Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros










Intervalo de año de publicación
1.
Braz J Med Biol Res ; 56: e12682, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37493770

RESUMEN

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.


Asunto(s)
Adenoviridae , Replicación Viral , Adenoviridae/genética , ARN Interferente Pequeño/genética , Vectores Genéticos/genética , Humanos , Células HEK293 , Transcripción Genética , Células Clonales
2.
Braz. j. med. biol. res ; 56: e12682, 2023. graf
Artículo en Inglés | LILACS-Express | LILACS | ID: biblio-1447681

RESUMEN

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.

3.
Braz J Med Biol Res ; 54(7): e10579, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34008754

RESUMEN

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.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , Cóclea , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Diferenciación Celular , Células Ciliadas Auditivas , Ratones , Órgano Espiral , Receptores Notch , Factor de Transcripción HES-1/genética
4.
Braz. j. med. biol. res ; 54(7): e10579, 2021. tab, graf
Artículo en Inglés | LILACS | ID: biblio-1249313

RESUMEN

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.


Asunto(s)
Animales , Ratas , Cóclea , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Órgano Espiral , Diferenciación Celular , Receptores Notch , Factor de Transcripción HES-1/genética , Células Ciliadas Auditivas
5.
Braz J Med Biol Res ; 53(3): e8876, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32077463

RESUMEN

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-ß (IFNß) 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 IFNß 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 IFNß production. MSCs used to produce IFNß inhibited B16 mouse melanoma cells in a co-culture assay. Moreover, the presence of p19Arf in the B16 cells sensitizes them to the IFNß produced by the MSCs. These data represent a critical demonstration of the use of MSCs as carriers of adenovirus encoding IFNß and applied as an anti-cancer strategy in combination with p19Arf gene therapy.


Asunto(s)
Inhibidor p16 de la Quinasa Dependiente de Ciclina/administración & dosificación , Interferón beta/metabolismo , Melanoma Experimental/terapia , Células Madre Mesenquimatosas/metabolismo , Animales , Línea Celular Tumoral , Proliferación Celular , Modelos Animales de Enfermedad , Terapia Genética , Masculino , Melanoma Experimental/metabolismo , Ratones , Ratones Endogámicos C57BL , Transducción Genética
6.
Braz. j. med. biol. res ; 53(3): e8876, 2020. graf
Artículo en Inglés | LILACS | ID: biblio-1089338

RESUMEN

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.


Asunto(s)
Animales , Masculino , Conejos , Melanoma Experimental/terapia , Interferón beta/metabolismo , Inhibidor p16 de la Quinasa Dependiente de Ciclina/administración & dosificación , Células Madre Mesenquimatosas/metabolismo , Transducción Genética , Melanoma Experimental/metabolismo , Terapia Genética , Línea Celular Tumoral , Proliferación Celular , Modelos Animales de Enfermedad , Ratones Endogámicos C57BL
7.
Braz J Med Biol Res ; 49(4): e5064, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27007652

RESUMEN

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 (250-350 g) 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.


Asunto(s)
Células Ciliadas Auditivas Internas/trasplante , Pérdida Auditiva Sensorineural/cirugía , Órgano Espiral/cirugía , Trasplante de Células Madre/métodos , Células Madre , Animales , Umbral Auditivo , Supervivencia Celular , Células Cultivadas , Potenciales Evocados Auditivos del Tronco Encefálico , Femenino , Cobayas , Inmunohistoquímica , Ratones Endogámicos BALB C , Neomicina , Inhibidores de la Síntesis de la Proteína , Reproducibilidad de los Resultados , Resultado del Tratamiento
8.
Braz. j. med. biol. res ; 49(4): e5064, 2016. tab, graf
Artículo en Inglés | LILACS | ID: biblio-951670

RESUMEN

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.


Asunto(s)
Animales , Femenino , Órgano Espiral/cirugía , Células Madre , Trasplante de Células Madre/métodos , Células Ciliadas Auditivas Internas/trasplante , Pérdida Auditiva Sensorineural/cirugía , Umbral Auditivo , Inmunohistoquímica , Inhibidores de la Síntesis de la Proteína , Neomicina , Supervivencia Celular , Células Cultivadas , Reproducibilidad de los Resultados , Potenciales Evocados Auditivos del Tronco Encefálico , Resultado del Tratamiento , Cobayas , Ratones Endogámicos BALB C
9.
Cancer Gene Ther ; 20(5): 317-25, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23618951

RESUMEN

Approximately 90% of melanomas retain wild-type p53, a characteristic that may help shape the development of novel treatment strategies. Here, we employed an adenoviral vector where transgene expression is controlled by p53 to deliver the p19 alternate reading frame (Arf) and interferon-ß (IFNß) complementary DNAs in the B16 mouse model of melanoma. In vitro, cell death was enhanced by combined gene transfer (63.82±15.30% sub-G0 cells); yet introduction of a single gene resulted in significantly fewer hypoploid cells (37.73±7.3% or 36.96±11.58%, p19Arf or IFNß, respectively, P<0.05). Annexin V staining and caspase-3 cleavage indicate a cell death mechanism consistent with apoptosis. Using reverse transcriptase quantitative PCR, we show that key transcriptional targets of p53 were upregulated in the presence of p19Arf, although treatment with IFNß did not alter expression of the genes studied. In situ gene therapy revealed significant inhibition of subcutaneous tumors by IFNß (571±25 mm3) or the combination of p19Arf and IFNß (489±124 mm3) as compared with the LacZ control (1875±33 mm3, P<0.001), whereas p19Arf yielded an intermediate result (1053±169 mm3, P<0.01 vs control). However, only the combination was associated with increased cell death and prolonged survival (P<0.01). As shown here, the combined transfer of p19Arf and IFNß using p53-responsive vectors enhanced cell death both in vitro and in vivo.


Asunto(s)
Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Interferón beta/genética , Melanoma Experimental/genética , Melanoma Experimental/terapia , Animales , Apoptosis/genética , Muerte Celular/genética , Línea Celular Tumoral , Inhibidor p16 de la Quinasa Dependiente de Ciclina/biosíntesis , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Modelos Animales de Enfermedad , Femenino , Interferón beta/biosíntesis , Interferón beta/metabolismo , Melanoma Experimental/patología , Ratones , Ratones Endogámicos C57BL , Transducción Genética
10.
In Vitro Cell Dev Biol Anim ; 49(4): 250-9, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23519559

RESUMEN

Genetically modified animals have numerous applications, ranging from basic research to livestock production and agriculture. Recent progress in animal cloning by nuclear transfer has made possible the production of transgenic animals using previously genetically modified cell lineages. However, to produce such lineages, an additional time for in vitro culturing and great manipulation is needed. Herein, we aimed to characterize different aspects of genetically modified cells compared to control cells, and we also analyzed the development rate of embryos produced by nuclear transfer by using them as nuclei donors after short or long periods of in vitro culturing (early versus late passages). We hypothesized that the genetic material inserted in the genome of these cells, associated with the prolonged time in culture, ultimately alters cell growth physiology and cell viability, which leads to impaired nuclei reprogramming potential and consequent reduction in the production of cloned blastocysts. Fetal fibroblasts expressing the enhanced Green Fluorescent Protein gene (eGFP) cultured for different periods in vitro were analyzed with respect to chromosomal numeric abnormalities, nuclear DNA fragmentation, the ratio of BAX and BCL2 gene transcripts, and the intensity of mitochondrial membrane potential, and they were then used as nuclei donors for somatic cell nuclear transfer (SCNT). Early passages were defined as fewer than 11 passages, and late passages were 18th passage (18(th)p) to 21(st)p. No differences were observed in the percentage of cells with chromosomal abnormalities or in the mitochondrial membrane potential analysis. eGFP cells in late passages and control cells in early passages were not different regarding DNA fragmentation; however, control cells in late passages presented higher fragmentation (P < 0.05). The Bax and Bcl2 gene expression ratio in control and transgenic cells presented different patterns regarding cell conditions during culture. For SCNT experiments, no difference was observed between groups reconstructed with early or late-passage cells when fusion (63.1% and 49%), cleavage (67.7% and 69.9%), eight-cell embryo (36.4% and 44.4%) and blastocyst (21.6% and 20.8%) rates were compared. In conclusion, culture behavior was different between control and eGFP cells. However, when different in vitro culturing periods were compared, long-term cultured transgenic fetal fibroblasts remained competent for blastocyst production when used as nuclei donors in the nuclear transfer technique, a feature needed for the genetic manipulation of cell culture experiments aiming for transgenic animal production.


Asunto(s)
Fibroblastos/citología , Transporte Activo de Núcleo Celular , Animales , Animales Modificados Genéticamente , Blastocisto/citología , Blastocisto/metabolismo , Bovinos , Linaje de la Célula , Supervivencia Celular , Clonación de Organismos , Embrión de Mamíferos/citología , Fibroblastos/metabolismo , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Técnicas In Vitro , Técnicas de Transferencia Nuclear , Factores de Tiempo
11.
Cell Transplant ; 19(3): 279-89, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-19995482

RESUMEN

Adipose tissue may represent a potential source of adult stem cells for tissue engineering applications in veterinary medicine. It can be obtained in large quantities, under local anesthesia, and with minimal discomfort. In this study, canine adipose tissue was obtained by biopsy from subcutaneous adipose tissue or by suction-assisted lipectomy (i.e., liposuction). Adipose tissue was processed to obtain a fibroblast-like population of cells similar to human adipose-derived stem cells (hASCs). These canine adipose-derived stem cells (cASCs) can be maintained in vitro for extended periods with stable population doubling and low levels of senescence. Immunofluorescence and flow cytometry show that the majority of cASCs are of mesodermal or mesenchymal origin. cASCs are able to differentiate in vitro into adipogenic, chondrogenic, myogenic, and osteogenic cells in the presence of lineage-specific induction factors. In conclusion, like human lipoaspirate, canine adipose tissue may also contain multipotent cells and represent an important stem cell source both for veterinary cell therapy as well as preclinical studies.


Asunto(s)
Tejido Adiposo/fisiología , Células Madre Adultas/fisiología , Diferenciación Celular/fisiología , Células Madre Multipotentes/fisiología , Tejido Adiposo/citología , Células Madre Adultas/citología , Células Madre Adultas/efectos de los fármacos , Animales , Técnicas de Cultivo de Célula , Diferenciación Celular/efectos de los fármacos , Linaje de la Célula/efectos de los fármacos , Linaje de la Célula/fisiología , Separación Celular/métodos , Células Cultivadas , Perros , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/fisiología , Mesodermo/citología , Mesodermo/fisiología , Células Madre Multipotentes/citología , Células Madre Multipotentes/efectos de los fármacos
12.
Genet Mol Res ; 7(3): 925-32, 2008 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-18949710

RESUMEN

Dermcidin (DCD) is a human gene mapped to chromosome 12q13 region, which is co-amplified with multiple oncogenes with a well-established role in the growth, survival and progression of breast cancers. Here, we present a summary of a DNA microarray-based study that identified the genes that are up- and down-regulated in a human MDA-361 pLKO control clone and three clones expressing short hairpin RNA against three different regions of DCD mRNA. A list of 235 genes was differentially expressed among independent clones (> 3-fold change and p < 0.005). The gene expression of 208 was reduced and of 27 was increased in the three DCD-RNAi clones compared to pLKO control clone. The expression of 77 genes (37%) encoding for enzymes involved in amino acid metabolism, glucose metabolism and oxidoreductase activity and several genes required for cell survival and DNA repair were decreased. The expression of EGFR/ErbB-1 gene, an important predictor of outcome in breast cancer, was reduced together with the genes for betacellulin and amphiregulin, two known ligands of EGFR/ErbB receptors. Many of the 27 genes up-regulated by DCD-RNAi expression have not yet been fully characterized; among those with known function, we identified the calcium-calmodulin-dependent protein kinase-II delta and calcineurin A alpha. We compared 132 up-regulated and 12 down-regulated genes in our dataset with those genes up- and down-regulated by inhibitors targeting various signaling pathway components. The analysis showed that the genes in the DCD pathway are aligned with those functionally influenced by the drugs sirolimus, LY-294002 and wortmannin. Therefore, DCD may exert its function by activating the PI3K/AKT/mTOR signaling pathway. Together, these bioinformatic approaches suggest the involvement of DCD in the regulation of genes for breast cancer cell metabolism, proliferation and survival.


Asunto(s)
Perfilación de la Expresión Génica , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , Péptidos/fisiología , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Línea Celular Tumoral , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Péptidos/genética , Interferencia de ARN
13.
Braz. j. med. biol. res ; 40(5): 601-613, May 2007. ilus, tab
Artículo en Inglés | LILACS | ID: lil-449078

RESUMEN

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.


Asunto(s)
Humanos , Terapia Genética , Leucemia/etiología , Enfermedades por Inmunodeficiencia Combinada Ligada al Cromosoma X/terapia , Transformación Celular Neoplásica , Ensayos Clínicos como Asunto , Terapia Genética/efectos adversos , Terapia Genética/métodos , Vectores Genéticos/genética , Factores de Riesgo , Transducción Genética , Enfermedades por Inmunodeficiencia Combinada Ligada al Cromosoma X/genética , Enfermedades por Inmunodeficiencia Combinada Ligada al Cromosoma X/inmunología
14.
Braz J Med Biol Res ; 40(5): 601-13, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17464421

RESUMEN

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 gammac 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.


Asunto(s)
Terapia Genética , Leucemia/etiología , Enfermedades por Inmunodeficiencia Combinada Ligada al Cromosoma X/terapia , Transformación Celular Neoplásica , Ensayos Clínicos como Asunto , Terapia Genética/efectos adversos , Terapia Genética/métodos , Vectores Genéticos/genética , Humanos , Factores de Riesgo , Transducción Genética , Enfermedades por Inmunodeficiencia Combinada Ligada al Cromosoma X/genética , Enfermedades por Inmunodeficiencia Combinada Ligada al Cromosoma X/inmunología
15.
Braz J Med Biol Res ; 32(7): 905-14, 1999 07.
Artículo en Inglés | MEDLINE | ID: mdl-10454751

RESUMEN

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.


Asunto(s)
Técnicas de Transferencia de Gen , Terapia Genética/métodos , Vectores Genéticos/genética , Glioblastoma/genética , Glioblastoma/terapia , Retroviridae/genética , Animales , Ciclo Celular/genética , Ciclo Celular/fisiología , Transformación Celular Neoplásica/genética , Ensayos Clínicos como Asunto , Modelos Animales de Enfermedad , Genes Supresores de Tumor , Humanos , Ratones , Ratas
16.
Braz. j. med. biol. res ; 32(7): 905-14, July 1999.
Artículo en Inglés | LILACS | ID: lil-234898

RESUMEN

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


Asunto(s)
Ratas , Ratones , Animales , Humanos , Técnicas de Transferencia de Gen , Terapia Genética/métodos , Vectores Genéticos/genética , Glioblastoma/genética , Glioblastoma/terapia , Retroviridae/genética , Ciclo Celular/genética , Ciclo Celular/fisiología , Transformación Celular Neoplásica/genética , Ensayos Clínicos como Asunto , Modelos Animales de Enfermedad , Genes Supresores de Tumor
17.
Exp Cell Res ; 238(1): 51-62, 1998 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-9457056

RESUMEN

The aim of this study was to demonstrate that the induction of growth arrest in human glioblastoma multiforme (GBM) cell lines by retrovirus-mediated transduction of growth control genes was dependent upon the integrity of specific endogenous control pathways. We assessed the status of the endogenous p16INK4A, p21CIP1, pRb, or p53 genes in eight GBM lines. As expected, we found varied combinations of gene defects. The outcome of transducing five of these cell lines with p16INK4A, p21CIP1, pRb, or p53 genes was not entirely predictable. The growth-inhibitory effects mediated by the transfer of the gene encoding p16 was dependent on the presence of the pRb protein, but was independent of p53 status. p21, a broadly active CDK inhibitor and a strong inducer of growth arrest, was not a universal growth suppressor in the group of glioblastoma cell lines analyzed. The suppression of GBM cell proliferation by viruses encoding pRb or p53 was generally predictable and appeared to be independent of the status of either p16 or p21. Suppression of cell growth was assessed by a colony formation assay, by observance of alterations in morphology, and by cell viability staining for trypan blue exclusion. Our findings suggest that to accomplish the suppression of GBM cell proliferation by the transduction of these cell-cycle control genes, the status of endogenous cell-cycle control genes must be taken into account.


Asunto(s)
Proteínas Portadoras/metabolismo , Proteínas de Ciclo Celular , Ciclo Celular/fisiología , Inhibidor p16 de la Quinasa Dependiente de Ciclina , Proteína de Retinoblastoma/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Proteínas Supresoras de Tumor , Proteínas Portadoras/biosíntesis , División Celular , Inhibidor p15 de las Quinasas Dependientes de la Ciclina , Inhibidor p21 de las Quinasas Dependientes de la Ciclina , Ciclinas/biosíntesis , Ciclinas/metabolismo , Inhibidores Enzimáticos/metabolismo , Glioblastoma , Humanos , Cinética , Proteínas Recombinantes/metabolismo , Proteína de Retinoblastoma/biosíntesis , Retroviridae , Transfección , Células Tumorales Cultivadas , Proteína p53 Supresora de Tumor/biosíntesis
18.
Biochem Biophys Res Commun ; 217(1): 333-40, 1995 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-8526930

RESUMEN

We have examined the response of the human multidrug resistance gene-1 (MDR1) downstream promoter to various mutants of human p53 in a reporter assay system. Our findings indicate that mutant 175H inhibits reporter activity driven by the MDR1 downstream promoter (base pairs -189 to +133 relative to the major transcriptional initiation site) in a dose-dependent manner in cotransfection assays in the BHK and the Saos-2 cell lines. A 123 base-pair segment of DNA (-119 to +4 relative to the major transcriptional initiation site) and a 193 base-pair segment (-189 to +4) have been isolated from the MDR1 downstream promoter which, like the full promoter, are negatively controlled by mutant 175H. However, a 135 base-pair segment (-2 to +133) of the promoter is activated by mutant 175H as well as mutant 248Q, but not by mutants 213Q and 234H. Thus some mutants of p53 are able to activate transcription from the 3' region of the MDR1 downstream promoter, an activity that characterizes these p53 mutants as "gain of function" mutants.


Asunto(s)
Resistencia a Múltiples Medicamentos/genética , Mutación , Regiones Promotoras Genéticas , Proteína p53 Supresora de Tumor/genética , Animales , Secuencia de Bases , Línea Celular , Cricetinae , Cartilla de ADN/genética , Genes Reporteros , Genes p53 , Vectores Genéticos , Humanos , Luciferasas/genética , Datos de Secuencia Molecular , Transfección
19.
Biochem Biophys Res Commun ; 217(3): 825-31, 1995 Dec 26.
Artículo en Inglés | MEDLINE | ID: mdl-8554604

RESUMEN

We have examined the interaction of the wild-type p53 protein with the downstream promoter of the human multidrug resistance gene-1 (MDR1). Our findings indicate that wild-type p53 inhibits reporter activity driven by the MDR1 downstream promoter (base pairs -189 to +133 relative to the major transcriptional initiation site) in a dose-dependent manner in cotransfection assays in the BHK and the Saos-2 cell lines. A 123 base-pair segment of DNA (-119 to +4 relative to the major transcriptional initiation site), a 193 base-pair segment (-189 to +4), and a 135 base-pair segment (-2 to +133) have been isolated from the MDR1 downstream promoter which, like the full promoter, are negatively controlled by wild-type p53. In addition, we show sequence-specific binding of wild-type p53 protein to the MDR1 downstream promoter. These in vitro results suggest that the presence of wild-type p53 negatively affects expression of the MDR1 gene product, p-glycoprotein, at the transcriptional level.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/genética , Resistencia a Múltiples Medicamentos , Regiones Promotoras Genéticas , Proteína p53 Supresora de Tumor/metabolismo , Secuencia de Bases , Sitios de Unión , Cartilla de ADN/química , Proteínas de Unión al ADN/metabolismo , Regulación de la Expresión Génica , Humanos , Datos de Secuencia Molecular , ARN Mensajero/genética
20.
Transfusion ; 24(6): 486-9, 1984.
Artículo en Inglés | MEDLINE | ID: mdl-6542262

RESUMEN

To investigate the clinical significance of HLA determinants expressed on red cells (RBCs), 51Cr survival studies were carried out in six women (four healthy individuals, two patients with solid carcinoma) immunized against HLA antigens by pregnancy or blood transfusions, respectively. Donors were selected who were compatible in typical RBC antigen systems assayed by conventional techniques but were mismatched for the HLA antigens in question. Crossmatches also were performed with RBC, as well as with lymphocytes, by means of a radioimmune anti-IgG test (RIAT). We found that RBC survival was shortened in all cases. The mean life-span of RBCs depended on antigen specificity rather than on the antibody strength. HLA incompatibility of RBCs could be monitored by the RIAT in all donor/recipient pairs. We conclude that a shortened mean life-span of RBC is to be expected by HLA antibodies, especially when HLA-B7 is involved, but the severity of an in vivo immune reaction in HLA incompatible transfusions cannot be predicted from the in vitro tests used.


Asunto(s)
Autoanticuerpos/inmunología , Envejecimiento Eritrocítico , Eritrocitos/inmunología , Antígenos HLA/análisis , Adulto , Citotoxicidad Inmunológica , Femenino , Humanos , Persona de Mediana Edad , Embarazo , Complicaciones Hematológicas del Embarazo/inmunología , Reacción a la Transfusión
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...