Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 179
Filtrar
1.
Cell Rep ; 43(4): 114050, 2024 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-38564336

RESUMO

Seo et al.1 shed light on virus-host interactions as they reveal how poxvirus A51R stabilizes microtubules in infected cells, which impacts vaccinia virus virulence in mice by potentially inhibiting reactive-oxygen-species-dependent antiviral responses in macrophages.


Assuntos
Microtúbulos , Vaccinia virus , Microtúbulos/metabolismo , Animais , Virulência , Vaccinia virus/patogenicidade , Vaccinia virus/fisiologia , Humanos , Camundongos , Macrófagos/virologia , Macrófagos/metabolismo , Poxviridae/patogenicidade , Poxviridae/genética , Poxviridae/fisiologia
2.
Viruses ; 13(10)2021 10 03.
Artigo em Inglês | MEDLINE | ID: mdl-34696416

RESUMO

Vaccinia virus (VACV) is an enveloped DNA virus from the Orthopoxvirus family, various strains of which were used in the successful eradication campaign against smallpox. Both original and newer VACV-based replicating vaccines reveal a risk of serious complications in atopic individuals. VACV encodes various factors interfering with host immune responses at multiple levels. In atopic skin, the production of type I interferon is compromised, while VACV specifically inhibits the phosphorylation of the Interferon Regulatory Factor 3 (IRF-3) and expression of interferons. To overcome this block, we generated a recombinant VACV-expressing murine IRF-3 (WR-IRF3) and characterized its effects on virus growth, cytokine expression and apoptosis in tissue cultures and in spontaneously atopic Nc/Nga and control Balb/c mice. Further, we explored the induction of protective immune responses against a lethal dose of wild-type WR, the surrogate of smallpox. We demonstrate that the overexpression of IRF-3 by WR-IRF3 increases the expression of type I interferon, modulates the expression of several cytokines and induces superior protective immune responses against a lethal poxvirus challenge in both Nc/Nga and Balb/c mice. Additionally, the results may be informative for design of other virus-based vaccines or for therapy of different viral infections.


Assuntos
Fator Regulador 3 de Interferon/imunologia , Infecções por Poxviridae/imunologia , Vaccinia virus/genética , Animais , Expressão Gênica/genética , Regulação Viral da Expressão Gênica/genética , Imunidade/imunologia , Fator Regulador 3 de Interferon/genética , Interferon Tipo I/metabolismo , Interleucina-1beta/imunologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Poxviridae/patogenicidade , Infecções por Poxviridae/prevenção & controle , Pele/imunologia , Vacínia/virologia , Vacinas Virais/imunologia , Replicação Viral/imunologia
3.
Virus Genes ; 56(2): 150-167, 2020 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-32076918

RESUMO

The viruses historically implicated or currently considered as candidates for misuse in bioterrorist events are poxviruses, filoviruses, bunyaviruses, orthomyxoviruses, paramyxoviruses and a number of arboviruses causing encephalitis, including alpha- and flaviviruses. All these viruses are of concern for public health services when they occur in natural outbreaks or emerge in unvaccinated populations. Recent events and intelligence reports point to a growing risk of dangerous biological agents being used for nefarious purposes. Public health responses effective in natural outbreaks of infectious disease may not be sufficient to deal with the severe consequences of a deliberate release of such agents. One important aspect of countermeasures against viral biothreat agents are the antiviral treatment options available for use in post-exposure prophylaxis. These issues were adressed by the organizers of the 16th Medical Biodefense Conference, held in Munich in 2018, in a special session on the development of drugs to treat infections with viruses currently perceived as a threat to societies or associated with a potential for misuse as biothreat agents. This review will outline the state-of-the-art methods in antivirals research discussed and provide an overview of antiviral compounds in the pipeline that are already approved for use or still under development.


Assuntos
Antivirais/uso terapêutico , Arbovírus/efeitos dos fármacos , Bioterrorismo/prevenção & controle , Viroses/tratamento farmacológico , Arbovírus/patogenicidade , Filoviridae/efeitos dos fármacos , Filoviridae/patogenicidade , Humanos , Orthobunyavirus/efeitos dos fármacos , Orthobunyavirus/patogenicidade , Orthomyxoviridae/efeitos dos fármacos , Orthomyxoviridae/patogenicidade , Paramyxovirinae/efeitos dos fármacos , Paramyxovirinae/patogenicidade , Poxviridae/efeitos dos fármacos , Poxviridae/patogenicidade , Viroses/virologia
4.
Methods Mol Biol ; 2023: 1-27, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31240668

RESUMO

Vaccinia virus, the prototype Orthopoxvirus, is widely used in the laboratory as a model system to study various aspects of viral biology and virus-host interactions, as a protein expression system, as a vaccine vector, and as an oncolytic agent. The ubiquitous use of vaccinia viruses in laboratories around the world raises certain safety concerns because the virus can be a pathogen in individuals with immunological and dermatological abnormalities, and on occasion can cause serious problems in normal hosts. This chapter reviews standard operating procedures when working with vaccinia virus and reviews published cases of accidental laboratory infections with poxviruses.


Assuntos
Infecção Laboratorial/prevenção & controle , Infecção Laboratorial/virologia , Poxviridae/patogenicidade , Vaccinia virus/patogenicidade , Humanos , Laboratórios/normas , Laboratórios/estatística & dados numéricos
5.
PLoS Pathog ; 15(2): e1007608, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30768651

RESUMO

Poxviruses employ sophisticated, but incompletely understood, signaling pathways that engage cellular defense mechanisms and simultaneously ensure viral factors are modulated properly. For example, the vaccinia B1 protein kinase plays a vital role in inactivating the cellular antiviral factor BAF, and likely orchestrates other pathways as well. In this study, we utilized experimental evolution of a B1 deletion virus to perform an unbiased search for suppressor mutations and identify novel pathways involving B1. After several passages of the ΔB1 virus we observed a robust increase in viral titer of the adapted virus. Interestingly, our characterization of the adapted viruses reveals that mutations correlating with a loss of function of the vaccinia B12 pseudokinase provide a striking fitness enhancement to this virus. In support of predictions that reductive evolution is a driver of poxvirus adaptation, this is clear experimental evidence that gene loss can be of significant benefit. Next, we present multiple lines of evidence demonstrating that expression of full length B12 leads to a fitness reduction in viruses with a defect in B1, but has no apparent impact on wild-type virus or other mutant poxviruses. From these data we infer that B12 possesses a potent inhibitory activity that can be masked by the presence of the B1 kinase. Further investigation of B12 attributes revealed that it primarily localizes to the nucleus, a characteristic only rarely found among poxviral proteins. Surprisingly, BAF phosphorylation is reduced under conditions in which B12 is present in infected cells without B1, indicating that B12 may function in part by enhancing antiviral activity of BAF. Together, our studies of B1 and B12 present novel evidence that a paralogous kinase-pseudokinase pair can exhibit a unique epistatic relationship in a virus, perhaps serving to enhance B1 conservation during poxvirus evolution and to orchestrate yet-to-be-discovered nuclear events during infection.


Assuntos
Vaccinia virus/metabolismo , Proteínas Virais/metabolismo , Replicação Viral/fisiologia , Células A549 , Animais , Linhagem Celular , Núcleo Celular , Chlorocebus aethiops , Replicação do DNA , DNA Viral/metabolismo , Proteínas de Ligação a DNA , Células HeLa , Interações Hospedeiro-Patógeno , Humanos , Camundongos , Proteínas Nucleares , Fosforilação , Poxviridae/metabolismo , Poxviridae/patogenicidade , Proteínas Quinases , Transdução de Sinais , Fatores de Transcrição , Vacínia , Vaccinia virus/patogenicidade , Proteínas Virais/genética , Proteínas Virais/fisiologia , Replicação Viral/genética
6.
PLoS Biol ; 17(1): e3000124, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30699104

RESUMO

The eradication of smallpox is one of the greatest medical successes in history. Vaccinia virus was made famous by being the virus used in the live vaccine that enabled this feat. Nearly 40 years on from that success, this prototypical poxvirus continues to empower the exploration of fundamental biology and the potential to develop therapeutics against some of the major causes of death and disease in the modern world.


Assuntos
Vacinação/métodos , Vacinação/tendências , Vaccinia virus/patogenicidade , Animais , Humanos , Poxviridae/patogenicidade , Infecções por Poxviridae/metabolismo , Pesquisa/tendências , Varíola/virologia , Vacinas Atenuadas , Vaccinia virus/metabolismo
7.
Nat Commun ; 9(1): 5440, 2018 12 21.
Artigo em Inglês | MEDLINE | ID: mdl-30575728

RESUMO

Soluble cytokine decoy receptors are potent immune modulatory reagents with therapeutic applications. Some virus-encoded secreted cytokine receptors interact with glycosaminoglycans expressed at the cell surface, but the biological significance of this activity in vivo is poorly understood. Here, we show the type I interferon binding protein (IFNα/ßBP) encoded by vaccinia and ectromelia viruses requires of this cell binding activity to confer full virulence to these viruses and to retain immunomodulatory activity. Expression of a variant form of the IFNα/ßBP that inhibits IFN activity, but does not interact with cell surface glycosaminoglycans, results in highly attenuated viruses with a virulence similar to that of the IFNα/ßBP deletion mutant viruses. Transcriptomics analysis and infection of IFN receptor-deficient mice confirmed that the control of IFN activity is the main function of the IFNα/ßBP in vivo. We propose that retention of secreted cytokine receptors at the cell surface may largely enhance their immunomodulatory activity.


Assuntos
Glicosaminoglicanos/metabolismo , Interferon Tipo I/metabolismo , Infecções por Poxviridae/imunologia , Poxviridae/patogenicidade , Proteínas Virais/metabolismo , Animais , Chlorocebus aethiops , Feminino , Células HeLa , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Poxviridae/imunologia , Poxviridae/metabolismo , Células Vero , Ligação Viral
8.
Nat Commun ; 9(1): 1790, 2018 05 03.
Artigo em Inglês | MEDLINE | ID: mdl-29724993

RESUMO

The role of cytokines and chemokines in anti-viral defense has been demonstrated, but their relative contribution to protective anti-viral responses in vivo is not fully understood. Cytokine response modifier D (CrmD) is a secreted receptor for TNF and lymphotoxin containing the smallpox virus-encoded chemokine receptor (SECRET) domain and is expressed by ectromelia virus, the causative agent of the smallpox-like disease mousepox. Here we show that CrmD is an essential virulence factor that controls natural killer cell activation and allows progression of fatal mousepox, and demonstrate that both SECRET and TNF binding domains are required for full CrmD activity. Vaccination with recombinant CrmD protects animals from lethal mousepox. These results indicate that a specific set of chemokines enhance the inflammatory and protective anti-viral responses mediated by TNF and lymphotoxin, and illustrate how viruses optimize anti-TNF strategies with the addition of a chemokine binding domain as soluble decoy receptors.


Assuntos
Quimiocinas/fisiologia , Ectromelia Infecciosa/imunologia , Ectromelia Infecciosa/prevenção & controle , Inflamação/etiologia , Fator de Necrose Tumoral alfa/fisiologia , Animais , Linfócitos T CD8-Positivos/imunologia , Linhagem Celular , Feminino , Inflamação/imunologia , Células Matadoras Naturais/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Poxviridae/patogenicidade , Fatores de Virulência/fisiologia , Replicação Viral
9.
J Virol ; 92(10)2018 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-29491158

RESUMO

Cytosolic recognition of DNA has emerged as a critical cellular mechanism of host immune activation upon pathogen invasion. The central cytosolic DNA sensor cGAS activates STING, which is phosphorylated, dimerizes and translocates from the endoplasmic reticulum (ER) to a perinuclear region to mediate IRF-3 activation. Poxviruses are double-stranded DNA viruses replicating in the cytosol and hence likely to trigger cytosolic DNA sensing. Here, we investigated the activation of innate immune signaling by 4 different strains of the prototypic poxvirus vaccinia virus (VACV) in a cell line proficient in DNA sensing. Infection with the attenuated VACV strain MVA activated IRF-3 via cGAS and STING, and accordingly STING dimerized and was phosphorylated during MVA infection. Conversely, VACV strains Copenhagen and Western Reserve inhibited STING dimerization and phosphorylation during infection and in response to transfected DNA and cyclic GMP-AMP, thus efficiently suppressing DNA sensing and IRF-3 activation. A VACV deletion mutant lacking protein C16, thought to be the only viral DNA sensing inhibitor acting upstream of STING, retained the ability to block STING activation. Similar inhibition of DNA-induced STING activation was also observed for cowpox and ectromelia viruses. Our data demonstrate that virulent poxviruses possess mechanisms for targeting DNA sensing at the level of the cGAS-STING axis and that these mechanisms do not operate in replication-defective strains such as MVA. These findings shed light on the role of cellular DNA sensing in poxvirus-host interactions and will open new avenues to determine its impact on VACV immunogenicity and virulence.IMPORTANCE Poxviruses are double-stranded DNA viruses infecting a wide range of vertebrates and include the causative agent of smallpox (variola virus) and its vaccine vaccinia virus (VACV). Despite smallpox eradication VACV remains of interest as a therapeutic. Attenuated strains are popular vaccine candidates, whereas replication-competent strains are emerging as efficient oncolytics in virotherapy. The successful therapeutic use of VACV depends on a detailed understanding of its ability to modulate host innate immune responses. DNA sensing is a critical cellular mechanism for pathogen detection and activation of innate immunity that is centrally coordinated by the endoplasmic reticulum-resident protein STING. Here, STING is shown to mediate immune activation in response to MVA, but not in response to virulent VACV strains or other virulent poxviruses, which prevent STING activation and DNA sensing during infection and after DNA transfection. These results provide new insights into poxvirus immune evasion and have implications in the rational design of VACV-based therapeutics.


Assuntos
Proteínas de Membrana/química , Proteínas de Membrana/metabolismo , Infecções por Poxviridae/metabolismo , Poxviridae/fisiologia , Linhagem Celular , Citosol/metabolismo , Citosol/virologia , Células HEK293 , Humanos , Fosforilação , Poxviridae/patogenicidade , Infecções por Poxviridae/virologia , Multimerização Proteica , Células THP-1 , Virulência , Replicação Viral
10.
PLoS Pathog ; 14(2): e1006884, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29447249

RESUMO

Host restriction factors constitute a formidable barrier for viral replication to which many viruses have evolved counter-measures. Human SAMD9, a tumor suppressor and a restriction factor for poxviruses in cell lines, is antagonized by two classes of poxvirus proteins, represented by vaccinia virus (VACV) K1 and C7. A paralog of SAMD9, SAMD9L, is also encoded by some mammals, while only one of two paralogs is retained by others. Here, we show that SAMD9L functions similarly to SAMD9 as a restriction factor and that the two paralogs form a critical host barrier that poxviruses must overcome to establish infection. In mice, which naturally lack SAMD9, overcoming SAMD9L restriction with viral inhibitors is essential for poxvirus replication and pathogenesis. While a VACV deleted of both K1 and C7 (vK1L-C7L-) was restricted by mouse cells and highly attenuated in mice, its replication and virulence were completely restored in SAMD9L-/- mice. In humans, both SAMD9 and SAMD9L are poxvirus restriction factors, although the latter requires interferon induction in many cell types. While knockout of SAMD9 with Crispr-Cas9 was sufficient for abolishing the restriction for vK1L-C7L- in many human cells, knockout of both paralogs was required for abolishing the restriction in interferon-treated cells. Both paralogs are antagonized by VACV K1, C7 and C7 homologs from diverse mammalian poxviruses, but mouse SAMD9L is resistant to the C7 homolog encoded by a group of poxviruses with a narrow host range in ruminants, indicating that host species-specific difference in SAMD9/SAMD9L genes serves as a barrier for cross-species poxvirus transmission.


Assuntos
Especificidade de Hospedeiro/genética , Infecções por Poxviridae/genética , Poxviridae/genética , Poxviridae/patogenicidade , Proteínas/fisiologia , Proteínas Supressoras de Tumor/fisiologia , Animais , Células Cultivadas , Chlorocebus aethiops , Células HEK293 , Células HeLa , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Mamíferos , Camundongos , Camundongos Knockout , Células NIH 3T3 , Infecções por Poxviridae/transmissão , Infecções por Poxviridae/virologia , Proteínas/genética , Homologia de Sequência , Proteínas Supressoras de Tumor/genética , Vaccinia virus/genética , Vaccinia virus/patogenicidade , Células Vero
11.
Viruses ; 9(8)2017 08 08.
Artigo em Inglês | MEDLINE | ID: mdl-28786952

RESUMO

Cells have multiple means to induce apoptosis in response to viral infection. Poxviruses must prevent activation of cellular apoptosis to ensure successful replication. These viruses devote a substantial portion of their genome to immune evasion. Many of these immune evasion products expressed during infection antagonize cellular apoptotic pathways. Poxvirus products target multiple points in both the extrinsic and intrinsic apoptotic pathways, thereby mitigating apoptosis during infection. Interestingly, recent evidence indicates that poxviruses also hijack cellular means of eliminating apoptotic bodies as a means to spread cell to cell through a process called apoptotic mimicry. Poxviruses are the causative agent of many human and veterinary diseases. Further, there is substantial interest in developing these viruses as vectors for a variety of uses including vaccine delivery and as oncolytic viruses to treat certain human cancers. Therefore, an understanding of the molecular mechanisms through which poxviruses regulate the cellular apoptotic pathways remains a top research priority. In this review, we consider anti-apoptotic strategies of poxviruses focusing on three relevant poxvirus genera: Orthopoxvirus, Molluscipoxvirus, and Leporipoxvirus. All three genera express multiple products to inhibit both extrinsic and intrinsic apoptotic pathways with many of these products required for virulence.


Assuntos
Apoptose , Interações Hospedeiro-Patógeno , Evasão da Resposta Imune , Infecções por Poxviridae/virologia , Poxviridae/fisiologia , Animais , Caspases/metabolismo , Humanos , Leporipoxvirus/patogenicidade , Leporipoxvirus/fisiologia , Molluscipoxvirus/patogenicidade , Molluscipoxvirus/fisiologia , Orthopoxvirus/patogenicidade , Orthopoxvirus/fisiologia , Poxviridae/genética , Poxviridae/patogenicidade , Infecções por Poxviridae/imunologia , Infecções por Poxviridae/fisiopatologia , Transdução de Sinais , Proteínas Virais/metabolismo , Virulência , Replicação Viral
12.
Methods Mol Biol ; 1581: 121-129, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28374246

RESUMO

Poxviruses cause many diseases in humans and animals worldwide, and there is a need for vaccines with improved safety and good efficacy. In addition, poxvirus vectors are widely used as recombinant vaccines for various infectious diseases and as recombinant and oncolytic vaccines for cancer. One concern with poxvirus vaccine vectors is that some poxviruses can infect a developing fetus and cause fetal loss or congenital disease. This can be an issue both for patients receiving a vaccine and for pregnant health care providers, including doctors, nurses, and veterinarians, who might receive accidental exposure to the poxvirus by injection or during patient care. We describe here a method for analyzing the safety of virus exposure in pregnant mammals using a mouse model testing vaccinia, canarypox, and raccoonpox virus vectors.


Assuntos
Infecções por Poxviridae/diagnóstico , Poxviridae/patogenicidade , Animais , Chlorocebus aethiops , Feminino , Camundongos , Orthopoxvirus/genética , Orthopoxvirus/patogenicidade , Orthopoxvirus/fisiologia , Poxviridae/genética , Poxviridae/fisiologia , Gravidez , Vacínia/diagnóstico , Células Vero , Carga Viral , Vacinas Virais/genética , Replicação Viral
13.
PLoS One ; 12(3): e0173697, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28282449

RESUMO

It is often not possible to demonstrate causality within the context of gut microbiota dysbiosis-linked diseases. Thus, we need a better understanding of the mechanisms whereby an altered host immunophysiology shapes its resident microbiota. In this regard, immune-modulating poxvirus strains and mutants could differentially alter gut mucosal immunity in the context of a natural immune response, providing a controlled natural in vivo setting to deepen our understanding of the immune determinants of microbiome composition. This study represents a proof-of-concept that the use of an existing collection of different immune-modulating poxviruses may represent an innovative tool in gut microbiome research. To this end, 16S rRNA amplicon sequencing and RNAseq transcriptome profiling were employed as proxies for microbiota composition and gut immunophysiological status in the analysis of caecal samples from control mice and mice infected with various poxvirus types. Our results show that different poxvirus species and mutants elicit different shifts in the mice mucosa-associated microbiota and, in some instances, significant concomitant shifts in gut transcriptome profiles, thus providing an initial validation to the proposed model.


Assuntos
Microbioma Gastrointestinal/fisiologia , Infecções por Poxviridae/imunologia , Poxviridae/patogenicidade , Animais , Vírus da Ectromelia/genética , Vírus da Ectromelia/patogenicidade , Feminino , Microbioma Gastrointestinal/imunologia , Interações Hospedeiro-Patógeno/imunologia , Camundongos Endogâmicos BALB C , Mutação , Poxviridae/genética , Poxviridae/imunologia , Infecções por Poxviridae/microbiologia , Infecções por Poxviridae/fisiopatologia , RNA Ribossômico 16S , Vaccinia virus/genética , Vaccinia virus/patogenicidade
14.
Virus Genes ; 53(2): 173-178, 2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-28000080

RESUMO

Proteins harbor domains or short linear motifs, which facilitate their functions and interactions. Finding functional motifs in protein sequences could predict the putative cellular roles or characteristics of hypothetical proteins. In this study, we present Shetti-Motif, which is an interactive tool to (i) map UniProt and PROSITE flat files, (ii) search for multiple pre-defined consensus patterns or experimentally validated functional motifs in large datasets protein sequences (proteome-wide), (iii) search for motifs containing repeated residues (low-complexity regions, e.g., Leu-, SR-, PEST-rich motifs, etc.). As proof of principle, using this comparative proteomics pipeline, eleven proteomes encoded by member of Poxviridae family were searched against about 100 experimentally validated functional motifs. The closely related viruses and viruses infect the same host cells (e.g. vaccinia and variola viruses) show similar motif-containing proteins profile. The motifs encoded by these viruses are correlated, which explains why poxviruses are able to interact with wide range of host cells. In conclusion, this in silico analysis is useful to establish a dataset(s) or potential proteins for further investigation or compare between species.


Assuntos
Motivos de Aminoácidos/genética , Biologia Computacional , Poxviridae/genética , Proteoma/genética , Sequência de Aminoácidos/genética , Interações Hospedeiro-Patógeno/genética , Poxviridae/patogenicidade , Domínios Proteicos/genética
17.
PLoS Pathog ; 10(5): e1004123, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24832205

RESUMO

Infections with monkeypox, cowpox and weaponized variola virus remain a threat to the increasingly unvaccinated human population, but little is known about their mechanisms of virulence and immune evasion. We now demonstrate that B22 proteins, encoded by the largest genes of these viruses, render human T cells unresponsive to stimulation of the T cell receptor by MHC-dependent antigen presentation or by MHC-independent stimulation. In contrast, stimuli that bypass TCR-signaling are not inhibited. In a non-human primate model of monkeypox, virus lacking the B22R homologue (MPXVΔ197) caused only mild disease with lower viremia and cutaneous pox lesions compared to wild type MPXV which caused high viremia, morbidity and mortality. Since MPXVΔ197-infected animals displayed accelerated T cell responses and less T cell dysregulation than MPXV US2003, we conclude that B22 family proteins cause viral virulence by suppressing T cell control of viral dissemination.


Assuntos
Evasão da Resposta Imune , Infecções por Poxviridae/imunologia , Poxviridae/patogenicidade , Linfócitos T/imunologia , Linfócitos T/virologia , Proteínas Virais/fisiologia , Animais , Células CHO , Células Cultivadas , Chlorocebus aethiops , Cricetinae , Cricetulus , Feminino , Células HEK293 , Humanos , Evasão da Resposta Imune/genética , Células Jurkat , Macaca mulatta , Camundongos , Camundongos Endogâmicos BALB C , Mpox/imunologia , Poxviridae/genética , Poxviridae/imunologia
18.
Infect Genet Evol ; 21: 15-40, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24161410

RESUMO

Poxviruses as a group can infect a large number of animals. However, at the level of individual viruses, even closely related poxviruses display highly diverse host ranges and virulence. For example, variola virus, the causative agent of smallpox, is human-specific and highly virulent only to humans, whereas related cowpox viruses naturally infect a broad spectrum of animals and only cause relatively mild disease in humans. The successful replication of poxviruses depends on their effective manipulation of the host antiviral responses, at the cellular-, tissue- and species-specific levels, which constitutes a molecular basis for differences in poxvirus host range and virulence. A number of poxvirus genes have been identified that possess host range function in experimental settings, and many of these host range genes target specific antiviral host pathways. Herein, we review the biology of poxviruses with a focus on host range, zoonotic infections, virulence, genomics and host range genes as well as the current knowledge about the function of poxvirus host range factors and how their interaction with the host innate immune system contributes to poxvirus host range and virulence. We further discuss the evolution of host range and virulence in poxviruses as well as host switches and potential poxvirus threats for human and animal health.


Assuntos
Evolução Biológica , Poxviridae/genética , Poxviridae/patogenicidade , Proteínas Virais/genética , Animais , Especificidade de Hospedeiro , Interações Hospedeiro-Patógeno , Humanos , Filogenia , Poxviridae/fisiologia , Infecções por Poxviridae/virologia , Proteínas Virais/metabolismo , Tropismo Viral , Virulência , Zoonoses/virologia
20.
Cell Host Microbe ; 13(2): 169-80, 2013 Feb 13.
Artigo em Inglês | MEDLINE | ID: mdl-23414757

RESUMO

Neutrophils mediate bacterial clearance through various mechanisms, including the release of mesh-like DNA structures or neutrophil extracellular traps (NETs) that capture bacteria. Although neutrophils are also recruited to sites of viral infection, their role in antiviral innate immunity is less clear. We show that systemic administration of virus analogs or poxvirus infection induces neutrophil recruitment to the liver microvasculature and the release of NETs that protect host cells from virus infection. After systemic intravenous poxvirus challenge, mice exhibit thrombocytopenia and the recruitment of both neutrophils and platelets to the liver vasculature. Circulating platelets interact with, roll along, and adhere to the surface of adherent neutrophils, forming large, dynamic aggregates. These interactions facilitate the release of NETs within the liver vasculature that are able to protect host cells from poxvirus infection. These findings highlight the role of NETs and early tissue-wide responses in preventing viral infection.


Assuntos
Neutrófilos/imunologia , Neutrófilos/virologia , Infecções por Poxviridae/imunologia , Poxviridae/patogenicidade , Animais , Plaquetas/metabolismo , Plaquetas/virologia , Antígeno CD11b/imunologia , Adesão Celular , Comunicação Celular , Citometria de Fluxo , Interações Hospedeiro-Patógeno , Células de Kupffer/imunologia , Lipopolissacarídeos/administração & dosagem , Lipopolissacarídeos/imunologia , Fígado/imunologia , Fígado/patologia , Fígado/virologia , Camundongos , Camundongos Endogâmicos C57BL , Neutrófilos/metabolismo , Poxviridae/imunologia , Infecções por Poxviridae/patologia , Infecções por Poxviridae/virologia , Trombocitopenia/patologia , Trombocitopenia/virologia , Receptor 3 Toll-Like/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...