Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Adicionar filtros








Intervalo de ano
1.
Braz. j. med. biol. res ; 56: e12682, 2023. graf
Artigo em Inglês | LILACS-Express | LILACS | ID: biblio-1447681

RESUMO

Preventing the replication of adenovirus could have practical uses, such as controlling infection with wild-type virus or in applications involving recombinant vectors. Mainly transient methods have been used to inhibit adenovirus replication, including siRNA or drugs. Here, we tested whether stable expression of shRNA designed to target hexon, Iva2, or pol can inhibit the replication of a recombinant adenoviral vector, Ad-LacZ (serotype 5, E1/E3 deleted), in 293T cells. Significant knockdown correlating with reduced Ad-LacZ replication was achieved only when hexon was targeted. Cell sorting and isolation of cellular clones further accentuated knockdown of the hexon transcript, reduced protein levels by more than 90%, and diminished adenovirus production. As visualized by transmission electron microscopy, the cellular clone expressing the hexon-specific shRNA yielded 89.2% fewer particles compared to the parental 293T cells. Full scale production followed by purification revealed a 90.2% reduction in Ad-LacZ biological titer. These results support the notion that stable expression of shRNA can be used as a means to control adenovirus replication.

2.
Braz. j. med. biol. res ; 54(7): e10579, 2021. tab, graf
Artigo em Inglês | LILACS | ID: biblio-1249313

RESUMO

NOTCH pathway proteins, including the transcriptional factor HES1, play crucial roles in the development of the inner ear by means of the lateral inhibition mechanism, in which supporting cells have their phenotype preserved while they are prevented from becoming hair cells. Genetic manipulation of this pathway has been demonstrated to increase hair cell number. The present study aimed to investigate gene expression effects in hair cells and supporting cells after Hes1-shRNA lentivirus transduction in organotypic cultures of the organ of Corti from postnatal-day-3 mice. Forty-eight hours after in vitro knockdown, Hes1 gene expression was reduced at both mRNA and protein levels. Myo7a (hair cell marker) and Sox2 (progenitor cell marker) mRNA levels also significantly increased. The modulation of gene expression in the organ of Corti upon Hes1 knockdown is consistent with cell phenotypes related to lateral inhibition mechanism interference in the inner ear. The lentivirus-based expression of Hes1-shRNA is a valuable strategy for genetic interference in the organ of Corti and for future evaluation of its efficacy in protocols aiming at the regeneration of hair cells in vivo.


Assuntos
Animais , Ratos , Cóclea , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Órgão Espiral , Diferenciação Celular , Receptores Notch , Fatores de Transcrição HES-1/genética , Células Ciliadas Auditivas
3.
Braz. j. med. biol. res ; 53(3): e8876, 2020. graf
Artigo em Inglês | LILACS | ID: biblio-1089338

RESUMO

The immune stimulatory and anti-neoplastic functions of type I interferon have long been applied for the treatment of melanoma. However, the systemic application of high levels of this recombinant protein is often met with toxicity. An approach that provides localized, yet transient, production of type I interferon may overcome this limitation. We propose that the use of mesenchymal stem cells (MSCs) as delivery vehicles for the production of interferon-β (IFNβ) may be beneficial when applied together with our cancer gene therapy approach. In our previous studies, we have shown that adenovirus-mediated gene therapy with IFNβ was especially effective in combination with p19Arf gene transfer, resulting in immunogenic cell death. Here we showed that MSCs derived from mouse adipose tissue were susceptible to transduction with adenovirus, expressed the transgene reliably, and yet were not especially sensitive to IFNβ production. MSCs used to produce IFNβ inhibited B16 mouse melanoma cells in a co-culture assay. Moreover, the presence of p19Arf in the B16 cells sensitizes them to the IFNβ produced by the MSCs. These data represent a critical demonstration of the use of MSCs as carriers of adenovirus encoding IFNβ and applied as an anti-cancer strategy in combination with p19Arf gene therapy.


Assuntos
Animais , Masculino , Coelhos , Melanoma Experimental/terapia , Interferon beta/metabolismo , Inibidor p16 de Quinase Dependente de Ciclina/administração & dosagem , Células-Tronco Mesenquimais/metabolismo , Transdução Genética , Melanoma Experimental/metabolismo , Terapia Genética , Linhagem Celular Tumoral , Proliferação de Células , Modelos Animais de Doenças , Camundongos Endogâmicos C57BL
4.
Braz. j. med. biol. res ; 49(4): e5064, 2016. tab, graf
Artigo em Inglês | LILACS | ID: biblio-951670

RESUMO

In mammals, damage to sensory receptor cells (hair cells) of the inner ear results in permanent sensorineural hearing loss. Here, we investigated whether postnatal mouse inner ear progenitor/stem cells (mIESCs) are viable after transplantation into the basal turns of neomycin-injured guinea pig cochleas. We also examined the effects of mIESC transplantation on auditory functions. Eight adult female Cavia porcellus guinea pigs (250-350g) were deafened by intratympanic neomycin delivery. After 7 days, the animals were randomly divided in two groups. The study group (n=4) received transplantation of LacZ-positive mIESCs in culture medium into the scala tympani. The control group (n=4) received culture medium only. At 2 weeks after transplantation, functional analyses were performed by auditory brainstem response measurement, and the animals were sacrificed. The presence of mIESCs was evaluated by immunohistochemistry of sections of the cochlea from the study group. Non-parametric tests were used for statistical analysis of the data. Intratympanic neomycin delivery damaged hair cells and increased auditory thresholds prior to cell transplantation. There were no significant differences between auditory brainstem thresholds before and after transplantation in individual guinea pigs. Some mIESCs were observed in all scalae of the basal turns of the injured cochleas, and a proportion of these cells expressed the hair cell marker myosin VIIa. Some transplanted mIESCs engrafted in the cochlear basilar membrane. Our study demonstrates that transplanted cells survived and engrafted in the organ of Corti after cochleostomy.


Assuntos
Animais , Feminino , Órgão Espiral/cirurgia , Células-Tronco , Transplante de Células-Tronco/métodos , Células Ciliadas Auditivas Internas/transplante , Perda Auditiva Neurossensorial/cirurgia , Limiar Auditivo , Imuno-Histoquímica , Inibidores da Síntese de Proteínas , Neomicina , Sobrevivência Celular , Células Cultivadas , Reprodutibilidade dos Testes , Potenciais Evocados Auditivos do Tronco Encefálico , Resultado do Tratamento , Cobaias , Camundongos Endogâmicos BALB C
5.
Braz. j. med. biol. res ; 40(5): 601-613, May 2007. ilus, tab
Artigo em Inglês | LILACS | ID: lil-449078

RESUMO

A successful gene therapy clinical trial that also encountered serious adverse effects has sparked extensive study and debate about the future directions for retrovirus-mediated interventions. Treatment of X-linked severe combined immunodeficiency with an oncoretrovirus harboring a normal copy of the gc gene was applied in two clinical trials, essentially curing 13 of 16 infants, restoring a normal immune system without the need for additional immune-related therapies. Approximately 3 years after their gene therapy, tragically, 3 of these children, all from the same trial, developed leukemia as a result of this experimental treatment. The current understanding of the mechanism behind this leukemogenesis involves three critical and cooperating factors, i.e., viral integration, oncogene activation, and the function of the therapeutic gene. In this review, we will explore the causes of this unwanted event and some of the possibilities for reducing the risk of its reoccurrence.


Assuntos
Humanos , Terapia Genética , Leucemia/etiologia , Doenças por Imunodeficiência Combinada Ligada ao Cromossomo X/terapia , Transformação Celular Neoplásica , Ensaios Clínicos como Assunto , Terapia Genética/efeitos adversos , Terapia Genética/métodos , Vetores Genéticos/genética , Fatores de Risco , Transdução Genética , Doenças por Imunodeficiência Combinada Ligada ao Cromossomo X/genética , Doenças por Imunodeficiência Combinada Ligada ao Cromossomo X/imunologia
6.
Braz. j. med. biol. res ; 32(7): 905-14, July 1999.
Artigo em Inglês | LILACS | ID: lil-234898

RESUMO

The use of gene therapy continues to be a promising, yet elusive, alternative for the treatment of cancer. The origins of cancer must be well understood so that the therapeutic gene can be chosen with the highest chance of successful tumor regression. The gene delivery system must be tailored for optimum transfer of the therapeutic gene to the target tissue. In order to accomplish this, we study models of G1 cell-cycle control in both normal and transformed cells in order to understand the reasons for uncontrolled cellular proliferation. We then use this information to choose the gene to be delivered to the cells. We have chosen to study p16, p21, p53 and pRb gene transfer using the pCL-retrovirus. Described here are some general concepts and specific results of our work that indicate continued hope for the development of genetically based cancer treatments


Assuntos
Ratos , Camundongos , Animais , Humanos , Técnicas de Transferência de Genes , Terapia Genética/métodos , Vetores Genéticos/genética , Glioblastoma/genética , Glioblastoma/terapia , Retroviridae/genética , Ciclo Celular/genética , Ciclo Celular/fisiologia , Transformação Celular Neoplásica/genética , Ensaios Clínicos como Assunto , Modelos Animais de Doenças , Genes Supressores de Tumor
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA