Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Am J Pathol ; 180(3): 1107-1120, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22214838

RESUMO

Coxsackieviruses are significant human pathogens causing myocarditis, meningitis, and encephalitis. We previously demonstrated the ability of coxsackievirus B3 (CVB3) to persist within the neonatal central nervous system (CNS) and to target neural stem cells. Given that CVB3 is a cytolytic virus and may therefore damage target cells, we characterized the potential reduction in neurogenesis within the developing brain and the subsequent developmental defects that occurred after the loss of these essential neural stem cells. Neonatal mice were inoculated with a recombinant CVB3 expressing eGFP (eGFP-CVB3), and alterations in neurogenesis and brain development were evaluated over time. We observed a reduction in proliferating cells in CNS neurogenic regions simultaneously with the presence of nestin(+) cells undergoing apoptosis. The size of the brain appeared smaller by histology, and a permanent decrease in brain wet weight was observed after eGFP-CVB3 infection. We also observed an inverse relationship between the amount of virus material and brain wet weight up to day 30 postinfection. In addition, signs of astrogliosis and a compaction of the cortical layers were observed at 90 days postinfection. Intriguingly, partial brain wet weight recovery was observed in mice treated with the antiviral drug ribavirin during the persistent stage of infection. Hence, long-term neurological sequelae might be expected after neonatal enteroviral infections, yet antiviral treatment initiated long after the end of acute infection might limit virus-mediated neuropathology.


Assuntos
Sistema Nervoso Central/virologia , Infecções por Coxsackievirus/complicações , Enterovirus Humano B , Células-Tronco Neurais/virologia , Neurogênese/fisiologia , Animais , Animais Recém-Nascidos , Antivirais/farmacologia , Apoptose/fisiologia , Astrócitos/virologia , Encéfalo/crescimento & desenvolvimento , Encéfalo/virologia , Divisão Celular , Proliferação de Células , Sistema Nervoso Central/crescimento & desenvolvimento , Proteínas de Fluorescência Verde/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Tamanho do Órgão , Proteínas Recombinantes , Carga Viral
2.
J Virol ; 85(12): 5718-32, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21471247

RESUMO

Enteroviruses, including coxsackieviruses, exhibit significant tropism for the central nervous system, and these viruses are commonly associated with viral meningitis and encephalitis. Previously, we described the ability of coxsackievirus B3 (CVB3) to infect proliferating neuronal progenitor cells located in the neonatal subventricular zone and persist in the adult murine central nervous system (CNS). Here, we demonstrate that cultured murine neurospheres, which comprise neural stem cells and their progeny at different stages of development, were highly susceptible to CVB3 infection. Neurospheres, or neural progenitor and stem cells (NPSCs), isolated from neonatal C57BL/6 mice, supported high levels of infectious virus production and high viral protein expression levels following infection with a recombinant CVB3 expressing enhanced green fluorescent protein (eGFP) protein. Similarly, NPSCs isolated from neonatal actin-promoter-GFP transgenic mice (actin-GFP NPSCs) were highly susceptible to infection with a recombinant CVB3 expressing DsRed (Discosoma sp. red fluorescent protein). Both nestin-positive and NG2(+) progenitor cells within neurospheres were shown to preferentially express high levels of viral protein as soon as 24 h postinfection (p.i.). By day 3 p.i., viral protein expression and viral titers increased dramatically in NPSCs with resultant cytopathic effects (CPE) and eventual cell death. In contrast, reduced viral replication, lower levels of CPE, and diminished viral protein expression levels were observed in NPSCs differentiated for 5 or 16 days in the presence of fetal bovine serum (FBS). Despite the presence of CPE and high levels of cell death following early CVB3 infection, surviving neurospheres were readily observed and continued to express detectable levels of viral protein as long as 37 days after initial infection. Also, CVB3 infection of actin-GFP NPSCs increased the percentage of cells expressing neuronal class III ß-tubulin following their differentiation in the presence of FBS. These results suggest that neural stem cells may be preferentially targeted by CVB3 and that neurogenic regions of the CNS may support persistent viral replication in the surviving host. In addition, normal progenitor cell differentiation may be altered in the host following infection.


Assuntos
Diferenciação Celular , Enterovirus Humano B/fisiologia , Enterovirus Humano B/patogenicidade , Células-Tronco Neurais/virologia , Animais , Células Cultivadas , Efeito Citopatogênico Viral , Enterovirus Humano B/genética , Enterovirus Humano B/ultraestrutura , Feminino , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microscopia de Fluorescência , Células-Tronco Neurais/citologia , Células-Tronco Neurais/ultraestrutura , Proteínas Virais/genética , Proteínas Virais/metabolismo , Replicação Viral , Proteína Vermelha Fluorescente
3.
J Neurosci ; 30(25): 8676-91, 2010 Jun 23.
Artigo em Inglês | MEDLINE | ID: mdl-20573913

RESUMO

Enterovirus infection in newborn infants is a significant cause of aseptic meningitis and encephalitis. Using a neonatal mouse model, we previously determined that coxsackievirus B3 (CVB3) preferentially targets proliferating neural stem cells located in the subventricular zone within 24 h after infection. At later time points, immature neuroblasts, and eventually mature neurons, were infected as determined by expression of high levels of viral protein. Here, we show that blood-derived Mac3(+) mononuclear cells were rapidly recruited to the CNS within 12 h after intracranial infection with CVB3. These cells displayed a myeloid-like morphology, were of a peripheral origin based on green fluorescent protein (GFP)-tagged adoptive cell transplant examination, and were highly susceptible to CVB3 infection during their migration into the CNS. Serial immunofluorescence images suggested that the myeloid cells enter the CNS via the choroid plexus, and that they may be infected during their extravasation and passage through the choroid plexus epithelium; these infected myeloid cells ultimately penetrate into the parenchyma of the brain. Before their migration through the ependymal cell layer, a subset of these infected myeloid cells expressed detectable levels of nestin, a marker for neural stem and progenitor cells. As these nestin(+) myeloid cells infected with CVB3 migrated through the ependymal cell layer, they revealed distinct morphological characteristics typical of type B neural stem cells. The recruitment of these novel myeloid cells may be specifically set in motion by the induction of a unique chemokine profile in the CNS induced very early after CVB3 infection, which includes upregulation of CCL12. We propose that intracranial CVB3 infection may lead to the recruitment of nestin(+) myeloid cells into the CNS which might represent an intrinsic host CNS repair response. In turn, the proliferative and metabolic status of recruited myeloid cells may render them attractive targets for CVB3 infection. Moreover, the migratory ability of these myeloid cells may point to a productive method of virus dissemination within the CNS.


Assuntos
Infecções por Coxsackievirus/virologia , Células Mieloides/virologia , Animais , Animais Recém-Nascidos , Plexo Corióideo/imunologia , Plexo Corióideo/virologia , Infecções por Coxsackievirus/imunologia , Imunofluorescência , Hibridização In Situ , Camundongos , Microscopia Confocal , Células Mieloides/imunologia , Neurônios/imunologia , Neurônios/virologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células-Tronco/imunologia , Células-Tronco/virologia
4.
J Virol ; 83(18): 9356-69, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19570873

RESUMO

Coxsackieviruses are significant human pathogens, and the neonatal central nervous system (CNS) is a major target for infection. Despite the extreme susceptibility of newborn infants to coxsackievirus infection and viral tropism for the CNS, few studies have been aimed at determining the long-term consequences of infection on the developing CNS. We previously described a neonatal mouse model of coxsackievirus B3 (CVB3) infection and determined that proliferating stem cells in the CNS were preferentially targeted. Here, we describe later stages of infection, the ensuing inflammatory response, and subsequent lesions which remain in the adult CNS of surviving animals. High levels of type I interferons and chemokines (in particular MCP-5, IP10, and RANTES) were upregulated following infection and remained at high levels up to day 10 postinfection (p.i). Chronic inflammation and lesions were observed in the hippocampus and cortex of surviving mice for up to 9 months p.i. CVB3 RNA was detected in the CNS up to 3 months p.i at high abundance ( approximately 10(6) genomes/mouse brain), and viral genomic material remained detectable in culture after two rounds of in vitro passage. These data suggest that CVB3 may persist in the CNS as a low-level, noncytolytic infection, causing ongoing inflammatory lesions. Thus, the effects of a relatively common infection during the neonatal period may be long lasting, and the prognosis for newborn infants recovering from acute infection should be reexplored.


Assuntos
Sistema Nervoso Central/virologia , Infecções por Coxsackievirus/patologia , Enterovirus/patogenicidade , Animais , Animais Recém-Nascidos , Córtex Cerebral/patologia , Córtex Cerebral/virologia , Quimiocinas/análise , Doença Crônica , Infecções por Coxsackievirus/imunologia , Enterovirus/genética , Enterovirus Humano B , Genoma Viral , Hipocampo/patologia , Hipocampo/virologia , Humanos , Inflamação , Interferon Tipo I/análise , Camundongos , RNA Viral/sangue , Fatores de Tempo , Regulação para Cima
5.
Gene ; 395(1-2): 160-9, 2007 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-17449199

RESUMO

RNA interference (RNAi)-mediated gene silencing has become a valuable tool for functional studies, reverse genomics, and drug discoveries. One major challenge of using RNAi is to identify the most effective short interfering RNAs (siRNAs) sites of a given gene. Although several published bioinformatic prediction models have proven useful, the process to select and validate optimal siRNA sites for a given gene remains empirical and laborious. Here, we developed a fluorescence-based selection system using a retroviral vector backbone, namely pSOS, which was based on the premise that candidate siRNAs would knockdown the chimeric transcript between GFP and target gene. The expression of siRNA was driven by the opposing convergent H1 and U6 promoters. This configuration simplifies the cloning of duplex siRNA oligonucleotide cassettes. We demonstrated that GFP signal reduction was closely correlated with siRNA knockdown efficiency of human beta-catenin, as well as with the inhibition of beta-catenin/Tcf4 signaling activity. The pSOS should not only facilitate the selection and validation of candidate siRNA sites, but also provide efficient delivery tools of siRNAs via viral vectors in mammalian cells. Thus, the pSOS system represents an efficient and user-friendly strategy to select and validate siRNA target sites.


Assuntos
Interferência de RNA , RNA Interferente Pequeno/genética , Sequência de Bases , Sítios de Ligação/genética , Linhagem Celular , DNA/genética , Expressão Gênica , Genes Reporter , Técnicas Genéticas , Vetores Genéticos , Proteínas de Fluorescência Verde/genética , Humanos , Regiões Promotoras Genéticas , Proteínas Recombinantes/genética , Transfecção , beta Catenina/genética
6.
J Bone Joint Surg Am ; 85(8): 1544-52, 2003 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12925636

RESUMO

BACKGROUND: Bone morphogenic proteins (BMPs) are known to promote osteogenesis, and clinical trials are currently underway to evaluate the ability of certain BMPs to promote fracture-healing and spinal fusion. The optimal BMPs to be used in different clinical applications have not been elucidated, and a comprehensive evaluation of the relative osteogenic activity of different BMPs is lacking. METHODS: To identify the BMPs that may possess the most osteoinductive activity, we analyzed the osteogenic activity of BMPs in mesenchymal progenitor and osteoblastic cells. Recombinant adenoviruses expressing fourteen human BMPs (BMP-2 to BMP-15) were constructed to infect pluripotent mesenchymal progenitor C3H10T1/2 cells, preosteoblastic C2C12 cells, and osteoblastic TE-85 cells. Osteogenic activity was determined by measuring the induction of alkaline phosphatase, osteocalcin, and matrix mineralization upon BMP stimulation. RESULTS: BMP-2, 6, and 9 significantly induced alkaline phosphatase activity in pluripotential C3H10T1/2 cells, while BMP-2, 4, 6, 7, and 9 significantly induced alkaline phosphatase activity in preosteoblastic C2C12 cells. In TE-85 osteoblastic cells, most BMPs (except BMP-3 and 12) were able to induce alkaline phosphatase activity. The results of alkaline phosphatase histochemical staining assays were consistent with those of alkaline phosphatase colorimetric assays. Furthermore, BMP-2, 6, and 9 (as well as BMP-4 and, to a lesser extent, BMP-7) significantly induced osteocalcin expression in C3H10T1/2 cells. In C2C12 cells, osteocalcin expression was strongly induced by BMP-2, 4, 6, 7, and 9. Mineralized nodules were readily detected in C3H10T1/2 cells infected with BMP-2, 6, and 9 (and, to a lesser extent, those infected with BMP-4 and 7). CONCLUSIONS: A comprehensive analysis of the osteogenic activity of fourteen types of BMPs in osteoblastic progenitor cells was conducted. Our results suggest an osteogenic hierarchical model in which BMP-2, 6, and 9 may play an important role in inducing osteoblast differentiation of mesenchymal stem cells. In contrast, most BMPs are able to stimulate osteogenesis in mature osteoblasts.


Assuntos
Proteínas Morfogenéticas Ósseas/farmacologia , Mesoderma/citologia , Osteoblastos/efeitos dos fármacos , Osteócitos/efeitos dos fármacos , Osteogênese/efeitos dos fármacos , Células-Tronco/efeitos dos fármacos , Adenoviridae/genética , Fosfatase Alcalina/metabolismo , Animais , Densidade Óssea/fisiologia , Proteínas Morfogenéticas Ósseas/classificação , Proteínas Morfogenéticas Ósseas/genética , Linhagem Celular , Técnicas de Transferência de Genes , Humanos , Camundongos , Osteossarcoma , Células Tumorais Cultivadas
7.
Cancer Lett ; 193(2): 161-70, 2003 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-12706873

RESUMO

Beta-Catenin is a critical transducer of the Wnt signal pathway and plays an important role in many developmental and cellular processes. Deregulation of beta-catenin signaling has been observed in a broad range of human tumors. In this report, we investigated whether tyrosine kinase inhibitor STI-571 could inhibit the beta-catenin signaling activity and hence suppress cell proliferation. Our results demonstrated that STI-571 effectively inhibited the constitutive activity of beta-catenin signaling in human colon cancer cells as well as the Wnt1-induced activation of beta-catenin signaling in HOS, HTB-94, and HEK 293 cells. Furthermore, STI-571 was shown to effectively suppress the proliferation of human colon cancer cells. Finally, we demonstrated that the Wnt1-mediated activation of a GAL4-beta-catenin heterologous transcription system was effectively inhibited by STI-571. Thus, our findings suggest that tyrosine phosphorylation may play an important role in regulating beta-catenin signaling activity, and inhibition of this signaling pathway by STI-571 may be further explored as an important target for alternative/adjuvant treatments for a broader range of human cancer.


Assuntos
Antineoplásicos/farmacologia , Proteínas do Citoesqueleto/biossíntese , Regulação para Baixo , Inibidores Enzimáticos/farmacologia , Piperazinas/farmacologia , Pirimidinas/farmacologia , Transdução de Sinais , Transativadores/biossíntese , Proteínas de Peixe-Zebra , Adenoviridae/genética , Benzamidas , Divisão Celular , Linhagem Celular , Neoplasias do Colo/tratamento farmacológico , Meios de Cultivo Condicionados/farmacologia , Proteínas do Citoesqueleto/metabolismo , Relação Dose-Resposta a Droga , Genes Reporter , Vetores Genéticos , Humanos , Mesilato de Imatinib , Proteínas Tirosina Quinases/antagonistas & inibidores , Proteínas Proto-Oncogênicas/biossíntese , Fatores de Tempo , Transativadores/metabolismo , Transcrição Gênica , Ativação Transcricional , Células Tumorais Cultivadas , Proteínas Wnt , Proteína Wnt1 , beta Catenina
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...