Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters











Publication year range
1.
Braz J Med Biol Res ; 56: e12682, 2023.
Article in English | MEDLINE | ID: mdl-37493770

ABSTRACT

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.


Subject(s)
Adenoviridae , Virus Replication , Adenoviridae/genetics , RNA, Small Interfering/genetics , Genetic Vectors/genetics , Humans , HEK293 Cells , Transcription, Genetic , Clone Cells
2.
Rev. bras. pesqui. méd. biol ; Braz. j. med. biol. res;56: e12682, 2023. graf
Article in English | LILACS-Express | LILACS | ID: biblio-1447681

ABSTRACT

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.
Article in English | MEDLINE | ID: mdl-34008754

ABSTRACT

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.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors , Cochlea , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Differentiation , Hair Cells, Auditory , Mice , Organ of Corti , Receptors, Notch , Transcription Factor HES-1/genetics
4.
Rev. bras. pesqui. méd. biol ; Braz. j. med. biol. res;54(7): e10579, 2021. tab, graf
Article in English | LILACS | ID: biblio-1249313

ABSTRACT

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.


Subject(s)
Animals , Rats , Cochlea , Basic Helix-Loop-Helix Transcription Factors/genetics , Organ of Corti , Cell Differentiation , Receptors, Notch , Transcription Factor HES-1/genetics , Hair Cells, Auditory
5.
Braz J Med Biol Res ; 53(3): e8876, 2020.
Article in English | MEDLINE | ID: mdl-32077463

ABSTRACT

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.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p16/administration & dosage , Interferon-beta/metabolism , Melanoma, Experimental/therapy , Mesenchymal Stem Cells/metabolism , Animals , Cell Line, Tumor , Cell Proliferation , Disease Models, Animal , Genetic Therapy , Male , Melanoma, Experimental/metabolism , Mice , Mice, Inbred C57BL , Transduction, Genetic
6.
Rev. bras. pesqui. méd. biol ; Braz. j. med. biol. res;53(3): e8876, 2020. graf
Article in English | LILACS | ID: biblio-1089338

ABSTRACT

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.


Subject(s)
Animals , Male , Rabbits , Melanoma, Experimental/therapy , Interferon-beta/metabolism , Cyclin-Dependent Kinase Inhibitor p16/administration & dosage , Mesenchymal Stem Cells/metabolism , Transduction, Genetic , Melanoma, Experimental/metabolism , Genetic Therapy , Cell Line, Tumor , Cell Proliferation , Disease Models, Animal , Mice, Inbred C57BL
7.
Braz J Med Biol Res ; 49(4): e5064, 2016.
Article in English | MEDLINE | ID: mdl-27007652

ABSTRACT

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.


Subject(s)
Hair Cells, Auditory, Inner/transplantation , Hearing Loss, Sensorineural/surgery , Organ of Corti/surgery , Stem Cell Transplantation/methods , Stem Cells , Animals , Auditory Threshold , Cell Survival , Cells, Cultured , Evoked Potentials, Auditory, Brain Stem , Female , Guinea Pigs , Immunohistochemistry , Mice, Inbred BALB C , Neomycin , Protein Synthesis Inhibitors , Reproducibility of Results , Treatment Outcome
8.
Rev. bras. pesqui. méd. biol ; Braz. j. med. biol. res;49(4): e5064, 2016. tab, graf
Article in English | LILACS | ID: biblio-951670

ABSTRACT

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.


Subject(s)
Animals , Female , Organ of Corti/surgery , Stem Cells , Stem Cell Transplantation/methods , Hair Cells, Auditory, Inner/transplantation , Hearing Loss, Sensorineural/surgery , Auditory Threshold , Immunohistochemistry , Protein Synthesis Inhibitors , Neomycin , Cell Survival , Cells, Cultured , Reproducibility of Results , Evoked Potentials, Auditory, Brain Stem , Treatment Outcome , Guinea Pigs , Mice, Inbred BALB C
9.
Cancer Gene Ther ; 20(5): 317-25, 2013 May.
Article in English | MEDLINE | ID: mdl-23618951

ABSTRACT

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.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p16/genetics , Interferon-beta/genetics , Melanoma, Experimental/genetics , Melanoma, Experimental/therapy , Animals , Apoptosis/genetics , Cell Death/genetics , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p16/biosynthesis , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Disease Models, Animal , Female , Interferon-beta/biosynthesis , Interferon-beta/metabolism , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Transduction, Genetic
10.
In Vitro Cell Dev Biol Anim ; 49(4): 250-9, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23519559

ABSTRACT

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.


Subject(s)
Fibroblasts/cytology , Active Transport, Cell Nucleus , Animals , Animals, Genetically Modified , Blastocyst/cytology , Blastocyst/metabolism , Cattle , Cell Lineage , Cell Survival , Cloning, Organism , Embryo, Mammalian/cytology , Fibroblasts/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , In Vitro Techniques , Nuclear Transfer Techniques , Time Factors
11.
Cell Transplant ; 19(3): 279-89, 2010.
Article in English | MEDLINE | ID: mdl-19995482

ABSTRACT

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.


Subject(s)
Adipose Tissue/physiology , Adult Stem Cells/physiology , Cell Differentiation/physiology , Multipotent Stem Cells/physiology , Adipose Tissue/cytology , Adult Stem Cells/cytology , Adult Stem Cells/drug effects , Animals , Cell Culture Techniques , Cell Differentiation/drug effects , Cell Lineage/drug effects , Cell Lineage/physiology , Cell Separation/methods , Cells, Cultured , Dogs , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/physiology , Mesoderm/cytology , Mesoderm/physiology , Multipotent Stem Cells/cytology , Multipotent Stem Cells/drug effects
12.
Genet Mol Res ; 7(3): 925-32, 2008 Sep 30.
Article in English | MEDLINE | ID: mdl-18949710

ABSTRACT

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.


Subject(s)
Gene Expression Profiling , Oligonucleotide Array Sequence Analysis/methods , Peptides/physiology , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic , Humans , Peptides/genetics , RNA Interference
13.
Rev. bras. pesqui. méd. biol ; Braz. j. med. biol. res;40(5): 601-613, May 2007. ilus, tab
Article in English | LILACS | ID: lil-449078

ABSTRACT

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.


Subject(s)
Humans , Genetic Therapy , Leukemia/etiology , X-Linked Combined Immunodeficiency Diseases/therapy , Cell Transformation, Neoplastic , Clinical Trials as Topic , Genetic Therapy/adverse effects , Genetic Therapy/methods , Genetic Vectors/genetics , Risk Factors , Transduction, Genetic , X-Linked Combined Immunodeficiency Diseases/genetics , X-Linked Combined Immunodeficiency Diseases/immunology
14.
Braz J Med Biol Res ; 40(5): 601-13, 2007 May.
Article in English | MEDLINE | ID: mdl-17464421

ABSTRACT

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.


Subject(s)
Genetic Therapy , Leukemia/etiology , X-Linked Combined Immunodeficiency Diseases/therapy , Cell Transformation, Neoplastic , Clinical Trials as Topic , Genetic Therapy/adverse effects , Genetic Therapy/methods , Genetic Vectors/genetics , Humans , Risk Factors , Transduction, Genetic , X-Linked Combined Immunodeficiency Diseases/genetics , X-Linked Combined Immunodeficiency Diseases/immunology
15.
Braz J Med Biol Res ; 32(7): 905-14, 1999 07.
Article in English | MEDLINE | ID: mdl-10454751

ABSTRACT

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.


Subject(s)
Gene Transfer Techniques , Genetic Therapy/methods , Genetic Vectors/genetics , Glioblastoma/genetics , Glioblastoma/therapy , Retroviridae/genetics , Animals , Cell Cycle/genetics , Cell Cycle/physiology , Cell Transformation, Neoplastic/genetics , Clinical Trials as Topic , Disease Models, Animal , Genes, Tumor Suppressor , Humans , Mice , Rats
16.
Rev. bras. pesqui. méd. biol ; Braz. j. med. biol. res;32(7): 905-14, July 1999.
Article in English | LILACS | ID: lil-234898

ABSTRACT

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


Subject(s)
Rats , Mice , Animals , Humans , Gene Transfer Techniques , Genetic Therapy/methods , Genetic Vectors/genetics , Glioblastoma/genetics , Glioblastoma/therapy , Retroviridae/genetics , Cell Cycle/genetics , Cell Cycle/physiology , Cell Transformation, Neoplastic/genetics , Clinical Trials as Topic , Disease Models, Animal , Genes, Tumor Suppressor
SELECTION OF CITATIONS
SEARCH DETAIL