Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
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
2.
BMC Genomics ; 21(1): 45, 2020 Jan 14.
Artigo em Inglês | MEDLINE | ID: mdl-31937237

RESUMO

BACKGROUND: Clostridium perfringens is a Gram-positive anaerobic pathogen that causes multiple diseases in humans and animals. C. perfringens lack flagella but have type IV pili (TFP) and can glide on agar surfaces. When C. perfringens bacteria are placed on surfaces, they become elongated, flexible and have TFP on their surface, traits not seen in liquid-grown cells. In addition, the main pilin in C. perfringens TFP, PilA2, undergoes differential post-translational modification when grown in liquid or on plates. To understand the mechanisms underlying these phenotypes, bacteria were grown in three types of liquid media and on agar plates with the same medium to compare gene expression using RNA-Seq. RESULTS: Hundreds of genes were differentially expressed, including transcriptional regulatory protein-encoding genes and genes associated with TFP functions, which were higher on plates than in liquid. Transcript levels of TFP genes reflected the proportion of each protein predicted to reside in a TFP assembly complex. To measure differences in rates of translation, the Escherichia coli reporter gene gusA gene (encoding ß-glucuronidase) was inserted into the chromosome downstream of TFP promoters and in-frame with the first gene of the operon. ß-glucuronidase expression was then measured in cells grown in liquid or on plates. ß-glucuronidase activity was proportional to mRNA levels in liquid-grown cells, but not plate-grown cells, suggesting significant levels of post-transcriptional regulation of these TFP-associated genes occurs when cells are grown on surfaces. CONCLUSIONS: This study reveals insights into how a non-flagellated pathogenic rod-shaped bacterium senses and responds to growth on surfaces, including inducing transcriptional regulators and activating multiple post-transcriptional regulatory mechanisms associated with TFP functions.


Assuntos
Clostridium perfringens/fisiologia , Proteínas de Fímbrias/genética , Regulação Bacteriana da Expressão Gênica , Animais , Aderência Bacteriana , Toxinas Bacterianas/genética , Sequência de Bases , Perfilação da Expressão Gênica , Ordem dos Genes , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Camundongos , Mioblastos/virologia , Óperon , Regiões Promotoras Genéticas , Temperatura , Transcriptoma
3.
Nature ; 574(7777): 259-263, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31554973

RESUMO

Chikungunya virus (CHIKV) is a re-emerging alphavirus that is transmitted to humans by mosquito bites and causes musculoskeletal and joint pain1,2. Despite intensive investigations, the human cellular factors that are critical for CHIKV infection remain unknown, hampering the understanding of viral pathogenesis and the development of anti-CHIKV therapies. Here we identified the four-and-a-half LIM domain protein 1 (FHL1)3 as a host factor that is required for CHIKV permissiveness and pathogenesis in humans and mice. Ablation of FHL1 expression results in the inhibition of infection by several CHIKV strains and o'nyong-nyong virus, but not by other alphaviruses and flaviviruses. Conversely, expression of FHL1 promotes CHIKV infection in cells that do not normally express it. FHL1 interacts directly with the hypervariable domain of the nsP3 protein of CHIKV and is essential for the replication of viral RNA. FHL1 is highly expressed in CHIKV-target cells and is particularly abundant in muscles3,4. Dermal fibroblasts and muscle cells derived from patients with Emery-Dreifuss muscular dystrophy that lack functional FHL15 are resistant to CHIKV infection. Furthermore,  CHIKV infection  is undetectable in Fhl1-knockout mice. Overall, this study shows that FHL1 is a key factor expressed by the host that enables CHIKV infection and identifies the interaction between nsP3 and FHL1 as a promising target for the development of anti-CHIKV therapies.


Assuntos
Febre de Chikungunya/virologia , Vírus Chikungunya/patogenicidade , Fatores Celulares Derivados do Hospedeiro/metabolismo , Interações Hospedeiro-Patógeno , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas com Domínio LIM/metabolismo , Proteínas Musculares/metabolismo , Animais , Células Cultivadas , Febre de Chikungunya/tratamento farmacológico , Vírus Chikungunya/efeitos dos fármacos , Vírus Chikungunya/genética , Vírus Chikungunya/crescimento & desenvolvimento , Feminino , Fibroblastos/virologia , Células HEK293 , Fatores Celulares Derivados do Hospedeiro/genética , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/deficiência , Peptídeos e Proteínas de Sinalização Intracelular/genética , Proteínas com Domínio LIM/deficiência , Proteínas com Domínio LIM/genética , Masculino , Camundongos , Proteínas Musculares/deficiência , Proteínas Musculares/genética , Mioblastos/virologia , Vírus O'nyong-nyong/crescimento & desenvolvimento , Vírus O'nyong-nyong/patogenicidade , Ligação Proteica , RNA Viral/biossíntese , Proteínas não Estruturais Virais/genética , Proteínas não Estruturais Virais/metabolismo , Replicação Viral
4.
J Fish Dis ; 41(9): 1331-1338, 2018 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-30003544

RESUMO

A continuous cell line consisting mostly of epithelioid cells was established from the caudal fin of marbled eels (Anguilla marmorata) and designated as marbled eel caudal fin (MECF)-1. The cells multiplied well in Leibovitz's L-15 medium containing 2% to 15% foetal bovine serum at temperatures of 20°C to 35°C and were subcultured for >90 passages during a 5-year period from 2012 to 2017. Transcripts of ictacalcin, keratin 13, cd146, nestin, ncam1 and myod1 were demonstrated in the cells using reverse transcription polymerase chain reaction. The results indicated that MECF-1 was composed of epidermal and mesenchyme stem and progenitor cells including myoblasts. MECF-1 was susceptible to Japanese eel herpesvirus HVA980811, marbled eel polyoma-like virus (MEPyV), aquabirnavirus MEIPNV1310 and aquareovirus CSV. By contrast, MECF-1 was noted refractory to megalocytiviruses RSIV-Ku and GSIV-K1 infection. Moreover, the cells were resistant to betanodavirus infection. In conclusion, MECF-1 derived from marbled eel is suitable for studies on anguillid viruses and interaction with host cells.


Assuntos
Anguilla/anatomia & histologia , Anguilla/virologia , Nadadeiras de Animais/citologia , Nadadeiras de Animais/virologia , Linhagem Celular/virologia , Técnicas de Cultura de Tecidos , Animais , Técnicas de Cultura de Células/veterinária , Linhagem Celular/citologia , Meios de Cultura/química , Suscetibilidade a Doenças , Células Epidérmicas , Epiderme/virologia , Doenças dos Peixes/virologia , Herpesviridae/fisiologia , Mioblastos/virologia , Polyomavirus/fisiologia , Reoviridae/fisiologia
5.
J Gen Virol ; 98(12): 3008-3025, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-29120300

RESUMO

Enterovirus 71 (EV71) has caused major outbreaks of hand, foot and mouth disease. EV71 infections increase the production of many host cytokines and pro-inflammatory factors, including interleukin (IL)-6, IL-10 and COX-2. Some of these molecules could stimulate the signal transducer and activator of transcription 3 (STAT3), which plays a key role in regulating host immune responses and several viral diseases. However, the role of STAT3 in EV71 infection remains unknown. This study found that the phosphorylation levels of STAT3 (pY705-STAT3) are closely related to EV71 infection. Further experiments revealed that STAT3 exerts an anti-EV71 activity. However, the antiviral activity of STAT3 is partially antagonized by EV71-induced miR-124, which directly targets STAT3 mRNA. Similarly, IL-6R, the α-subunit of the IL-6 receptor complex, exhibits anti-EV71 activity and is directly targeted by the virus-induced miR-124. These results indicate that EV71 can evade host IL-6R- and STAT3-mediated antiviral activities by EV71-induced miR-124. This suggests that controlling miR-124 and the downstream targets, IL-6R and STAT3, might benefit the antiviral treatment of EV71 infection.


Assuntos
Enterovirus Humano A/genética , Evasão da Resposta Imune , MicroRNAs/genética , RNA Mensageiro/genética , Receptores de Interleucina-6/genética , Fator de Transcrição STAT3/genética , Sequência de Bases , Sítios de Ligação , Linhagem Celular Tumoral , Ciclo-Oxigenase 2/genética , Ciclo-Oxigenase 2/imunologia , Enterovirus Humano A/crescimento & desenvolvimento , Enterovirus Humano A/imunologia , Regulação da Expressão Gênica , Células HEK293 , Células HeLa , Humanos , Interleucina-10/genética , Interleucina-10/imunologia , Interleucina-6/genética , Interleucina-6/imunologia , MicroRNAs/imunologia , Mioblastos/imunologia , Mioblastos/virologia , Neurônios/imunologia , Neurônios/virologia , Fosforilação , RNA Mensageiro/imunologia , Receptores de Interleucina-6/imunologia , Fator de Transcrição STAT3/imunologia , Transdução de Sinais , Replicação Viral
6.
Am J Physiol Regul Integr Comp Physiol ; 313(3): R240-R250, 2017 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-28637658

RESUMO

Skeletal muscle stem cells play a critical role in regeneration of myofibers. We previously demonstrated that chronic binge alcohol (CBA) markedly attenuates myoblast differentiation potential and myogenic gene expression. Muscle-specific microRNAs (miRs) are implicated in regulation of myogenic genes. The aim of this study was to determine whether myoblasts isolated from asymptomatic CBA-administered simian immunodeficiency virus (SIV)-infected macaques treated with antiretroviral therapy (ART) showed similar impairments and, if so, to elucidate potential underlying mechanisms. Myoblasts were isolated from muscle at 11 mo after SIV infection from CBA/SIV macaques and from time-matched sucrose (SUC)-treated SIV-infected (SUC/SIV) animals and age-matched controls. Myoblast differentiation and myogenic gene expression were significantly decreased in myoblasts from SUC/SIV and CBA/SIV animals compared with controls. SIV and CBA decreased muscle-specific miR-206 in plasma and muscle and SIV decreased miR-206 expression in myoblasts, with no statistically significant changes in other muscle-specific miRs. These findings were associated with a significant increase in histone deacetylase 4 (HDAC4) and decrease in myogenic enhancer factor 2C (MEF2C) expression in CBA/SIV muscle. Transfection with miR-206 inhibitor decreased myotube differentiation, increased expression of HDAC4, and decreased MEF2C, suggesting a critical role of miR-206 in myogenesis. Moreover, HDAC4 was confirmed to be a direct miR-206 target. These results support a mechanistic role for decreased miR-206 in suppression of myoblast differentiation resulting from chronic alcohol and SIV infection. The parallel changes in skeletal muscle and circulating levels of miR-206 warrant studies to establish the possible use of plasma miR-206 as an indicator of impaired muscle function.


Assuntos
Consumo Excessivo de Bebidas Alcoólicas/fisiopatologia , Diferenciação Celular , MicroRNAs/metabolismo , Desenvolvimento Muscular , Mioblastos/citologia , Mioblastos/patologia , Síndrome de Imunodeficiência Adquirida dos Símios/fisiopatologia , Animais , Consumo Excessivo de Bebidas Alcoólicas/patologia , Consumo Excessivo de Bebidas Alcoólicas/virologia , Regulação para Baixo , Macaca mulatta , Masculino , MicroRNAs/genética , Mioblastos/virologia , Síndrome de Imunodeficiência Adquirida dos Símios/patologia , Síndrome de Imunodeficiência Adquirida dos Símios/virologia
7.
Skelet Muscle ; 7(1): 7, 2017 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-28506310

RESUMO

BACKGROUND: shRNA lentiviral vectors are extensively used for gene knockdowns in mammalian cells, and non-target shRNAs typically are considered the proper experimental control for general changes caused by RNAi. However, the effects of non-target lentivirus controls on the modulation of cell signaling pathways remain largely unknown. In this study, we evaluated the effect of control lentiviral transduction on oxytocin receptor (OXTR) expression through the ERK/MAPK pathway in mouse and human skeletal muscle cells, on myogenic activity, and in vivo on mouse muscle regeneration. Furthermore, we mined published data for the influence of viral infections on OXTR levels in human populations and found that unrelated viral pathologies have a common consequence: diminished levels of OXTR. METHODS: We examined the change in OXTR mRNA expression upon transduction with control and Smad3-targeting viral vectors through real time RT-PCR and Western blotting, and confirmed with immunofluorescence. Changes in Smad3 and OXTR expression were examined both in vitro with mouse and human myoblasts and in vivo in mouse satellite cells. The general effects of viral infections on OXTR downregulation in humans were also examined by analyzing published Gene Expression Omnibus (GEO) datasets. The change in myoblast myogenic activity caused by the viral transduction (the percent of Pax7 + Ki67+ cells) was examined by immunofluorescence. RESULTS: Results shown in this work establish that lentiviral control vectors significantly downregulate OXTR expression at mRNA and protein levels and diminish key downstream effectors of OXTR, ERK signaling, reducing the myogenic proliferation of infected cells. This effect is evolutionarily conserved between mouse and human myogenic cells, and it manifests in satellite cells after control lentiviral transduction of mice in vivo. Furthermore, an examination of published datasets uncovered similar OXTR downregulation in humans that are afflicted with different viral infections. Additionally, cells transduced with Smad3-targeting shRNA downregulate OXTR even more than cells transduced with control viruses. CONCLUSIONS: Our work suggests that experimental cohorts transduced with control viruses may not behave the same as un-transduced cells and animals, specifically that control viral vectors significantly change the intensity of key cell-signaling pathways, such as OXTR/ERK. Our results further demonstrate that lentiviral transduction significantly decreases myogenic proliferation and suggest that viral infections in general may play a role in decreasing muscle health and regeneration, a decline in metabolic health, and a lower sense of well-being, as these rely on effective OXTR signaling. Additionally, our data suggest pathway crosstalk between TGF-ß/pSmad3 and OXTR, implying that sustained attenuation of the TGF-ß/pSmad3 pathway will reduce pro-regenerative OXTR/pERK signaling.


Assuntos
Receptores de Ocitocina/genética , Transdução de Sinais , Proteína Smad3/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Viroses/metabolismo , Animais , Células Cultivadas , Regulação para Baixo , Evolução Molecular , Humanos , Lentivirus/patogenicidade , Camundongos , Camundongos Endogâmicos C57BL , Mioblastos/metabolismo , Mioblastos/virologia , Receptores de Ocitocina/metabolismo , Células Satélites de Músculo Esquelético/metabolismo , Células Satélites de Músculo Esquelético/virologia , Viroses/genética
8.
PLoS One ; 12(1): e0170080, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28095450

RESUMO

Infectious bursal disease virus (IBDV), a member of the Birnaviridae family, is responsible for a devastating immunosuppressive disease affecting juvenile domestic chickens. IBDV particles are naked icosahedrons enclosing a bipartite double-stranded RNA genome harboring three open reading frames (ORF). One of these ORFs codes for VP5, a non-structural polypeptide dispensable for virus replication in tissue culture but essential for IBDV pathogenesis. Using two previously described recombinant viruses, whose genomes differ in a single nucleotide, expressing or not the VP5 polypeptide, we have analyzed the role of this polypeptide during the IBDV replication process. Here, we show that VP5 is not involved in house-keeping steps of the virus replication cycle; i.e. genome transcription/replication, protein translation and virus assembly. Although infection with the VP5 expressing and non-expressing viruses rendered similar intracellular infective progeny yields, striking differences were detected on the ability of their progenies to exiting infected cells. Experimental data shows that the bulk of the VP5-expressing virus progeny efficiently egresses infected cells during the early phase of the infection, when viral metabolism is peaking and virus-induced cell death rates are as yet minimal, as determined by qPCR, radioactive protein labeling and quantitative real-time cell death analyses. In contrast, the release of the VP5-deficient virus progeny is significantly abridged and associated to cell death. Taken together, data presented in this report show that IBDV uses a previously undescribed VP5-dependent non-lytic egress mechanism significantly enhancing the virus dissemination speed. Ultrastructural analyses revealed that newly assembled IBDV virions associate to a vesicular network apparently facilitating their trafficking from virus assembly factories to the extracellular milieu, and that this association requires the expression of the VP5 polypeptide.


Assuntos
Infecções por Birnaviridae/virologia , Vírus da Doença Infecciosa da Bursa/patogenicidade , Proteínas não Estruturais Virais/metabolismo , Vírion/metabolismo , Liberação de Vírus/fisiologia , Replicação Viral , Animais , Infecções por Birnaviridae/metabolismo , Células Cultivadas , Músculo Esquelético/citologia , Músculo Esquelético/metabolismo , Músculo Esquelético/virologia , Mioblastos/citologia , Mioblastos/metabolismo , Mioblastos/virologia , Codorniz
9.
Antimicrob Agents Chemother ; 60(9): 5357-67, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27353263

RESUMO

Enterovirus 71 (EV-A71) is a major causative pathogen of hand, foot, and mouth disease (HFMD) epidemics. No antiviral therapies are currently available for treating EV-A71 infections. Here, we selected five reported enterovirus inhibitors (suramin, itraconazole [ITZ], GW5074, rupintrivir, and favipiravir) with different mechanisms of action to test their abilities to inhibit EV-A71 replication alone and in combination. All selected compounds have anti-EV-A71 activities in cell culture. The combination of rupintrivir and ITZ or favipiravir was synergistic, while the combination of rupintrivir and suramin was additive. The combination of suramin and favipiravir exerted a strong synergistic antiviral effect. The observed synergy was not due to cytotoxicity, as there was no significant increase in cytotoxicity when compounds were used in combinations at the tested doses. To investigate the potential inhibitory mechanism of favipiravir against enterovirus, two favipiravir-resistant EV-A71 variants were independently selected, and both of them carried an S121N mutation in the finger subdomain of the 3D polymerase. Reverse engineering of this 3D S121N mutation into an infectious clone of EV-A71 confirmed the resistant phenotype. Moreover, viruses resistant to ITZ or favipiravir remained susceptible to other inhibitors. Most notably, combined with ITZ, rupintrivir prevented the development of ITZ-resistant variants. Taken together, these results provide a rational basis for the design of combination regimens for use in the treatment of EV-A71 infections.


Assuntos
Antivirais/farmacologia , Enterovirus Humano A/efeitos dos fármacos , Isoxazóis/farmacologia , Itraconazol/farmacologia , Pirrolidinonas/farmacologia , Suramina/farmacologia , Proteínas não Estruturais Virais/genética , Amidas/farmacologia , Sequência de Aminoácidos , Animais , Sítios de Ligação , Linhagem Celular Tumoral , Chlorocebus aethiops , Combinação de Medicamentos , Farmacorresistência Viral/genética , Sinergismo Farmacológico , Enterovirus Humano A/genética , Enterovirus Humano A/crescimento & desenvolvimento , Humanos , Indóis/farmacologia , Simulação de Acoplamento Molecular , Mutação , Mioblastos/efeitos dos fármacos , Mioblastos/virologia , Fenóis/farmacologia , Fenilalanina/análogos & derivados , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Estrutura Secundária de Proteína , Pirazinas/farmacologia , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Valina/análogos & derivados , Células Vero , Proteínas não Estruturais Virais/antagonistas & inibidores , Proteínas não Estruturais Virais/metabolismo , Replicação Viral/efeitos dos fármacos
10.
PLoS One ; 10(7): e0132099, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26154387

RESUMO

Myogenesis is defined as growth, differentiation and repair of muscles where cell fusion of myoblasts to multinucleated myofibers is one major characteristic. Other cell fusion events in humans are found with bone resorbing osteoclasts and placental syncytiotrophoblasts. No unifying gene regulation for natural cell fusions has been found. We analyzed skeletal muscle biopsies of competitive cyclists for muscle-specific attributes and expression of human endogenous retrovirus (ERV) envelope genes due to their involvement in cell fusion of osteoclasts and syncytiotrophoblasts. Comparing muscle biopsies from post- with the pre-competitive seasons a significant 2.25-fold increase of myonuclei/mm fiber, a 2.38-fold decrease of fiber area/nucleus and a 3.1-fold decrease of satellite cells (SCs) occurred. We propose that during the pre-competitive season SC proliferation occurred following with increased cell fusion during the competitive season. Expression of twenty-two envelope genes of muscle biopsies demonstrated a significant increase of putative muscle-cell fusogenic genes Syncytin-1 and Syncytin-3, but also for the non-fusogenic erv3. Immunohistochemistry analyses showed that Syncytin-1 mainly localized to the sarcolemma of myofibers positive for myosin heavy-chain isotypes. Cellular receptors SLC1A4 and SLC1A5 of Syncytin-1 showed significant decrease of expression in post-competitive muscles compared with the pre-competitive season, but only SLC1A4 protein expression localized throughout the myofiber. Erv3 protein was strongly expressed throughout the myofiber, whereas envK1-7 localized to SC nuclei and myonuclei. Syncytin-1 transcription factors, PPARγ and RXRα, showed no protein expression in the myofiber, whereas the pCREB-Ser133 activator of Syncytin-1 was enriched to SC nuclei and myonuclei. Syncytin-1, Syncytin-3, SLC1A4 and PAX7 gene regulations along with MyoD1 and myogenin were verified during proliferating or actively-fusing human primary myoblast cell cultures, resembling muscle biopsies of cyclists. Myoblast treatment with anti-Synycytin-1 abrogated cell fusion in vitro. Our findings support functional roles for ERV envelope proteins, especially Syncytin-1, contributing to cell fusion of myotubes.


Assuntos
Retrovirus Endógenos/genética , Exercício Físico , Genes Virais , Mioblastos/citologia , Mioblastos/virologia , Resistência Física , Adolescente , Ciclismo , Fusão Celular , Células Cultivadas , Crioultramicrotomia , Imunofluorescência , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Humanos , Masculino , Fibras Musculares Esqueléticas/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Células Satélites de Músculo Esquelético/metabolismo , Fatores de Tempo
11.
J Med Virol ; 87(5): 733-9, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25656089

RESUMO

In addition to fever, rash, and arthralgia/arthritis, myalgia is another dominant symptom in Chikungunya virus (CHIKV) infection. How CHIKV induces myalgia is unclear. To better understand the viral factors involved in CHIKV-induced myalgia, CHIKVs were isolated from patients with and without myalgia designated myalgia-CHIKV and mild-CHIKV, respectively. The response of myoblasts to infection by the two groups of clinical isolates of CHIKV was investigated. Both groups of CHIKV replicated well in primary human myoblasts. However, the myalgia-CHIKVs replicated to a higher titer and caused the death of infected myoblast more rapidly than the mild-CHIKVs. CHIKV-infected myoblasts increased production of four out of five inflammatory cytokines examined (MCP-1, IP-10, MIP-1α, and IL-8) in comparison to mock-infected cells. Comparison between the myoblast inflammatory cytokine responses showed that myalgia-CHIKVs were stronger activators of cytokines than mild-CHIKVs. This means that recent epidemic strains of CHIKV exhibited different degrees of myoblast permissiveness as evidenced by differences in the ability to replicate and to stimulate inflammatory responses in myoblasts. This data suggest that the myopathic syndrome in recent epidemics is dependent upon the strain of CHIKV.


Assuntos
Febre de Chikungunya/virologia , Vírus Chikungunya/fisiologia , Mialgia/virologia , Mioblastos/imunologia , Mioblastos/virologia , Replicação Viral , Adulto , Células Cultivadas , Febre de Chikungunya/epidemiologia , Febre de Chikungunya/patologia , Vírus Chikungunya/crescimento & desenvolvimento , Vírus Chikungunya/isolamento & purificação , Citocinas/metabolismo , Humanos , Carga Viral
12.
Avian Pathol ; 43(5): 414-9, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25204414

RESUMO

Marek's disease virus (MDV; also known as Gallid herpesvirus 2, MDV-1) causes oncogenic disease in chickens producing clinical signs that include lymphomas, visceral tumours, nerve lesions, and immunosuppression. MDV vaccines are widely used and mostly produced using primary cells: chicken embryo fibroblast cells, duck embryo fibroblast cells, chicken embryo kidney cells or chicken kidney cells. An immortalized cell line that can be used to manufacture the virus has long been desired. In this report, we demonstrate that QM7 cells were susceptible to infection with either MDV or herpesvirus of turkey (HVT; also known as Meleagrid herpesvirus 1, MDV-3). Polymerase chain reaction analysis with primers amplifying selected MDV genes revealed that QM7 cells did not contain these sequences. However, MDV genes were detected in QT35 cells, which have been reported to harbour latent MDV virus. Transfection of naked MDV DNA initiated efficient infection of QM7 cells. In addition, QM7 cell lysate, clarified supernatant, and QM7 cell pellet infected with MDV were negative for reverse transcriptase activity, indicating absence of endogenous retrovirus. QM7 cells were also found to be free of other avian pathogens in a chick embryo inoculation test. In vivo studies of MDV growing in QM7 cells showed the virus retained its pathogenicity and virulence. In ovo experiments demonstrated that both HVT and MDV propagated in QM7 cells did not interfere with hatchability of injected eggs, and viruses could be re-isolated from hatched chicks. The results suggest that QM7 could be a good host cell line for growing both MDV and HVT.


Assuntos
Herpesvirus Meleagrídeo 1/fisiologia , Mardivirus/fisiologia , Mioblastos/virologia , Cultura de Vírus , Animais , Linhagem Celular , Galinhas , Genoma Viral , Mardivirus/patogenicidade , Doença de Marek/virologia , Codorniz , Organismos Livres de Patógenos Específicos , Latência Viral
13.
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
14.
J Virol ; 87(22): 12327-38, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24027304

RESUMO

Rabies virus (RABV), which is transmitted via a bite wound caused by a rabid animal, infects peripheral nerves and then spreads to the central nervous system (CNS) before causing severe neurological symptoms and death in the infected individual. Despite the importance of this ability of the virus to spread from a peripheral site to the CNS (neuroinvasiveness) in the pathogenesis of rabies, little is known about the mechanism underlying the neuroinvasiveness of RABV. In this study, to obtain insights into the mechanism, we conducted comparative analysis of two fixed RABV strains, Nishigahara and the derivative strain Ni-CE, which cause lethal and asymptomatic infections, respectively, in mice after intramuscular inoculation. Examination of a series of chimeric viruses harboring the respective genes from Nishigahara in the genetic background of Ni-CE revealed that the Nishigahara phosphoprotein (P) gene plays a major role in the neuroinvasiveness by mediating infection of peripheral nerves. The results obtained from both in vivo and in vitro experiments strongly suggested that the Nishigahara P gene, but not the Ni-CE P gene, is important for stable viral replication in muscle cells. Further investigation based on the previous finding that RABV phosphoprotein counteracts the host interferon (IFN) system demonstrated that the Nishigahara P gene, but not the Ni-CE P gene, functions to suppress expression of the beta interferon (IFN-ß) gene (Ifn-ß) and IFN-stimulated genes in muscle cells. In conclusion, we provide the first data strongly suggesting that RABV phosphoprotein assists viral replication in muscle cells by counteracting the host IFN system and, consequently, enhances infection of peripheral nerves.


Assuntos
Células Musculares/virologia , Mioblastos/virologia , Nervos Periféricos/virologia , Fosfoproteínas/metabolismo , Vírus da Raiva/patogenicidade , Raiva/virologia , Proteínas Estruturais Virais/metabolismo , 2',5'-Oligoadenilato Sintetase/genética , 2',5'-Oligoadenilato Sintetase/metabolismo , Animais , Western Blotting , Células Cultivadas , Feminino , Fator Regulador 1 de Interferon/genética , Fator Regulador 1 de Interferon/metabolismo , Interferons/farmacologia , Camundongos , Chaperonas Moleculares , Células Musculares/metabolismo , Células Musculares/patologia , Mioblastos/metabolismo , Mioblastos/patologia , Proteínas de Resistência a Myxovirus/genética , Proteínas de Resistência a Myxovirus/metabolismo , Neuroblastoma/genética , Neuroblastoma/patologia , Neuroblastoma/virologia , Nervos Periféricos/metabolismo , Nervos Periféricos/patologia , Fosfoproteínas/genética , RNA Mensageiro/genética , Raiva/genética , Raiva/patologia , Vírus da Raiva/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Rabdomiossarcoma/genética , Rabdomiossarcoma/patologia , Rabdomiossarcoma/virologia , Proteínas Estruturais Virais/genética , Virulência , Replicação Viral
15.
Virology ; 434(1): 27-37, 2012 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-22889614

RESUMO

Human 2',5'-oligoadenylate synthetase 3 (OAS3) exerts antiviral effect against alphaviruses including Chikungunya virus (CHIKV) by inhibiting viral RNA accumulation. Here, we identified a CHIKV variant exhibiting a remarkable resistance to the antiviral action of OAS3 in human epithelial HeLa cells. Using a molecular clone of CHIKV with Renilla luciferase inserted as a reporter gene in the non-structural region, we demonstrated that a single glutamine-to-lysine amino acid change at position 166 of the envelope E2 glycoprotein restores CHIKV replication in OAS3 expressing HeLa cells. Viral entry assays showed that CHIKV with a lysine at position E2-166 was more efficient at entering the replicative pathway. The E2-E166K substitution promotes a greater efficiency of CHIKV replication in human myoblasts leading to severe apoptosis through a more robust activation of the PKR pathway. These observations provide a new insight into the role of E2 into the pathogenicity of CHIKV in human cells.


Assuntos
2',5'-Oligoadenilato Sintetase/biossíntese , Vírus Chikungunya/fisiologia , Proteínas Virais/genética , Proteínas Virais/metabolismo , Internalização do Vírus , 2',5'-Oligoadenilato Sintetase/imunologia , Animais , Apoptose , Fusão Gênica Artificial , Vírus Chikungunya/genética , Vírus Chikungunya/crescimento & desenvolvimento , Vírus Chikungunya/imunologia , Genes Reporter , Células HeLa , Humanos , Luciferases/análise , Luciferases/genética , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Mioblastos/fisiologia , Mioblastos/virologia , Renilla/enzimologia
16.
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
17.
Med Sci Monit ; 17(12): BR364-71, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22129894

RESUMO

BACKGROUND: The expression of microRNA-206 (miR-206) is high in skeletal muscle but low in most other tissues. The expression of miR-206 is increased in muscular dystrophy, suggesting its involvement in the pathogenesis of muscle diseases. To determine the role of miR-206 in muscle cell differentiation and explore a possible gene therapy vector, we constructed a miR-206 adenoviral expression vector (AdvmiR-206) and tested for transfection into C2C12 stem cells. MATERIAL/METHODS: A 355-bp PCR amplicon from C57B6 mouse skeletal muscle genomic DNA was inserted into the adenoviral shuttle vector pAdTrack-CMV, which was then co-transformed with the adenoviral backbone plasmid pAdEasy-1 into competent E. coli BJ5183 bacteria. The specificity and function of this recombinant adenoviral MiR-206 were studied in C2C12 cells by Northern blot, immunofluorescence, Western blot, and flow cytometry. RESULTS: Increased expression of miR-206 in AdvmiR-206 transfected C2C12 cells (P < 0.001) and resulted in morphological and biochemical changes over time that were similar to serum deprivation, including elongated cells and increased myosin heavy chain proteins. Even in the absence of serum deprivation, miR-206 overexpression accounted for a 50% reduction of S-phase cells (P < 0.01). Moreover, in untransfected C2C12 cells, the introduction of miR-206-specific antisense oligoribonucleotides inhibited the normal response to serum deprivation. Twenty-four hours after lipofection of antisense oligoribonucleotides, the number of elongated cells was reduced by half (P < 0.01). CONCLUSIONS: Collectively, these data support a role for miR-206 in myoblast differentiation. We foresee potential applications for the AdvmiR-206 vector in research and therapy.


Assuntos
Adenoviridae/genética , MicroRNAs/genética , Desenvolvimento Muscular/genética , Mioblastos/metabolismo , Mioblastos/virologia , Recombinação Genética/genética , Animais , Ciclo Celular , Diferenciação Celular/genética , Linhagem Celular , Regulação da Expressão Gênica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Músculo Esquelético , Especificidade de Órgãos/genética , Transfecção
18.
Wei Sheng Wu Xue Bao ; 49(9): 1259-64, 2009 Sep.
Artigo em Chinês | MEDLINE | ID: mdl-20030068

RESUMO

OBJECTIVE: To construct the recombinant baculovirus expressing Infectious bursal disease (IBDV) VP2 gene in the chicken primary myoblast cells. METHODS: A proteinase K digestion and phenol-chloroform extraction method was used to extract dsRNA genome from IBDV. VP2 gene was amplified by Reverse Transcription Polymerase Chain Reaction (RT-PCR) with the genome RNA as template. The pFastBac-pCMV-VP2 baculovirus transfer vector was constructed by inserting VP2 gene under the immediate-early promoter of cytomegalovirus. The VP2 recombinant bacmid was obtained by Bac-to-Bac system and transfected sf9 insect cell to acquire VP2 recombinant baculovirus. After amplification of recombinant baculovirus on cell passages, the recombinant virus was seeded on chicken primary myoblast cells with 50 multiplicity of infection (MOI), and the cells were harvested at 72 hours after infection. RESULTS: Sodium Dodecyl Sulphate Poly-Acrylamide Gel Electrophoresis (SDS-PAGE) and Western blot results showed that the VP2 gene was successfully expressed in chicken primary myoblast cells. The product was a 48kDa protein and could be recognized by anti-IBDV serum. CONCLUSION: The recombinant baculovirus could efficiently delivery IBDV VP2 gene into chicken primary cells and that CMV, a mammalian-cell-active promoter, was functional in chicken primary cells and could direct the expression of VP2 antigen protein. The research can be a potential basis for the development of baculovirus vector vaccines for IBDV and other avian infectious disease.


Assuntos
Baculoviridae/genética , Expressão Gênica , Vetores Genéticos/genética , Mioblastos/metabolismo , Proteínas Estruturais Virais/genética , Animais , Baculoviridae/fisiologia , Linhagem Celular , Células Cultivadas , Galinhas , Clonagem Molecular , Vetores Genéticos/fisiologia , Vírus da Doença Infecciosa da Bursa/genética , Vírus da Doença Infecciosa da Bursa/metabolismo , Mioblastos/virologia , Spodoptera , Proteínas Estruturais Virais/metabolismo
19.
Hum Gene Ther ; 20(9): 1013-21, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19534598

RESUMO

In vivo gene transduction with adeno-associated virus (AAV)-based vectors depends on laborious procedures for the production of high-titer vector stocks. Purification steps for efficient clearance of impurities such as host cell proteins and empty vector particles are required to meet end-product specifications. Therefore, the development of alternative, realistic methods to facilitate a scalable virus recovery procedure is critical to promote in vivo investigations. However, the conventional purification procedure with resin-based packed-bed chromatography suffers from a number of limitations, including variations in pressure, slow pore diffusion, and large bed volumes. Here we have employed disposable high-performance anion- and cation-exchange membrane adsorbers to effectively purify recombinant viruses. As a result of isoelectric focusing analysis, the isoelectric point of empty particles was found to be significantly higher than that of packaged virions. Therefore, AAV vector purification with the membrane adsorbers was successful and allowed higher levels of gene transfer in vivo without remarkable signs of toxicity or inflammation. Electron microscopy of the AAV vector stocks obtained revealed highly purified virions with as few as 0.8% empty particles. Furthermore, the membrane adsorbers enabled recovery of AAV vectors in the transduced culture supernatant. Also, the ion-exchange enrichment of retroviral vectors bearing the amphotropic envelope was successful. This rapid and scalable viral purification protocol using disposable membrane adsorbers is particularly promising for in vivo experimentation and clinical investigations.


Assuntos
Cromatografia Líquida/métodos , Dependovirus/isolamento & purificação , Vetores Genéticos , Troca Iônica , Recombinação Genética , Adsorção , Animais , Linhagem Celular , Dependovirus/classificação , Dependovirus/genética , Dependovirus/fisiologia , Cães , Humanos , Focalização Isoelétrica , Rim/citologia , Rim/virologia , Masculino , Membranas , Mioblastos/citologia , Mioblastos/virologia , Ratos , Ratos Wistar , Sorotipagem , Transfecção
20.
Zhongguo Yi Xue Ke Xue Yuan Xue Bao ; 30(4): 498-505, 2008 Aug.
Artigo em Chinês | MEDLINE | ID: mdl-18795628

RESUMO

OBJECTIVE: To construct the recombinant adenovirus containing herpes simplex virus-1 virion protein (VP) 22 and human microdystrophin gene, then the adenovirus was transfected into C2C12 myoblast and studied on the property of protein transduction with VP22-mediated microdystrophin in C2C12 myoblast. METHODS: The full-length VP22 cDNA was obtained from recombinant plasmid pSINrep5-VP22 with PCR, and the product was directionally inserted into pShuttle-CMV to acquire the plasmid pCMV-VP22. Microdystrophin cDNA was obtained from recombinant plasmid pBSK-micro digested with restrictive endonuclease NotI, and the product was directionally inserted into pCMV-VP22 to acquire the plasmid pCMV-VP22-MICDYS. The plasmid of pCMV-VP22-MICDYS was lined with Pme I, and the fragment containing VP22-microdystrophin was reclaimed and transfected into E1 coli BJ5183 with plasmid pAdeasy-1. After having been screened by selected media, the extracted plasmid of positive bacteria was transfected into HEK293 cells with liposome and was identified by observing the cytopathic effect of cells and by PCR method to acquire the recombinant adenovirus Ad-VP22-MICDYS. Finally, the C2C12 myoblast were transfected with the recombinant adenovirus Ad-VP22-MICDYS and Ad-MICDYS, and the expression of microdystrophin was detected by RT-PCR, Western blot and immunocytochemistry. RESULTS: The recombinant adenovirus including VP22 and microdystrophin gene was successfully constructed. VP22 transferred VP22-microdystrophin fused protein from infected C2C12 myoblast into uninfected cells and enhance the expression of microdystrophin in myoblast. CONCLUSIONS: Recombinant adenovirus containing VP22 and microdystrophin gene was constructed successfully. VP22 can enhance the expression with microdystrophin in myoblast. It lays the foundation for further studying on VP22-mediated recombinant including microdystrophin gene to cure Duchenne muscular dystrophy.


Assuntos
Adenoviridae/genética , Distrofina/genética , Vetores Genéticos/genética , Transdução Genética , Proteínas Estruturais Virais/genética , Adenoviridae/fisiologia , Animais , Linhagem Celular , Distrofina/metabolismo , Vetores Genéticos/metabolismo , Humanos , Camundongos , Mioblastos/metabolismo , Mioblastos/virologia , Simplexvirus/genética , Simplexvirus/metabolismo , Proteínas Estruturais Virais/metabolismo , Vírion/genética , Vírion/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...