Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
1.
Cell Host Microbe ; 31(6): 874-889, 2023 06 14.
Artigo em Inglês | MEDLINE | ID: mdl-37321171

RESUMO

Recombination is thought to be a mechanism that facilitates cross-species transmission in coronaviruses, thus acting as a driver of coronavirus spillover and emergence. Despite its significance, the mechanism of recombination is poorly understood, limiting our potential to estimate the risk of novel recombinant coronaviruses emerging in the future. As a tool for understanding recombination, here, we outline a framework of the recombination pathway for coronaviruses. We review existing literature on coronavirus recombination, including comparisons of naturally observed recombinant genomes as well as in vitro experiments, and place the findings into the recombination pathway framework. We highlight gaps in our understanding of coronavirus recombination illustrated by the framework and outline how further experimental research is critical for disentangling the molecular mechanism of recombination from external environmental pressures. Finally, we describe how an increased understanding of the mechanism of recombination can inform pandemic predictive intelligence, with a retrospective emphasis on SARS-CoV-2.


Assuntos
COVID-19 , SARS-CoV-2 , Humanos , SARS-CoV-2/genética , Estudos Retrospectivos , Filogenia , Recombinação Genética
2.
Viruses ; 15(3)2023 03 09.
Artigo em Inglês | MEDLINE | ID: mdl-36992420

RESUMO

Human cytomegalovirus (HCMV) is a ubiquitous pathogen that infects the majority of the world population and causes lifelong latent infection. HCMV has been shown to exacerbate cardiovascular diseases, including myocarditis, vascular sclerosis, and transplant vasculopathy. Recently, we have shown that murine CMV (MCMV) recapitulates the cardiovascular dysfunction observed in patients with HCMV-induced myocarditis. To understand the viral mechanisms involved in CMV-induced heart dysfunction, we further characterized cardiac function in response to MCMV and examined virally encoded G-protein-coupled receptor homologs (vGPCRs) US28 and M33 as potential factors that promote infection in the heart. We hypothesized that the CMV-encoded vGPCRs could exacerbate cardiovascular damage and dysfunction. Three viruses were used to evaluate the role of vGPCRs in cardiac dysfunction: wild-type MCMV, a M33-deficient virus (∆M33), and a virus with the M33 open reading frame (ORF) replaced with US28, an HCMV vGPCR (i.e., US28+). Our in vivo studies revealed that M33 plays a role in promoting cardiac dysfunction by increasing viral load and heart rate during acute infection. During latency, ΔM33-infected mice demonstrated reduced calcification, altered cellular gene expression, and less cardiac hypertrophy compared with wild-type MCMV-infected mice. Ex vivo viral reactivation from hearts was less efficient in ΔM33-infected animals. HCMV protein US28 expression restored the ability of the M33-deficient virus to reactivate from the heart. US28+ MCMV infection caused damage to the heart comparable with wild-type MCMV infection, suggesting that the US28 protein is sufficient to complement the function of M33 in the heart. Altogether, these data suggest a role for vGPCRs in viral pathogenesis in the heart and thus suggest that vGPCRs promote long-term cardiac damage and dysfunction.


Assuntos
Infecções por Citomegalovirus , Cardiopatias , Muromegalovirus , Miocardite , Humanos , Animais , Camundongos , Muromegalovirus/fisiologia , Receptores de Quimiocinas/genética , Proteínas Virais/metabolismo , Citomegalovirus/fisiologia , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo
3.
PLoS One ; 17(8): e0265479, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35976883

RESUMO

The salivary glands are a crucial site of replication for human cytomegalovirus (HCMV) and its murine counterpart, murine cytomegalovirus (MCMV). Studies of MCMV often involve the use of BALB/c strain mice, but most in vitro assays are carried out in the NIH 3T3 cell line, which is derived from Swiss Albino mice. This report describes a BALB/c-derived mouse salivary gland cell line immortalized using the SV40 large T antigen. Cells stained positive for PDGFR1 and negative for E-cadherin and PECAM-1, indicating mesenchymal origin. This cell line, which has been named murine salivary gland mesenchymal (mSGM), shows promise as a tool for ex vivo immunological assays due to its MHC haplotype match with the BALB/c mouse strain. In addition, plaque assays using mSGM rather than NIH 3T3 cells are significantly more sensitive for detecting low concentrations of MCMV particles. Finally, it is demonstrated that mSGM cells express all 3 BALB/c MHC class I isotypes and are susceptible to T cell-mediated ex vivo cytotoxicity assays, leading to many possible uses in immunological studies of MCMV.


Assuntos
Muromegalovirus , Animais , Citomegalovirus , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Células NIH 3T3 , Glândulas Salivares , Células-Tronco
4.
Front Immunol ; 13: 1047299, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36569845

RESUMO

Introduction: Human cytomegalovirus (HCMV) is a global health threat due to its ubiquity and lifelong persistence in infected people. During latency, host CD8+ T cell responses to HCMV continue to increase in a phenomenon known as memory inflation. We used murine CMV (MCMV) as a model for HCMV to characterize the memory inflation response to wild-type MCMV (KP) and a latency-defective mutant (ΔM33stop), which lacks M33, an MCMV chemokine receptor homolog. M33 is essential for normal reactivation from latency and this was leveraged to determine whether reactivation in vivo contributes to T cell memory inflation. Methods: Mice were infected with wild-type or mutant MCMV and T cell responses were analyzed by flow cytometry at acute and latent time points. Ex vivo reactivation and cytotoxicity assays were carried out to further investigate immunity and virus replication. Quantitative reverse-transcriptase polymerase chain reaction (q-RTPCR) was used to examine gene expression during reactivation. MHC expression on infected cells was analyzed by flow cytometry. Finally, T cells were depleted from latently-infected B cell-deficient mice to examine the in vivo difference in reactivation between wild-type and ΔM33stop. Results: We found that ΔM33stop triggers memory inflation specific for peptides derived from the immediate-early protein IE1 but not the early protein m164, in contrast to wild-type MCMV. During ex vivo reactivation, gene expression in DM33stop-infected lung tissues was delayed compared to wild-type virus. Normal gene expression was partially rescued by substitution of the HCMV US28 open reading frame in place of the M33 gene. In vivo depletion of T cells in immunoglobulin heavy chain-knockout mice resulted in reactivation of wild-type MCMV, but not ΔM33stop, confirming the role of M33 during reactivation from latency. Further, we found that M33 induces isotype-specific downregulation of MHC class I on the cell surface suggesting previously unappreciated roles in immune evasion. Discussion: Our results indicate that M33 is more polyfunctional than previously appreciated. In addition to its role in reactivation, which had been previously described, we found that M33 alters viral gene expression, host T cell memory inflation, and MHC class I expression. US28 was able to partially complement most functions of M33, suggesting that its role in HCMV infection may be similarly pleotropic.


Assuntos
Infecções por Citomegalovirus , Evasão da Resposta Imune , Humanos , Animais , Camundongos , Latência Viral/fisiologia , Citomegalovirus/fisiologia , Receptores Acoplados a Proteínas G , Linfócitos T CD8-Positivos , Infecções por Citomegalovirus/genética
5.
Pathogens ; 10(5)2021 May 18.
Artigo em Inglês | MEDLINE | ID: mdl-34069957

RESUMO

Human Cytomegalovirus (HCMV) is a widespread pathogen that causes lifelong latent infection and is associated with the exacerbation of chronic inflammatory diseases in seropositive individuals. Of particular impact, HCMV infection is known to worsen many cardiovascular diseases including myocarditis, atherosclerosis, hypertension, and transplant vasculopathy. Due to its similarity to HCMV, murine CMV (MCMV) is an appropriate model to understand HCMV-induced pathogenesis in the heart and vasculature. MCMV shares similar sequence homology and recapitulates much of the HCMV pathogenesis, including HCMV-induced cardiovascular diseases. This review provides insight into HCMV-associated cardiovascular diseases and the murine model of MCMV infection, which has been used to study the viral pathogenesis and mechanisms contributing to cardiovascular diseases. Our new functional studies using echocardiography demonstrate tachycardia and hypertrophy in the mouse, similar to HCMV-induced myocarditis in humans. For the first time, we show long term heart dysfunction and that MCMV reactivates from latency in the heart, which raises the intriguing idea that HCMV latency and frequent virus reactivation perturbs long term cardiovascular function.

6.
J Virol Methods ; 297: 114270, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34461152

RESUMO

Human cytomegalovirus (HCMV) is a widespread pathogen that causes lifelong latent infection in the majority of the world population. HCMV is associated with increased incidence and severity of many cardiovascular diseases including myocarditis, atherosclerosis, and transplant vasculopathy. Due to the species-restricted nature of cytomegalovirus infection, murine cytomegalovirus (MCMV) is a useful model that recapitulates many of the features of HCMV infection of the cardiovascular system. While in vivo MCMV studies are able to answer many questions regarding pathogenesis of infection, in vitro experiments using cell lines are useful tools to further understand the potential underlying mechanisms. In this study, we characterize MCMV infection of the murine aortic smooth muscle cell line (MOVAS). Our findings demonstrate that MOVAS cells are permissive for MCMV infection, form plaques under carboxymethyl cellulose overlay, and produce progeny virus similar to NIH 3T3 murine embryonic fibroblasts. In addition, MCMV infection induces calcification in MOVAS cells similar to that seen in the epicardium of MCMV-infected hearts. We conclude that MOVAS cells are a useful in vitro tool for studying CMV-mediated cardiac calcification.


Assuntos
Infecções por Citomegalovirus , Muromegalovirus , Animais , Citomegalovirus , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Músculo Liso Vascular , Miócitos de Músculo Liso
7.
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
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA