Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 56
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
J Gene Med ; 20(7-8): e3028, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29935087

RESUMEN

BACKGROUND: Previous studies have shown that foamy viral (FV) vectors are a promising alternative to gammaretroviral and lentiviral vectors and also that insulators can improve FV vector safety. However, in a previous analysis of insulator effects on FV vector safety, strong viral promoters were used to elicit genotoxic events. In the present study, we developed and analyzed the efficacy and safety of a high-titer, clinically relevant FV vector driven by the housekeeping promoter elongation factor-1α and insulated with an enhancer blocking A1 insulator (FV-EGW-A1). METHODS: Human CD34+ cord blood cells were exposed to an enhanced green fluorescent protein expressing vector, FV-EGW-A1, at a multiplicity of infection of 10 and then maintained in vitro or transplanted into immunodeficient mice. Flow cytometry was used to measure engraftment and marking in vivo. FV vector integration sites were analyzed to assess safety. RESULTS: FV-EGW-A1 resulted in high-marking, multilineage engraftment of human repopulating cells with no evidence of silencing. Engraftment was highly polyclonal with no clonal dominance and a promising safety profile based on integration site analysis. CONCLUSIONS: An FV vector with an elongation factor-1α promoter and an A1 insulator is a promising vector design for use in the clinic.


Asunto(s)
Técnicas de Transferencia de Gen , Vectores Genéticos/genética , Células Madre Hematopoyéticas/metabolismo , Spumavirus/genética , Transducción Genética , Animales , Expresión Génica , Orden Génico , Genes Reporteros , Terapia Genética , Vectores Genéticos/administración & dosificación , Vectores Genéticos/efectos adversos , Trasplante de Células Madre Hematopoyéticas , Xenoinjertos , Humanos , Ratones , Transgenes
2.
Mol Ther ; 24(7): 1237-46, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27058824

RESUMEN

Lentiviral vectors (LVs) pseudotyped with vesicular stomatitis virus envelope glycoprotein (VSV-G) have demonstrated great promise in gene therapy trials employing hematopoietic stem cell and T-cells. The VSV-G envelope confers broad tropism and stability to the vector but is toxic when constitutively expressed, which has impeded efforts to generate stable producer cell lines. We previously showed that cocal pseudotyped LVs offer an excellent alternative to VSV-G vectors because of their broad tropism and resistance to human serum inactivation. In this study, we demonstrate that cocal LVs transduce CD34(+) and CD4(+) T-cells more efficiently than VSV-G LVs and share the same receptor(s) for cell entry. 293T-cells stably expressing the cocal envelope produced significantly higher LV titers than VSV-G expressing cells. We developed cocal pseudotyped, third-generation, self-inactivating LV producer cell lines for a GFP reporter and for a WT1 tumor-specific T-cell receptor, which achieved concentrated titers above 10(8) IU/ml and were successfully adapted for growth in suspension, serum-free culture. The resulting LVs were at least as effective as standard LVs in transducing CD34(+) and CD4(+) T-cells. Our stable cocal LV producer cell lines should facilitate the production of large-scale, high titer clinical grade vectors.


Asunto(s)
Vectores Genéticos/genética , Células Madre Hematopoyéticas/metabolismo , Lentivirus/genética , Glicoproteínas de Membrana/genética , Linfocitos T/metabolismo , Transducción Genética , Técnicas de Cultivo de Célula , Expresión Génica , Genes Reporteros , Ingeniería Genética , Células HEK293 , Humanos , Lentivirus/metabolismo , Glicoproteínas de Membrana/metabolismo , Receptores de LDL/metabolismo , Receptores Virales/metabolismo , Transgenes , Virus de la Estomatitis Vesicular Indiana/genética , Virus de la Estomatitis Vesicular Indiana/metabolismo , Proteínas del Envoltorio Viral/genética
3.
Mol Carcinog ; 55(11): 1761-1771, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26512949

RESUMEN

Prostate cancer (PC) is the second leading cause of cancer related deaths in US men. Androgen deprivation therapy (ADT) improves clinical outcome, but tumors often recur and progress to androgen independent prostate cancer (AIPC) which no longer responds to ADT. The progression to AIPC is due to genetic alterations that allow PC cancer cells to grow in the absence of androgen. Here we performed an insertional mutagenesis screen using a replication-incompetent lentiviral vector (LV) to identify the genes that promote AIPC in an orthotopic mouse model. Androgen sensitive PC cells, LNCaP, were mutagenized with LV and injected into the prostate of male mice. After tumor development, mice were castrated to select for cells that proliferate in the absence of androgen. Proviral integration sites and nearby dysregulated genes were identified in tumors developed in an androgen deficient environment. Using publically available datasets, the expression of these candidate androgen independence genes in human PC tissues were analyzed. A total of 11 promising candidate AIPC genes were identified: GLYATL1, FLNA, OBSCN, STRA13, WHSC1, ARFGAP3, KDM2A, FAM83H, CLDN7, CNOT6, and B3GNT9. Seven out the 11 candidate genes; GLYATL1, OBSCN, STRA13, KDM2A, FAM83H, CNOT6, and B3GNT6, have not been previously implicated in PC. An in vitro clonogenic assay showed that knockdown of KDM2A, FAM83H, and GLYATL1 genes significantly inhibited the colony forming ability of LNCaP cells. Additionally, we showed that a combination of four genes, OBSCN, FAM83H, CLDN7, and ARFGAP3 could significantly predicted the recurrence risk in PC patients after prostatectomy (P = 5.3 × 10-5 ). © 2015 Wiley Periodicals, Inc.


Asunto(s)
Andrógenos/metabolismo , Genes Relacionados con las Neoplasias , Lentivirus/genética , Mutagénesis Insercional/métodos , Neoplasias de la Próstata/genética , Animales , Línea Celular Tumoral , Progresión de la Enfermedad , Perfilación de la Expresión Génica/métodos , Vectores Genéticos/farmacología , Humanos , Masculino , Ratones , Trasplante de Neoplasias , Neoplasias de la Próstata/metabolismo
4.
Blood ; 123(23): 3578-84, 2014 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-24642749

RESUMEN

Current approaches to hematopoietic stem cell (HSC) gene therapy involve the collection and ex vivo manipulation of HSCs, a process associated with loss of stem cell multipotency and engraftment potential. An alternative approach for correcting blood-related diseases is the direct intravenous administration of viral vectors, so-called in vivo gene therapy. In this study, we evaluated the safety and efficacy of in vivo gene therapy using a foamy virus vector for the correction of canine X-linked severe combined immunodeficiency (SCID-X1). In newborn SCID-X1 dogs, injection of a foamy virus vector expressing the human IL2RG gene resulted in an expansion of lymphocytes expressing the common γ chain and the development of CD3(+) T lymphocytes. CD3(+) cells expressed CD4 and CD8 coreceptors, underwent antigen receptor gene rearrangement, and demonstrated functional maturity in response to T-cell mitogens. Retroviral integration site analysis in 4 animals revealed a polyclonal pattern of integration in all dogs with evidence for dominant clones. These results demonstrate that a foamy virus vector can be administered with therapeutic benefit in the SCID-X1 dog, a clinically relevant preclinical model for in vivo gene therapy.


Asunto(s)
Terapia Genética/métodos , Vectores Genéticos/administración & dosificación , Spumavirus , Enfermedades por Inmunodeficiencia Combinada Ligada al Cromosoma X/terapia , Animales , Células Sanguíneas/metabolismo , Linaje de la Célula/genética , Modelos Animales de Enfermedad , Perros , Células HEK293 , Humanos , Inyecciones Intravenosas , Integración Viral/genética
5.
BMC Bioinformatics ; 16: 212, 2015 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-26150117

RESUMEN

BACKGROUND: Analyzing the integration profile of retroviral vectors is a vital step in determining their potential genotoxic effects and developing safer vectors for therapeutic use. Identifying retroviral vector integration sites is also important for retroviral mutagenesis screens. RESULTS: We developed VISA, a vector integration site analysis server, to analyze next-generation sequencing data for retroviral vector integration sites. Sequence reads that contain a provirus are mapped to the human genome, sequence reads that cannot be localized to a unique location in the genome are filtered out, and then unique retroviral vector integration sites are determined based on the alignment scores of the remaining sequence reads. CONCLUSIONS: VISA offers a simple web interface to upload sequence files and results are returned in a concise tabular format to allow rapid analysis of retroviral vector integration sites.


Asunto(s)
Vectores Genéticos , Genoma Humano , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Internet , Retroviridae/genética , Programas Informáticos , Integración Viral/genética , Humanos
6.
Blood ; 122(2): 179-87, 2013 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-23719296

RESUMEN

Despite continued progress in the development of novel antiretroviral therapies, it has become increasingly evident that drug-based treatments will not lead to a functional or sterilizing cure for HIV(+) patients. In 2009, an HIV(+) patient was effectively cured of HIV following allogeneic transplantation of hematopoietic stem cells (HSCs) from a CCR5(-/-) donor. The utility of this approach, however, is severely limited because of the difficulty in finding matched donors. Hence, we studied the potential of HIV-resistant stem cells in the autologous setting in a nonhuman primate AIDS model and incorporated a fusion inhibitor (mC46) as the means for developing infection-resistant cells. Pigtail macaques underwent identical transplants and Simian-Human Immunodeficiency Virus (SHIV) challenge procedures with the only variation between control and mC46 macaques being the inclusion of a fusion-inhibitor expression cassette. Following SHIV challenge, mC46 macaques, but not control macaques, showed a positive selection of gene-modified CD4(+) T cells in peripheral blood, gastrointestinal tract, and lymph nodes, accounting for >90% of the total CD4(+) T-cell population. mC46 macaques also maintained high frequencies of SHIV-specific, gene-modified CD4(+) T cells, an increase in nonmodified CD4(+) T cells, enhanced cytotoxic T lymphocyte function, and antibody responses. These data suggest that HSC protection may be a potential alternative to conventional antiretroviral therapy in patients with HIV/AIDS.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Proteínas Recombinantes de Fusión/genética , Síndrome de Inmunodeficiencia Adquirida del Simio/genética , Síndrome de Inmunodeficiencia Adquirida del Simio/inmunología , Virus de la Inmunodeficiencia de los Simios/inmunología , Animales , Antígenos Virales/inmunología , Linfocitos B/inmunología , Recuento de Linfocito CD4 , Relación CD4-CD8 , Linfocitos T CD4-Positivos/virología , Tratamiento Basado en Trasplante de Células y Tejidos , Expresión Génica , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/metabolismo , Macaca nemestrina , Síndrome de Inmunodeficiencia Adquirida del Simio/terapia , Síndrome de Inmunodeficiencia Adquirida del Simio/virología , Carga Viral , Viremia/inmunología , Viremia/virología
7.
Mol Cancer ; 13: 120, 2014 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-24885513

RESUMEN

BACKGROUND: Insertional mutagenesis screens have been used with great success to identify oncogenes and tumor suppressor genes. Typically, these screens use gammaretroviruses (γRV) or transposons as insertional mutagens. However, insertional mutations from replication-competent γRVs or transposons that occur later during oncogenesis can produce passenger mutations that do not drive cancer progression. Here, we utilized a replication-incompetent lentiviral vector (LV) to perform an insertional mutagenesis screen to identify genes in the progression to androgen-independent prostate cancer (AIPC). METHODS: Prostate cancer cells were mutagenized with a LV to enrich for clones with a selective advantage in an androgen-deficient environment provided by a dysregulated gene(s) near the vector integration site. We performed our screen using an in vitro AIPC model and also an in vivo xenotransplant model for AIPC. Our approach identified proviral integration sites utilizing a shuttle vector that allows for rapid rescue of plasmids in E. coli that contain LV long terminal repeat (LTR)-chromosome junctions. This shuttle vector approach does not require PCR amplification and has several advantages over PCR-based techniques. RESULTS: Proviral integrations were enriched near prostate cancer susceptibility loci in cells grown in androgen-deficient medium (p < 0.001), and five candidate genes that influence AIPC were identified; ATPAF1, GCOM1, MEX3D, PTRF, and TRPM4. Additionally, we showed that RNAi knockdown of ATPAF1 significantly reduces growth (p < 0.05) in androgen-deficient conditions. CONCLUSIONS: Our approach has proven effective for use in PCa, identifying a known prostate cancer gene, PTRF, and also several genes not previously associated with prostate cancer. The replication-incompetent shuttle vector approach has broad potential applications for cancer gene discovery, and for interrogating diverse biological and disease processes.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Lentivirus/genética , Mutagénesis Insercional/métodos , Próstata/metabolismo , Neoplasias de la Próstata/genética , Receptores Androgénicos/genética , Animales , Línea Celular Tumoral , Proliferación Celular , Células Clonales , Escherichia coli/genética , Escherichia coli/metabolismo , Sitios Genéticos , Vectores Genéticos , Ensayos Analíticos de Alto Rendimiento , Humanos , Lentivirus/metabolismo , Masculino , Ratones , ATPasas de Translocación de Protón Mitocondriales/antagonistas & inhibidores , ATPasas de Translocación de Protón Mitocondriales/genética , ATPasas de Translocación de Protón Mitocondriales/metabolismo , Chaperonas Moleculares/antagonistas & inhibidores , Chaperonas Moleculares/genética , Chaperonas Moleculares/metabolismo , Próstata/patología , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Receptores Androgénicos/deficiencia , Transducción de Señal , Secuencias Repetidas Terminales , Replicación Viral , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Mol Ther ; 21(6): 1270-8, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23587923

RESUMEN

Umbilical cord blood (CB) transplantation is a promising therapeutic approach but continues to be associated with delayed engraftment and infections. Here, we explored in our macaque CB transplant model expansion and engraftment kinetics of cells expanded with the combination of HOXB4 and Delta-1. CB cells were divided into two equal fractions; one fraction was transduced with HOXB4 yellow fluorescent protein (YFP) and expanded on control OP9 cells, and the other was transduced with HOXB4 green fluorescent protein (GFP) and expanded on Delta-expressing OP9 cells (OP9-DL1). Both fractions were transplanted into myeloablated subjects. Neutrophil and platelet recovery occurred within 7 and 19 days respectively, which was significantly earlier than in our previous study using cells expanded with HOXB4 alone, which resulted in neutrophil recovery within 12 days (P = 0.05) and platelet recovery within 37 days (P = 0.02). Furthermore, two of three animals in the current study remained fully transfusion-independent after transplantation. By day 30, reconstitution of lymphocytes was significantly greater with the HOXB4/OP9-DL1 expanded cells in all animals (P = 0.05). In conclusion, our data show that the combination of OP9-DL1 and HOXB4 can result in increased numbers of repopulating cells, thus leading to rapid engraftment and transfusion independence in macaques transplanted with autologous, expanded CB cells.


Asunto(s)
Proteínas de Homeodominio/genética , Péptidos y Proteínas de Señalización Intracelular/genética , Macaca nemestrina/genética , Proteínas de la Membrana/genética , Regiones Promotoras Genéticas , Factores de Transcripción/genética , Animales , Antígenos CD34/metabolismo , Plaquetas/metabolismo , Transfusión Sanguínea/métodos , Diferenciación Celular , Células Cultivadas , Ensayo de Unidades Formadoras de Colonias , Femenino , Sangre Fetal/citología , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Trasplante de Células Madre Hematopoyéticas/métodos , Proteínas de Homeodominio/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Macaca nemestrina/metabolismo , Masculino , Proteínas de la Membrana/metabolismo , Ratones , Factores de Transcripción/metabolismo
9.
Nucleic Acids Res ; 40(2): e14, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22110042

RESUMEN

Two major limitations to achieve efficient homing endonuclease-stimulated gene correction using retroviral vectors are low frequency of gene targeting and random integration of the targeting vectors. To overcome these issues, we developed a reporter system for quick and facile testing of novel strategies to promote the selection of cells that undergo targeted gene repair and to minimize the persistence of random integrations and non-homologous end-joining events. In this system, the gene target has an I-SceI site upstream of an EGFP reporter; and the repair template includes a non-functional EGFP gene, the positive selection transgene MGMTP140K tagged with mCherry, and the inducible Caspase-9 suicide gene. Using this dual fluorescent reporter system it is possible to detect properly targeted integration. Furthermore, this reporter system provides an efficient approach to enrich for gene correction events and to deplete events produced by random integration. We have also developed a second reporter system containing MGMTP140K in the integrated target locus, which allows for selection of primary cells with the integrated gene target after transplantation. This system is particularly useful for testing repair strategies in primary hematopoietic stem cells. Thus, our reporter systems should allow for more efficient gene correction with less unwanted off target effects.


Asunto(s)
Endodesoxirribonucleasas/metabolismo , Marcación de Gen/métodos , Genes Reporteros , Línea Celular , Colorantes Fluorescentes/análisis , Genoma , Proteínas Fluorescentes Verdes/análisis , Proteínas Fluorescentes Verdes/genética , Células HEK293 , Humanos , Proteína Metiltransferasas/análisis , Proteína Metiltransferasas/genética
10.
Biol Blood Marrow Transplant ; 19(8): 1210-9, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23707854

RESUMEN

Hepatic iron overload is common in patients undergoing hematopoietic cell transplantation. We showed previously in a murine model that transplantation of allogeneic T cells induced iron deposition and down-regulation of hepcidin (Hamp) in hepatocytes. We hypothesized that hepatic injury was related to disrupted iron homeostasis triggered by the interaction of Fas-ligand, expressed on activated T cells, with Fas on hepatocytes. In the current study, we determined the effects of modified expression of the Flice inhibitory protein (FLIP long [FLIPL]), which interferes with Fas signaling, on the impact of Fas-initiated signals on the expression of IL-6 and Stat3 and their downstream target, Hamp. To exclude a possible contribution by other pathways, we used agonistic anti-Fas antibodies (rather than allogeneic T cells) to trigger Fas signaling. Inhibition of FLIPL by RNA interference resulted, as expected, not only in enhanced hepatocyte apoptosis in response to Fas signals, but also in decreased levels of IL-6, Stat3, and Hamp. In contrast, overexpression of FLIPL protected hepatocytes against agonistic anti-Fas antibody-mediated apoptosis and increased the levels of IL-6 and Stat3, thereby maintaining the expression of Hamp in an NF-κB-dependent fashion. In vivo overexpression of FLIPL in the liver via hydrodynamic transfection, similarly, interfered with Fas-initiated apoptosis and prevented down-regulation of IL-6, Stat3, and Hamp. These data indicate that Fas-dependent signals alter the regulation of iron homeostasis and suggest that signals initiated by Fas may contribute to peritransplantation iron accumulation.


Asunto(s)
Hepcidinas/metabolismo , Sobrecarga de Hierro/etiología , Linfocitos T/trasplante , Receptor fas/metabolismo , Animales , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/farmacología , Apoptosis/fisiología , Proteína Reguladora de Apoptosis Similar a CASP8 y FADD/antagonistas & inhibidores , Proteína Reguladora de Apoptosis Similar a CASP8 y FADD/biosíntesis , Proteína Reguladora de Apoptosis Similar a CASP8 y FADD/metabolismo , Línea Celular Tumoral , Proteína Ligando Fas/metabolismo , Femenino , Hepatocitos/inmunología , Hepatocitos/metabolismo , Hepcidinas/biosíntesis , Hepcidinas/genética , Humanos , Interleucina-6/biosíntesis , Interleucina-6/genética , Sobrecarga de Hierro/genética , Sobrecarga de Hierro/metabolismo , Ratones , Ratones Endogámicos C57BL , Factor de Transcripción STAT3/biosíntesis , Factor de Transcripción STAT3/genética , Transducción de Señal , Linfocitos T/inmunología , Linfocitos T/metabolismo , Transfección , Trasplante Homólogo , Resultado del Tratamiento , Receptor fas/inmunología
11.
Biochem Biophys Res Commun ; 427(1): 36-40, 2012 Oct 12.
Artículo en Inglés | MEDLINE | ID: mdl-22995402

RESUMEN

AMP-activated protein kinase (AMPK) is a master regulator of energy metabolism in skeletal muscle; AMPK induces muscle protein degradation but the underlying mechanisms are unclear. Myostatin is a powerful negative regulator of skeletal muscle mass and growth in mammalian species. We hypothesized that AMPK stimulates myostatin expression, which provides an explanation for the negative role of AMPK in muscle growth. The objective of this study is to demonstrate that AMPK stimulates myostatin expression using C2C12 cells as a model. Activation of AMPK by 5-aminoimidazole-4-carboxamide-1-ß-d-riboruranoside (AICAR) dramatically increased the mRNA expression and protein content of myostatin in C2C12 myotubes, and to a lesser degree in myoblasts. Metformin, another AMPK activator, also stimulated myostatin expression at low concentrations. In addition, ectopic expression of AMPK wild-type α subunit (enhancing AMPK activity) and AMPK K45R mutant (knockdown AMPK activity) enhanced and reduced myostatin expression, respectively. These results indicate that AMPK stimulates myostatin expression in C2C12 cells, providing an explanation for the negative effect of AMPK on muscle growth.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Músculo Esquelético/crecimiento & desarrollo , Miostatina/biosíntesis , Proteínas Quinasas Activadas por AMP/genética , Aminoimidazol Carboxamida/análogos & derivados , Aminoimidazol Carboxamida/farmacología , Animales , Línea Celular , Activación Enzimática , Metformina/farmacología , Ratones , Ratones Noqueados , Desarrollo de Músculos , Fibras Musculares Esqueléticas/efectos de los fármacos , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/metabolismo , Miostatina/genética , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Ribonucleótidos/farmacología
12.
J Clin Invest ; 118(4): 1502-10, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18357342

RESUMEN

Retroviral vector-mediated HSC gene therapy has been used to treat individuals with a number of life-threatening diseases. However, some patients with SCID-X1 developed retroviral vector-mediated leukemia after treatment. The selective growth advantage of gene-modified cells in patients with SCID-X1 suggests that the transgene may have played a role in leukemogenesis. Here we report that 2 of 2 dogs and 1 of 2 macaques developed myeloid leukemia approximately 2 years after being transplanted with cells that overexpressed homeobox B4 (HOXB4) and cells transduced with a control gammaretroviral vector that did not express HOXB4. The leukemic cells had dysregulated expression of oncogenes, a block in myeloid differentiation, and overexpression of HOXB4. HOXB4 knockdown restored differentiation in leukemic cells, suggesting involvement of HOXB4. In contrast, leukemia did not arise from the cells carrying the control gammaretroviral vector. In addition, leukemia did not arise in 5 animals with high-level marking and polyclonal long-term repopulation following transplantation with cells transduced with an identical gammaretrovirus vector backbone expressing methylguanine methyltransferase. These findings, combined with the absence of leukemia in many other large animals transplanted with cells transduced with gammaretroviral vectors expressing genes other than HOXB4, show that HOXB4 overexpression poses a significant risk of leukemogenesis. Our data thus suggest the continued need for caution in genetic manipulation of repopulating cells, particularly when the transgene might impart an intrinsic growth advantage.


Asunto(s)
Gammaretrovirus/genética , Terapia Genética/efectos adversos , Vectores Genéticos/genética , Proteínas de Homeodominio/metabolismo , Leucemia Mieloide/etiología , Macaca nemestrina , Trasplante de Células Madre/efectos adversos , Animales , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Perros , Regulación de la Expresión Génica , Marcadores Genéticos/genética , Hematopoyesis , Proteínas de Homeodominio/genética , Leucemia Mieloide/genética , Leucemia Mieloide/metabolismo , Leucemia Mieloide/patología , Mutación/genética , Factores de Tiempo
13.
Blood ; 113(21): 5094-103, 2009 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-19336761

RESUMEN

Overexpression of methylguanine methyltransferase P140K (MGMTP140K) has been successfully used for in vivo selection and chemoprotection in mouse and large animal studies, and has promise for autologous and allogeneic gene therapy. We examined the long-term safety of MGMTP140K selection in a clinically relevant dog model. Based on the association of provirus integration and proto-oncogene activation leading to leukemia in the X-linked immunodeficiency trial, we focused our analysis on the distribution of retrovirus integration sites (RIS) relative to proto-oncogene transcription start sites (TSS). We analyzed RIS near proto-oncogene TSS before (n = 157) and after (n = 129) chemotherapy in dogs that received MGMTP140K gene-modified cells and identified no overall increase of RIS near proto-oncogene TSS after chemotherapy. We also wanted to determine whether in vivo selected cells retained fundamental characteristics of hematopoietic stem cells. To that end, we performed secondary transplantation of MGMTP140K gene-modified cells after in vivo selection in dog leukocyte antigen (DLA)-matched dogs. Gene-modified cells achieved multilineage repopulation, and we identified the same gene-modified clone in both dogs more than 800 and 900 days after transplantation. These data suggest that MGMTP140K selection is well tolerated and should allow clinically for selection of gene-corrected cells in genetic or infectious diseases or chemoprotection for treatment of malignancy.


Asunto(s)
Terapia Genética/métodos , Supervivencia de Injerto , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/metabolismo , O(6)-Metilguanina-ADN Metiltransferasa/genética , Animales , Linaje de la Célula , Perros , Vectores Genéticos , Retroviridae/genética
14.
Mol Ther ; 18(4): 725-33, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19997089

RESUMEN

Lentiviral vectors are established as efficient and convenient vehicles for gene transfer. They are almost always pseudotyped with the envelope glycoprotein of vesicular stomatitis virus (VSV-G) due to the high titers that can be achieved, their stability, and broad tropism. We generated a novel cocal vesiculovirus envelope glycoprotein plasmid and compared the properties of lentiviral vectors pseudotyped with cocal, VSV-G, and a modified feline endogenous retrovirus envelope glycoprotein (RD114/TR). Cocal-pseudotyped lentiviral vectors can be produced at titers as high as with VSV-G, have a broad tropism, and are stable, allowing for efficient concentration by centrifugation. Additionally, cocal vectors are more resistant to inactivation by human serum than VSV-G-pseudotyped vectors, and efficiently transduce human CD34(+) nonobese diabetic/severe combined immunodeficient (NOD/SCID) mouse-repopulating cells (SRCs), and long-term primate hematopoietic repopulating cells. These studies establish the potential of cocal-pseudotyped lentiviral vectors for a variety of scientific and therapeutic gene transfer applications, including in vivo gene delivery and hematopoietic stem cell (HSC) gene therapy.


Asunto(s)
Terapia Genética/métodos , Vectores Genéticos/inmunología , Células Madre Hematopoyéticas , Lentivirus/inmunología , Glicoproteínas de Membrana/inmunología , Células Receptoras Sensoriales , Animales , Antígenos CD34/genética , Gatos , Vectores Genéticos/genética , Humanos , Lentivirus/genética , Macaca , Glicoproteínas de Membrana/genética , Ratones , Ratones SCID , Transducción Genética , Proteínas del Envoltorio Viral/genética , Proteínas del Envoltorio Viral/inmunología
15.
Blood ; 111(12): 5537-43, 2008 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-18388180

RESUMEN

Lentiviral vectors are attractive for hematopoietic stem cell (HSC) gene therapy because they do not require mitosis for nuclear entry, they efficiently transduce hematopoietic repopulating cells, and self-inactivating (SIN) designs can be produced at high titer. Experiments to evaluate HIV-derived lentiviral vectors in nonhuman primates prior to clinical trials have been hampered by low transduction frequencies due in part to host restriction by TRIM5alpha. We have established conditions for efficient transduction of pigtailed macaque (Macaca nemestrina) long-term repopulating cells using VSV-G-pseudotyped HIV-based lentiviral vectors. Stable, long-term, high-level gene marking was observed in 3 macaques using relatively low MOIs (5-10) in a 48-hour ex vivo transduction protocol. All animals studied had rapid neutrophil engraftment with a median of 10.3 days to a count greater than 0.5 x 10(9)/L (500/microL). Expression was detected in all lineages, with long-term marking levels in granulocytes at approximately 20% to 30%, and in lymphocytes at approximately 12% to 23%. All animals had polyclonal engraftment as determined by analysis of vector integration sites. These data suggest that lentiviral vectors should be highly effective for HSC gene therapy, particularly for diseases in which maintaining the engraftment potential of stem cells using short-term ex vivo transduction protocols is critical.


Asunto(s)
Vectores Genéticos , VIH-1/genética , Células Madre Hematopoyéticas/fisiología , Lentivirus/genética , Transducción Genética/métodos , Animales , Antígenos CD34/metabolismo , Linaje de la Célula/fisiología , Expresión Génica , Terapia Genética/métodos , Supervivencia de Injerto , Trasplante de Células Madre Hematopoyéticas , Técnicas In Vitro , Macaca nemestrina , Transgenes/genética
16.
J Med Primatol ; 38(6): 438-43, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19793180

RESUMEN

BACKGROUND: Macaca nemestrina is a nonhuman primate used as a model in preclinical studies of hematopoietic stem cell transplantation and adoptive transfer of T cells. Adoptive T cell transfer studies typically require ex vivo expansion of substantial numbers of T cells prior to their reinfusion into the subject. METHODS: Pigtailed macaque peripheral blood CD4(+) cells were expanded using CD3 and CD28 antibody-coated beads. These cells were transformed using Herpesvirus saimiri and were also transduced with HIV-1 based lentiviral vectors. RESULTS: We report an efficient method for the ex vivo expansion of CD4(+) T cells from Macaca nemestrina peripheral blood. With this protocol, primary CD4(+) T cells can be expanded between 300- to 6000-fold during 24-day period and can be efficiently transduced with lentiviral vectors. Furthermore, these T cells can be transformed by Herpesvirus saimiri and maintained in culture for several months. The transformed T cell lines can be productively infected with the simian immunodeficiency virus (SIV) strain SIV(mac239). CONCLUSIONS: We have established methods for the expansion and transformation of primary M. nemestrina CD4(+) T cells and demonstrated the utility of these methods for several applications.


Asunto(s)
Linfocitos T CD4-Positivos , Técnicas de Cultivo de Célula , Macaca nemestrina/inmunología , Animales , Antígenos CD28/inmunología , Complejo CD3/inmunología , Linfocitos T CD4-Positivos/virología , Línea Celular Transformada , Humanos , Separación Inmunomagnética , Síndrome de Inmunodeficiencia Adquirida del Simio , Virus de la Inmunodeficiencia de los Simios
17.
Methods Mol Biol ; 1907: 103-114, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30542994

RESUMEN

Prostate cancer (PC) is the second leading cause of cancer-related deaths in US men, and progression to androgen-independent PC (AIPC) typically results in metastasis and is lethal. However, the mechanisms whereby PC progresses from androgen dependence to androgen independence are not completely understood. Mutagenesis screens to identify novel genes involved in the progression to AIPC have been performed using replication-incompetent lentiviral vectors (LVs). In this approach the LV acts both as a mutagen and as molecular tag to identify nearby genes that may have been dysregulated by the vector provirus, and are candidate AIPC genes. Here we describe protocols for generation of replication-incompetent LV preparations and performing a mutagenesis screen to identify AIPC genes in vitro.


Asunto(s)
Andrógenos/metabolismo , Genes Relacionados con las Neoplasias , Lentivirus/genética , Mutagénesis , Neoplasias de la Próstata/diagnóstico , Neoplasias de la Próstata/genética , Replicación Viral , Progresión de la Enfermedad , Vectores Genéticos , Ensayos Analíticos de Alto Rendimiento , Humanos , Masculino , Células Tumorales Cultivadas
18.
Mol Ther ; 15(7): 1356-65, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17440443

RESUMEN

The potential for leukemia caused by retroviral vector integration has become a significant concern for hematopoietic stem cell gene therapy. We analyzed the distribution of vector integrants in pigtailed macaque and baboon repopulating cells for the two most commonly used retroviral vector systems, human immunodeficiency virus (HIV)-based lentiviral vectors and murine leukemia virus (MLV)-based gammaretroviral vectors, to help define their relative genotoxicity. All animals had polyclonal engraftment with no apparent adverse effects from transplantation with gene-modified cells. In all, 380 MLV and 235 HIV unique vector integration sites were analyzed and had distinct distribution patterns in relation to genes and CpG islands as observed in previous in vitro studies. Both vector types were found more frequently in and near proto-oncogenes in repopulating cells than in a random dataset. Analysis of functional classes of genes with integrants within 100 kilobases (kb) of their transcription start sites showed an over-representation of genes involved in growth or survival near both lentiviral and gammaretroviral integrants. Microarray analysis showed that both gammaretroviral and lentiviral vectors were found close to genes with high expression levels in primitive cells enriched for hematopoietic stem cells. These data help define the relative risk of insertional mutagenesis with MLV-, HIV-, and simian immunodeficiency virus (SIV)-based vectors in a highly relevant primate model.


Asunto(s)
Vectores Genéticos/genética , VIH/genética , Virus de la Leucemia Murina/genética , Macaca nemestrina/genética , Papio/genética , Integración Viral/genética , Animales , Células Sanguíneas/metabolismo , Islas de CpG , Expresión Génica , Genoma/genética , Proto-Oncogenes/genética , Transcripción Genética/genética
19.
Oncotarget ; 9(21): 15451-15463, 2018 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-29643985

RESUMEN

Replication-incompetent gammaretroviral (γRV) and lentiviral (LV) vectors have both been used in insertional mutagenesis screens to identify cancer drivers. In this approach the vectors stably integrate in the host cell genome and induce cancers by dysregulating nearby genes. The cells that contain a retroviral vector provirus in or near a proto-oncogene or tumor suppressor are preferentially enriched in a tumor. γRV and LV vectors have different integration profiles and genotoxic potential, making them potentially complementary tools for insertional mutagenesis screens. We performed screens using both γRV and LV vectors to identify driver genes that mediate progression of androgen-independent prostate cancer (AIPC) using a xenotransplant mouse model. Vector transduced LNCaP cells were injected orthotopically into the prostate gland of immunodeficient mice. Mice that developed tumors were castrated to create an androgen-deficient environment and metastatic tumors that developed were analyzed. A high-throughput modified genomic sequencing PCR (MGS-PCR) approach identified the positions of vector integrations in these metastatic tumors. OR2A14, FER1L6, TAOK3, MAN1A2, MBNL2, SERBP1, PLEKHA2, SPTAN1, ADAMTS1, SLC30A5, ABCC1, SLC7A1 and SLC25A24 were identified as candidate prostate cancer (PC) progression genes. TAOK3 and ABCC1 expression in PC patients predicted the risk of recurrence after androgen deprivation therapy. Our data shows that γRV and LV vectors are complementary approaches to identify cancer driver genes which may be promising potential biomarkers and therapeutic targets.

20.
Blood Adv ; 2(9): 987-999, 2018 05 08.
Artículo en Inglés | MEDLINE | ID: mdl-29720491

RESUMEN

Hematopoietic stem-cell gene therapy is a promising treatment of X-linked severe combined immunodeficiency disease (SCID-X1), but currently, it requires recipient conditioning, extensive cell manipulation, and sophisticated facilities. With these limitations in mind, we explored a simpler therapeutic approach to SCID-X1 treatment by direct IV administration of foamy virus (FV) vectors in the canine model. FV vectors were used because they have a favorable integration site profile and are resistant to serum inactivation. Here, we show improved efficacy of our in vivo gene therapy platform by mobilization with granulocyte colony-stimulating factor (G-CSF) and AMD3100 before injection of an optimized FV vector incorporating the human phosphoglycerate kinase enhancerless promoter. G-CSF/AMD3100 mobilization before FV vector delivery accelerated kinetics of CD3+ lymphocyte recovery, promoted thymopoiesis, and increased immune clonal diversity. Gene-corrected T lymphocytes exhibited a normal CD4:CD8 ratio and a broad T-cell receptor repertoire and showed restored γC-dependent signaling function. Treated animals showed normal primary and secondary antibody responses to bacteriophage immunization and evidence for immunoglobulin class switching. These results demonstrate safety and efficacy of an accessible, portable, and translatable platform with no conditioning regimen for the treatment of SCID-X1 and other genetic diseases.


Asunto(s)
Enfermedades de los Perros , Terapia Genética , Vectores Genéticos/farmacología , Factor Estimulante de Colonias de Granulocitos/farmacología , Movilización de Célula Madre Hematopoyética , Compuestos Heterocíclicos/farmacología , Spumavirus , Enfermedades por Inmunodeficiencia Combinada Ligada al Cromosoma X , Animales , Bencilaminas , Relación CD4-CD8 , Ciclamas , Modelos Animales de Enfermedad , Enfermedades de los Perros/sangre , Enfermedades de los Perros/genética , Enfermedades de los Perros/terapia , Perros , Humanos , Fosfoglicerato Quinasa/genética , Enfermedades por Inmunodeficiencia Combinada Ligada al Cromosoma X/sangre , Enfermedades por Inmunodeficiencia Combinada Ligada al Cromosoma X/genética , Enfermedades por Inmunodeficiencia Combinada Ligada al Cromosoma X/terapia , Enfermedades por Inmunodeficiencia Combinada Ligada al Cromosoma X/veterinaria
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA