Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
J Virol ; 95(22): e0090421, 2021 10 27.
Artigo em Inglês | MEDLINE | ID: mdl-34468171

RESUMO

Zika virus (ZIKV) infection became a worldwide concern due to its correlation with the development of microcephaly and other neurological disorders. ZIKV neurotropism is well characterized, but the role of peripheral viral amplification to brain infection remains unknown. Here, we found that ZIKV replicates in human primary skeletal muscle myoblasts, impairing its differentiation into myotubes but not interfering with the integrity of the already-formed muscle fibers. Using mouse models, we showed ZIKV tropism to muscle tissue either during embryogenesis after maternal transmission or when infection occurred after birth. Interestingly, ZIKV replication in the mouse skeletal muscle started immediately after ZIKV inoculation, preceding viral RNA detection in the brain and causing no disruption to the integrity of the blood brain barrier, and remained active for more than 2 weeks, whereas replication in the spleen and liver were not sustained over time. In addition, ZIKV infection of the skeletal muscle induces necrotic lesions, inflammation, and fiber atrophy. We also found a reduction in the expression of regulatory myogenic factors that are essential for muscle repair after injury. Taken together, our results indicate that the skeletal muscle is an early site of viral amplification and lesion that may result in late consequences in muscle development after ZIKV infection. IMPORTANCE Zika Virus (ZIKV) neurotropism and its deleterious effects on central nervous system have been well characterized. However, investigations of the initial replication sites for the establishment of infection and viral spread to neural tissues remain underexplored. A complete description of the range of ZIKV-induced lesions and others factors that can influence the severity of the disease is necessary to prevent ZIKV's deleterious effects. ZIKV has been shown to access the central nervous system without significantly affecting blood-brain barrier permeability. Here, we demonstrated that skeletal muscle is an earlier site of ZIKV replication, contributing to the increase of peripheral ZIKV load. ZIKV replication in muscle promotes necrotic lesions and inflammation and also impairs myogenesis. Overall, our findings showed that skeletal muscle is involved in pathogenesis and opens new fields in the investigation of the long-term consequences of early infection.


Assuntos
Fibras Musculares Esqueléticas/virologia , Infecção por Zika virus/virologia , Zika virus/fisiologia , Aedes , Animais , Animais Recém-Nascidos , Linhagem Celular , Feminino , Humanos , Transmissão Vertical de Doenças Infecciosas , Camundongos , Camundongos Knockout , Fibras Musculares Esqueléticas/citologia , Mioblastos , Replicação Viral
3.
PLoS Negl Trop Dis ; 14(8): e0008282, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32817655

RESUMO

Muscle cells are potential targets of many arboviruses, such as Ross River, Dengue, Sindbis, and chikungunya viruses, that may be involved in the physiopathological course of the infection. During the recent outbreak of Zika virus (ZIKV), myalgia was one of the most frequently reported symptoms. We investigated the susceptibility of human muscle cells to ZIKV infection. Using an in vitro model of human primary myoblasts that can be differentiated into myotubes, we found that myoblasts can be productively infected by ZIKV. In contrast, myotubes were shown to be resistant to ZIKV infection, suggesting a differentiation-dependent susceptibility. Infection was accompanied by a caspase-independent cytopathic effect, associated with paraptosis-like cytoplasmic vacuolization. Proteomic profiling was performed 24h and 48h post-infection in cells infected with two different isolates. Proteome changes indicate that ZIKV infection induces an upregulation of proteins involved in the activation of the Interferon type I pathway, and a downregulation of protein synthesis. This work constitutes the first observation of primary human muscle cells susceptibility to ZIKV infection, and differentiation-dependent restriction of infection from myoblasts to myotubes. Since myoblasts constitute the reservoir of stem cells involved in reparation/regeneration in muscle tissue, the infection of muscle cells and the viral-induced alterations observed here could have consequences in ZIKV infection pathogenesis.


Assuntos
Diferenciação Celular , Células Musculares/metabolismo , Células Musculares/virologia , Proteômica , Infecção por Zika virus , Morte Celular , Linhagem Celular , Efeito Citopatogênico Viral , Suscetibilidade a Doenças , Interações Hospedeiro-Patógeno , Humanos , Interferon Tipo I/metabolismo , Células Musculares/patologia , Fibras Musculares Esqueléticas/metabolismo , Fibras Musculares Esqueléticas/virologia , Mioblastos/metabolismo , Mioblastos/virologia , Proteínas/metabolismo , Células-Tronco , Replicação Viral , Zika virus/patogenicidade , Infecção por Zika virus/patologia , Infecção por Zika virus/virologia
4.
PLoS Pathog ; 15(8): e1007993, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31465513

RESUMO

Chikungunya virus (CHIKV) is an arthritogenic alphavirus that acutely causes fever as well as severe joint and muscle pain. Chronic musculoskeletal pain persists in a substantial fraction of patients for months to years after the initial infection, yet we still have a poor understanding of what promotes chronic disease. While replicating virus has not been detected in joint-associated tissues of patients with persistent arthritis nor in various animal models at convalescent time points, viral RNA is detected months after acute infection. To identify the cells that might contribute to pathogenesis during this chronic phase, we developed a recombinant CHIKV that expresses Cre recombinase (CHIKV-3'-Cre). CHIKV-3'-Cre replicated in myoblasts and fibroblasts, and it induced arthritis during the acute phase in mice. Importantly, it also induced chronic disease, including persistent viral RNA and chronic myositis and synovitis similar to wild-type virus. CHIKV-3'-Cre infection of tdTomato reporter mice resulted in a population of tdTomato+ cells that persisted for at least 112 days. Immunofluorescence and flow cytometric profiling revealed that these tdTomato+ cells predominantly were myofibers and dermal and muscle fibroblasts. Treatment with an antibody against Mxra8, a recently defined host receptor for CHIKV, reduced the number of tdTomato+ cells in the chronic phase and diminished the levels of chronic viral RNA, implicating these tdTomato+ cells as the reservoir of chronic viral RNA. Finally, isolation and flow cytometry-based sorting of the tdTomato+ fibroblasts from the skin and ankle and analysis for viral RNA revealed that the tdTomato+ cells harbor most of the persistent CHIKV RNA at chronic time points. Therefore, this CHIKV-3'-Cre and tdTomato reporter mouse system identifies the cells that survive CHIKV infection in vivo and are enriched for persistent CHIKV RNA. This model represents a useful tool for studying CHIKV pathogenesis in the acute and chronic stages of disease.


Assuntos
Artrite Experimental/virologia , Febre de Chikungunya/virologia , Vírus Chikungunya/patogenicidade , Derme/patologia , Fibroblastos/patologia , Músculo Esquelético/patologia , RNA Viral/metabolismo , Animais , Artrite Experimental/metabolismo , Artrite Experimental/patologia , Febre de Chikungunya/metabolismo , Vírus Chikungunya/genética , Derme/metabolismo , Derme/virologia , Modelos Animais de Doenças , Fibroblastos/metabolismo , Fibroblastos/virologia , Camundongos , Camundongos Endogâmicos C57BL , Fibras Musculares Esqueléticas/metabolismo , Fibras Musculares Esqueléticas/patologia , Fibras Musculares Esqueléticas/virologia , Músculo Esquelético/metabolismo , Músculo Esquelético/virologia , RNA Viral/genética , Replicação Viral
6.
Cell Biol Int ; 39(11): 1307-16, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26123964

RESUMO

We have utilized the enveloped viral model to study the effect of fluvastatin on membrane trafficking in isolated rat myofibers. Our immunofluorescence studies constantly showed that infections in myofibers, which were treated with fluvastatin prior and during the infection with either vesicular stomatitis virus (VSV) or influenza A virus, propagated more slowly than in control myofibers without drug treatment. Experiments with a virus expressing Dad1 tagged with green fluorescent protein (GFP-Dad1) showed that fluvastatin did not affect its distribution within the ER/SR network and immunofluorescence staining for GM130 did not show any marked effect on the structure of the Golgi components. Furthermore, fluvastatin did not inhibit trafficking of the chimeric transport marker VSV temperature sensitive G protein (tsG-GFP) from the ER to the Golgi. We next subjected VSV infected myofibers for pulse-chase labeling experiments and found that fluvastatin did not slow down the ER-to-Golgi trafficking or Golgi to plasma membrane trafficking of the viral glycoprotein. These studies show that fluvastatin inhibited the propagation of viral infection in skeletal myofibers but no adverse effect on the exocytic trafficking could be demonstrated. These results suggest that other effects of statins rather than inhibition of ER-to-Golgi trafficking might be behind the myotoxic effects of the statins.


Assuntos
Ácidos Graxos Monoinsaturados/farmacologia , Indóis/farmacologia , Fibras Musculares Esqueléticas/efeitos dos fármacos , Fibras Musculares Esqueléticas/virologia , Infecções por Orthomyxoviridae/tratamento farmacológico , Estomatite Vesicular/tratamento farmacológico , Animais , Modelos Animais de Doenças , Retículo Endoplasmático/metabolismo , Fluvastatina , Proteínas de Fluorescência Verde/biossíntese , Proteínas de Fluorescência Verde/química , Proteínas de Fluorescência Verde/genética , Vírus da Influenza A/efeitos dos fármacos , Vírus da Influenza A/crescimento & desenvolvimento , Masculino , Glicoproteínas de Membrana/biossíntese , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/genética , Fibras Musculares Esqueléticas/metabolismo , Infecções por Orthomyxoviridae/metabolismo , Infecções por Orthomyxoviridae/virologia , Transporte Proteico/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Estomatite Vesicular/metabolismo , Estomatite Vesicular/virologia , Vírus da Estomatite Vesicular Indiana/efeitos dos fármacos , Vírus da Estomatite Vesicular Indiana/crescimento & desenvolvimento , Proteínas do Envelope Viral/biossíntese , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/genética
7.
Immunology ; 146(2): 312-26, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26173587

RESUMO

Self-amplifying mRNAs (SAM(®) ) are a novel class of nucleic acid vaccines, delivered by a non-viral delivery system. They are effective at eliciting potent and protective immune responses and are being developed as a platform technology with potential to be used for a broad range of targets. However, their mechanism of action has not been fully elucidated. To date, no evidence of in vivo transduction of professional antigen-presenting cells (APCs) by SAM vector has been reported, while the antigen expression has been shown to occur mostly in the muscle fibres. Here we show that bone-marrow-derived APCs rather than muscle cells are responsible for induction of MHC class-I restricted CD8 T cells in vivo, but direct transfection of APCs by SAM vectors is not required. Based on all our in vivo and in vitro data we propose that upon SAM vaccination the antigen is expressed within muscle cells and then transferred to APCs, suggesting cross-priming as the prevalent mechanism for priming the CD8 T-cell response by SAM vaccines.


Assuntos
Células Apresentadoras de Antígenos/imunologia , Células da Medula Óssea/imunologia , Linfócitos T CD8-Positivos/imunologia , Apresentação Cruzada , Vírus da Influenza A Subtipo H1N1/imunologia , Vacinas contra Influenza/imunologia , Fibras Musculares Esqueléticas/imunologia , RNA Mensageiro/imunologia , RNA Viral/imunologia , Proteínas de Ligação a RNA/imunologia , Proteínas do Core Viral/imunologia , Animais , Células Apresentadoras de Antígenos/virologia , Células da Medula Óssea/virologia , Transplante de Medula Óssea , Linfócitos T CD8-Positivos/virologia , Comunicação Celular , Linhagem Celular , Cricetinae , Feminino , Antígenos de Histocompatibilidade Classe I/genética , Antígenos de Histocompatibilidade Classe I/imunologia , Vírus da Influenza A Subtipo H1N1/genética , Vacinas contra Influenza/genética , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fibras Musculares Esqueléticas/virologia , Proteínas do Nucleocapsídeo , RNA Mensageiro/genética , RNA Viral/genética , Proteínas de Ligação a RNA/genética , Transfecção , Quimeras de Transplante , Proteínas do Core Viral/genética
8.
J Virol ; 89(5): 2494-506, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25540384

RESUMO

UNLABELLED: Skeletal muscle, at 30 to 40% of body mass, is the most abundant soft tissue in the body. Besides its primary function in movement and posture, skeletal muscle is a significant innate immune organ with the capacity to produce cytokines and chemokines and respond to proinflammatory cytokines. Little is known about the role of skeletal muscle during systemic influenza A virus infection in any host and particularly avian species. Here we used primary chicken and duck multinucleated myotubes to examine their susceptibility and innate immune response to influenza virus infections. Both chicken and duck myotubes expressed avian and human sialic acid receptors and were readily susceptible to low-pathogenicity (H2N3 A/mallard duck/England/7277/06) and high-pathogenicity (H5N1 A/turkey/England/50-92/91 and H5N1 A/turkey/Turkey/1/05) avian and human H1N1 (A/USSR/77) influenza viruses. Both avian host species produced comparable levels of progeny H5N1 A/turkey/Turkey/1/05 virus. Notably, the rapid accumulation of viral nucleoprotein and matrix (M) gene RNA in chicken and duck myotubes was accompanied by extensive cytopathic damage with marked myotube apoptosis (widespread microscopic blebs, caspase 3/7 activation, and annexin V binding at the plasma membrane). Infected chicken myotubes produced significantly higher levels of proinflammatory cytokines than did the corresponding duck cells. Additionally, in chicken myotubes infected with H5N1 viruses, the induction of interferon beta (IFN-ß) and IFN-inducible genes, including the melanoma differentiation-associated protein 5 (MDA-5) gene, was relatively weak compared to infection with the corresponding H2N3 virus. Our findings highlight that avian skeletal muscle fibers are capable of productive influenza virus replication and are a potential tissue source of infection. IMPORTANCE: Infection with high-pathogenicity H5N1 viruses in ducks is often asymptomatic, and skeletal muscle from such birds could be a source of infection of humans and animals. Little is known about the ability of influenza A viruses to replicate in avian skeletal muscle fibers. We show here that cultured chicken and duck myotubes were highly susceptible to infection with both low- and high-pathogenicity avian influenza viruses. Infected myotubes of both avian species displayed rapid virus accumulation, apoptosis, and extensive cellular damage. Our results indicate that avian skeletal muscle fibers of chicken and duck could be significant contributors to progeny production of highly pathogenic H5N1 viruses.


Assuntos
Vírus da Influenza A Subtipo H1N1/crescimento & desenvolvimento , Vírus da Influenza A Subtipo H3N2/crescimento & desenvolvimento , Virus da Influenza A Subtipo H5N1/crescimento & desenvolvimento , Fibras Musculares Esqueléticas/virologia , Animais , Apoptose , Células Cultivadas , Galinhas , Citocinas/metabolismo , Efeito Citopatogênico Viral , Patos , Perfilação da Expressão Gênica , RNA Mensageiro/biossíntese , RNA Viral/biossíntese , Receptores Virais/análise , Ácidos Siálicos/análise
9.
Virology ; 458-459: 22-32, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24928036

RESUMO

In mice persistently infected since birth with the prototypic arenavirus lymphocytic choriomeningitis viurs, viral antigen and RNA are readily detected in most organs and cell types but remarkably absent in skeletal muscle. Here we report that mouse C2C12 myoblasts that are readily infected by LCMV, become highly refractory to LCMV infection upon their differentiation into myotubes. Myotube's resistance to LCMV was not due to an intracellular restriction of virus replication but rather an impaired cell entry mediated by the LCMV surface glycoprotein. Our findings provide an explanation for the observation that in LCMV carrier mice myotubes, which are constantly exposed to blood-containing virus, remain free of viral antigen and RNA despite myotubes express high levels of the LCMV receptor alpha dystroglycan and do not pose an intracellular blockade to LCMV multiplication.


Assuntos
Vírus da Coriomeningite Linfocítica/fisiologia , Fibras Musculares Esqueléticas/virologia , Internalização do Vírus , Replicação Viral/fisiologia , Animais , Caveolina 3/genética , Caveolina 3/metabolismo , Linhagem Celular , Regulação da Expressão Gênica , Camundongos , MicroRNAs/genética , MicroRNAs/metabolismo
10.
J Virol ; 88(5): 2414-25, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24335291

RESUMO

UNLABELLED: Chikungunya virus (CHIKV) is an alphavirus transmitted by mosquitoes that is known to cause severe arthritis and myositis in affected patients. The ongoing epidemic began in eastern Africa in 2004 and then spread to islands of the Indian Ocean, India, and Southeast Asia, ultimately afflicting millions. During this outbreak, more severe disease manifestations, including fatalities, have been documented. The reasons for this change in pathogenesis are multifactorial but likely include mutations that have arisen in the viral genome which could alter disease pathogenesis. To test this hypothesis, we used a murine model of CHIKV to compare the disease pathogeneses of two recombinant strains of CHIKV, the first derived from the La Reunion outbreak in 2006 (LR2006 OPY1) and the second isolated from Senegal in 1983 (37997). While the two strains exhibited similar growth in mammalian cells in vitro, we observed more severe clinical disease and pathology in mice infected with the LR2006 OPY1 strain of CHIKV, which included prolonged viremia and elevated viral titers and persistence in the muscle, resulting in devastating myonecrosis. Both CHIKV strains infected connective tissue fibroblasts of the muscle, but only the LR2006 OPY1 strain replicated within myofibers in vivo, despite similar growth of the two strains in these cell types in vitro. However, when the 37997 strain was administered directly into muscle, myofiber infection was comparable to that in LR2006 OPY1-infected mice. These results indicate that differences in the ability of the strain of CHIKV to establish infection in myofibers may contribute to the increased disease severity. IMPORTANCE: CHIKV is an emerging pathogen that causes significant morbidity. Little is known about the pathogenesis of the disease, and this study suggests that the ability of a recent epidemic strain to infect myofibers results in increased disease severity. Better understanding of how CHIKV causes disease contributes to the ultimate goal of creating therapeutics to alleviate the impact of this debilitating virus.


Assuntos
Infecções por Alphavirus/virologia , Vírus Chikungunya/fisiologia , Fibras Musculares Esqueléticas/virologia , Infecções por Alphavirus/metabolismo , Animais , Febre de Chikungunya , Vírus Chikungunya/efeitos dos fármacos , Vírus Chikungunya/patogenicidade , Citocinas/metabolismo , Modelos Animais de Doenças , Fibroblastos/virologia , Genoma Viral , Mediadores da Inflamação/metabolismo , Interferon beta/biossíntese , Interferon beta/farmacologia , Camundongos , Camundongos Knockout , Fibras Musculares Esqueléticas/patologia , Recombinação Genética
11.
PLoS One ; 8(11): e79628, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24223983

RESUMO

Besides the classical respiratory and systemic symptoms, unusual complications of influenza A infection in humans involve the skeletal muscles. Numerous cases of acute myopathy and/or rhabdomyolysis have been reported, particularly following the outbreak of pandemic influenza A(H1N1) in 2009. The pathogenesis of these influenza-associated myopathies (IAM) remains unkown, although the direct infection of muscle cells is suspected. Here, we studied the susceptibility of cultured human primary muscle cells to a 2009 pandemic and a 2008 seasonal influenza A(H1N1) isolate. Using cells from different donors, we found that differentiated muscle cells (i. e. myotubes) were highly susceptible to infection by both influenza A(H1N1) isolates, whereas undifferentiated cells (i. e. myoblasts) were partially resistant. The receptors for influenza viruses, α2-6 and α2-3 linked sialic acids, were detected on the surface of myotubes and myoblasts. Time line of viral nucleoprotein (NP) expression and nuclear export showed that the first steps of the viral replication cycle could take place in muscle cells. Infected myotubes and myoblasts exhibited budding virions and nuclear inclusions as observed by transmission electron microscopy and correlative light and electron microscopy. Myotubes, but not myoblasts, yielded infectious virus progeny that could further infect naive muscle cells after proteolytic treatment. Infection led to a cytopathic effect with the lysis of muscle cells, as characterized by the release of lactate dehydrogenase. The secretion of proinflammatory cytokines by muscle cells was not affected following infection. Our results are compatible with the hypothesis of a direct muscle infection causing rhabdomyolysis in IAM patients.


Assuntos
Vírus da Influenza A Subtipo H1N1/fisiologia , Músculo Esquelético/citologia , Músculo Esquelético/virologia , Pandemias , Estações do Ano , Morte Celular , Diferenciação Celular , Proliferação de Células , Humanos , Fibras Musculares Esqueléticas/citologia , Fibras Musculares Esqueléticas/metabolismo , Fibras Musculares Esqueléticas/virologia , Mioblastos/citologia , Mioblastos/virologia , Receptores de Superfície Celular/metabolismo , Replicação Viral
12.
Mol Ther ; 20(11): 2087-97, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22850678

RESUMO

Gene therapy vectors based on the adeno-associated virus (AAV) are extremely efficient for gene transfer into post-mitotic cells of heart, muscle, brain, and retina. The reason for their exquisite tropism for these cells has long remained elusive. Here, we show that upon terminal differentiation, cardiac and skeletal myocytes downregulate proteins of the DNA damage response (DDR) and that this markedly induces permissivity to AAV transduction. We observed that expression of members of the MRN complex (Mre11, Rad50, Nbs1), which bind the incoming AAV genomes, faded in cardiomyocytes at ~2 weeks after birth, as well as upon myoblast differentiation in vitro; in both cases, withdrawal of the cells from the cell cycle coincided with increased AAV permissivity. Treatment of proliferating cells with short-interfering RNAs (siRNAs) against the MRN proteins, or with microRNA-24, which is normally upregulated upon terminal differentiation and negatively controls the Nbs1 levels, significantly increased permissivity to AAV transduction. Consistently, delivery of these small RNAs to the juvenile liver concomitant with AAV markedly improved in vivo hepatocyte transduction. Collectively, these findings support the conclusion that cellular DDR proteins inhibit AAV transduction and that terminal cell differentiation relieves this restriction.


Assuntos
Diferenciação Celular , Dependovirus/genética , Regulação da Expressão Gênica no Desenvolvimento , Fibras Musculares Esqueléticas/fisiologia , Miócitos Cardíacos/fisiologia , Transdução Genética , Transportadores de Cassetes de Ligação de ATP/genética , Transportadores de Cassetes de Ligação de ATP/metabolismo , Hidrolases Anidrido Ácido , Animais , Ciclo Celular , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Proliferação de Células , Dano ao DNA , Enzimas Reparadoras do DNA/genética , Enzimas Reparadoras do DNA/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Regulação para Baixo , Técnicas de Silenciamento de Genes , Células HeLa , Coração/crescimento & desenvolvimento , Humanos , Fígado/metabolismo , Fígado/virologia , Proteína Homóloga a MRE11 , Camundongos , MicroRNAs/genética , MicroRNAs/metabolismo , Fibras Musculares Esqueléticas/virologia , Miócitos Cardíacos/virologia , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , RNA Interferente Pequeno/genética , Ratos , beta-Galactosidase/biossíntese , beta-Galactosidase/genética
13.
J Med Virol ; 84(8): 1254-66, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22711354

RESUMO

Human adenovirus type-36 (HAdV-36) is a specific pathogen that may lead to increased adiposity and obesity. In order to evaluate the effects of HAdV-36 on gene transcription, a microarray analysis of muscle cells infected with HAdV-36 was performed. Gene expression profile was determined by microarray analysis in cultured human skeletal muscle cells with or without HAdV-36 infection. Quantitative real-time PCR (qPCR) assay was performed in selected 35 genes to verify the results of the microarray analysis. A total of 13,060 unique genes were detected in the HAdV-36 infected muscle cells infected with HAdV-36. Among them, 1,004 genes were significantly altered by using a cut-off point at fold change ≥1.5 and P value <0.05. Most of the principal 100 altered genes were involved in development, immune response, signal transduction, transcriptional regulation as well as carbohydrate, lipid and protein metabolism. Thirty-two genes (91.4%) from the 35 selected genes were confirmed by qPCR assay. In addition, HAdV-36 altered 252 genes that are associated with cancer. The study showed HAdV-36 infection upregulated host cell antiviral defense. HAdV-36 also induces changes in gene expression related to cellular signaling pathways of signal transduction, transcriptional regulation as well as carbohydrate, lipid and protein metabolism. However, it remains to be investigated if HAdV-36 infection could lead to oncogenesis.


Assuntos
Infecções por Adenovirus Humanos/virologia , Adenovírus Humanos/fisiologia , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Fibras Musculares Esqueléticas/metabolismo , Fibras Musculares Esqueléticas/virologia , Proteínas/metabolismo , Adenovírus Humanos/genética , Células Cultivadas , Humanos , Obesidade , Análise de Sequência com Séries de Oligonucleotídeos , Proteínas/genética
14.
J Infect Dis ; 206(3): 407-14, 2012 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-22615321

RESUMO

BACKGROUND: Sindbis virus (SINV) is a mosquito-borne alphavirus found in Eurasia, Africa, and Oceania. Clinical SINV infection is characterized by febrile rash and arthritis and sometimes prolonged arthralgia and myalgia. The pathophysiological mechanisms of musculoskeletal and rheumatic disease caused by SINV are inadequately understood. METHODS: We studied the muscle pathology of SINV infection ex vivo by examining a unique muscle biopsy obtained from a patient with chronic myalgia and arthralgia 6 months after acute SINV infection and assessed potential genetic predisposing factors by determining the human leukocyte antigen (HLA) and complement factor C4 genes and proteins. In addition, we performed in vitro SINV infections of primary human myoblasts and myotubes. RESULTS: In the muscle biopsy we found evidence of muscle regeneration due to previous necrotic lesions likely caused by earlier SINV infection. We showed that human myoblasts and myotubes were susceptible in vitro for SINV infection as the cells became immunoreactive for viral antigens and cytopathic effect was observed. The patient was homozygous for HLA-B*35 alleles and heterozygous for HLA-DRB1*01 and HLA-DRB1*03 alleles and had total deficiency of C4B protein. CONCLUSIONS: This study provides new insights concerning pathological processes leading to chronic symptoms in SINV infection and demonstrates for the first time the susceptibility of human myogenic cells to SINV infection.


Assuntos
Infecções por Alphavirus/complicações , Fibras Musculares Esqueléticas/virologia , Doenças Musculares/virologia , Mioblastos/virologia , Dor/complicações , Sindbis virus , Humanos , Masculino , Pessoa de Meia-Idade , Dor/virologia , RNA Viral/isolamento & purificação , Reação em Cadeia da Polimerase em Tempo Real/métodos , Reação em Cadeia da Polimerase Via Transcriptase Reversa/métodos
15.
Exp Cell Res ; 316(11): 1784-94, 2010 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-20362571

RESUMO

We examined the progression of the WSN influenza virus infection in isolated, multinucleated rat skeletal myofibers. Contrary to mononucleated cells, the adsorbed virions showed markedly delayed entry kinetics. Viral budding occurred on the sarcolemma, but the hemagglutinin envelope glycoprotein matured inefficiently and was poorly cleaved. Compatible with this, plaque assays indicated that infective viral particles were not formed. In situ hybridization studies showed that at low-dose infection, viral RNA production was restricted to one or a few nuclei within a myofiber. Dual in situ hybridization indicated that two different viral RNAs usually co-localized in the same nucleus or nuclei, suggesting that different viral genome segments replicated in the same nucleus. Newly synthesized viral ribonucleoprotein particles (vRNPs) did not re-enter virgin nuclei. Therefore, a single infected nucleus was able to support viral protein production, and notably, these proteins could reach hundreds of micrometers from the nucleus of origin. These results suggest that after viral disassembly in the endosome, the genome segments remained glued together and entered a myonucleus as a package. Spreading of the infection into virgin nuclei either by vRNPs or newly made virions did not occur, and thus the infection was abortive.


Assuntos
Fibras Musculares Esqueléticas/virologia , Infecções por Orthomyxoviridae/virologia , Orthomyxoviridae/patogenicidade , Animais , Sequência de Bases , Núcleo Celular/virologia , Feminino , Hibridização In Situ , Técnicas In Vitro , Microscopia Eletrônica de Transmissão , Modelos Biológicos , Fibras Musculares Esqueléticas/ultraestrutura , Orthomyxoviridae/genética , Orthomyxoviridae/fisiologia , Orthomyxoviridae/ultraestrutura , Infecções por Orthomyxoviridae/patologia , RNA Viral/genética , RNA Viral/metabolismo , Ratos , Sarcolema/ultraestrutura , Sarcolema/virologia , Proteínas Virais/metabolismo , Internalização do Vírus , Liberação de Vírus
16.
Am J Vet Res ; 69(12): 1637-45, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19046012

RESUMO

OBJECTIVE: To develop a reliable method for converting cultured equine skin-derived fibroblasts into muscle cells. SAMPLE POPULATION: Equine skin-derived fibroblasts. PROCEDURES: The equine myogenic differentiation 1 (eqMyoD) genomic sequence was obtained by use of equine bacterial artificial chromosome screening and PCR sequencing. Total mRNA was extracted from foal skeletal muscle, and eqMyoD cDNA was cloned into a plasmid vector with an internal ribosomal entry site to express bicistronic eqMyoD or enhanced green fluorescent protein (EGFP). Transient expression was confirmed by immunocytochemical analysis and western immunoblots in equine fibroblasts and fibroblasts from National Institutes of Health Swiss mouse embryos, prior to generation of a lentiviral vector containing the same coding sequences. Transformation of equine skin-derived cells into skeletal myotubes was examined by use of immunohistochemical analysis, western immunoblotting, and periodic acid-Schiff staining. RESULTS: eqMyoD mRNA consists of 960 bp and shares high homology with myogenic differentiation 1 from other mammals. Transfection confirmed the expression of a 53-kd protein with mainly nuclear localization. Lentiviral transduction was efficient, with approximately 80% of EGFP-positive cells transformed into multinucleated myotubes during 15 days, as determined by expression of the muscle-specific proteins desmin, troponin-T, and sarcomeric myosin and by cytoplasmic storage of glycogen. CONCLUSIONS AND CLINICAL RELEVANCE: Equine primary fibroblasts were transformed by lentiviral transduction of eqMyoD into fusion-competent myoblasts. This may offer a preferable alternative to primary myoblast cultures for the investigation of cellular defects associated with muscle diseases of horses, such as recurrent exertional rhabdomyolysis and polysaccharide storage myopathy.


Assuntos
Fibroblastos/citologia , Cavalos , Lentivirus/fisiologia , Fibras Musculares Esqueléticas/citologia , Proteína MyoD/metabolismo , Pele/citologia , Células 3T3 , Sequência de Aminoácidos , Animais , Regulação da Expressão Gênica/fisiologia , Humanos , Camundongos , Dados de Sequência Molecular , Fibras Musculares Esqueléticas/metabolismo , Fibras Musculares Esqueléticas/virologia , Proteína MyoD/genética
17.
J Fish Dis ; 31(11): 825-34, 2008 11.
Artigo em Inglês | MEDLINE | ID: mdl-19238757

RESUMO

Two new cell lines have been established from the muscle and swim bladder tissues of barramundi, Lates calcarifer, and designated as BM (barramundi muscle) and BSB (barramundi swimbladder), respectively. The cells multiplied well at 28 degrees C in Leibovitz's L-15 medium supplemented with 10% foetal bovine serum, and have been continuously subcultured more than 100 times to date. Morphologically, BM cells were mostly fibroblastic, whereas BSB were mostly epithelial. Both cell lines were susceptible to grouper iridovirus (GIV) and displayed characteristics of apoptosis after viral infection. The induction of apoptosis was further assayed in GIV-infected BM and BSB cells by various methods. The inhibition of cell growth by GIV was demonstrated by MTT [3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide] assay. Morphological observations revealed typical apoptotic features in the infected cells, including cell shrinkage and rounding, chromosome condensation and formation of apoptotic body-like vesicles. Chromosome fragmentation was detected by DNA laddering and TUNEL assays. Finally, the appearance of phosphotidylserine on the outer leaflet of apoptotic cell membranes was confirmed by annexin V staining. This is the first report of apoptosis induced by GIV in fish cells.


Assuntos
Sacos Aéreos/citologia , Apoptose/fisiologia , Efeito Citopatogênico Viral/fisiologia , Iridovirus/classificação , Fibras Musculares Esqueléticas/virologia , Perciformes/fisiologia , Animais , Linhagem Celular , Fatores de Tempo
18.
Gene Ther ; 14(17): 1249-60, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17581597

RESUMO

Using murine models, we have previously demonstrated that recombinant adeno-associated virus (rAAV)-mediated microdystrophin gene transfer is a promising approach to treatment of Duchenne muscular dystrophy (DMD). To examine further therapeutic effects and the safety issue of rAAV-mediated microdystrophin gene transfer using larger animal models, such as dystrophic dog models, we first investigated transduction efficiency of rAAV in wild-type canine muscle cells, and found that rAAV2 encoding beta-galactosidase effectively transduces canine primary myotubes in vitro. Subsequent rAAV2 transfer into skeletal muscles of normal dogs, however, resulted in low and transient expression of beta-galactosidase together with intense cellular infiltrations in vivo, where cellular and humoral immune responses were remarkably activated. In contrast, rAAV2 expressing no transgene elicited no cellular infiltrations. Co-administration of immunosuppressants, cyclosporine and mycophenolate mofetil could partially improve rAAV2 transduction. Collectively, these results suggest that immune responses against the transgene product caused cellular infiltration and eliminated transduced myofibers in dogs. Furthermore, in vitro interferon-gamma release assay showed that canine splenocytes respond to immunogens or mitogens more susceptibly than murine ones. Our results emphasize the importance to scrutinize the immune responses to AAV vectors in larger animal models before applying rAAV-mediated gene therapy to DMD patients.


Assuntos
Dependovirus/genética , Terapia Genética/efeitos adversos , Vetores Genéticos/efeitos adversos , Músculo Esquelético/imunologia , Distrofia Muscular Animal/terapia , Distrofia Muscular de Duchenne/terapia , Animais , Sequência de Bases , Calmodulina/genética , Ciclosporina/administração & dosagem , Cães , Distrofina/genética , Distrofina/metabolismo , Engenharia Genética , Terapia Genética/métodos , Vetores Genéticos/genética , Imunossupressores/administração & dosagem , Injeções Intramusculares , Interferon gama/imunologia , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos C57BL , Modelos Animais , Dados de Sequência Molecular , Fibras Musculares Esqueléticas/imunologia , Fibras Musculares Esqueléticas/virologia , Distrofia Muscular Animal/imunologia , Distrofia Muscular de Duchenne/imunologia , Infecções por Parvoviridae/imunologia , RNA Mensageiro/análise , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Especificidade da Espécie , Linfócitos T Citotóxicos/imunologia , Transdução Genética/métodos , Transgenes , beta-Galactosidase/genética
19.
Am J Med Sci ; 332(2): 88-89, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16909056

RESUMO

A patient with rhabdomyolysis-induced acute renal failure due to influenza B virus infection is presented. Influenza B infection caused rhabdomyolysis with efflux of myoglobin from myocytes, causing acute renal failure. In conclusion, influenza virus type B can cause severe rhabdomyolysis leading to acute renal failure.


Assuntos
Injúria Renal Aguda/etiologia , Vírus da Influenza B , Influenza Humana/complicações , Rabdomiólise/etiologia , Injúria Renal Aguda/sangue , Injúria Renal Aguda/virologia , Adulto , Humanos , Influenza Humana/sangue , Masculino , Fibras Musculares Esqueléticas/metabolismo , Fibras Musculares Esqueléticas/virologia , Mioglobina/metabolismo , Rabdomiólise/sangue , Rabdomiólise/virologia
20.
Hum Gene Ther ; 17(2): 193-205, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16454653

RESUMO

Until recently, adenovirus-based gene therapy has been almost exclusively based on human adenovirus serotype 5 (Ad5). The aim of this study was to systematically compare the efficiency of transduction of primary muscle cells from various species by two adenoviral vectors from subgroups C and D. Transduction of a panel of myoblasts demonstrated a striking specificity of an Ad19a-based replication-defective E1-deleted vector (Ad19aEGFP) for human cells, whereas the Ad5-based vector had high affinity for nonhuman primate myoblasts. Transgene expression correlated well with cell-associated vector genomes. Up to 6.59% of the initially applied Ad19aEGFP vector particles were taken up by human myoblasts, as compared with 0.1% of the corresponding Ad5 vector. Remarkably, Ad19aEGFP but not Ad5EGFP efficiently transduced differentiated human myotubes, an in vitro model for skeletal muscle transduction. Uptake of Ad19aEGFP vector particles in human myotubes was 12-fold more efficient than that of Ad5EGFP. Moreover, both vectors demonstrated an early block at the level of vector uptake in mouse myoblasts and rat L6 cells. Investigation of the underlying mechanism for binding and uptake of the two vectors by human myoblasts showed high susceptibility for Ad19a to neuraminidase and wheat germ agglutinin (WGA) lectin, whereas Ad5-mediated transduction was dependent on binding to the coxsackie-adenovirus receptor (CAR) and sensitive to soluble RGD peptide and heparin. Our study offers insights into species-dependent factors that determine Ad tropism and, moreover, provides a basis for application of the novel Ad19a-based vector for gene transfer into human skeletal muscle.


Assuntos
Adenovírus Humanos/genética , Terapia Genética/métodos , Vetores Genéticos/farmacologia , Músculo Esquelético/virologia , Animais , Células Cultivadas , Proteína de Membrana Semelhante a Receptor de Coxsackie e Adenovirus , Epitopos/química , Epitopos/metabolismo , Citometria de Fluxo/métodos , Vetores Genéticos/genética , Heparitina Sulfato/metabolismo , Humanos , Camundongos , Fibras Musculares Esqueléticas/citologia , Fibras Musculares Esqueléticas/virologia , Músculo Esquelético/citologia , Músculo Esquelético/fisiologia , Mioblastos/citologia , Mioblastos/virologia , Ácido N-Acetilneuramínico/química , Ácido N-Acetilneuramínico/metabolismo , Ratos , Receptores Virais/química , Receptores Virais/metabolismo , Especificidade da Espécie , Transdução Genética , Tropismo , Replicação Viral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA