Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 91
Filtrar
1.
Virol J ; 20(1): 244, 2023 10 26.
Artigo em Inglês | MEDLINE | ID: mdl-37885034

RESUMO

BACKGROUND: Foamy viruses (FVs) are unique nonpathogenic retroviruses, which remain latent in the host for a long time. Therefore, they may be safe, effective gene transfer vectors. In this study, were assessed FV-host cell interactions and the molecular mechanisms underlying FV latent infection. METHODS: We used the prototype FV (PFV) to infect HT1080 cells and a PFV indicator cell line (PFVL) to measure virus titers. After 48 h of infection, the culture supernatant (i.e., cell-free PFV particles) and transfected cells (i.e., cell-associated PFV particles) were harvested and incubated with PFVL. After another 48 h, the luciferase activity was used to measure virus titers. RESULTS: Through transcriptomics sequencing, we found that PREB mRNA expression was significantly upregulated. Moreover, PREB overexpression reduced PFV replication, whereas endogenous PREB knockdown increased PFV replication. PREB interacted with the Tas DNA-binding and transcriptional activation domains and interfered with its binding to the PFV long terminal repeat and internal promoter, preventing the recruitment of transcription factors and thereby inhibiting the transactivation function of Tas. PREB C-terminal 329-418 aa played a major role in inhibiting PFV replication; PREB also inhibited bovine FV replication. Therefore, PREB has a broad-spectrum inhibitory effect on FV replication. CONCLUSIONS: Our results demonstrated that PREB inhibits PFV replication by impeding its transcription.


Assuntos
Spumavirus , Animais , Bovinos , Spumavirus/genética , Spumavirus/metabolismo , Fatores de Transcrição/metabolismo , Linhagem Celular , Domínios Proteicos , Retroviridae , Replicação Viral
2.
Virol J ; 19(1): 195, 2022 11 22.
Artigo em Inglês | MEDLINE | ID: mdl-36419065

RESUMO

BACKGROUND: Foamy viruses (FVs) are retroviruses with unique replication strategies that cause lifelong latent infections in their hosts. FVs can also produce foam-like cytopathic effects in vitro. However, the effect of host cytokines on FV replication requires further investigation. Although interferon induced transmembrane (IFITMs) proteins have become the focus of antiviral immune response research due to their broad-spectrum antiviral ability, it remains unclear whether IFITMs can affect FV replication. METHOD: In this study, the PFV virus titer was characterized by measuring luciferase activity after co-incubation of PFVL cell lines with the cell culture supernatants (cell-free PFV) or the cells transfected with pcPFV plasmid/infected with PFV (cell-associated PFV). The foam-like cytopathic effects of PFV infected cells was observed to reflect the virus replication. The total RNA of PFV infected cells was extracted, and the viral genome was quantified by Quantitative reverse transcription PCR to detect the PFV entry into target cells. RESULTS: In the present study, we demonstrated that IFITM1-3 overexpression inhibited prototype foamy virus (PFV) replication. In addition, an IFITM3 knockdown by small interfering RNA increased PFV replication. We further demonstrated that IFITM3 inhibited PFV entry into host cells. Moreover, IFITM3 also reduced the number of PFV envelope proteins, which was related to IFITM3 promoted envelope degradation through the lysosomal pathway. CONCLUSIONS: Taken together, these results demonstrate that IFITM3 inhibits PFV replication by inhibiting PFV entry into target cells and reducing the number of PFV envelope.


Assuntos
Spumavirus , Viroses , Humanos , Antivirais/metabolismo , Spumavirus/genética , Replicação Viral , Linhagem Celular , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo
3.
Virol J ; 18(1): 255, 2021 12 20.
Artigo em Inglês | MEDLINE | ID: mdl-34930370

RESUMO

BACKGROUND: Enterovirus 71 (EV71) usually infects infants causing hand-foot-mouth disease (HFMD), even fatal neurological disease like aseptic meningitis. Effective drug for preventing and treating EV71 infection is unavailable currently. EV71 3C mediated the cleavage of many proteins and played an important role in viral inhibiting host innate immunity. Promyelocytic leukemia (PML) protein, the primary organizer of PML nuclear bodies (PML-NBs), can be induced by interferon and is involved in antiviral activity. PML inhibits EV71 replication, and EV71 infection reduces PML expression, but the molecular mechanism is unclear. METHODS: The cleavage of PMLIII and IV was confirmed by co-transfection of EV71 3C protease and PML. The detailed cleavage sites were evaluated further by constructing the Q to A mutant of PML. PML knockout cells were infected with EV71 to identify the effect of cleavage on EV71 replication. Immunofluorescence analysis to examine the interference of EV71 3C on the formation of PML-NBs. RESULTS: EV71 3C directly cleaved PMLIII and IV. Furthermore, 3C cleaved PMLIV at the sites of Q430-A431 and Q444-S445 through its protease activity. Overexpression of PMLIV Q430A/Q444A variant exhibited stronger antiviral potential than the wild type. PMLIV Q430A/Q444A formed normal nuclear bodies that were not affected by 3C, suggesting that 3C may impair PML-NBs production via PMLIV cleavage and counter its antiviral activities. PML, especially PMLIV, which sequesters viral proteins in PML-NBs and inhibits viral production, is a novel target of EV71 3C cleavage. CONCLUSIONS: EV71 3C cleaves PMLIV at Q430-A431 and Q444-S445. Cleavage reduces the antiviral function of PML and decomposes the formation of PML-NBs, which is conducive to virus replication.


Assuntos
Enterovirus Humano A , Enterovirus , Proteases Virais 3C , Peptídeo Hidrolases , Proteína da Leucemia Promielocítica/genética
4.
Appl Opt ; 58(1): 94-101, 2019 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-30645517

RESUMO

The transmission properties of one-dimensional photonic crystals (PCs) containing a metamaterial (MM) defect layer are investigated using the transfer matrix method. The MM is composed of alternating layers of a dielectric material and a Dirac semimetal (DS) material. Numerical results show that the defective PCs possess a tunable defect mode, which is significantly dependent on the Fermi level of the DS as well as the structural parameters of the MM defect layer. The defect mode properties under different incident angles for TE and TM polarizations are also studied. Such defective structures have potential applications in tunable filters and sensors in terahertz regions.

5.
Retrovirology ; 15(1): 65, 2018 09 21.
Artigo em Inglês | MEDLINE | ID: mdl-30241541

RESUMO

BACKGROUND: Human immunodeficiency virus type 1 (HIV-1) Tat protein plays an essential role in HIV-1 gene transcription. Tat transactivates HIV-1 long terminal repeat (LTR)-directed gene expression through direct interactions with the transactivation-responsive region (TAR) element and other cis elements in the LTR. The TAR-independent Tat-mediated LTR transactivation is modulated by several host factors, but the mechanism is not fully understood. RESULTS: Here, we report that Tat interacts with the Rel homology domain of RelB through its core region. Furthermore, RelB significantly increases Tat-mediated transcription of the HIV-1 LTR and viral gene expression, which is independent of the TAR. Both Tat and RelB are recruited to the HIV-1 promoter, of which RelB facilitates the recruitment of Tat to the viral LTR. The NF-κB elements are key to the accumulation of Tat and RelB on the LTR. Knockout of RelB reduces the accumulation of RNA polymerase II on the LTR, and decreases HIV-1 gene transcription. Together, our data suggest that RelB contributes to HIV-1 transactivation. CONCLUSIONS: Our results demonstrate that RelB interacts with Tat and enhances TAR-independent activation of HIV-1 LTR promoter, which adds new insights into the multi-layered mechanisms of Tat in regulating the gene expression of HIV-1.


Assuntos
HIV-1/genética , HIV-1/metabolismo , Fator de Transcrição RelB/metabolismo , Transcrição Gênica , Produtos do Gene tat do Vírus da Imunodeficiência Humana/metabolismo , Linhagem Celular , Técnicas de Inativação de Genes , Repetição Terminal Longa de HIV/genética , Humanos , NF-kappa B/metabolismo , Regiões Promotoras Genéticas , Ligação Proteica , Domínios Proteicos , Provírus/genética , Provírus/metabolismo , Fator de Transcrição RelB/antagonistas & inibidores , Fator de Transcrição RelB/química , Fator de Transcrição RelB/genética , Ativação Transcricional , Produtos do Gene tat do Vírus da Imunodeficiência Humana/química , Produtos do Gene tat do Vírus da Imunodeficiência Humana/genética
6.
J Virol ; 91(2)2017 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-27847364

RESUMO

Enterovirus 71 (EV71) is an emerging pathogen causing hand, foot, and mouth disease (HFMD) and fatal neurological diseases in infants and young children due to their underdeveloped immunocompetence. EV71 infection can induce cellular apoptosis through a variety of pathways, which promotes EV71 release. The viral protease 3C plays an important role in EV71-induced apoptosis. However, the molecular mechanism responsible for 3C-triggered apoptosis remains elusive. Here, we found that EV71 3C directly interacted with PinX1, a telomere binding protein. Furthermore, 3C cleaved PinX1 at the site of Q50-G51 pair through its protease activity. Overexpression of PinX1 reduced the level of EV71-induced apoptosis and EV71 release, whereas depletion of PinX1 by small interfering RNA promoted apoptosis induced by etoposide and increased EV71 release. Taken together, our study uncovered a mechanism that EV71 utilizes to promote host cell apoptosis through cleavage of cellular protein PinX1 by 3C. IMPORTANCE: EV71 3C plays an important role in processing viral proteins and interacting with host cells. In this study, we showed that 3C promoted apoptosis through cleaving PinX1, a telomere binding protein, and that this cleavage facilitated EV71 release. Our study demonstrated that PinX1 plays an important role in EV71 release and revealed a novel mechanism that EV71 utilizes to induce apoptosis. This finding is important in understanding EV71-host cell interactions and has potential impact on understanding other enterovirus-host cell interactions.


Assuntos
Apoptose , Infecções por Coxsackievirus/metabolismo , Infecções por Coxsackievirus/virologia , Cisteína Endopeptidases/metabolismo , Enterovirus Humano A/fisiologia , Proteínas Supressoras de Tumor/metabolismo , Proteínas Virais/metabolismo , Proteases Virais 3C , Apoptose/efeitos dos fármacos , Proteínas de Ciclo Celular , Linhagem Celular , Infecções por Coxsackievirus/genética , Etoposídeo/farmacologia , Humanos , Ligação Proteica , Proteólise , Proteínas de Ligação a Telômeros/genética , Proteínas de Ligação a Telômeros/metabolismo , Proteínas Supressoras de Tumor/genética , Liberação de Vírus
7.
Mol Cell Biochem ; 441(1-2): 165-171, 2018 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-28913576

RESUMO

N-Myc interactor (Nmi) is reported to participate in many activities, such as signaling transduction, transcription regulation, and antiviral responses. As Nmi may play important roles in interferon (IFN)-induced responses, we investigated the mechanism how Nmi protein is regulated. We identified and cloned the promoter of Nmi gene. Sequence analysis and luciferase assays shown that an IFN-stimulated response element (ISRE) and a GC box in the promoter were essential for the basal transcription activity of Nmi gene. We also found that interferon regulatory factor 1 (IRF-1) could activate transcription of Nmi by binding to the ISRE in the promoter. Knockdown of IRF-1 decreases IFN-induced Nmi transcription. These results revealed that IRF-1 is involved in the IFN-inducible expression of Nmi.


Assuntos
Regulação da Expressão Gênica/efeitos dos fármacos , Fator Regulador 1 de Interferon/metabolismo , Interferons/farmacologia , Peptídeos e Proteínas de Sinalização Intracelular/biossíntese , Regiões Promotoras Genéticas/fisiologia , Elementos de Resposta/fisiologia , Transcrição Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/fisiologia , Células HeLa , Humanos , Fator Regulador 1 de Interferon/genética , Peptídeos e Proteínas de Sinalização Intracelular/genética , Transcrição Gênica/fisiologia
8.
J Biol Chem ; 291(52): 26670-26685, 2016 Dec 23.
Artigo em Inglês | MEDLINE | ID: mdl-27784779

RESUMO

In the HIV-1 replication cycle, the endosomal sorting complex required for transport (ESCRT) machinery promotes viral budding and release in the late stages. In this process, the ESCRT proteins, ALIX and TSG101, are recruited through interactions with HIV-1 Gag p6. ALG-2, also known as PDCD6, interacts with both ALIX and TSG101 and bridges ESCRT-III and ESCRT-I. In this study, we show that ALG-2 affects HIV-1 production negatively at both the exogenous and endogenous levels. Through a yeast two-hybrid screen, we identified HEBP2 as the binding partner of ALG-2, and we solved the crystal structure of the ALG-2·HEBP2 complex. The function of ALG-2·HEBP2 complex in HIV-1 replication was further explored. ALG-2 inhibits HIV-1 production by affecting Gag expression and distribution, and HEBP2 might aid this process by tethering ALG-2 in the cytoplasm.


Assuntos
Proteínas Reguladoras de Apoptose/metabolismo , Proteínas de Ligação ao Cálcio/metabolismo , Proteínas de Ciclo Celular/metabolismo , Proteínas de Ligação a DNA/metabolismo , Complexos Endossomais de Distribuição Requeridos para Transporte/metabolismo , Infecções por HIV/metabolismo , HIV-1/fisiologia , Hemeproteínas/metabolismo , Proteínas da Gravidez/metabolismo , Fatores de Transcrição/metabolismo , Replicação Viral/fisiologia , Apoptose , Proteínas Reguladoras de Apoptose/química , Proteínas Reguladoras de Apoptose/genética , Proteínas de Ligação ao Cálcio/química , Proteínas de Ligação ao Cálcio/genética , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/genética , Cristalização , Cristalografia por Raios X , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/genética , Complexos Endossomais de Distribuição Requeridos para Transporte/química , Complexos Endossomais de Distribuição Requeridos para Transporte/genética , Infecções por HIV/virologia , Células HeLa , Proteínas Ligantes de Grupo Heme , Hemeproteínas/química , Hemeproteínas/genética , Humanos , Proteínas da Gravidez/química , Proteínas da Gravidez/genética , Ligação Proteica , Conformação Proteica , Transporte Proteico , Fatores de Transcrição/química , Fatores de Transcrição/genética
9.
Virol J ; 14(1): 173, 2017 09 06.
Artigo em Inglês | MEDLINE | ID: mdl-28877726

RESUMO

BACKGROUND: Human BST2 (hBST2, also called Tetherin) is a host restriction factor that blocks the release of various enveloped viruses. BST2s from different mammals also possess antiviral activity. Bovine BST2s (bBST2s), bBST2A1 and bBST2A2, reduce production of cell-free bovine leukemia virus (BLV) and vesicular stomatitis virus (VSV). However, the effect of bBST2 on other retroviruses remains unstudied. RESULTS: Here, we studied the antiviral activity of wildtype and mutant bBST2A1 proteins on retroviruses including human immunodeficiency virus type 1 (HIV-1), prototypic foamy virus (PFV), bovine foamy virus (BFV) and bovine immunodeficiency virus (BIV). The results showed that wildtype bBST2A1 suppressed the release of HIV-1, PFV and BFV. We also generated bBST2A1 mutants, and found that GPI anchor and dimerization, but not glycosylation, are essential for antiviral activity of bBST2A1. Moreover, unlike hBST2, bBST2A1 displayed no inhibitory effect on cell-to-cell transmission of PFV, BFV and BIV. CONCLUSIONS: Our data suggested that bBST2A1 inhibited retrovirus release, however, had no effect on cell-to-cell transmission of retroviruses.


Assuntos
Antígeno 2 do Estroma da Médula Óssea/genética , Antígeno 2 do Estroma da Médula Óssea/metabolismo , Infecções por Retroviridae/transmissão , Retroviridae/fisiologia , Liberação de Vírus/genética , Animais , Antivirais/metabolismo , Bovinos , Linhagem Celular , Dimerização , Proteínas Ligadas por GPI/metabolismo , Humanos , Mutação
10.
J Biol Chem ; 290(7): 4248-59, 2015 Feb 13.
Artigo em Inglês | MEDLINE | ID: mdl-25527505

RESUMO

The interferon-induced transmembrane proteins (IFITMs) broadly inhibit virus infections, particularly at the viral entry level. However, despite this shared ability to inhibit fusion, IFITMs differ in the potency and breadth of viruses restricted, an anomaly that is not fully understood. Here, we show that differences in the range of viruses restricted by IFITM1 are regulated by a C-terminal non-canonical dibasic sorting signal KRXX that suppresses restriction of some viruses by governing its intracellular distribution. Replacing the two basic residues with alanine (KR/AA) increased restriction of jaagsiekte sheep retrovirus and 10A1 amphotropic murine leukemia virus. Deconvolution microscopy revealed an altered subcellular distribution for KR/AA, with fewer molecules in LAMP1-positive lysosomes balanced by increased levels in CD63-positive multivesicular bodies, where jaagsiekte sheep retrovirus pseudovirions are colocalized. IFITM1 binds to cellular adaptor protein complex 3 (AP-3), an association that is lost when the dibasic motif is altered. Although knockdown of AP-3 itself decreases some virus entry, expression of parental IFITM1, but not its KR/AA mutant, potentiates inhibition of viral infections in AP-3 knockdown cells. By using the substituted cysteine accessibility method, we provide evidence that IFITM1 adopts more than one membrane topology co-existing in cellular membranes. Because the C-terminal dibasic sorting signal is unique to human IFITM1, our results provide novel insight into understanding the species- and virus-specific antiviral effect of IFITMs.


Assuntos
Complexo 3 de Proteínas Adaptadoras/metabolismo , Antígenos de Diferenciação/metabolismo , Membrana Celular/metabolismo , Retrovirus Jaagsiekte de Ovinos/fisiologia , Sinais Direcionadores de Proteínas/fisiologia , Internalização do Vírus , Animais , Antígenos de Diferenciação/genética , Western Blotting , Fusão Celular , Células Cultivadas , Humanos , Imunoprecipitação , Lisossomos/metabolismo , Mutação/genética , Transporte Proteico , Ovinos , Viroses/virologia , Replicação Viral
11.
Virol J ; 13: 117, 2016 06 30.
Artigo em Inglês | MEDLINE | ID: mdl-27363487

RESUMO

BACKGROUND: Bovine foamy virus (BFV) encodes the transactivator BTas, which enhances viral gene transcription by binding to the long terminal repeat promoter and the internal promoter. In this study, we investigated the different replication capacities of two similar BFV full-length DNA clones, pBS-BFV-Y and pBS-BFV-B. RESULTS: Here, functional analysis of several chimeric clones revealed a major role for the C-terminal region of the viral genome in causing this difference. Furthermore, BTas-B, which is located in this C-terminal region, exhibited a 20-fold higher transactivation activity than BTas-Y. Sequence alignment showed that these two sequences differ only at amino acid 108, with BTas-B containing N108 and BTas-Y containing D108 at this position. Results of mutagenesis studies demonstrated that residue N108 is important for BTas binding to viral promoters. In addition, the N108D mutation in pBS-BFV-B reduced the viral replication capacity by about 1.5-fold. CONCLUSIONS: Our results suggest that residue N108 is important for BTas binding to BFV promoters and has a major role in BFV replication. These findings not only advances our understanding of the transactivation mechanism of BTas, but they also highlight the importance of certain sequence polymorphisms in modulating the replication capacity of isolated BFV clones.


Assuntos
Doenças dos Bovinos/virologia , Regulação Viral da Expressão Gênica , Regiões Promotoras Genéticas , Infecções por Retroviridae/veterinária , Spumavirus/metabolismo , Transativadores/química , Transativadores/metabolismo , Proteínas Virais/química , Proteínas Virais/metabolismo , Motivos de Aminoácidos , Animais , Bovinos , Infecções por Retroviridae/virologia , Spumavirus/química , Spumavirus/genética , Transativadores/genética , Proteínas Virais/genética
12.
J Immunol ; 193(8): 4125-36, 2014 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-25225669

RESUMO

Owing to ongoing recognition of pathogen-associated molecular patterns, immune activation and upregulation of IFN-stimulated genes (ISGs) are sustained in the chronically infected host. Albeit most ISGs are important effectors for containing viral replication, some might exert compensatory immune suppression to limit pathological dysfunctions, although the mechanisms are not fully understood. In this study, we report that the ISG lymphocyte Ag 6 complex, locus E (LY6E) is a negative immune regulator of monocytes. LY6E in monocytes negatively modulated CD14 expression and subsequently dampened the responsiveness to LPS stimulation in vitro. In the setting of chronic HIV infection, the upregulation of LY6E was correlated with reduced CD14 level on monocytes; however, the immunosuppressive effect of LY6E was not adequate to remedy the hyperresponsiveness of activated monocytes. Taken together, the regulatory LY6E pathway in monocytes represents one of negative feedback mechanisms that counterbalance monocyte activation, which might be caused by LPS translocation through the compromised gastrointestinal tract during persistent HIV-1 infection and may serve as a potential target for immune intervention.


Assuntos
Antígenos de Superfície/imunologia , Infecções por HIV/imunologia , HIV-1/imunologia , Interferon-alfa/imunologia , Receptores de Lipopolissacarídeos/imunologia , Monócitos/imunologia , Receptor 4 Toll-Like/imunologia , Adulto , Antígenos de Superfície/genética , Linhagem Celular Tumoral , Feminino , Proteínas Ligadas por GPI/genética , Proteínas Ligadas por GPI/imunologia , Trato Gastrointestinal/imunologia , Infecções por HIV/virologia , Células HeLa , Humanos , Tolerância Imunológica , Fatores Reguladores de Interferon/genética , Interferon-alfa/farmacologia , Receptores de Lipopolissacarídeos/biossíntese , Lipopolissacarídeos/imunologia , Masculino , Pessoa de Meia-Idade , Interferência de RNA , RNA Interferente Pequeno , Transdução de Sinais/imunologia , Fator de Necrose Tumoral alfa/biossíntese , Regulação para Cima , Replicação Viral/imunologia
13.
Retrovirology ; 12: 1, 2015 Jan 09.
Artigo em Inglês | MEDLINE | ID: mdl-25571928

RESUMO

BACKGROUND: The human myxovirus-resistance protein B (MxB, also called Mx2) was recently reported to inhibit HIV-1 infection by impeding the nuclear import and integration of viral DNA. However, it is currently unknown whether there exist MxB-resistant HIV-1 strains in the infected individuals. Answer to this question should address whether MxB exerts an inhibitory pressure on HIV-1 in vivo and whether HIV-1 has evolved to evade MxB inhibition. FINDINGS: We have examined ten transmitted founder (T/F) HIV-1 strains for their sensitivity to MxB inhibition by infecting CD4+ T cell lines SupT1 and PM1 that were stably transduced to express MxB. Two T/F stains, CH040.c and RHPA.c, were found resistant and this resistance phenotype was mapped to the amino acid positions 87 and 208 in viral capsid. The H87Q mutation is located in the cyclophilin A (CypA) binding loop and has a prevalence of 21% in HIV-1 sequences registered in HIV database. This finding prompted us to test other frequent amino acid variants in the CypA-binding region and the results revealed MxB-resistant mutations at amino acid positions 86, 87, 88 and 92 in capsid. All these mutations diminished the interaction of HIV-1 capsid with CypA. CONCLUSIONS: Our results demonstrate the existence of MxB-resistant T/F HIV-1 strains. The high prevalence of MxB-resistant mutations in the CypA-binding loop indicates the significant selective pressure of MxB on HIV-1 replication in vivo especially given that this viral resistance mechanism operates at expense of losing CypA.


Assuntos
Ciclofilina A/metabolismo , Proteína do Núcleo p24 do HIV/metabolismo , HIV-1/imunologia , Interações Hospedeiro-Patógeno , Evasão da Resposta Imune , Proteínas de Resistência a Myxovirus/metabolismo , Proteína do Núcleo p24 do HIV/genética , Humanos , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Mutação de Sentido Incorreto , Ligação Proteica , Seleção Genética
14.
J Virol ; 88(12): 7036-44, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24719420

RESUMO

UNLABELLED: Foamy viruses (FVs) are complex retroviruses that establish lifelong persistent infection without evident pathology. However, the roles of cellular factors in FV latency are poorly understood. This study revealed that N-Myc interactor (Nmi) could inhibit the replication of prototype foamy virus (PFV). Overexpression of Nmi reduced PFV replication, whereas its depletion by small interfering RNA increased PFV replication. The Nmi-mediated impairment of PFV replication resulted from the diminished transactivation by PFV Tas of the viral long terminal repeat (LTR) and an internal promoter (IP). Nmi was determined to interact with Tas and abrogate its function by sequestration in the cytoplasm. In addition, human and bovine Nmi proteins were found to inhibit the replication of bovine foamy virus (BFV) and PFV. Together, these results indicate that Nmi inhibits both human and bovine FVs by interfering with the transactivation function of Tas and may have a role in the host defense against FV infection. IMPORTANCE: From this study, we report that the N-Myc interactor (Nmi), an interferon-induced protein, can interact with the regulatory protein Tas of the prototype foamy virus and sequester it in the cytoplasm. The results of this study suggest that Nmi plays an important role in maintaining foamy virus latency and may reveal a new pathway in the interferon-mediated antiviral barrier against viruses. These findings are important for understanding virus-host relationships not only with FVs but potentially for other retroviruses as well.


Assuntos
Proteínas Proto-Oncogênicas c-myc/metabolismo , Infecções por Retroviridae/metabolismo , Spumavirus/metabolismo , Proteínas Virais/metabolismo , Linhagem Celular , Interações Hospedeiro-Patógeno , Humanos , Ligação Proteica , Proteínas Proto-Oncogênicas c-myc/genética , Infecções por Retroviridae/genética , Infecções por Retroviridae/virologia , Spumavirus/genética , Sequências Repetidas Terminais , Proteínas Virais/genética
15.
Cell Microbiol ; 16(7): 1080-93, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24521078

RESUMO

Members of the interferon-induced transmembrane (IFITM) protein family inhibit the entry of a wide range of viruses. Viruses often exploit the endocytosis pathways to invade host cells and escape from the endocytic vesicles often in response to low pH. Localization to these endocytic vesicles is essential for IFITM3 to interfere with the cytosolic entry of pH-dependent viruses. However, the nature of the sorting signal that targets IFITM3 to these vesicles is poorly defined. In this study, we report that IFITM3 possesses a YxxΦ sorting motif, i.e. 20-YEML-23, that enables IFITM3 to undergo endocytosis through binding to the µ2 subunit of the AP-2 complex. IFITM3 accumulates at the plasma membrane as a result of either mutating 20-YEML-23, depleting the µ2 subunit or overexpressing µ2 mutants. Importantly, blocking endocytosis of IFITM3 abrogates its ability to inhibit pH-dependent viruses. We have therefore identified a critical sorting signal, namely 20-YEML-23, that controls both the endocytic trafficking and the antiviral action of IFITM3. This finding also reveals that as an endocytic protein, IFITM3 first arrives at the plasma membrane before it is endocytosed and further traffics to the late endosomes where it acts to impede virus entry.


Assuntos
Endossomos/metabolismo , Proteínas de Membrana/metabolismo , Proteínas de Ligação a RNA/metabolismo , Complexo 2 de Proteínas Adaptadoras/metabolismo , Subunidades mu do Complexo de Proteínas Adaptadoras/metabolismo , Motivos de Aminoácidos , Membrana Celular/metabolismo , Sequência Conservada , Endocitose , Células HEK293 , Humanos , Vírus da Influenza A Subtipo H1N1/fisiologia , Proteínas de Membrana/química , Dados de Sequência Molecular , Ligação Proteica , Sinais Direcionadores de Proteínas , Subunidades Proteicas , Transporte Proteico , Proteínas de Ligação a RNA/química , Internalização do Vírus
16.
BMC Vet Res ; 11: 224, 2015 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-26307352

RESUMO

BACKGROUND: Bovine herpesvirus 4 (BoHV-4) is a gammaherpesvirus whose genome was cloned as Bacterial Artificial Chromosome (BAC) and exploited as a gene delivery vector for vaccine purposes. Although BoHV-4 genome has been completely sequenced and its open reading frames (ORFs) structurally defined in silico, most of them are not functionally characterized. In BoHV-4 genome two major immediate early genes (IE) are present, IE1 and IE2. IE2 is an essential gene because its removal from the viral genome renders the virus unable to replicate, whereas for IE1 no many functional information are available. RESULTS: In this work, IE1 contribution in initiating and maintaining BoHV-4 lytic replication was assessed generating a recombinant BoHV-4 genome lacking of IE1 gene, BoHV-4ΔIE1. In contrast to BoHV-4IE2 deleted mutant, BoHV-4ΔIE1 infectious replicating viral particles (IRVPs) could be reconstituted following viral DNA electroporation in permissive cells. However the titer of BoHV-4ΔIE1 IRVPs produced into the cell supernatant and BoHV-4ΔIE1 plaques size were reduced respect to BoHV-4 undeleted control. Further the impaired BoHV-4ΔIE1 IRVPs produced into the cell supernatant could be rescued by expressing IE1 gene product in trans, confirming the implication of IE1 in BoHV-4 lytic replication. Next, the possible role of BoHV-4IE1 as bone marrow stromal cell antigen 2 (BST-2) counteracting factor, as hypothesized by IE1 amino-terminal gene product homology with Kaposi Sarcoma Associated Herpesvirus (KSHV) K5, was excluded too. CONCLUSIONS: Although the real function of BoHV-4IE1 is still elusive, a new BoHV-4 genome gene locus as a target site for the insertion of foreign DNA and resulting in the attenuation of the virus has been revealed. These data can be considered of relevance to improve BoHV-4 gene delivery properties.


Assuntos
Regulação Viral da Expressão Gênica/fisiologia , Genes Precoces/fisiologia , Herpesvirus Bovino 4/metabolismo , Animais , Linhagem Celular , Cromossomos Artificiais Bacterianos , Deleção de Genes , Genoma Viral , Herpesvirus Bovino 4/genética , Humanos , Células-Tronco Mesenquimais , Ensaio de Placa Viral , Replicação Viral/fisiologia
17.
J Biol Chem ; 288(29): 21148-21160, 2013 Jul 19.
Artigo em Inglês | MEDLINE | ID: mdl-23754279

RESUMO

LINE-1 (long interspersed element 1) is an autonomous non-long terminal repeat retrotransposon. Its replication often causes mutation and rearrangement of host genomic DNA. Accordingly, host cells have evolved mechanisms to control LINE-1 mobility. Here, we report that a helicase named MOV10 effectively suppresses LINE-1 transposition. Mutating the helicase motifs impairs this function of MOV10, suggesting that MOV10 requires its helicase activity to suppress LINE-1 replication. Further studies show that MOV10 post-transcriptionally diminishes the level of LINE-1 RNA. The association of MOV10 with both LINE-1 RNA and ORF1 suggests that MOV10 interacts with LINE-1 RNP and consequently causes its RNA degradation. These data demonstrate collectively that MOV10 contributes to the cellular control of LINE-1 replication.


Assuntos
Elementos Nucleotídeos Longos e Dispersos/genética , RNA Helicases/metabolismo , Regiões 5' não Traduzidas/genética , Animais , Proteínas Argonautas/metabolismo , DNA Complementar/genética , Técnicas de Silenciamento de Genes , Células HEK293 , Células HeLa , Humanos , Camundongos , Regiões Promotoras Genéticas/genética , RNA/biossíntese , Estabilidade de RNA/genética
18.
Retrovirology ; 11: 45, 2014 Jun 09.
Artigo em Inglês | MEDLINE | ID: mdl-24912525

RESUMO

BACKGROUND: The Vpr protein of human immunodeficiency virus type 1 (HIV-1) plays an important role in viral replication. It has been reported that Vpr stimulates the nuclear factor-κB (NF-κB) and activator protein 1 (AP-1) signaling pathways, and thereby regulates viral and host cell gene expression. However, the molecular mechanism behind this function of Vpr is not fully understood. RESULTS: Here, we have identified transforming growth factor-ß-activated kinase 1 (TAK1) as the important upstream signaling molecule that Vpr associates with in order to activate NF-κB and AP-1 signaling. HIV-1 virion-associated Vpr is able to stimulate phosphorylation of TAK1. This activity of Vpr depends on its association with TAK1, since the S79A Vpr mutant lost interaction with TAK1 and was unable to activate TAK1. This association allows Vpr to promote the interaction of TAB3 with TAK1 and increase the polyubiquitination of TAK1, which renders TAK1 phosphorylation. In further support of the key role of TAK1 in this function of Vpr, knockdown of endogenous TAK1 significantly attenuated the ability of Vpr to activate NF-κB and AP-1 as well as the ability to stimulate HIV-1 LTR promoter. CONCLUSIONS: HIV-1 Vpr enhances the phosphorylation and polyubiquitination of TAK1, and as a result, activates NF-κB and AP-1 signaling pathways and stimulates HIV-1 LTR promoter.


Assuntos
Produtos do Gene vpr/metabolismo , HIV-1/fisiologia , MAP Quinase Quinase Quinases/genética , NF-kappa B/metabolismo , Fator de Transcrição AP-1/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Linhagem Celular , Linhagem Celular Tumoral , Produtos do Gene vpr/genética , Células HEK293 , HIV-1/genética , HIV-1/metabolismo , Células HeLa , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Células Jurkat , MAP Quinase Quinase Quinases/metabolismo , NF-kappa B/genética , Fosforilação , Regiões Promotoras Genéticas , Transdução de Sinais , Fator de Transcrição AP-1/genética , Ubiquitinação , Replicação Viral
19.
Retrovirology ; 11: 26, 2014 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-24656154

RESUMO

BACKGROUND: The equine infectious anemia virus (EIAV) is a lentivirus of the Retrovirus family, which causes persistent infection in horses often characterized by recurrent episodes of high fever. It has a similar morphology and life cycle to the human immunodeficiency virus (HIV). Its transmembrane glycoprotein, gp45 (analogous to gp41 in HIV), mediates membrane fusion during the infection. However, the post-fusion conformation of EIAV gp45 has not yet been determined. EIAV is the first member of the lentiviruses for which an effective vaccine has been successfully developed. The attenuated vaccine strain, FDDV, has been produced from a pathogenic strain by a series of passages in donkey dermal cells. We have previously reported that a V/I505T mutation in gp45, in combination with other mutations in gp90, may potentially contribute to the success of the vaccine strain. To this end, we now report on our structural and biochemical studies of the gp45 protein from both wide type and vaccine strain, providing a valuable structural model for the advancement of the EIAV vaccine. RESULTS: We resolved crystal structures of the ecto-domain of gp45 from both the wild-type EIAV and the vaccine strain FDDV. We found that the V/I505T mutation in gp45 was located in a highly conserved d position within the heptad repeat, which protruded into a 3-fold symmetry axis within the six-helix bundle. Our crystal structure analyses revealed a shift of a hydrophobic to hydrophilic interaction due to this specific mutation, and further biochemical and virological studies confirmed that the mutation reduced the overall stability of the six-helix bundle in post-fusion conformation. Moreover, we found that altering the temperatures drastically affected the viral infectivity. CONCLUSIONS: Our high-resolution crystal structures of gp45 exhibited high conservation between the gp45/gp41 structures of lentiviruses. In addition, a hydrophobic to hydrophilic interaction change in the EIAV vaccine strain was found to modulate the stability and thermal-sensitivity of the overall gp45 structure. Our observations suggest that lowering the stability of the six-helix bundle (post-fusion), which may stabilizes the pre-fusion conformation, might be one of the reasons of acquired dominance for FDDV in viral attenuation.


Assuntos
Vírus da Anemia Infecciosa Equina/fisiologia , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Mutação de Sentido Incorreto , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/metabolismo , Animais , Cristalografia por Raios X , Cavalos , Vírus da Anemia Infecciosa Equina/química , Vírus da Anemia Infecciosa Equina/genética , Proteínas Mutantes/genética , Conformação Proteica , Estabilidade Proteica/efeitos da radiação , Temperatura , Proteínas do Envelope Viral/genética , Vacinas Virais/química , Vacinas Virais/genética
20.
Virus Genes ; 48(3): 464-73, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24615636

RESUMO

Foamy virus (FV) establishes persistent infection in the host without causing apparent disease. Besides the transactivator Tas protein, another auxiliary protein--Bet--has been reported in prototype foamy virus, equine foamy virus, and feline foamy virus. Here, we found the putative bbet gene in clone C74 from a cDNA library of bovine foamy virus strain 3026 (BFV3026) by comparison of gene localization, composition, and splicing features with other known bet genes. Subsequently, BBet protein was detected in BFV3026-infected cells by Western blot and immunofluorescence analyses. Analysis of the BBet mutant infectious clone (pBS-BFVdelBBet) revealed that BBet could inhibit BFV3026 replication. Consistent with this result, overexpression of BBet in Cf2Th cells reduced BFV replication by approximately threefold. Furthermore, virus replication levels similarly were reduced by approximately threefold in pBS-BFV-transfected and BFV3026-infected Cf2Th cells stably expressing BBet compared with control cells. After three passages, BFV3026 replicated more slowly in BBet-expressing cells. This study implicates BBet as a negative regulator of BFV replication and provides a resource for future studies on the function of this protein in the virus lifecycle.


Assuntos
Doenças dos Bovinos/virologia , Regulação para Baixo , Infecções por Retroviridae/veterinária , Spumavirus/fisiologia , Proteínas Virais/metabolismo , Replicação Viral , Animais , Bovinos , Linhagem Celular , Feminino , Regulação Viral da Expressão Gênica , Camundongos , Camundongos Endogâmicos BALB C , Infecções por Retroviridae/virologia , Spumavirus/genética , Proteínas Virais/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA