tetherin, during CIV infection for the first time.


Assuntos
Antígeno 2 do Estroma da Médula Óssea/imunologia , Doenças do Cão/imunologia , Infecções por Orthomyxoviridae/veterinária , Orthomyxoviridae/fisiologia , Animais , Antígeno 2 do Estroma da Médula Óssea/genética , Doenças do Cão/genética , Doenças do Cão/virologia , Cães , Interferon-alfa/genética , Interferon-alfa/imunologia , Orthomyxoviridae/genética , Orthomyxoviridae/imunologia , Infecções por Orthomyxoviridae/genética , Infecções por Orthomyxoviridae/imunologia , Infecções por Orthomyxoviridae/virologia , Replicação Viral
6.
Nat Commun ; 9(1): 1371, 2018 04 10.
Artigo em Inglês | MEDLINE | ID: mdl-29636452

RESUMO

HIV-1 causes chronic inflammation and AIDS in humans, whereas related simian immunodeficiency viruses (SIVs) replicate efficiently in their natural hosts without causing disease. It is currently unknown to what extent virus-specific properties are responsible for these different clinical outcomes. Here, we incorporate two putative HIV-1 virulence determinants, i.e., a Vpu protein that antagonizes tetherin and blocks NF-κB activation and a Nef protein that fails to suppress T cell activation via downmodulation of CD3, into a non-pathogenic SIVagm strain and test their impact on viral replication and pathogenicity in African green monkeys. Despite sustained high-level viremia over more than 4 years, moderately increased immune activation and transcriptional signatures of inflammation, the HIV-1-like SIVagm does not cause immunodeficiency or any other disease. These data indicate that species-specific host factors rather than intrinsic viral virulence factors determine the pathogenicity of primate lentiviruses.


Assuntos
HIV-1/patogenicidade , Especificidade de Hospedeiro , Proteínas do Vírus da Imunodeficiência Humana/imunologia , Lentivirus de Primatas/crescimento & desenvolvimento , Vírus da Imunodeficiência Símia/patogenicidade , Proteínas Virais Reguladoras e Acessórias/imunologia , Produtos do Gene nef do Vírus da Imunodeficiência Humana/imunologia , Sequência de Aminoácidos , Animais , Antígeno 2 do Estroma da Médula Óssea/genética , Antígeno 2 do Estroma da Médula Óssea/imunologia , Complexo CD3/genética , Complexo CD3/imunologia , Chlorocebus aethiops , Feminino , Regulação da Expressão Gênica , HIV-1/crescimento & desenvolvimento , Proteínas do Vírus da Imunodeficiência Humana/genética , Humanos , Lentivirus de Primatas/patogenicidade , Ativação Linfocitária , NF-kappa B/genética , NF-kappa B/imunologia , Alinhamento de Sequência , Transdução de Sinais , Vírus da Imunodeficiência Símia/crescimento & desenvolvimento , Transcrição Gênica , Carga Viral , Proteínas Virais Reguladoras e Acessórias/genética , Virulência , Replicação Viral , Produtos do Gene nef do Vírus da Imunodeficiência Humana/genética
7.
J Virol ; 92(5)2018 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-29237831

RESUMO

Nef-specific CD8+ T lymphocytes (CD8TL) are linked to extraordinary control of primate lentiviral replication, but the mechanisms underlying their efficacy remain largely unknown. The immunodominant, Mamu-B*017:01+-restricted Nef195-203MW9 epitope in SIVmac239 partially overlaps a sorting motif important for interactions with host AP-2 proteins and, hence, downmodulation of several host proteins, including Tetherin (CD317/BST-2), CD28, CD4, SERINC3, and SERINC5. We reasoned that CD8TL-driven evolution in this epitope might compromise Nef's ability to modulate these important molecules. Here, we used deep sequencing of SIV from nine B*017:01+ macaques throughout infection with SIVmac239 to characterize the patterns of viral escape in this epitope and then assayed the impacts of these variants on Nef-mediated modulation of multiple host molecules. Acute variation in multiple Nef195-203MW9 residues significantly compromised Nef's ability to downregulate surface Tetherin, CD4, and CD28 and reduced its ability to prevent SERINC5-mediated reduction in viral infectivity but did not impact downregulation of CD3 or major histocompatibility complex class I, suggesting the selective disruption of immunomodulatory pathways involving Nef AP-2 interactions. Together, our data illuminate a pattern of viral escape dictated by a selective balance to maintain AP-2-mediated downregulation while evading epitope-specific CD8TL responses. These data could shed light on mechanisms of both CD8TL-driven viral control generally and on Mamu-B*017:01-mediated viral control specifically.IMPORTANCE A rare subset of humans infected with HIV-1 and macaques infected with SIV can control the virus without aid of antiviral medications. A common feature of these individuals is the ability to mount unusually effective CD8 T lymphocyte responses against the virus. One of the most formidable aspects of HIV is its ability to evolve to evade immune responses, particularly CD8 T lymphocytes. We show that macaques that target a specific peptide in the SIV Nef protein are capable of better control of the virus and that, as the virus evolves to escape this response, it does so at a cost to specific functions performed by the Nef protein. Our results help show how the virus can be controlled by an immune response, which could help in designing effective vaccines.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/virologia , Epitopos de Linfócito T/imunologia , Evasão da Resposta Imune/imunologia , Síndrome de Imunodeficiência Adquirida dos Símios/imunologia , Proteínas Virais Reguladoras e Acessórias/imunologia , Animais , Evolução Biológica , Antígeno 2 do Estroma da Médula Óssea/imunologia , Antígeno 2 do Estroma da Médula Óssea/metabolismo , Epitopos de Linfócito T/genética , Antígenos de Histocompatibilidade Classe I/imunologia , Humanos , Macaca/virologia , Glicoproteínas de Membrana , Proteínas de Membrana , Mutação , Proteínas de Neoplasias , RNA Viral , Receptores de Superfície Celular , Análise de Sequência , Síndrome de Imunodeficiência Adquirida dos Símios/virologia , Vírus da Imunodeficiência Símia/genética , Vírus da Imunodeficiência Símia/imunologia , Vírus da Imunodeficiência Símia/patogenicidade , Proteínas do Envelope Viral/imunologia , Proteínas Virais Reguladoras e Acessórias/genética , Replicação Viral , Produtos do Gene nef do Vírus da Imunodeficiência Humana/imunologia
8.
PLoS One ; 12(12): e0189073, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29216247

RESUMO

Vesicular stomatitis virus (VSV) release from infected cells is inhibited by the interferon (IFN)-inducible antiviral host cell factor tetherin (BST-2, CD317). However, several viruses encode tetherin antagonists and it is at present unknown whether residual VSV spread in tetherin-positive cells is also promoted by a virus-encoded tetherin antagonist. Here, we show that the viral glycoprotein (VSV-G) antagonizes tetherin in transfected cells, although with reduced efficiency as compared to the HIV-1 Vpu protein. Tetherin antagonism did not involve alteration of tetherin expression and was partially dependent on a GXXXG motif in the transmembrane domain of VSV-G. However, mutation of the GXXXG motif did not modulate tetherin sensitivity of infectious VSV. These results identify VSV-G as a tetherin antagonist in transfected cells but fail to provide evidence for a contribution of tetherin antagonism to viral spread.


Assuntos
Antígeno 2 do Estroma da Médula Óssea/metabolismo , Vesiculovirus/metabolismo , Proteínas Virais/fisiologia , Vírion/fisiologia , Animais , Antígeno 2 do Estroma da Médula Óssea/imunologia , Linhagem Celular , Proteínas de Ligação ao GTP/imunologia , Humanos , Suínos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA