Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 293
Filtrar
1.
Cell Rep ; 37(5): 109926, 2021 11 02.
Artigo em Inglês | MEDLINE | ID: mdl-34731629

RESUMO

Interferon regulatory factor 3 (IRF3) is an essential transductor for initiation of many immune responses. Here, we show that lncRNA-ISIR directly binds IRF3 to promote its phosphorylation, dimerization, and nuclear translocation, along with enhanced target gene productions. In vivo lncRNA-ISIR deficiency results in reduced IFN production, uncontrolled viral replication, and increased mortality. The human homolog, AK131315, also binds IRF3 and promotes its activation. More important, AK131315 expression is positively correlated with type I interferon (IFN-I) level and severity in patients with lupus. Mechanistically, in resting cells, IRF3 is bound to suppressor protein Flightless-1 (Fli-1), which keeps its inactive state. Upon infection, IFN-I-induced lncRNA-ISIR binds IRF3 at DNA-binding domain in cytoplasm and removes Fli-1's association from IRF3, consequently facilitating IRF3 activation. Our results demonstrate that IFN-I-inducible lncRNA-ISIR feedback strengthens IRF3 activation by removing suppressive Fli-1 in immune responses, revealing a method of lncRNA-mediated modulation of transcription factor (TF) activation.


Assuntos
Fator Regulador 3 de Interferon/metabolismo , Lúpus Eritematoso Sistêmico/metabolismo , Macrófagos Peritoneais/metabolismo , RNA Longo não Codificante/metabolismo , Estomatite Vesicular/metabolismo , Animais , Estudos de Casos e Controles , Chlorocebus aethiops , Modelos Animais de Doenças , Inativação Gênica , Células HEK293 , Humanos , Fator Regulador 3 de Interferon/genética , Lúpus Eritematoso Sistêmico/genética , Lúpus Eritematoso Sistêmico/imunologia , Macrófagos Peritoneais/imunologia , Macrófagos Peritoneais/virologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína Proto-Oncogênica c-fli-1/genética , Proteína Proto-Oncogênica c-fli-1/metabolismo , Células RAW 264.7 , RNA Longo não Codificante/genética , Receptor de Interferon alfa e beta/genética , Receptor de Interferon alfa e beta/metabolismo , Células Vero , Estomatite Vesicular/genética , Estomatite Vesicular/imunologia , Estomatite Vesicular/virologia , Vírus da Estomatite Vesicular Indiana/imunologia , Vírus da Estomatite Vesicular Indiana/patogenicidade
2.
Viruses ; 13(8)2021 08 07.
Artigo em Inglês | MEDLINE | ID: mdl-34452429

RESUMO

As a result of a viral infection, viral genomes are not only recognized by RIG-I, but also lead to the activation of RNase L, which cleaves cellular RNA to generate the endogenous RIG-I ligand (eRL). The eRL was previously identified as a specific sequence derived from the internal transcribed spacer region 2, which bears a 2'3' cyclic phosphate instead of the common 5' triphosphate. By now, the generation of the eRL and its immunostimulatory effect were shown both in vitro and in reporter systems. In this work, we aimed to elucidate whether the eRL is also generated in Influenza A (IAV) and vesicular stomatitis virus (VSV) infected cells. RNA was extracted from virus-infected cells and used for immunostimulations as well as specific PCR-strategies to detect eRL cleavage. We show that the eRL is generated in IAV infected HEK293 cells, but we could not detect specific eRL fragments in VSV infected cells. Further, RIG-I mediated IFN-response depends not only on viral genomes but also on the eRL, as immunostimulatory properties remain present under 5'triphosphate degrading conditions. In summary, we prove the IAV infection induced eRL generation in HEK293 cells, amplifying the innate immune response.


Assuntos
Proteína DEAD-box 58/genética , Interações Hospedeiro-Patógeno/genética , Imunidade Inata , Vírus da Influenza A/imunologia , Receptores Imunológicos/genética , Células A549 , Proteína DEAD-box 58/imunologia , Genoma Viral , Células HEK293 , Interações Hospedeiro-Patógeno/imunologia , Humanos , Vírus da Influenza A/patogenicidade , Influenza Humana/virologia , Interferon Tipo I/genética , Interferon Tipo I/imunologia , Receptores Imunológicos/imunologia , Vírus da Estomatite Vesicular Indiana/imunologia , Vírus da Estomatite Vesicular Indiana/patogenicidade , Replicação Viral/imunologia
3.
STAR Protoc ; 2(3): 100708, 2021 09 17.
Artigo em Inglês | MEDLINE | ID: mdl-34386779

RESUMO

Innate immunity is the first line of host defense against viral infection. As one of the innate immune cell types, antigen-presenting cells play an important role in the process of antiviral immunity. This protocol describes the analysis of innate immunity induced by vesicular stomatitis virus infection of peritoneal macrophages in vitro and in vivo detection of IFN-ß production and lung injury. For complete details on the use and execution of this protocol, please refer to Shen et al. (2021).


Assuntos
Separação Celular/métodos , Imunidade Inata/fisiologia , Viroses/diagnóstico por imagem , Animais , Células Apresentadoras de Antígenos/imunologia , Interferon Tipo I/imunologia , Macrófagos/imunologia , Macrófagos Peritoneais/citologia , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Serina-Treonina Quinases , Estomatite Vesicular/imunologia , Vírus da Estomatite Vesicular Indiana/imunologia , Vírus da Estomatite Vesicular Indiana/patogenicidade , Viroses/imunologia , Replicação Viral/imunologia
4.
Biotechniques ; 68(6): 305-310, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32202142

RESUMO

Viral infection of cultured cells induces changes in the biophysical characteristics of the affected cells. Advanced microscopic cameras such as Ovizio's QMod, coupled with the appropriate software, can measure a variety of characteristics on a per-cell basis. We have employed this system to monitor the progression of vesicular stomatitis virus infection in Vero cells and to describe the cellular changes associated with advancing vesicular stomatitis virus infection. The measurements of cellular characteristics are operator-independent, and the goal is to establish a robust method to mathematically determine viral infection levels in a given sample. This will provide a means to measure viral titer in a faster and less subjective way than manual reading of plaque assays or tissue culture infectious dose 50 assays.


Assuntos
Estomatite Vesicular/diagnóstico , Vírus da Estomatite Vesicular Indiana/isolamento & purificação , Viroses/virologia , Animais , Linhagem Celular , Chlorocebus aethiops/virologia , Microscopia , Células Vero/virologia , Estomatite Vesicular/diagnóstico por imagem , Estomatite Vesicular/virologia , Vírus da Estomatite Vesicular Indiana/patogenicidade , Viroses/diagnóstico , Viroses/diagnóstico por imagem
5.
Nat Immunol ; 21(3): 321-330, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-32066949

RESUMO

Differentiation of CD4+ T cells into either follicular helper T (TFH) or type 1 helper T (TH1) cells influences the balance between humoral and cellular adaptive immunity, but the mechanisms whereby pathogens elicit distinct effector cells are incompletely understood. Here we analyzed the spatiotemporal dynamics of CD4+ T cells during infection with recombinant vesicular stomatitis virus (VSV), which induces early, potent neutralizing antibodies, or recombinant lymphocytic choriomeningitis virus (LCMV), which induces a vigorous cellular response but inefficient neutralizing antibodies, expressing the same T cell epitope. Early exposure of dendritic cells to type I interferon (IFN), which occurred during infection with VSV, induced production of the cytokine IL-6 and drove TFH cell polarization, whereas late exposure to type I IFN, which occurred during infection with LCMV, did not induce IL-6 and allowed differentiation into TH1 cells. Thus, tight spatiotemporal regulation of type I IFN shapes antiviral CD4+ T cell differentiation and might instruct vaccine design strategies.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Interferon Tipo I/metabolismo , Imunidade Adaptativa , Transferência Adotiva , Animais , Linfócitos T CD4-Positivos/classificação , Diferenciação Celular/imunologia , Feminino , Interleucina-6/biossíntese , Vírus da Coriomeningite Linfocítica/imunologia , Vírus da Coriomeningite Linfocítica/patogenicidade , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Análise Espaço-Temporal , Linfócitos T Auxiliares-Indutores/imunologia , Células Th1/imunologia , Vírus da Estomatite Vesicular Indiana/imunologia , Vírus da Estomatite Vesicular Indiana/patogenicidade , Vírus da Estomatite Vesicular New Jersey/imunologia , Vírus da Estomatite Vesicular New Jersey/patogenicidade
6.
Viruses ; 12(2)2020 01 22.
Artigo em Inglês | MEDLINE | ID: mdl-31979016

RESUMO

Vesicular stomatitis virus (VSV) is a zoonotic, negative-stranded RNA virus of the family Rhabdoviridae. The nucleoprotein (N) of VSV protects the viral genomic RNA and plays an essential role in viral transcription and replication, which makes the nucleoprotein an ideal target of host defense. However, whether and how host innate/intrinsic immunity limits VSV infection by targeting the N protein are unknown. In this study, we found that the N protein of VSV (VSV-N) interacted with a ubiquitin E3 ligase, tripartite motif protein 41 (TRIM41). Overexpression of TRIM41 inhibited VSV infection. Conversely, the depletion of TRIM41 increased host susceptibility to VSV. Furthermore, the E3 ligase defective mutant of TRIM41 failed to limit VSV infection, suggesting the requirement of the E3 ligase activity of TRIM41 in viral restriction. Indeed, TRIM41 ubiquitinated VSV-N in cells and in vitro. TRIM41-mediated ubiquitination leads to the degradation of VSV-N through proteasome, thereby limiting VSV infection. Taken together, our study identifies TRIM41 as a new intrinsic immune factor against VSV by targeting the viral nucleoprotein for ubiquitination and subsequent protein degradation.


Assuntos
Imunidade Inata , Proteínas do Nucleocapsídeo/química , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitinação , Vírus da Estomatite Vesicular Indiana/patogenicidade , Linhagem Celular , Células HEK293 , Humanos , Proteínas do Nucleocapsídeo/genética , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteólise , Ubiquitina-Proteína Ligases/genética , Vírus da Estomatite Vesicular Indiana/genética , Replicação Viral
7.
Sci Rep ; 9(1): 7755, 2019 05 23.
Artigo em Inglês | MEDLINE | ID: mdl-31123310

RESUMO

Crimean-Congo hemorrhagic fever virus (CCHFV), a tick-borne bunyavirus, can cause a life-threatening hemorrhagic syndrome in humans but not in its animal host. The virus is widely distributed throughout southeastern Europe, the Middle East, Africa, and Asia. Disease management has proven difficult and there are no broadly licensed vaccines or therapeutics. Recombinant vesicular stomatitis viruses (rVSV) expressing foreign glycoproteins (GP) have shown promise as experimental vaccines for several viral hemorrhagic fevers. Here, we developed and assessed a replication competent rVSV vector expressing the CCHFV glycoprotein precursor (GPC), which encodes CCHFV structural glycoproteins. This construct drives strong expression of CCHFV-GP, in vitro. Using these vectors, we vaccinated STAT-1 knock-out mice, an animal model for CCHFV. The vector was tolerated and 100% efficacious against challenge from a clinical strain of CCHFV. Anti-CCHFV-GP IgG and neutralizing antibody titers were observed in surviving animals. This study demonstrates that a rVSV expressing only the CCHFV-GP has the potential to serve as a replication competent vaccine platform against CCHF infections.


Assuntos
Glicoproteínas/imunologia , Febre Hemorrágica da Crimeia/prevenção & controle , Vírus da Estomatite Vesicular Indiana/imunologia , Animais , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Linhagem Celular , Modelos Animais de Doenças , Feminino , Vírus da Febre Hemorrágica da Crimeia-Congo/imunologia , Vírus da Febre Hemorrágica da Crimeia-Congo/patogenicidade , Febre Hemorrágica da Crimeia/imunologia , Camundongos , Camundongos Knockout , Fator de Transcrição STAT1/genética , Vacinação/métodos , Vacinas/imunologia , Vírus da Estomatite Vesicular Indiana/patogenicidade , Vesiculovirus/imunologia
8.
Nat Microbiol ; 4(6): 1006-1013, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30833734

RESUMO

Antiviral immunity has been studied extensively from the perspective of virus-cell interactions, yet the role of virus-virus interactions remains poorly addressed. Here, we demonstrate that viral escape from interferon (IFN)-based innate immunity is a social process in which IFN-stimulating viruses determine the fitness of neighbouring viruses. We propose a general and simple social evolution framework to analyse how natural selection acts on IFN shutdown and validate it in cell cultures and mice infected with vesicular stomatitis virus. Furthermore, we find that IFN shutdown is costly because it reduces short-term viral progeny production, thus fulfilling the definition of an altruistic trait. Hence, in well-mixed populations, the IFN-blocking wild-type virus is susceptible to invasion by IFN-stimulating variants and spatial structure consequently determines whether IFN shutdown can evolve. Our findings reveal that fundamental social evolution rules govern viral innate immunity evasion and virulence and suggest possible antiviral interventions.


Assuntos
Antivirais/imunologia , Evolução Biológica , Evasão da Resposta Imune , Imunidade Inata , Animais , Encéfalo/patologia , Encéfalo/virologia , RNA Polimerases Dirigidas por DNA , Modelos Animais de Doenças , Feminino , Interações Hospedeiro-Patógeno/imunologia , Interferons/farmacologia , Camundongos , Camundongos Endogâmicos BALB C , Modelos Biológicos , Vírus da Estomatite Vesicular Indiana/efeitos dos fármacos , Vírus da Estomatite Vesicular Indiana/patogenicidade , Proteínas Virais
9.
Mol Cell ; 73(4): 803-814.e6, 2019 02 21.
Artigo em Inglês | MEDLINE | ID: mdl-30639243

RESUMO

Intron retention (IR) has emerged as an important mechanism of gene expression control, but the factors controlling IR events remain poorly understood. We observed consistent IR in one intron of the Irf7 gene and identified BUD13 as an RNA-binding protein that acts at this intron to increase the amount of successful splicing. Deficiency in BUD13 was associated with increased IR, decreased mature Irf7 transcript and protein levels, and consequently a dampened type I interferon response, which compromised the ability of BUD13-deficient macrophages to withstand vesicular stomatitis virus (VSV) infection. Global analysis of BUD13 knockdown and BUD13 cross-linking to RNA revealed a subset of introns that share many characteristics with the one found in Irf7 and are spliced in a BUD13-dependent manner. Deficiency of BUD13 led to decreased mature transcript from genes containing such introns. Thus, by acting as an antagonist to IR, BUD13 facilitates the expression of genes at which IR occurs.


Assuntos
Fator Regulador 7 de Interferon/metabolismo , Interferon Tipo I/metabolismo , Íntrons , Macrófagos/metabolismo , Proteínas de Ligação a RNA/metabolismo , Estomatite Vesicular/metabolismo , Vírus da Estomatite Vesicular Indiana/patogenicidade , Animais , Sítios de Ligação , Chlorocebus aethiops , Sequência Rica em GC , Células HEK293 , Interações Hospedeiro-Patógeno , Humanos , Fator Regulador 7 de Interferon/genética , Interferon Tipo I/imunologia , Macrófagos/imunologia , Macrófagos/virologia , Camundongos Endogâmicos C57BL , Ligação Proteica , Sítios de Splice de RNA , Splicing de RNA , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteínas de Ligação a RNA/genética , Células Vero , Estomatite Vesicular/genética , Estomatite Vesicular/imunologia , Estomatite Vesicular/virologia , Vírus da Estomatite Vesicular Indiana/imunologia
10.
Nucleic Acids Res ; 47(1): 299-309, 2019 01 10.
Artigo em Inglês | MEDLINE | ID: mdl-30395342

RESUMO

The L proteins of rhabdoviruses, such as vesicular stomatitis virus (VSV) and rabies virus (RABV), possess an unconventional mRNA capping enzyme (GDP polyribonucleotidyltransferase, PRNTase) domain with a loop structure protruding into an active site cavity of the RNA-dependent RNA polymerase (RdRp) domain. Here, using complementary VSV and RABV systems, we show that the loop governs RNA synthesis and capping during the dynamic stop-start transcription cycle. A conserved tryptophan residue in the loop was identified as critical for terminal de novo initiation from the genomic promoter to synthesize the leader RNA and virus replication in host cells, but not for internal de novo initiation or elongation from the gene-start sequence for mRNA synthesis or pre-mRNA capping. The co-factor P protein was found to be essential for both terminal and internal initiation. A conserved TxΨ motif adjacent the tryptophan residue in the loop was required for pre-mRNA capping in the step of the covalent enzyme-pRNA intermediate formation, but not for either terminal or internal transcription initiation. These results provide insights into the regulation of stop-start transcription by the interplay between the RdRp active site and the dual-functional priming-capping loop of the PRNTase domain in non-segmented negative strand RNA viruses.


Assuntos
RNA Polimerases Dirigidas por DNA/química , RNA Polimerase Dependente de RNA/química , Transcrição Gênica , Vírus da Estomatite Vesicular Indiana/genética , Proteínas Virais/química , Domínio Catalítico/genética , RNA Polimerases Dirigidas por DNA/genética , Humanos , Capuzes de RNA/genética , RNA Mensageiro/genética , RNA Viral/genética , RNA Polimerase Dependente de RNA/genética , Vírus da Raiva/genética , Vírus da Raiva/patogenicidade , Rhabdoviridae/genética , Triptofano , Vírus da Estomatite Vesicular Indiana/patogenicidade , Proteínas Virais/genética , Replicação Viral/genética
11.
Vaccine ; 36(41): 6061-6069, 2018 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-30219365

RESUMO

The ability to rapidly and accurately determine viral infectivity can help improve the speed of vaccine product development and manufacturing. Current methods to determine infectious viral titers, such as the end-point dilution (50% tissue culture infective dose, TCID50) and plaque assays are slow, labor intensive, and often subjective. In order to accelerate virus quantification, Laser Force Cytology (LFC) was used to monitor vesicular stomatitis virus (VSV) infection in Vero (African green monkey kidney) cells. LFC uses a combination of optical and fluidic forces to interrogate single cells without the use of labels or antibodies. Using a combination of variables measured by the Radiance™ LFC instrument (LumaCyte), an infection metric was developed that correlates well with the viral titer as measured by TCID50 and shortens the timeframe from infection to titer determination from 3 days to 16 h (a 4.5 fold reduction). A correlation was also developed between in-process cellular measurements and the viral titer of collected supernatant, demonstrating the potential for real-time infectivity measurements. Overall, these results demonstrate the utility of LFC as a tool for rapid infectivity measurements throughout the vaccine development process.


Assuntos
Estomatite Vesicular/virologia , Vesiculovirus/isolamento & purificação , Vesiculovirus/patogenicidade , Animais , Anticorpos Antivirais/imunologia , Chlorocebus aethiops , Técnicas Citológicas , Células Vero , Vírus da Estomatite Vesicular Indiana/imunologia , Vírus da Estomatite Vesicular Indiana/isolamento & purificação , Vírus da Estomatite Vesicular Indiana/patogenicidade , Vesiculovirus/imunologia
12.
Sci Rep ; 8(1): 10669, 2018 Jul 13.
Artigo em Inglês | MEDLINE | ID: mdl-30006542

RESUMO

Viral fusion proteins are essential for enveloped virus infection. These proteins mediate fusion between the virus envelope and host cellular membrane to release the viral genome into cells. Vesicular stomatitis virus G (VSV G) protein is a typical type III viral fusion protein. To study the mechanism of VSV G protein mediated membrane fusion, we set up a cell-cell fusion system in which cells are marked by different fluorescent proteins. Taking advantage of this system, we performed real-time recording and quantitative analysis of the cell fusion mediated by VSV G. We found that the time scale required for VSV G mediated cell-cell fusion was approximately 1-2 minutes. Next, we specifically examined the function of the transmembrane (TM) region of VSV G protein in membrane fusion by replacing the TM region with those of other fusion proteins. The TM region replacements dramatically impaired VSV G protein function in the cell-cell fusion assay and diminished VSV G mediated lentivirus and recombinant VSV infection efficiency. Further experiments implied that the TM region played a role in the transition from hemi-fusion to full fusion. Several residues within the TM region were identified as important for membrane fusion. Overall, our findings unraveled the important function of the TM region in VSV G mediated viral fusion.


Assuntos
Fusão de Membrana , Glicoproteínas de Membrana/metabolismo , Estomatite Vesicular/virologia , Vírus da Estomatite Vesicular Indiana/patogenicidade , Proteínas do Envelope Viral/metabolismo , Proteínas Virais de Fusão/metabolismo , Animais , Células CHO , Membrana Celular/metabolismo , Membrana Celular/virologia , Chlorocebus aethiops , Cricetulus , Células HEK293 , Células HeLa , Humanos , Microscopia Intravital , Glicoproteínas de Membrana/genética , Microscopia Confocal , Mutação , Domínios Proteicos/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Células Vero , Vírus da Estomatite Vesicular Indiana/genética , Proteínas do Envelope Viral/genética , Proteínas Virais de Fusão/genética
13.
Vet Microbiol ; 219: 30-39, 2018 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-29778202

RESUMO

Vesicular stomatitis virus (VSV) can cause serious vesicular lesions in pigs, and the matrix (M) protein is its predominant virulence factor. Dendritic cells (DCs) act as the bridge between innate and adaptive immune responses. However, the susceptibility of porcine DCs to VSV infection and the role of M protein in modulating the function of infected DCs are still poorly defined. Thus, this study aimed to determine the ability of virulent wild-type VSV(wtVSV) and two attenuated M protein variants (VSVΔM51 and VSVMT) to induce maturation of porcine monocyte-derived DCs (MoDCs) in vitro. It was found that both wtVSV and the M protein mutant VSVs could productively replicate in porcine MoDCs. Infection with wtVSV resulted in weak proinflammatory cytokine responses and interfered with DC maturation via downregulation of the costimulatory molecule complex CD80/86. Whilst VSVΔM51 could activate porcine MoDCs, VSVMT, a highly attenuated recombinant VSV with triple mutations in the M protein, induced a potent maturation of MoDCs, as evidenced by efficient cytokine induction, and upregulation of CD80/86 and MHC class II. Overall, our findings reveal that porcine MoDCs are differentially activated by VSV, dependent on the presence of a functional M protein. M protein plays a crucial role in modulating porcine DC-VSV interactions. The data further support the potential use of VSVMT as a vaccine vector for pigs.


Assuntos
Células Dendríticas/virologia , Monócitos/virologia , Vírus da Estomatite Vesicular Indiana/genética , Proteínas da Matriz Viral/farmacologia , Animais , Moléculas de Adesão Celular/imunologia , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/imunologia , Interleucina-1beta/biossíntese , Interleucina-1beta/imunologia , Monócitos/imunologia , Monócitos/fisiologia , Proteínas Mutantes/genética , Proteínas Mutantes/imunologia , Proteínas Mutantes/farmacologia , Suínos , Estomatite Vesicular/virologia , Vírus da Estomatite Vesicular Indiana/efeitos dos fármacos , Vírus da Estomatite Vesicular Indiana/imunologia , Vírus da Estomatite Vesicular Indiana/patogenicidade , Proteínas da Matriz Viral/genética
14.
PLoS Pathog ; 13(12): e1006773, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-29281729

RESUMO

The histone demethylase LSD1 has been known as a key transcriptional coactivator for DNA viruses such as herpes virus. Inhibition of LSD1 was found to block viral genome transcription and lytic replication of DNA viruses. However, RNA virus genomes do not rely on chromatin structure and histone association, and the role of demethylase activity of LSD1 in RNA virus infections is not anticipated. Here, we identify that, contrary to its role in enhancing DNA virus replication, LSD1 limits RNA virus replication by demethylating and activating IFITM3 which is a host restriction factor for many RNA viruses. We have found that LSD1 is recruited to demethylate IFITM3 at position K88 under IFNα treatment. However, infection by either Vesicular Stomatitis Virus (VSV) or Influenza A Virus (IAV) triggers methylation of IFITM3 by promoting its disassociation from LSD1. Accordingly, inhibition of the enzymatic activity of LSD1 by Trans-2-phenylcyclopropylamine hydrochloride (TCP) increases IFITM3 monomethylation which leads to more severe disease outcomes in IAV-infected mice. In summary, our findings highlight the opposite role of LSD1 in fighting RNA viruses comparing to DNA viruses infection. Our data suggest that the demethylation of IFITM3 by LSD1 is beneficial for the host to fight against RNA virus infection.


Assuntos
Histona Desmetilases/metabolismo , Vírus da Influenza A/patogenicidade , Proteínas de Membrana/metabolismo , Proteínas de Ligação a RNA/metabolismo , Animais , Sítios de Ligação , Progressão da Doença , Inibidores Enzimáticos/farmacologia , Feminino , Células HEK293 , Histona Desmetilases/antagonistas & inibidores , Interações Hospedeiro-Patógeno , Humanos , Vírus da Influenza A/fisiologia , Proteínas de Membrana/química , Proteínas de Membrana/deficiência , Proteínas de Membrana/genética , Metilação , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Modelos Biológicos , Infecções por Orthomyxoviridae/etiologia , Infecções por Orthomyxoviridae/metabolismo , Proteínas de Ligação a RNA/química , Tranilcipromina/farmacologia , Vírus da Estomatite Vesicular Indiana/patogenicidade , Vírus da Estomatite Vesicular Indiana/fisiologia , Replicação Viral , Zika virus/patogenicidade , Zika virus/fisiologia
15.
Cell Host Microbe ; 22(1): 48-60.e5, 2017 Jul 12.
Artigo em Inglês | MEDLINE | ID: mdl-28704652

RESUMO

Asymptomatic infections often proceed undetected, yet can still prime the host to be sensitive to secondary environmental stress. While the mechanisms underlying disease caused by asymptomatic infections are unknown, it is believed that productive pathogen replication is required. We report that the environmental stress of carbon dioxide (CO2) anesthesia converts an asymptomatic rhabdovirus infection in Drosophila to one that is lethal. This lethality results from a pool of infectious virus in glial cells and is regulated by the antiviral RNAi pathway of the host. CO2 sensitivity is caused by the fusogenic activity of the viral glycoprotein, which results in fusion of neurons and glia. Expression of the viral glycoprotein, but not a fusion defective mutant, is sufficient to cause CO2 sensitivity, which can occur even in the absence of productive viral replication. These findings highlight how viral proteins, independent of pathogen replication, may predispose hosts to life-threatening environmental stress.


Assuntos
Dióxido de Carbono/farmacologia , Meio Ambiente , Neurônios/virologia , Estresse Fisiológico , Vírus da Estomatite Vesicular Indiana/efeitos dos fármacos , Vírus da Estomatite Vesicular Indiana/patogenicidade , Viroses , Animais , Animais Geneticamente Modificados , Anopheles/imunologia , Anopheles/virologia , Antivirais/farmacologia , Comportamento Animal , Drosophila melanogaster/imunologia , Drosophila melanogaster/virologia , Glicoproteínas/metabolismo , Concentração de Íons de Hidrogênio , Imunidade Inata , Neuroglia/imunologia , Neuroglia/virologia , Neurônios/imunologia , Interferência de RNA , Sindbis virus/imunologia , Sindbis virus/patogenicidade , Ensaio de Placa Viral , Proteínas Virais/metabolismo , Viroses/imunologia , Viroses/virologia , Replicação Viral/efeitos dos fármacos
16.
Sci Signal ; 10(482)2017 Jun 06.
Artigo em Inglês | MEDLINE | ID: mdl-28588082

RESUMO

The unfolded protein response (UPR) is an ancient cellular pathway that detects and alleviates protein-folding stresses. The UPR components X-box binding protein 1 (XBP1) and inositol-requiring enzyme 1α (IRE1α) promote type I interferon (IFN) responses. We found that Xbp1-deficient mouse embryonic fibroblasts and macrophages had impaired antiviral resistance. However, this was not because of a defect in type I IFN responses but rather an inability of Xbp1-deficient cells to undergo viral-induced apoptosis. The ability to undergo apoptosis limited infection in wild-type cells. Xbp1-deficient cells were generally resistant to the intrinsic pathway of apoptosis through an indirect mechanism involving activation of the nuclease IRE1α. We observed an IRE1α-dependent reduction in the abundance of the proapoptotic microRNA miR-125a and a corresponding increase in the amounts of the members of the antiapoptotic Bcl-2 family. The activation of IRE1α by the hepatitis C virus (HCV) protein NS4B in XBP1-proficient cells also conferred apoptosis resistance and promoted viral replication. Furthermore, we found evidence of IRE1α activation and decreased miR-125a abundance in liver biopsies from patients infected with HCV compared to those in the livers of healthy controls. Our results reveal a prosurvival role for IRE1α in virally infected cells and suggest a possible target for IFN-independent antiviral therapy.


Assuntos
Apoptose , Endorribonucleases/metabolismo , Hepatite C/virologia , Herpes Simples/virologia , MicroRNAs/genética , Proteínas Serina-Treonina Quinases/metabolismo , Estomatite Vesicular/virologia , Animais , Estudos de Casos e Controles , Células Cultivadas , Feminino , Hepacivirus/patogenicidade , Hepatite C/metabolismo , Hepatite C/patologia , Herpes Simples/metabolismo , Herpes Simples/patologia , Humanos , Fígado/virologia , Masculino , Camundongos , Camundongos Knockout , Simplexvirus/patogenicidade , Estomatite Vesicular/metabolismo , Estomatite Vesicular/patologia , Vírus da Estomatite Vesicular Indiana/patogenicidade , Proteínas não Estruturais Virais/metabolismo , Replicação Viral , Proteína 1 de Ligação a X-Box/fisiologia
17.
Hum Gene Ther Clin Dev ; 28(2): 108-115, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28514874

RESUMO

Vesicular stomatitis virus (VSV) is a negative-stranded RNA virus that naturally causes disease in livestock including horses, cattle and pigs. The two main identified VSV serotypes are New Jersey (VSNJV) and Indiana (VSIV). VSV is a rapidly replicating, potently immunogenic virus that has been engineered to develop novel oncolytic therapies for cancer treatment. Swine are a natural host for VSV and provide a relevant and well-established model, amenable to biological sampling to monitor virus shedding and neutralizing antibodies. Previous reports have documented the pathogenicity and transmissibility of wild-type isolates and recombinant strains of VSIV and VSNJV using the swine model. Oncolytic VSV engineered to express interferon-beta (IFNß) and the sodium iodide symporter (NIS), VSV-IFNß-NIS, has been shown to be a potent new therapeutic agent inducing rapid and durable tumor remission following systemic therapy in preclinical mouse models. VSV-IFNß-NIS is currently undergoing clinical evaluation for the treatment of advanced cancer in human and canine patients. To support clinical studies and comprehensively assess the risk of transmission to susceptible species, we tested the pathogenicity and transmissibility of oncolytic VSV-IFNß-NIS using the swine model. Following previously established protocols to evaluate VSV pathogenicity, intradermal inoculation with 107 TCID50 VSV-IFNß-NIS caused no observable symptoms in pigs. There was no detectable shedding of infectious virus in VSV-IFNß-NIS in biological excreta of inoculated pigs or exposed naive pigs kept in direct contact throughout the experiment. VSV-IFNß-NIS inoculated pigs became seropositive for VSV antibodies, while contact pigs displayed no symptoms of VSV infection, and importantly did not seroconvert. These data indicate that oncolytic VSV is both nonpathogenic and not transmissible in pigs, a natural host. These findings support further clinical development of oncolytic VSV-IFNß-NIS as a safe therapeutic for human and canine cancer.


Assuntos
Terapia Viral Oncolítica/efeitos adversos , Vírus Oncolíticos/patogenicidade , Vírus da Estomatite Vesicular Indiana/patogenicidade , Vírus da Estomatite Vesicular New Jersey/patogenicidade , Animais , Interferon gama/genética , Interferon gama/metabolismo , Vírus Oncolíticos/genética , Suínos , Simportadores/genética , Simportadores/metabolismo , Vírus da Estomatite Vesicular Indiana/genética , Vírus da Estomatite Vesicular New Jersey/genética , Eliminação de Partículas Virais
18.
Nat Commun ; 8: 14128, 2017 01 03.
Artigo em Inglês | MEDLINE | ID: mdl-28045099

RESUMO

Apoptosis is a genetically regulated cell suicide programme mediated by activation of the effector caspases 3, 6 and 7. If apoptotic cells are not scavenged, they progress to a lytic and inflammatory phase called secondary necrosis. The mechanism by which this occurs is unknown. Here we show that caspase-3 cleaves the GSDMD-related protein DFNA5 after Asp270 to generate a necrotic DFNA5-N fragment that targets the plasma membrane to induce secondary necrosis/pyroptosis. Cells that express DFNA5 progress to secondary necrosis, when stimulated with apoptotic triggers such as etoposide or vesicular stomatitis virus infection, but disassemble into small apoptotic bodies when DFNA5 is deleted. Our findings identify DFNA5 as a central molecule that regulates apoptotic cell disassembly and progression to secondary necrosis, and provide a molecular mechanism for secondary necrosis. Because DFNA5-induced secondary necrosis and GSDMD-induced pyroptosis are dependent on caspase activation, we propose that they are forms of programmed necrosis.


Assuntos
Proteínas Reguladoras de Apoptose/genética , Apoptose/genética , Caspase 3/genética , Necrose/genética , Piroptose/genética , Receptores de Estrogênio/genética , Animais , Apoptose/efeitos dos fármacos , Proteínas Reguladoras de Apoptose/metabolismo , Caspase 3/metabolismo , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Membrana Celular/virologia , Etoposídeo/farmacologia , Regulação da Expressão Gênica , Células HEK293 , Células Hep G2 , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Macrófagos/virologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Necrose/induzido quimicamente , Necrose/metabolismo , Proteínas de Ligação a Fosfato , Cultura Primária de Células , Piroptose/efeitos dos fármacos , Receptores de Estrogênio/metabolismo , Vírus da Estomatite Vesicular Indiana/crescimento & desenvolvimento , Vírus da Estomatite Vesicular Indiana/patogenicidade
19.
PLoS One ; 10(6): e0131137, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26091335

RESUMO

Three different matrix (M) proteins termed M1, M2 and M3 have been described in cells infected with vesicular stomatitis virus (VSV). Individual expression of VSV M proteins induces an evident cytopathic effect including cell rounding and detachment, in addition to a partial inhibition of cellular protein synthesis, likely mediated by an indirect mechanism. Analogous to viroporins, M1 promotes the budding of new virus particles; however, this process does not produce an increase in plasma membrane permeability. In contrast to M1, M2 and M3 neither interact with the cellular membrane nor promote the budding of double membrane vesicles at the cell surface. Nonetheless, all three species of M protein interfere with the transport of cellular mRNAs from the nucleus to the cytoplasm and also modulate the redistribution of the splicing factor. The present findings indicate that all three VSV M proteins share some activities that interfere with host cell functions.


Assuntos
Vírus da Estomatite Vesicular Indiana/fisiologia , Vírus da Estomatite Vesicular Indiana/patogenicidade , Proteínas da Matriz Viral/fisiologia , Animais , Linhagem Celular , Permeabilidade da Membrana Celular , Sistema Livre de Células , Cricetinae , Efeito Citopatogênico Viral , Ribonucleoproteínas Nucleares Heterogêneas Grupo F-H/metabolismo , Interações Hospedeiro-Patógeno/fisiologia , Microscopia Eletrônica de Transmissão , Biossíntese de Proteínas , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Viral/genética , RNA Viral/metabolismo , Vírus da Estomatite Vesicular Indiana/genética , Proteínas da Matriz Viral/genética , Liberação de Vírus
20.
Evolution ; 69(1): 117-25, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25311513

RESUMO

Life-history theory predicts that traits for survival and reproduction cannot be simultaneously maximized in evolving populations. For this reason, in obligate parasites such as infectious viruses, selection for improved between-host survival during transmission may lead to evolution of decreased within-host reproduction. We tested this idea using experimental evolution of RNA virus populations, passaged under differing transmission times in the laboratory. A single ancestral genotype of vesicular stomatitis virus (VSV), a negative-sense RNA Rhabdovirus, was used to found multiple virus lineages evolved in either ordinary 24-h cell-culture passage, or in delayed passages of 48 h. After 30 passages (120 generations of viral evolution), we observed that delayed transmission selected for improved extracellular survival, which traded-off with lowered viral fecundity (slower exponential population growth and smaller mean plaque size). To further examine the confirmed evolutionary trade-off, we obtained consensus whole-genome sequences of evolved virus populations, to infer phenotype-genotype associations. Results implied that increased virus survival did not occur via convergence; rather, improved virion stability was gained via independent mutations in various VSV structural proteins. Our study suggests that RNA viruses can evolve different molecular solutions for enhanced survival despite their limited genetic architecture, but suffer generalized reproductive trade-offs that limit overall fitness gains.


Assuntos
Evolução Molecular , Seleção Genética , Vírus da Estomatite Vesicular Indiana/genética , Animais , Linhagem Celular , Cricetinae , Aptidão Genética , Genoma Viral , Especificidade de Hospedeiro , Vírus da Estomatite Vesicular Indiana/patogenicidade , Virulência/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA