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








Base de dados
Intervalo de ano de publicação
1.
Nat Commun ; 15(1): 6778, 2024 Aug 08.
Artigo em Inglês | MEDLINE | ID: mdl-39117661

RESUMO

Multiple omics analyzes of Vaccinia virus (VACV) infection have defined molecular characteristics of poxvirus biology. However, little is known about the monkeypox (mpox) virus (MPXV) in humans, which has a different disease manifestation despite its high sequence similarity to VACV. Here, we perform an in-depth multi-omics analysis of the transcriptome, proteome, and phosphoproteome signatures of MPXV-infected primary human fibroblasts to gain insights into the virus-host interplay. In addition to expected perturbations of immune-related pathways, we uncover regulation of the HIPPO and TGF-ß pathways. We identify dynamic phosphorylation of both host and viral proteins, which suggests that MAPKs are key regulators of differential phosphorylation in MPXV-infected cells. Among the viral proteins, we find dynamic phosphorylation of H5 that influenced the binding of H5 to dsDNA. Our extensive dataset highlights signaling events and hotspots perturbed by MPXV, extending the current knowledge on poxviruses. We use integrated pathway analysis and drug-target prediction approaches to identify potential drug targets that affect virus growth. Functionally, we exemplify the utility of this approach by identifying inhibitors of MTOR, CHUK/IKBKB, and splicing factor kinases with potent antiviral efficacy against MPXV and VACV.


Assuntos
Fibroblastos , Monkeypox virus , Mpox , Proteínas Virais , Humanos , Monkeypox virus/genética , Fosforilação , Mpox/virologia , Mpox/metabolismo , Fibroblastos/virologia , Fibroblastos/metabolismo , Proteínas Virais/metabolismo , Proteínas Virais/genética , Proteoma/metabolismo , Interações Hospedeiro-Patógeno , Transdução de Sinais , Proteômica/métodos , Transcriptoma , Antivirais/farmacologia , Multiômica
2.
Cell Host Microbe ; 31(6): 928-936.e4, 2023 06 14.
Artigo em Inglês | MEDLINE | ID: mdl-37236191

RESUMO

Mpox represents a persistent health concern with varying disease severity. Reinfections with mpox virus (MPXV) are rare, possibly indicating effective memory responses to MPXV or related poxviruses, notably vaccinia virus (VACV) from smallpox vaccination. We assessed cross-reactive and virus-specific CD4+ and CD8+ T cells in healthy individuals and mpox convalescent donors. Cross-reactive T cells were most frequently observed in healthy donors over 45 years. Notably, long-lived memory CD8+ T cells targeting conserved VACV/MPXV epitopes were identified in older individuals more than four decades after VACV exposure and exhibited stem-like characteristics, defined by T cell factor-1 (TCF-1) expression. In mpox convalescent donors, MPXV-reactive CD4+ and CD8+ T cells were more prevalent than in controls, demonstrating enhanced functionality and skewing toward effector phenotypes, which correlated with milder disease. Collectively, we report robust effector memory MPXV-specific T cell responses in mild mpox and long-lived TCF-1+ VACV/MPXV-specific CD8+ T cells decades after smallpox vaccination.


Assuntos
Mpox , Poxviridae , Varíola , Humanos , Linfócitos T CD8-Positivos , Mpox/metabolismo , Varíola/metabolismo , Vaccinia virus
3.
FASEB J ; 37(5): e22902, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-37014316

RESUMO

The monkeypox epidemic has attracted global attention to poxviruses. The cytoplasmic replication of poxviruses requires extensive protein synthesis, challenging the capacity of the endoplasmic reticulum (ER). However, the role of the ER in the life cycle of poxviruses is unclear. In this study, we demonstrate that infection with the lumpy skin disease virus (LSDV), a member of the poxvirus family, causes ER stress in vivo and in vitro, further facilitating the activation of the unfolded protein response (UPR). Although UPR activation aids in the restoration of the cellular environment, its significance in the LSDV life cycle remains unclear. Furthermore, the significance of ER imbalance for viral replication is also unknown. We show that LSDV replication is hampered by an unbalanced ER environment. In addition, we verify that the LSDV replication depends on the activation of PERK-eIF2α and IRE1-XBP1 signaling cascades rather than ATF6, implying that global translation and reduced XBP1 cleavage are deleterious to LSDV replication. Taken together, these findings indicate that LSDV is involved in the repression of global translational signaling, ER chaperone transcription, and ATF6 cleavage from the Golgi into the nucleus, thereby maintaining cell homeostasis; moreover, PERK and IRE1 activation contribute to LSDV replication. Our findings suggest that targeting UPR elements may be applied in response to infection from LSDV or even other poxviruses, such as monkeypox.


Assuntos
Vírus da Doença Nodular Cutânea , Mpox , Animais , Bovinos , Humanos , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Vírus da Doença Nodular Cutânea/metabolismo , Mpox/metabolismo , Transdução de Sinais , Resposta a Proteínas não Dobradas , Estresse do Retículo Endoplasmático/fisiologia , Retículo Endoplasmático/metabolismo , Fator 6 Ativador da Transcrição/metabolismo
4.
Nat Microbiol ; 5(7): 955-965, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32341480

RESUMO

Monkeypox is a viral zoonotic disease on the rise across endemic habitats. Despite the growing importance of monkeypox virus, our knowledge on its host spectrum and sylvatic maintenance is limited. Here, we describe the recent repeated emergence of monkeypox virus in a wild, human-habituated western chimpanzee (Pan troglodytes verus, hereafter chimpanzee) population from Taï National Park, Ivory Coast. Through daily monitoring, we show that further to causing its typical exanthematous syndrome, monkeypox can present itself as a severe respiratory disease without a diffuse rash. By analysing 949 non-invasively collected samples, we identify the circulation of at least two distinct monkeypox virus lineages and document the shedding of infectious particles in faeces and flies, suggesting that they could mediate indirect transmission. We also show that the carnivorous component of the Taï chimpanzees' diet, mainly consisting of the sympatric monkeys they regularly hunt, did not change nor shift towards rodent consumption (the presumed reservoir) before the outbreaks, suggesting that the sudden emergence of monkeypox virus in this population is probably due to changes in the ecology of the virus itself. Using long-term mortality surveillance data from Taï National Park, we provide evidence of little to no prior viral activity over at least two decades. We conclude that great ape sentinel systems devoted to the longitudinal collection of behavioural and health data can help clarify the epidemiology and clinical presentation of zoonotic pathogens.


Assuntos
Animais Selvagens , Monkeypox virus/fisiologia , Mpox/virologia , Pan troglodytes/virologia , Animais , Ecossistema , Exantema/etiologia , Exantema/metabolismo , Exantema/patologia , Espaço Extracelular/metabolismo , Fezes/virologia , Genoma Viral , Genômica/métodos , Glutationa/metabolismo , Sequenciamento de Nucleotídeos em Larga Escala , Mpox/complicações , Mpox/metabolismo , Mpox/mortalidade , Monkeypox virus/classificação , Monkeypox virus/isolamento & purificação , Pan troglodytes/metabolismo , Filogenia , Transtornos Respiratórios/etiologia , Transtornos Respiratórios/metabolismo
5.
Virology ; 475: 129-38, 2015 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-25462353

RESUMO

Monkeypox virus (MPXV) is an emerging pathogen from Africa that causes disease similar to smallpox. Two clades with different geographic distributions and virulence have been described. Here, we utilized bioinformatic tools to identify genomic regions in MPXV containing multiple virulence genes and explored their roles in pathogenicity; two selected regions were then deleted singularly or in combination. In vitro and in vivo studies indicated that these regions play a significant role in MPXV replication, tissue spread, and mortality in mice. Interestingly, while deletion of either region led to decreased virulence in mice, one region had no effect on in vitro replication. Deletion of both regions simultaneously also reduced cell culture replication and significantly increased the attenuation in vivo over either single deletion. Attenuated MPXV with genomic deletions present a safe and efficacious tool in the study of MPX pathogenesis and in the identification of genetic factors associated with virulence.


Assuntos
Monkeypox virus/classificação , Mpox/virologia , Animais , Anticorpos Antivirais , Linhagem Celular , Feminino , Regulação Viral da Expressão Gênica , Humanos , Camundongos , Camundongos Endogâmicos , Mpox/imunologia , Mpox/metabolismo , Monkeypox virus/genética , Monkeypox virus/patogenicidade , Mutação , Reação em Cadeia da Polimerase em Tempo Real , Vírus Reordenados , Ensaio de Placa Viral , Virulência , Replicação Viral/genética , Replicação Viral/fisiologia
6.
Mol Cell Proteomics ; 11(6): M111.015701, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22205724

RESUMO

Monkeypox virus (MPXV) is comprised of two clades: Congo Basin MPXV, with an associated case fatality rate of 10%, and Western African MPXV, which is associated with less severe infection and minimal lethality. We thus postulated that Congo Basin and West African MPXV would differentially modulate host cell responses and, as many host responses are regulated through phosphorylation independent of transcription or translation, we employed systems kinomics with peptide arrays to investigate these functional host responses. Using this approach we have demonstrated that Congo Basin MPXV infection selectively down-regulates host responses as compared with West African MPXV, including growth factor- and apoptosis-related responses. These results were confirmed using fluorescence-activated cell sorting analysis demonstrating that West African MPXV infection resulted in a significant increase in apoptosis in human monocytes as compared with Congo Basin MPXV. Further, differentially phosphorylated kinases were identified through comparison of our MPXV data sets and validated as potential targets for pharmacological inhibition of Congo Basin MPXV infection, including increased Akt S473 phosphorylation and decreased p53 S15 phosphorylation. Inhibition of Akt S473 phosphorylation resulted in a significant decrease in Congo Basin MPXV virus yield (261-fold) but did not affect West African MPXV. In addition, treatment with staurosporine, an apoptosis activator resulted in a 49-fold greater decrease in Congo Basin MPXV yields as compared with West African MPXV. Thus, using a systems kinomics approach, our investigation demonstrates that West African and Congo Basin MPXV differentially modulate host cell responses and has identified potential host targets of therapeutic interest.


Assuntos
Monkeypox virus/fisiologia , Mpox/metabolismo , Fosfoproteínas/metabolismo , Proteínas Quinases/metabolismo , Proteoma/metabolismo , Animais , Apoptose , Linhagem Celular , Chlorocebus aethiops , Análise por Conglomerados , Interações Hospedeiro-Patógeno , Humanos , Imidazóis/farmacologia , Monócitos/enzimologia , Monócitos/metabolismo , Monócitos/virologia , Fosforilação , Inibidores de Proteínas Quinases/farmacologia , Processamento de Proteína Pós-Traducional , Proteínas Proto-Oncogênicas c-met/metabolismo , Piridinas/farmacologia , Transdução de Sinais , Replicação Viral/efeitos dos fármacos
7.
Mol Cell Proteomics ; 9(12): 2760-71, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20736407

RESUMO

Understanding viral pathogenesis is challenging because of confounding factors, including nonabrasive access to infected tissues and high abundance of inflammatory mediators that may mask mechanistic details. In diseases such as influenza and smallpox where the primary cause of mortality results from complications in the lung, the characterization of lung fluid offers a unique opportunity to study host-pathogen interactions with minimal effect on infected animals. This investigation characterizes the global proteome response in the pulmonary fluid, bronchoalveolar lavage fluid, of macaques during upper respiratory infection by monkeypox virus (MPXV), a close relative of the causative agent of smallpox, variola virus. These results are compared and contrasted against infections by vaccinia virus (VV), a low pathogenic relative of MPXV, and with extracellular fluid from MPXV-infected HeLa cells. To identify changes in the pulmonary protein compartment, macaque lung fluid was sampled twice prior to infection, serving as base line, and up to six times following intrabronchial infection with either MPXV or VV. Increased expression of inflammatory proteins was observed in response to both viruses. Although the increased expression resolved for a subset of proteins, such as C-reactive protein, S100A8, and S100A9, high expression levels persisted for other proteins, including vitamin D-binding protein and fibrinogen γ. Structural and metabolic proteins were substantially decreased in lung fluid exclusively during MPXV and not VV infection. Decreases in structural and metabolic proteins were similarly observed in the extracellular fluid of MPXV-infected HeLa cells. Results from this study suggest that the host inflammatory response may not be the only facilitator of viral pathogenesis, but rather maintaining pulmonary structural integrity could be a key factor influencing disease progression and mortality.


Assuntos
Líquidos Corporais/metabolismo , Pulmão/metabolismo , Mpox/metabolismo , Proteoma , Proteínas Virais/metabolismo , Sequência de Aminoácidos , Animais , Células HeLa , Humanos , Macaca , Dados de Sequência Molecular , Mpox/imunologia , Reação em Cadeia da Polimerase , Espectrometria de Massas em Tandem , Proteínas Virais/química
8.
J Cutan Pathol ; 32(1): 28-34, 2005 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15660652

RESUMO

BACKGROUND: Human monkeypox, an emerging viral zoonosis first recognized in Africa, has recently emerged in the mid-western US. Initially, it presents with skin eruptions and fevers with diaphoresis and rigors. Clinically, the skin lesions progress from papules to vesiculopustules to resolving eschars. METHODS: Three cutaneous biopsy specimens from two patients with polymerase chain reaction (PCR)-proven monkeypox were available for review. The histologic, immunohistochemical and electron-microscopic features were identified. RESULTS: The clinical progression of lesions is mirrored histologically with ballooning degeneration of basal keratinocytes and spongiosis of a mildly acanthotic epidermis progressing to full thickness necrosis of a markedly acanthotic epidermis containing few viable keratinocytes. A lichenoid-mixed inflammatory cell infiltrate is present, which exhibits progressive exocytosis with the keratinocyte necrosis. Inflammation of the superficial and deep vascular plexes, eccrine units and follicles is also present. Viral cytopathic effect is manifest by multinucleated syncytial keratinocytes. Immunohistochemically, viral antigen is detected within keratinocytes of the lesional epidermis, follicular and eccrine epithelium and few dermal mononuclear cells. Electron microscopy reveals virions at various stages of assembly within the keratinocyte cytoplasm. CONCLUSIONS: The histologic differential diagnosis includes herpes simplex virus, varicella and other pox viruses, such as smallpox. The first one may be differentiated histologically, immunohistochemically and electron microscopically. The last two may be differentiated using PCR assay for the monkeypox extracellular-envelope virus protein gene.


Assuntos
Queratinócitos/ultraestrutura , Monkeypox virus/isolamento & purificação , Mpox/patologia , Dermatopatias Vesiculobolhosas/patologia , Antígenos Virais/análise , Varicela/diagnóstico , DNA Viral/análise , Diagnóstico Diferencial , Herpes Simples/diagnóstico , Humanos , Queratinócitos/virologia , Microscopia Eletrônica de Transmissão , Mpox/metabolismo , Monkeypox virus/genética , Monkeypox virus/ultraestrutura , Reação em Cadeia da Polimerase , Polimorfismo de Fragmento de Restrição , Dermatopatias Vesiculobolhosas/metabolismo , Varíola/diagnóstico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA