Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 196
Filtrar
1.
Viruses ; 13(11)2021 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-34835083

RESUMO

Human cytomegalovirus (HCMV) tegument protein pp150 is essential for the completion of the final steps in virion maturation. Earlier studies indicated that three pp150nt (N-terminal one-third of pp150) conformers cluster on each triplex (Tri1, Tri2A and Tri2B), and extend towards small capsid proteins atop nearby major capsid proteins, forming a net-like layer of tegument densities that enmesh and stabilize HCMV capsids. Based on this atomic detail, we designed several peptides targeting pp150nt. Our data show significant reduction in virus growth upon treatment with one of these peptides (pep-CR2) with an IC50 of 1.33 µM and no significant impact on cell viability. Based on 3D modeling, pep-CR2 specifically interferes with the pp150-capsid binding interface. Cells pre-treated with pep-CR2 and infected with HCMV sequester pp150 in the nucleus, indicating a mechanistic disruption of pp150 loading onto capsids and subsequent nuclear egress. Furthermore, pep-CR2 effectively inhibits mouse cytomegalovirus (MCMV) infection in cell culture, paving the way for future animal testing. Combined, these results indicate that CR2 of pp150 is amenable to targeting by a peptide inhibitor, and can be developed into an effective antiviral.


Assuntos
Proteínas do Capsídeo/ultraestrutura , Fosfoproteínas/metabolismo , Fosfoproteínas/fisiologia , Proteínas da Matriz Viral/metabolismo , Proteínas da Matriz Viral/fisiologia , Animais , Capsídeo , Proteínas do Capsídeo/metabolismo , Microscopia Crioeletrônica/métodos , Citomegalovirus/genética , Citomegalovirus/metabolismo , Citomegalovirus/patogenicidade , Infecções por Citomegalovirus/metabolismo , Humanos , Camundongos , Muromegalovirus/metabolismo , Muromegalovirus/patogenicidade , Fosfoproteínas/ultraestrutura , Proteínas da Matriz Viral/ultraestrutura , Vírion , Montagem de Vírus
2.
Viruses ; 13(8)2021 08 14.
Artigo em Inglês | MEDLINE | ID: mdl-34452475

RESUMO

Human cytomegalovirus causes diseases in individuals with insufficient immunity. Cytomegaloviruses exploit the ubiquitin proteasome pathway to manipulate the proteome of infected cells. The proteasome degrades ubiquitinated proteins. The family of cullin RING ubiquitin ligases (CRL) regulates the stability of numerous important proteins. If the cullin within the CRL is modified with Nedd8 ("neddylated"), the CRL is enzymatically active, while CRLs lacking Nedd8 modifications are inactive. The Nedd8-activating enzyme (NAE) is indispensable for neddylation. By binding to NAE and inhibiting neddylation, the drug MLN4924 (pevonedistat) causes CRL inactivation and stabilization of CRL target proteins. We showed that MLN4924 elicits potent antiviral activity against cytomegaloviruses, suggesting that NAE might be a druggable host dependency factor (HDF). However, MLN4924 is a nucleoside analog related to AMP, and the antiviral activity of MLN4924 may have been influenced by off-target effects in addition to NAE inhibition. To test if NAE is indeed an HDF, we assessed the novel NAE inhibitor TAS4464 and observed potent antiviral activity against mouse and human cytomegalovirus. Additionally, we raised an MLN4924-resistant cell clone and showed that MLN4924 as well as TAS4464 lose their antiviral activity in these cells. Our results indicate that NAE, the neddylation process, and CRLs are druggable HDFs of cytomegaloviruses.


Assuntos
Antivirais/farmacologia , Citomegalovirus/efeitos dos fármacos , Interações entre Hospedeiro e Microrganismos/efeitos dos fármacos , Muromegalovirus/efeitos dos fármacos , Enzimas Ativadoras de Ubiquitina/antagonistas & inibidores , Enzimas Ativadoras de Ubiquitina/metabolismo , Animais , Linhagem Celular , Linhagem Celular Tumoral , Proteínas Culina/metabolismo , Ciclopentanos/metabolismo , Citomegalovirus/patogenicidade , Humanos , Camundongos , Muromegalovirus/patogenicidade , Proteína NEDD8/metabolismo , Processamento de Proteína Pós-Traducional , Proteoma , Pirimidinas/metabolismo , Pirimidinas/farmacologia , Pirróis/farmacologia , Enzimas Ativadoras de Ubiquitina/genética , Ubiquitina-Proteína Ligases/metabolismo
3.
Nat Commun ; 12(1): 4181, 2021 07 07.
Artigo em Inglês | MEDLINE | ID: mdl-34234136

RESUMO

Nucleobase and nucleoside analogs (NNA) are widely used as anti-viral and anti-cancer agents, and NNA phosphorylation is essential for the activity of this class of drugs. Recently, diphosphatase NUDT15 was linked to thiopurine metabolism with NUDT15 polymorphism associated with drug toxicity in patients. Profiling NNA drugs, we identify acyclovir (ACV) and ganciclovir (GCV) as two new NNAs metabolized by NUDT15. NUDT15 hydrolyzes ACV and GCV triphosphate metabolites, reducing their effects against cytomegalovirus (CMV) in vitro. Loss of NUDT15 potentiates cytotoxicity of ACV and GCV in host cells. In hematopoietic stem cell transplant patients, the risk of CMV viremia following ACV prophylaxis is associated with NUDT15 genotype (P = 0.015). Donor NUDT15 deficiency is linked to graft failure in patients receiving CMV-seropositive stem cells (P = 0.047). In conclusion, NUDT15 is an important metabolizing enzyme for ACV and GCV, and NUDT15 variation contributes to inter-patient variability in their therapeutic effects.


Assuntos
Aciclovir/farmacologia , Antivirais/farmacologia , Infecções por Citomegalovirus/prevenção & controle , Ganciclovir/análogos & derivados , Pirofosfatases/genética , Aciclovir/uso terapêutico , Adolescente , Adulto , Idoso , Animais , Antibioticoprofilaxia , Antivirais/uso terapêutico , Variação Biológica da População/genética , Linhagem Celular , Criança , Pré-Escolar , Cristalografia por Raios X , Citomegalovirus/efeitos dos fármacos , Citomegalovirus/genética , Citomegalovirus/isolamento & purificação , Infecções por Citomegalovirus/diagnóstico , Infecções por Citomegalovirus/etiologia , Infecções por Citomegalovirus/virologia , DNA Viral/sangue , DNA Viral/isolamento & purificação , Modelos Animais de Doenças , Farmacorresistência Viral , Feminino , Ganciclovir/farmacologia , Ganciclovir/uso terapêutico , Transplante de Células-Tronco Hematopoéticas/efeitos adversos , Interações entre Hospedeiro e Microrganismos/genética , Humanos , Lactente , Recém-Nascido , Masculino , Pessoa de Meia-Idade , Muromegalovirus/isolamento & purificação , Muromegalovirus/patogenicidade , Variantes Farmacogenômicos , Polimorfismo de Nucleotídeo Único , Pirofosfatases/metabolismo , Pirofosfatases/ultraestrutura , Resultado do Tratamento , Adulto Jovem
4.
Front Immunol ; 12: 668885, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33968074

RESUMO

Acute infection with murine cytomegalovirus (mCMV) is controlled by CD8+ T cells and develops into a state of latent infection, referred to as latency, which is defined by lifelong maintenance of viral genomes but absence of infectious virus in latently infected cell types. Latency is associated with an increase in numbers of viral epitope-specific CD8+ T cells over time, a phenomenon known as "memory inflation" (MI). The "inflationary" subset of CD8+ T cells has been phenotyped as KLRG1+CD62L- effector-memory T cells (iTEM). It is agreed upon that proliferation of iTEM requires repeated episodes of antigen presentation, which implies that antigen-encoding viral genes must be transcribed during latency. Evidence for this has been provided previously for the genes encoding the MI-driving antigenic peptides IE1-YPHFMPTNL and m164-AGPPRYSRI of mCMV in the H-2d haplotype. There exist two competing hypotheses for explaining MI-driving viral transcription. The "reactivation hypothesis" proposes frequent events of productive virus reactivation from latency. Reactivation involves a coordinated gene expression cascade from immediate-early (IE) to early (E) and late phase (L) transcripts, eventually leading to assembly and release of infectious virus. In contrast, the "stochastic transcription hypothesis" proposes that viral genes become transiently de-silenced in latent viral genomes in a stochastic fashion, not following the canonical IE-E-L temporal cascade of reactivation. The reactivation hypothesis, however, is incompatible with the finding that productive virus reactivation is exceedingly rare in immunocompetent mice and observed only under conditions of compromised immunity. In addition, the reactivation hypothesis fails to explain why immune evasion genes, which are regularly expressed during reactivation in the same cells in which epitope-encoding genes are expressed, do not prevent antigen presentation and thus MI. Here we show that IE, E, and L genes are transcribed during latency, though stochastically, not following the IE-E-L temporal cascade. Importantly, transcripts that encode MI-driving antigenic peptides rarely coincide with those that encode immune evasion proteins. As immune evasion can operate only in cis, that is, in a cell that simultaneously expresses antigenic peptides, the stochastic transcription hypothesis explains why immune evasion is not operative in latently infected cells and, therefore, does not interfere with MI.


Assuntos
Linfócitos T CD4-Positivos/virologia , Infecções por Herpesviridae/virologia , Evasão da Resposta Imune , Memória Imunológica , Infecção Latente/virologia , Pulmão/virologia , Muromegalovirus/patogenicidade , Ativação Viral , Latência Viral , Animais , Antígenos Virais/genética , Antígenos Virais/metabolismo , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Modelos Animais de Doenças , Feminino , Regulação Viral da Expressão Gênica , Infecções por Herpesviridae/imunologia , Infecções por Herpesviridae/metabolismo , Interações Hospedeiro-Patógeno , Proteínas Imediatamente Precoces/genética , Proteínas Imediatamente Precoces/metabolismo , Infecção Latente/imunologia , Infecção Latente/metabolismo , Pulmão/imunologia , Pulmão/metabolismo , Camundongos Endogâmicos BALB C , Modelos Imunológicos , Muromegalovirus/genética , Muromegalovirus/imunologia , Fenótipo , Processos Estocásticos , Fatores de Tempo , Transcrição Gênica
5.
Biosci Rep ; 40(12)2020 12 23.
Artigo em Inglês | MEDLINE | ID: mdl-33245094

RESUMO

INTRODUCTION: Multiple studies have suggested an association between cytomegalovirus (CMV) infection and essential hypertension (EH). MicroRNAs (miRNAs) play a critical role in the development of EH by regulating the expression of specific target genes. However, little is known about the role of miRNAs in CMV-induced EH. In the present study, we compared the miRNA expression profiles of samples from normal and murine cytomegalovirus (MCMV)-infected C57BL/6 mice using high-throughput sequencing analysis. METHODS: We collected the thoracic aorta, heart tissues, and peripheral blood from 20 normal mice and 20 MCMV-infected mice. We identified differentially expressed miRNAs in the peripheral blood samples and predicted their target genes using bioinformatics tools. We then experimentally validated them using quantitative reverse transcription polymerase chain reaction (qRT-PCR) and the target genes with double luciferase reporter gene assay. RESULTS: We found 118 differentially expressed miRNAs, among which 9 miRNAs were identified as potential MCMV infection-induced hypertension regulators. We then validated the expression of two candidate miRNAs, mmu-miR-1929-3p and mcmv-miR-m01-4-5p, using qRT-PCR. Furthermore, the dual-luciferase reporter gene assay revealed that the 3'-untranslated region (UTR) of endothelin A receptor (Ednra) messenger RNA (mRNA) contained a binding site for mmu-miR-1929-3p. Collectively, our data suggest that MCMV infection can raise the blood pressure and reduce mmu-miR-1929-3p expression in C57BL/6 mice. Moreover, we found that mmu-miR-1929-3p targets the 3'-UTR of the Ednra mRNA. CONCLUSION: This novel regulatory axis could aid the development of new approaches for the clinical prevention and control of EH.


Assuntos
Pressão Sanguínea , Hipertensão Essencial/metabolismo , Infecções por Herpesviridae/complicações , MicroRNAs/metabolismo , Regiões 3' não Traduzidas , Animais , Sítios de Ligação , Pressão Sanguínea/genética , Biologia Computacional , Modelos Animais de Doenças , Hipertensão Essencial/genética , Hipertensão Essencial/fisiopatologia , Hipertensão Essencial/virologia , Regulação da Expressão Gênica , Infecções por Herpesviridae/virologia , Sequenciamento de Nucleotídeos em Larga Escala , Camundongos , Camundongos Endogâmicos C57BL , MicroRNAs/genética , Muromegalovirus/patogenicidade , Células NIH 3T3 , Receptor de Endotelina A/genética , Receptor de Endotelina A/metabolismo , Transdução de Sinais
6.
Viruses ; 12(11)2020 10 28.
Artigo em Inglês | MEDLINE | ID: mdl-33126536

RESUMO

Cytomegaloviruses all encode the viral inhibitor of caspase-8-induced apoptosis (vICA). After binding to this initiator caspase, vICA blocks caspase-8 proteolytic activity and ability to activate caspase-3 and/or caspase-7. In this manner, vICA has long been known to prevent apoptosis triggered via tumor necrosis factor (TNF) family death receptor-dependent extrinsic signaling. Here, we employ fully wild-type murine cytomegalovirus (MCMV) and vICA-deficient MCMV (∆M36) to investigate the contribution of TNF signaling to apoptosis during infection of different cell types. ∆M36 shows the expected ability to kill mouse splenic hematopoietic cells, bone marrow-derived macrophages (BMDM), and dendritic cells (BMDC). Antibody blockade or genetic elimination of TNF protects myeloid cells from death, and caspase-8 activation accompanies cell death. Interferons, necroptosis, and pyroptotic gasdermin D (GSDMD) do not contribute to myeloid cell death. Human and murine fibroblasts or murine endothelial cells (SVEC4-10) normally insensitive to TNF become sensitized to ∆M36-induced apoptosis when treated with TNF or TNF-containing BMDM-conditioned medium. We demonstrate that myeloid cells are the natural source of TNF that triggers apoptosis in either myeloid (autocrine) or non-myeloid cells (paracrine) during ∆M36 infection of mice. Caspase-8 suppression by vICA emerges as key to subverting innate immune elimination of a wide variety of infected cell types.


Assuntos
Apoptose/genética , Caspase 8/metabolismo , Muromegalovirus/patogenicidade , Transdução de Sinais , Fator de Necrose Tumoral alfa/imunologia , Proteínas Virais/genética , Animais , Caspase 8/genética , Sobrevivência Celular , Células Dendríticas/imunologia , Células Dendríticas/virologia , Células Endoteliais/imunologia , Células Endoteliais/virologia , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Macrófagos/imunologia , Macrófagos/virologia , Camundongos , Camundongos Knockout , Muromegalovirus/imunologia , Células Mieloides/imunologia , Células Mieloides/virologia , Células NIH 3T3 , Fator de Necrose Tumoral alfa/genética
7.
Cell Rep ; 32(4): 107969, 2020 07 28.
Artigo em Inglês | MEDLINE | ID: mdl-32726632

RESUMO

Major histocompatibility complex class I (MHC-I)-restricted immune responses are largely attributed to cytotoxic T lymphocytes (CTLs). However, natural killer (NK) cells, as predicted by the missing-self hypothesis, have opposing requirements for MHC-I, suggesting that they may also demonstrate MHC-I-restricted effects. In mice, the Ly49 inhibitory receptors prevent NK cell killing of missing-self targets in effector responses, and they have a proposed second function in licensing or educating NK cells via self-MHC-I in vivo. Here we show MHC-I-restricted control of murine cytomegalovirus (MCMV) infection in vivo that is NK cell dependent. Using mice lacking specific Ly49 receptors, we show that control of MCMV requires inhibitory Ly49 receptors and an inhibitory signaling motif and the capacity for MCMV to downregulate MHC-I. Taken together, these data provide definitive evidence that the inhibitory receptors are required for missing-self rejection and are relevant to MHC-I-restricted NK cell control of a viral infection in vivo.


Assuntos
Antígenos de Histocompatibilidade Classe I/metabolismo , Células Matadoras Naturais/metabolismo , Subfamília A de Receptores Semelhantes a Lectina de Células NK/metabolismo , Animais , Antígenos Ly , Infecções por Citomegalovirus/imunologia , Antígenos de Histocompatibilidade Classe I/imunologia , Células Matadoras Naturais/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Muromegalovirus/imunologia , Muromegalovirus/patogenicidade , Subfamília A de Receptores Semelhantes a Lectina de Células NK/imunologia , Receptores de Células Matadoras Naturais , Viroses
8.
Viral Immunol ; 33(3): 237-245, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32286167

RESUMO

Human cytomegalovirus (HCMV) is associated with increased risk of chronic diseases of the heart and vasculature, including myocarditis, atherosclerosis, and transplant vasculopathy. To investigate CMV infection of the heart, murine cytomegalovirus (MCMV) was used to evaluate both acute and latent infection and the subsequent phenotypic and functional consequences of infection. Female BALB/c mice were intraperitoneally (i.p.) inoculated with 1 × 106 pfu of MCMV and evaluated at 14 and 50 days postinfection (dpi). At each time point, echocardiography was used to evaluate cardiac function and histology was conducted for phenotypic evaluation. MCMV replication in the heart was detected as early as 3 dpi and was no longer detectable at 14 dpi. Infected animals had significant cardiac pathology at 14 and 50 dpi when compared to uninfected controls. Histology revealed fibrosis of the heart as early as 14 dpi and the presence of white fibrous deposits on the surface of the heart. Functional evaluation showed significantly increased heart rate and muscle thickening in the latently infected animals when compared to the control animals. At 50 dpi, latent virus was measured by explant reactivation assay, demonstrating that MCMV establishes latency and is capable of reactivation from the heart, similar to other tissues such as spleen and salivary glands. Collectively, these studies illustrate that MCMV infection results in phenotypic alterations within the heart as early as 14 dpi, which progress to functional abnormalities during latency. These findings are similar to sinus tachycardia and hypertrophy of the heart muscle observed in cases of HCMV-induced acute myocarditis.


Assuntos
Coração/fisiopatologia , Coração/virologia , Infecções por Herpesviridae/complicações , Infecções por Herpesviridae/fisiopatologia , Hipertrofia/virologia , Muromegalovirus/patogenicidade , Animais , Feminino , Fibrose/virologia , Humanos , Camundongos Endogâmicos BALB C , Miocardite/fisiopatologia , Miocardite/virologia , Miocárdio/patologia , Glândulas Salivares/virologia , Baço/virologia , Fatores de Tempo , Ativação Viral , Latência Viral , Replicação Viral
9.
J Pathol ; 251(2): 200-212, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32243583

RESUMO

Age-related macular degeneration (AMD) is a complex, multifactorial, progressive disease which represents a leading cause of irreversible visual impairment and blindness in older individuals. Human cytomegalovirus (HCMV), which infects 50-80% of humans, is usually acquired during early life and persists in a latent state for the life of the individual. In view of its previously described pro-angiogenic properties, we hypothesized that cytomegalovirus might be a novel risk factor for progression to an advanced form, neovascular AMD, which is characterized by choroidal neovascularization (CNV). The purpose of this study was to investigate if latent ocular murine cytomegalovirus (MCMV) infection exacerbated the development of CNV in vascular endothelial growth factor (VEGF)-overexpressing VEGF-Ahyper mice. Here we show that neonatal infection with MCMV resulted in dissemination of virus to various organs throughout the body including the eye, where it localized principally to the choroid in both VEGF-overexpressingVEGF-Ahyper and wild-type(WT) 129 mice. By 6 months post-infection, no replicating virus was detected in eyes and extraocular tissues, although virus DNA was still present in all eyes and extraocular tissues of both VEGF-Ahyper and WT mice. Expression of MCMV immediate early (IE) 1 mRNA was detected only in latently infected eyes of VEGF-Ahyper mice, but not in eyes of WT mice. Significantly increased CNV was observed in eyes of MCMV-infected VEGF-Ahyper mice compared to eyes of uninfected VEGF-Ahyper mice, while no CNV lesions were observed in eyes of either infected or uninfected WT mice. Protein levels of several inflammatory/angiogenic factors, particularly VEGF and IL-6, were significantly higher in eyes of MCMV-infected VEGF-Ahyper mice, compared to uninfected controls. Initial studies of ocular tissue from human cadavers revealed that HCMV DNA was present in four choroid/retinal pigment epithelium samples from 24 cadavers. Taken together, our data suggest that ocular HCMV latency could be a significant risk factor for the development of AMD. © 2020 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Assuntos
Neovascularização de Coroide/virologia , Retinite por Citomegalovirus/virologia , Degeneração Macular/virologia , Muromegalovirus/patogenicidade , Retina/virologia , Latência Viral , Idoso , Idoso de 80 Anos ou mais , Animais , Neovascularização de Coroide/genética , Neovascularização de Coroide/metabolismo , Neovascularização de Coroide/patologia , Retinite por Citomegalovirus/genética , Retinite por Citomegalovirus/metabolismo , Retinite por Citomegalovirus/patologia , Modelos Animais de Doenças , Progressão da Doença , Feminino , Humanos , Proteínas Imediatamente Precoces/metabolismo , Mediadores da Inflamação/metabolismo , Interleucina-6/metabolismo , Degeneração Macular/genética , Degeneração Macular/metabolismo , Degeneração Macular/patologia , Masculino , Camundongos da Linhagem 129 , Camundongos Transgênicos , Pessoa de Meia-Idade , Retina/metabolismo , Retina/ultraestrutura , Fatores de Risco , Transdução de Sinais , Fatores de Tempo , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo
10.
PLoS Genet ; 16(3): e1008604, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-32130224

RESUMO

The influence of environmental insults on the onset and progression of mitochondrial diseases is unknown. To evaluate the effects of infection on mitochondrial disease we used a mouse model of Leigh Syndrome, where a missense mutation in the Taco1 gene results in the loss of the translation activator of cytochrome c oxidase subunit I (TACO1) protein. The mutation leads to an isolated complex IV deficiency that mimics the disease pathology observed in human patients with TACO1 mutations. We infected Taco1 mutant and wild-type mice with a murine cytomegalovirus and show that a common viral infection exacerbates the complex IV deficiency in a tissue-specific manner. We identified changes in neuromuscular morphology and tissue-specific regulation of the mammalian target of rapamycin pathway in response to viral infection. Taken together, we report for the first time that a common stress condition, such as viral infection, can exacerbate mitochondrial dysfunction in a genetic model of mitochondrial disease.


Assuntos
Deficiência de Citocromo-c Oxidase/genética , Infecções por Citomegalovirus/genética , Complexo IV da Cadeia de Transporte de Elétrons/genética , Doenças Mitocondriais/genética , Proteínas Mitocondriais/genética , Muromegalovirus/patogenicidade , Animais , Deficiência de Citocromo-c Oxidase/virologia , Infecções por Citomegalovirus/virologia , Modelos Animais de Doenças , Doença de Leigh/genética , Doença de Leigh/virologia , Camundongos , Camundongos Endogâmicos C57BL , Doenças Mitocondriais/virologia , Mutação/genética , Serina-Treonina Quinases TOR/genética
11.
Nat Rev Immunol ; 20(2): 113-127, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31666730

RESUMO

The coordinated activities of innate and adaptive immunity are critical for effective protection against viruses. To counter this, some viruses have evolved sophisticated strategies to circumvent immune cell recognition. In particular, cytomegaloviruses encode large arsenals of molecules that seek to subvert T cell and natural killer cell function via a remarkable array of mechanisms. Consequently, these 'immunoevasins' play a fundamental role in shaping the nature of the immune system by driving the evolution of new immune receptors and recognition mechanisms. Here, we review the diverse strategies adopted by cytomegaloviruses to target immune pathways and outline the host's response.


Assuntos
Imunidade Adaptativa/imunologia , Infecções por Citomegalovirus/imunologia , Citomegalovirus/patogenicidade , Evasão da Resposta Imune/imunologia , Imunidade Inata/imunologia , Complexo Principal de Histocompatibilidade/imunologia , Proteínas Virais/imunologia , Animais , Betaherpesvirinae/patogenicidade , Proteínas do Capsídeo/imunologia , Infecções por Herpesviridae/imunologia , Interações entre Hospedeiro e Microrganismos/imunologia , Humanos , Camundongos , Muromegalovirus/patogenicidade , Proteínas de Ligação a RNA/imunologia , Proteínas do Envelope Viral/imunologia
12.
Nat Microbiol ; 5(2): 331-342, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31844296

RESUMO

Viruses manipulate cellular signalling by inducing the degradation of crucial signal transducers, usually via the ubiquitin-proteasome pathway. Here, we show that the murine cytomegalovirus (Murid herpesvirus 1) M45 protein induces the degradation of two cellular signalling proteins, the nuclear factor κ-light-chain-enhancer of activated B cells (NF-κB) essential modulator (NEMO) and the receptor-interacting protein kinase 1 (RIPK1), via a different mechanism: it induces their sequestration as insoluble protein aggregates and subsequently facilitates their degradation by autophagy. Aggregation of target proteins requires a distinct sequence motif in M45, which we termed 'induced protein aggregation motif'. In a second step, M45 recruits the retromer component vacuolar protein sorting 26B (VPS26B) and the microtubule-associated protein light chain 3 (LC3)-interacting adaptor protein TBC1D5 to facilitate degradation of aggregates by selective autophagy. The induced protein aggregation motif is conserved in M45-homologous proteins of several human herpesviruses, including herpes simplex virus, Epstein-Barr virus and Kaposi's sarcoma-associated herpesvirus, but is only partially conserved in the human cytomegalovirus UL45 protein. We further show that the HSV-1 ICP6 protein induces RIPK1 aggregation and degradation in a similar fashion to M45. These data suggest that induced protein aggregation combined with selective autophagy of aggregates (aggrephagy) represents a conserved viral immune-evasion mechanism.


Assuntos
Herpesviridae/imunologia , Peptídeos e Proteínas de Sinalização Intracelular/imunologia , Proteína Serina-Treonina Quinases de Interação com Receptores/imunologia , Animais , Autofagia/imunologia , Proteína 5 Relacionada à Autofagia/deficiência , Proteína 5 Relacionada à Autofagia/genética , Células Cultivadas , Células HEK293 , Herpesviridae/metabolismo , Herpesviridae/patogenicidade , Herpesvirus Humano 1/imunologia , Herpesvirus Humano 1/metabolismo , Herpesvirus Humano 1/patogenicidade , Interações entre Hospedeiro e Microrganismos/imunologia , Humanos , Evasão da Resposta Imune , Peptídeos e Proteínas de Sinalização Intracelular/química , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Modelos Biológicos , Muromegalovirus/imunologia , Muromegalovirus/metabolismo , Muromegalovirus/patogenicidade , Agregados Proteicos/imunologia , Proteólise , Proteína Serina-Treonina Quinases de Interação com Receptores/química , Proteína Serina-Treonina Quinases de Interação com Receptores/metabolismo , Ribonucleotídeo Redutases/genética , Ribonucleotídeo Redutases/imunologia , Ribonucleotídeo Redutases/metabolismo , Proteínas Virais/genética , Proteínas Virais/imunologia , Proteínas Virais/metabolismo
13.
Curr Med Sci ; 39(6): 899-905, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-31845220

RESUMO

Absent in melanoma 2 (AIM2) inflammasome is a crucial link bridging the innate host defense and the subsequent adaptive immunity when activated by exogenous double stranded DNA (dsDNA). Through establishing models of disseminated murine cytomegalovirus (MCMV) infection in BALB/c and C57BL/6 mice, we evaluated dynamic expression of AIM2 inflammasome components and its relationship with pathological damage and viral replication, trying to figure out whether AIM2 inflammasome is related to the chronic mechanism of MCMV. BALB/c and C57BL/6 mice were sacrificed on day 0, 1, 3, 7, 14 and 28 post infection. Expression levels of AIM2, pro-caspase-1, caspase-1 p20, pro-IL1ß and mature IL1ß in primary peritoneal macrophages (PMs) and spleens were detected by Western blotting. Contents of IL18 in the serum were detected by ELISA. Pathological examinations of livers were performed, and mRNA levels of MCMV glycoprotein B (gB) in salivary glands also assessed. Results showed that expression levels of AIM2 in PMs and spleens of C57BL/6 mice increased on day 3, even continued to day 28; caspase-1 p20 and mature IL1ß increased on day 7, 14 and 28; the persistently high expression of IL18 in the serum started on day 1, showing a double peak curve. As for BALB/c mice, expression of AIM2 in PMs increased on day 1 and day 7, while contents of AIM2 in spleens increased on day 1 and day 3; caspase-1 p20 and mature IL1ß merely increased 7 days fter infection. Thereafter, expression levels of AIM2, caspase-1 p20, mature IL1ß and IL18 were limited; the duration of AIM2 inflammasome activation in BALB/c mice was much shorter than that in C57BL/6 mice. The severer pathological damage and more viral replications in BALB/c mice further proved the deficient antiviral immunity to MCMV. In conclusion, the activation of AIM2 inflammasome in BALB/c mice was short-lived, which is quite possibly related to the chronicity of MCMV infection.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Infecções por Herpesviridae/imunologia , Inflamassomos/metabolismo , Muromegalovirus/patogenicidade , Animais , Caspase 1/metabolismo , Células Cultivadas , Modelos Animais de Doenças , Feminino , Regulação da Expressão Gênica , Interleucina-1beta/metabolismo , Macrófagos Peritoneais/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Muromegalovirus/genética , Muromegalovirus/imunologia , Baço/imunologia , Proteínas do Envelope Viral/genética
14.
J Virol ; 94(1)2019 12 12.
Artigo em Inglês | MEDLINE | ID: mdl-31597762

RESUMO

Natural killer (NK) cells are a subset of innate lymphoid cells (ILC) capable of recognizing stressed and infected cells through multiple germ line-encoded receptor-ligand interactions. Missing-self recognition involves NK cell sensing of the loss of host-encoded inhibitory ligands on target cells, including MHC class I (MHC-I) molecules and other MHC-I-independent ligands. Mouse cytomegalovirus (MCMV) infection promotes a rapid host-mediated loss of the inhibitory NKR-P1B ligand Clr-b (encoded by Clec2d) on infected cells. Here we provide evidence that an MCMV m145 family member, m153, functions to stabilize cell surface Clr-b during MCMV infection. Ectopic expression of m153 in fibroblasts augments Clr-b cell surface levels. Moreover, infections using m153-deficient MCMV mutants (Δm144-m158 and Δm153) show an accelerated and exacerbated Clr-b downregulation. Importantly, enhanced loss of Clr-b during Δm153 mutant infection reverts to wild-type levels upon exogenous m153 complementation in fibroblasts. While the effects of m153 on Clr-b levels are independent of Clec2d transcription, imaging experiments revealed that the m153 and Clr-b proteins only minimally colocalize within the same subcellular compartments, and tagged versions of the proteins were refractory to coimmunoprecipitation under mild-detergent conditions. Surprisingly, the Δm153 mutant possesses enhanced virulence in vivo, independent of both Clr-b and NKR-P1B, suggesting that m153 potentially targets additional host factors. Nevertheless, the present data highlight a unique mechanism by which MCMV modulates NK ligand expression.IMPORTANCE Cytomegaloviruses are betaherpesviruses that in immunocompromised individuals can lead to severe pathologies. These viruses encode various gene products that serve to evade innate immune recognition. NK cells are among the first immune cells that respond to CMV infection and use germ line-encoded NK cell receptors (NKR) to distinguish healthy from virus-infected cells. One such axis that plays a critical role in NK recognition involves the inhibitory NKR-P1B receptor, which engages the host ligand Clr-b, a molecule commonly lost on stressed cells ("missing-self"). In this study, we discovered that mouse CMV utilizes the m153 glycoprotein to circumvent host-mediated Clr-b downregulation, in order to evade NK recognition. These results highlight a novel MCMV-mediated immune evasion strategy.


Assuntos
Interações Hospedeiro-Patógeno/genética , Células Matadoras Naturais/virologia , Lectinas Tipo C/genética , Muromegalovirus/genética , Subfamília B de Receptores Semelhantes a Lectina de Células NK/genética , Receptores Imunológicos/genética , Proteínas da Matriz Viral/genética , Animais , Regulação da Expressão Gênica/imunologia , Teste de Complementação Genética , Infecções por Herpesviridae , Interações Hospedeiro-Patógeno/imunologia , Imunidade Inata , Células Matadoras Naturais/imunologia , Lectinas Tipo C/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Muromegalovirus/imunologia , Muromegalovirus/patogenicidade , Células NIH 3T3 , Subfamília B de Receptores Semelhantes a Lectina de Células NK/imunologia , Receptores Imunológicos/imunologia , Transdução de Sinais , Carga Viral , Proteínas da Matriz Viral/deficiência , Proteínas da Matriz Viral/imunologia , Replicação Viral
15.
Proc Natl Acad Sci U S A ; 116(30): 15170-15177, 2019 07 23.
Artigo em Inglês | MEDLINE | ID: mdl-31285326

RESUMO

The magnitude of CD8 T cell responses against viruses is checked by the balance of proliferation and death. Caspase-8 (CASP8) has the potential to influence response characteristics through initiation of apoptosis, suppression of necroptosis, and modulation of cell death-independent signal transduction. Mice deficient in CASP8 and RIPK3 (Casp8-/-Ripk3-/- ) mount enhanced peak CD8 T cell levels against the natural mouse pathogen murine cytomegalovirus (MCMV) or the human pathogen herpes simplex virus-1 compared with littermate control RIPK3-deficient or WT C57BL/6 mice, suggesting an impact of CASP8 on the magnitude of antiviral CD8 T cell expansion and not on contraction. The higher peak response to MCMV in Casp8-/-Ripk3-/- mice resulted from accumulation of greater numbers of terminally differentiated KLRG1hi effector CD8 T cell subsets. Antiviral Casp8-/-Ripk3-/- T cells exhibited enhanced proliferation when splenocytes were transferred into WT recipient mice. Thus, cell-autonomous CASP8 normally restricts CD8 T cell proliferation following T cell receptor activation in response to foreign antigen. Memory inflation is a hallmark quality of the T cell response to cytomegalovirus infection. Surprisingly, MCMV-specific memory inflation was not sustained long-term in Casp8-/-Ripk3-/- mice even though these mice retained immunity to secondary challenge. In addition, the accumulation of abnormal B220+CD3+ T cells in these viable CASP8-deficient mice was reduced by chronic MCMV infection. Combined, these data brings to light the cell death-independent role of CASP8 during CD8 T cell expansion in mice lacking the confounding impact of RIPK3-mediated necroptosis.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Caspase 8/genética , Infecções por Citomegalovirus/imunologia , Muromegalovirus/imunologia , Proteína Serina-Treonina Quinases de Interação com Receptores/genética , Subpopulações de Linfócitos T/imunologia , Transferência Adotiva , Animais , Linfócitos T CD8-Positivos/virologia , Caspase 8/imunologia , Proliferação de Células , Infecções por Citomegalovirus/genética , Infecções por Citomegalovirus/virologia , Feminino , Regulação da Expressão Gênica , Herpes Simples/genética , Herpes Simples/imunologia , Herpes Simples/virologia , Herpesvirus Humano 1/imunologia , Herpesvirus Humano 1/patogenicidade , Memória Imunológica , Lectinas Tipo C/genética , Lectinas Tipo C/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Muromegalovirus/patogenicidade , Proteína Serina-Treonina Quinases de Interação com Receptores/deficiência , Proteína Serina-Treonina Quinases de Interação com Receptores/imunologia , Receptores de Antígenos de Linfócitos T/genética , Receptores de Antígenos de Linfócitos T/imunologia , Receptores Imunológicos/genética , Receptores Imunológicos/imunologia , Transdução de Sinais , Subpopulações de Linfócitos T/virologia
16.
mBio ; 10(3)2019 06 25.
Artigo em Inglês | MEDLINE | ID: mdl-31239384

RESUMO

Human cytomegalovirus (HCMV) is a betaherpesvirus that is a significant pathogen within newborn and immunocompromised populations. Morbidity associated with HCMV infection is the consequence of viral dissemination. HCMV has evolved to manipulate the host immune system to enhance viral dissemination and ensure long-term survival within the host. The immunomodulatory protein vCXCL-1, a viral chemokine functioning primarily through the CXCR2 chemokine receptor, is hypothesized to attract CXCR2+ neutrophils to infection sites, aiding viral dissemination. Neutrophils harbor HCMV in vivo; however, the interaction between vCXCL-1 and the neutrophil has not been evaluated in vivo Using the mouse model and mouse cytomegalovirus (MCMV) infection, we show that murine neutrophils harbor and transfer infectious MCMV and that virus replication initiates within this cell type. Utilizing recombinant MCMVs expressing vCXCL-1 from the HCMV strain (Toledo), we demonstrated that vCXCL-1 significantly enhances MCMV dissemination kinetics. Through cellular depletion experiments, we observe that neutrophils impact dissemination but that overall dissemination is largely neutrophil independent. This work adds neutrophils to the list of innate cells (i.e., dendritic and macrophages/monocytes) that contribute to MCMV dissemination but refutes the hypothesis that neutrophils are the primary cell responding to vCXCL-1.IMPORTANCE An adequate in vivo analysis of HCMV's viral chemokine vCXCL-1 has been lacking. Here we generate recombinant MCMVs expressing vCXCL-1 to study vCXCL-1 function in vivo using MCMV as a surrogate. We demonstrate that vCXCL-1 increases MCMV dissemination kinetics for both primary and secondary dissemination. Additionally, we provide evidence, that the murine neutrophil is largely a bystander in the mouse's response to vCXCL-1. We confirm the hypothesis that vCXCL-1 is a HCMV virulence factor. Infection of severely immunocompromised mice with MCMVs expressing vCXCL-1 was lethal in more than 50% of infected animals, while all animals infected with parental virus survived during a 12-day period. This work provides needed insights into vCXCL-1 function in vivo.


Assuntos
Quimiocina CXCL1/imunologia , Infecções por Citomegalovirus/imunologia , Citomegalovirus/imunologia , Muromegalovirus/imunologia , Neutrófilos/virologia , Animais , Quimiocina CXCL1/genética , Interações Hospedeiro-Patógeno/imunologia , Humanos , Cinética , Camundongos , Camundongos Endogâmicos BALB C , Muromegalovirus/patogenicidade , Neutrófilos/imunologia , Receptores de Interleucina-8B/genética , Receptores de Interleucina-8B/imunologia , Fatores de Virulência/imunologia , Replicação Viral
17.
PLoS Pathog ; 15(2): e1007615, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30779794

RESUMO

Cytomegalovirus (CMV) infection causes birth defects and life-threatening complications in immunosuppressed patients. Lack of vaccine and need for more effective drugs have driven widespread ongoing therapeutic development efforts against human CMV (HCMV), mostly using murine CMV (MCMV) as the model system for preclinical animal tests. The recent publication (Yu et al., 2017, DOI: 10.1126/science.aam6892) of an atomic model for HCMV capsid with associated tegument protein pp150 has infused impetus for rational design of novel vaccines and drugs, but the absence of high-resolution structural data on MCMV remains a significant knowledge gap in such development efforts. Here, by cryoEM with sub-particle reconstruction method, we have obtained the first atomic structure of MCMV capsid with associated pp150. Surprisingly, the capsid-binding patterns of pp150 differ between HCMV and MCMV despite their highly similar capsid structures. In MCMV, pp150 is absent on triplex Tc and exists as a "Λ"-shaped dimer on other triplexes, leading to only 260 groups of two pp150 subunits per capsid in contrast to 320 groups of three pp150 subunits each in a "Δ"-shaped fortifying configuration. Many more amino acids contribute to pp150-pp150 interactions in MCMV than in HCMV, making MCMV pp150 dimer inflexible thus incompatible to instigate triplex Tc-binding as observed in HCMV. While pp150 is essential in HCMV, our pp150-deletion mutant of MCMV remained viable though with attenuated infectivity and exhibiting defects in retaining viral genome. These results thus invalidate targeting pp150, but lend support to targeting capsid proteins, when using MCMV as a model for HCMV pathogenesis and therapeutic studies.


Assuntos
Proteínas do Capsídeo/ultraestrutura , Fosfoproteínas/metabolismo , Fosfoproteínas/fisiologia , Proteínas da Matriz Viral/metabolismo , Proteínas da Matriz Viral/fisiologia , Animais , Capsídeo , Proteínas do Capsídeo/metabolismo , Microscopia Crioeletrônica/métodos , Citomegalovirus/genética , Citomegalovirus/metabolismo , Citomegalovirus/patogenicidade , Infecções por Citomegalovirus/metabolismo , Genoma Viral/genética , Humanos , Camundongos , Muromegalovirus/metabolismo , Muromegalovirus/patogenicidade , Fosfoproteínas/ultraestrutura , Deleção de Sequência/genética , Proteínas da Matriz Viral/ultraestrutura , Vírion , Montagem de Vírus
18.
EMBO J ; 38(5)2019 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-30696688

RESUMO

Cytomegaloviruses (CMVs) are master manipulators of the host immune response. Here, we reveal that the murine CMV (MCMV) protein m152 specifically targets the type I interferon (IFN) response by binding to stimulator of interferon genes (STING), thereby delaying its trafficking to the Golgi compartment from where STING initiates type I IFN signaling. Infection with an MCMV lacking m152 induced elevated type I IFN responses and this leads to reduced viral transcript levels both in vitro and in vivo This effect is ameliorated in the absence of STING Interestingly, while m152 inhibits STING-mediated IRF signaling, it did not affect STING-mediated NF-κB signaling. Analysis of how m152 targets STING translocation reveals that STING activates NF-κB signaling already from the ER prior to its trafficking to the Golgi. Strikingly, this response is important to promote early MCMV replication. Our results show that MCMV has evolved a mechanism to specifically antagonize the STING-mediated antiviral IFN response, while preserving its pro-viral NF-κB response, providing an advantage in the establishment of an infection.


Assuntos
Infecções por Citomegalovirus/imunologia , Interações Hospedeiro-Patógeno/imunologia , Fatores Reguladores de Interferon/metabolismo , Interferon Tipo I/metabolismo , Glicoproteínas de Membrana/metabolismo , Proteínas de Membrana/fisiologia , NF-kappa B/metabolismo , Proteínas Virais/metabolismo , Animais , Infecções por Citomegalovirus/metabolismo , Infecções por Citomegalovirus/virologia , Fatores Reguladores de Interferon/genética , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Muromegalovirus/genética , Muromegalovirus/isolamento & purificação , Muromegalovirus/patogenicidade , NF-kappa B/genética , Ligação Proteica , Proteínas Virais/genética , Replicação Viral
19.
J Virol ; 93(3)2019 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-30404805

RESUMO

Cytomegaloviruses (CMVs) establish systemic infections across diverse cell types. Glycoproteins that alter tropism can potentially guide their spread. Glycoprotein O (gO) is a nonessential fusion complex component of both human CMV (HCMV) and murine CMV (MCMV). We tested its contribution to MCMV spread from the respiratory tract. In vitro, MCMV lacking gO poorly infected fibroblasts and epithelial cells. Cell binding was intact, but penetration was delayed. In contrast, myeloid infection was preserved, and in the lungs, where myeloid and type 2 alveolar epithelial cells are the main viral targets, MCMV lacking gO showed a marked preference for myeloid infection. Its poor epithelial cell infection was associated with poor primary virus production and reduced virulence. Systemic spread, which proceeds via infected CD11c+ myeloid cells, was initially intact but then diminished, because less epithelial infection led ultimately to less myeloid infection. Thus, the tight linkage between peripheral and systemic MCMV infections gave gO-dependent infection a central role in host colonization.IMPORTANCE Human cytomegalovirus is a leading cause of congenital disease. This reflects its capacity for systemic spread. A vaccine is needed, but the best viral targets are unclear. Attention has focused on the virion membrane fusion complex. It has 2 forms, so we need to know what each contributes to host colonization. One includes the virion glycoprotein O. We used murine cytomegalovirus, which has equivalent fusion complexes, to determine the importance of glycoprotein O after mucosal infection. We show that it drives local virus replication in epithelial cells. It was not required to infect myeloid cells, which establish systemic infection, but poor local replication reduced systemic spread as a secondary effect. Therefore, targeting glycoprotein O of human cytomegalovirus has the potential to reduce both local and systemic infections.


Assuntos
Células Epiteliais/virologia , Fibroblastos/virologia , Infecções por Herpesviridae/virologia , Pulmão/virologia , Glicoproteínas de Membrana/metabolismo , Muromegalovirus/patogenicidade , Proteínas do Envelope Viral/metabolismo , Replicação Viral , Animais , Células Cultivadas , Células Epiteliais/metabolismo , Fibroblastos/metabolismo , Infecções por Herpesviridae/metabolismo , Pulmão/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Internalização do Vírus
20.
Theranostics ; 8(20): 5634-5644, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30555569

RESUMO

Rationales: Gene-targeting ribozymes represent promising nucleic acid-based gene interference agents for therapeutic application. We previously used an in vitro selection procedure to engineer novel RNase P-based ribozyme variants with enhanced targeting activity. However, it has not been reported whether these ribozyme variants also exhibit improved activity in blocking gene expression in animals. Methods and Results: In this report, R388-AS, a new engineered ribozyme variant, was designed to target the mRNA of assemblin (AS) of murine cytomegalovirus (MCMV), which is essential for viral progeny production. Variant R338-AS cleaved AS mRNA sequence in vitro at least 200 times more efficiently than ribozyme M1-AS, which originated from the wild type RNase P catalytic RNA sequence. In cultured MCMV-infected cells, R338-AS exhibited better antiviral activity than M1-AS and decreased viral AS expression by 98-99% and virus production by 15,000 fold. In MCMV-infected mice, R388-AS was more active in inhibiting AS expression, blocking viral replication, and improving animal survival than M1-AS. Conclusions: Our results provide the first direct evidence that novel engineered RNase P ribozyme variants with more active catalytic activity in vitro are also more effective in inhibiting viral gene expression in animals. Moreover, our studies imply the potential of engineering novel RNase P ribozyme variants with unique mutations to improve ribozyme activity for therapeutic application.


Assuntos
Terapia Genética/métodos , Muromegalovirus/efeitos dos fármacos , Muromegalovirus/patogenicidade , RNA Catalítico/genética , Ribonuclease P/metabolismo , Animais , Citomegalovirus/genética , Camundongos , RNA Antissenso/genética , RNA Mensageiro/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...