Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Blood ; 117(10): 2817-26, 2011 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-21156846

RESUMEN

ß-Thalassemia major results from severely reduced or absent expression of the ß-chain of adult hemoglobin (α2ß2;HbA). Increased levels of fetal hemoglobin (α2γ2;HbF), such as occurs with hereditary persistence of HbF, ameliorate the severity of ß-thalassemia, raising the potential for genetic therapy directed at enhancing HbF. We used an in vitro model of human erythropoiesis to assay for enhanced production of HbF after gene delivery into CD34(+) cells obtained from mobilized peripheral blood of normal adults or steady-state bone marrow from patients with ß-thalassemia major. Lentiviral vectors encoding (1) a human γ-globin gene with or without an insulator, (2) a synthetic zinc-finger transcription factor designed to interact with the γ-globin gene promoters, or (3) a short-hairpin RNA targeting the γ-globin gene repressor, BCL11A, were tested. Erythroid progeny of normal CD34(+) cells demonstrated levels of HbF up to 21% per vector copy. For ß-thalassemic CD34(+) cells, similar gene transfer efficiencies achieved HbF production ranging from 45% to 60%, resulting in up to a 3-fold increase in the total cellular Hb content. These observations suggest that both lentiviral-mediated γ-globin gene addition and genetic reactivation of endogenous γ-globin genes have potential to provide therapeutic HbF levels to patients with ß-globin deficiency.


Asunto(s)
Células Precursoras Eritroides/metabolismo , Hemoglobina Fetal/biosíntesis , Técnicas de Transferencia de Gen , Terapia Genética , Talasemia beta/terapia , gamma-Globinas/genética , Antígenos CD34/metabolismo , Southern Blotting , Western Blotting , Separación Celular , Eritropoyesis/fisiología , Hemoglobina Fetal/genética , Citometría de Flujo , Vectores Genéticos , Humanos , Lentivirus/genética , Reacción en Cadena de la Polimerasa
2.
Mol Ther ; 20(10): 1882-92, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22871664

RESUMEN

Human immunodeficiency virus type 1 (HIV1) vectors poorly transduce rhesus hematopoietic cells due to species-specific restriction factors, including the tripartite motif-containing 5 isoformα (TRIM5α) which targets the HIV1 capsid. We previously developed a chimeric HIV1 (χHIV) vector system wherein the vector genome is packaged with the simian immunodeficiency virus (SIV) capsid for efficient transduction of both rhesus and human CD34(+) cells. To evaluate whether χHIV vectors could efficiently transduce rhesus hematopoietic repopulating cells, we performed a competitive repopulation assay in rhesus macaques, in which half of the CD34(+) cells were transduced with standard SIV vectors and the other half with χHIV vectors. As compared with SIV vectors, χHIV vectors achieved higher vector integration, and the transgene expression rates were two- to threefold higher in granulocytes and red blood cells and equivalent in lymphocytes and platelets for 2 years. A recipient of χHIV vector-only transduced cells reached up to 40% of transgene expression rates in granulocytes and lymphocytes and 20% in red blood cells. Similar to HIV1 and SIV vectors, χHIV vector frequently integrated into gene regions, especially into introns. In summary, our χHIV vector demonstrated efficient transduction for rhesus long-term repopulating cells, comparable with SIV vectors. This χHIV vector should allow preclinical testing of HIV1-based therapeutic vectors in large animal models.


Asunto(s)
Vectores Genéticos/genética , VIH-1/genética , Células Madre Hematopoyéticas , Transducción Genética , Animales , Antígenos CD34/metabolismo , Southern Blotting , Cápside/metabolismo , Proteínas de la Cápside/genética , Proteínas de la Cápside/metabolismo , Línea Celular , Trasplante de Células Madre Hematopoyéticas , Humanos , Macaca mulatta , Reacción en Cadena en Tiempo Real de la Polimerasa , Virus de la Inmunodeficiencia de los Simios/genética , Transgenes
3.
Blood ; 115(15): 3033-41, 2010 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-20190190

RESUMEN

Fetal hemoglobin (HbF) is a potent genetic modifier of the severity of beta-thalassemia and sickle cell anemia. We used an in vitro culture model of human erythropoiesis in which late-stage erythroblasts are derived directly from human CD34(+) hematopoietic cells to evaluate HbF production. This system recapitulates expression of globin genes according to the developmental stage of the originating cell source. When cytokine-mobilized peripheral blood CD34(+) cells from adults were cultured, background levels of HbF were 2% or less. Cultured cells were readily transduced with lentiviral vectors when exposed to vector particles between 48 and 72 hours. Among the genetic elements that may enhance fetal hemoglobin production is an artificial zinc-finger transcription factor, GG1-VP64, designed to interact with the proximal gamma-globin gene promoters. Our data show that lentiviral-mediated, enforced expression of GG1-VP64 under the control of relatively weak erythroid-specific promoters induced significant amounts of HbF (up to 20%) in erythroblasts derived from adult CD34(+) cells without altering their capacity for erythroid maturation and only modestly reducing the total numbers of cells that accumulate in culture after transduction. These observations demonstrate the potential for sequence-specific enhancement of HbF in patients with beta-thalassemia or sickle cell anemia.


Asunto(s)
Eritroblastos/metabolismo , Hemoglobina Fetal/biosíntesis , Regiones Promotoras Genéticas/genética , Transactivadores/metabolismo , Dedos de Zinc , gamma-Globinas/genética , Adulto , Antígenos CD34/metabolismo , Biomarcadores/metabolismo , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Cromatina/metabolismo , Citocinas/farmacología , Eritroblastos/citología , Eritroblastos/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Vectores Genéticos/genética , Movilización de Célula Madre Hematopoyética , Humanos , Lentivirus/genética , Modelos Genéticos , Especificidad de Órganos/efectos de los fármacos , Especificidad de Órganos/genética , Unión Proteica/efectos de los fármacos , Transducción Genética
4.
Brain Res ; 1793: 148053, 2022 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-35973608

RESUMEN

The UBTF E210K neuroregression syndrome is caused by de novo dominant mutations in UBTF (NM_014233.3:c.628G > A, p.Glu210Lys). In humans, onset is typically at 2.5 to 3 years and characterized by slow progression of global motor, cognitive and behavioral dysfunction. Other potentially pathogenic UBTF variants have been reported in humans with severe neurological disease and it remains undetermined if the UBTF E210K mutation operates via gain- and/or loss-of-function. Here we examine the behavioral, cognitive, motor, and molecular effects of Ubtf knockout and knockdown in mice as a means of gauging the role of loss-of-function in humans. Ubtf+/- mice show progression of behavioral (dominance tube), cognitive (cross maze), and mild motor abnormalities from 3 to 18 months. At 18 months, Ubtf+/- mice had more slips on a raised 9-mm round beam task, shorter latencies to fall on the accelerated rotarod, reduced open field vertical and jump counts, and significant deficits in spatial learning and memory. Via crosses to Nestin-Cre (NesCre) mice we found that homozygous Ubtf deletion limited to the central nervous system was embryonic lethal. Tamoxifen-induced homozygous knockdown of Ubtf in adult mice with the Cre-ERT2 system was associated with precipitous deterioration in neurological functioning. At the molecular level, 18-month-old Ubtf+/- mice showed mild increases in cerebellar 53BP1 immunoreactivity. These findings show that UBTF is essential for embryogenesis and survival in adults, and the deleterious effects of UBTF haploinsufficiency progress with age. Loss-of-function mechanisms may contribute, in part, to the human UBTF E210K neuroregression syndrome.


Asunto(s)
Enfermedades del Sistema Nervioso , Adulto , Animales , Modelos Animales de Enfermedad , Humanos , Lactante , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación
5.
Blood ; 113(23): 5747-56, 2009 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-19365082

RESUMEN

Correction of murine models of beta-thalassemia has been achieved through high-level globin lentiviral vector gene transfer into mouse hematopoietic stem cells (HSCs). However, transduction of human HSCs is less robust and may be inadequate to achieve therapeutic levels of genetically modified erythroid cells. We therefore developed a double gene lentiviral vector encoding both human gamma-globin under the transcriptional control of erythroid regulatory elements and methylguanine methyltransferase (MGMT), driven by a constitutive cellular promoter. MGMT expression provides cellular resistance to alkylator drugs, which can be administered to kill residual untransduced, diseased HSCs, whereas transduced cells are protected. Mice transplanted with beta-thalassemic HSCs transduced with a gamma-globin/MGMT vector initially had subtherapeutic levels of red cells expressing gamma-globin. To enrich gamma-globin-expressing cells, transplanted mice were treated with the alkylator agent 1,3-bis-chloroethyl-1-nitrosourea. This resulted in significant increases in the number of gamma-globin-expressing red cells and the amount of fetal hemoglobin, leading to resolution of anemia. Selection of transduced HSCs was also obtained when cells were drug-treated before transplantation. Mice that received these cells demonstrated reconstitution with therapeutic levels of gamma-globin-expressing cells. These data suggest that MGMT-based drug selection holds promise as a modality to improve gene therapy for beta-thalassemia.


Asunto(s)
Metilasas de Modificación del ADN/metabolismo , Enzimas Reparadoras del ADN/metabolismo , Vectores Genéticos/genética , Células Madre Hematopoyéticas/metabolismo , Lentivirus/genética , Proteínas Supresoras de Tumor/metabolismo , Talasemia beta/metabolismo , gamma-Globinas/metabolismo , Animales , Metilasas de Modificación del ADN/genética , Enzimas Reparadoras del ADN/genética , Resistencia a Medicamentos , Eritrocitos/metabolismo , Femenino , Terapia Genética , Trasplante de Células Madre Hematopoyéticas , Masculino , Ratones , Proteínas Supresoras de Tumor/genética , Talasemia beta/genética , Talasemia beta/patología , Talasemia beta/terapia , gamma-Globinas/genética
6.
Mol Ther ; 17(2): 245-52, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19050697

RESUMEN

Increased levels of red cell fetal hemogloblin, whether due to hereditary persistence of expression or from induction with hydroxyurea therapy, effectively ameliorate sickle cell disease (SCD). Therefore, we developed erythroid-specific, gamma-globin lentiviral vectors for hematopoietic stem cell (HSC)-targeted gene therapy with the goal of permanently increasing fetal hemoglobin (HbF) production in sickle red cells. We evaluated two different gamma-globin lentiviral vectors for therapeutic efficacy in the BERK sickle cell mouse model. The first vector, V5, contained the gamma-globin gene driven by 3.1 kb of beta-globin regulatory sequences and a 130-bp beta-globin promoter. The second vector, V5m3, was identical except that the gamma-globin 3'-untranslated region (3'-UTR) was replaced with the beta-globin 3'-UTR. Adult erythroid cells have beta-globin mRNA 3'-UTR-binding proteins that enhance beta-globin mRNA stability and we postulated this design might enhance gamma-globin expression. Stem cell gene transfer was efficient and nearly all red cells in transplanted mice expressed human gamma-globin. Both vectors demonstrated efficacy in disease correction, with the V5m3 vector producing a higher level of gamma-globin mRNA which was associated with high-level correction of anemia and secondary organ pathology. These data support the rationale for a gene therapy approach to SCD by permanently enhancing HbF using a gamma-globin lentiviral vector.


Asunto(s)
Anemia de Células Falciformes/terapia , Hemoglobina Fetal/fisiología , Terapia Genética/métodos , Vectores Genéticos/genética , Lentivirus/genética , gamma-Globinas/genética , Animales , Southern Blotting , Células Cultivadas , Electroforesis , Hemoglobina Fetal/genética , Hemoglobina Fetal/metabolismo , Citometría de Flujo , Trasplante de Células Madre Hematopoyéticas , Ratones , Modelos Genéticos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Bazo/metabolismo , Bazo/patología
7.
Mol Ther ; 16(3): 525-33, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18195719

RESUMEN

Although hematopoietic cell gene therapy using retroviral vectors has recently achieved success in clinical trials, safety issues regarding vector insertional mutagenesis have emerged. Vector insertion, resulting in transcriptional activation of proto-oncogenes, played a role in the development of lymphoid leukemia in an X-linked severe combined immunodeficiency trial, and caused myeloid clonal dominance in a trial for chronic granulomatous disease. These events have raised the question of whether gene therapy for other disorders such as beta-thalassemia and sickle cell disease may hold a similar risk. In this study, we prospectively evaluated whether gamma-globin lentiviral vectors containing enhancer elements from the beta-globin locus control region could alter the expression of genes near the vector insertion. We studied this question in primary, clonal murine beta-thalassemic erythroid cells, where globin regulatory elements are highly active. We found an overall incidence of perturbed expression in 28% of the transduced clones, with 11% of all genes contained within a 600-kilobase region surrounding the vector-insertion site demonstrating altered expression. This rate was higher than that observed for a lentiviral vector containing a viral long-terminal repeat (LTR). This is the first direct evidence that lentiviral vectors can cause insertional dysregulation of cellular genes at a frequent rate.


Asunto(s)
Eritrocitos/metabolismo , Vectores Genéticos/genética , Lentivirus/genética , Talasemia beta/sangre , Animales , Células Cultivadas , Eritrocitos/citología , Expresión Génica , Globinas/genética , Globinas/metabolismo , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Ratones , Mutagénesis Insercional , Análisis de Secuencia por Matrices de Oligonucleótidos , Talasemia beta/patología
8.
Mol Ther Methods Clin Dev ; 2: 15045, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26665131

RESUMEN

Patients with severe sickle cell disease (SCD) are candidates for gene therapy using autologous hematopoietic stem cells (HSCs), but concomitant multi-organ disease may contraindicate pretransplant conditioning with full myeloablation. We tested whether nonmyeloablative conditioning, a regimen used successfully for allogeneic bone marrow transplantation of adult SCD patients, allows engraftment of γ-globin gene-corrected cells to a therapeutic level in the Berkeley mouse model of SCD. Animals transplanted according to this regimen averaged 35% engraftment of transduced hematopoietic stem cells with an average vector copy < 2.0. Fetal hemoglobin (HbF) levels ranged from 20 to 44% of total hemoglobin and approximately two-thirds of circulating red blood cells expressed HbF detected by immunofluorescence (F-cells). Gene therapy treatment of SCD mice ameliorated anemia, reduced hyperleukocytosis, improved renal function, and reduced iron accumulation in liver, spleen, and kidneys. Thus, modest levels of chimerism with donor cells expressing high levels of HbF from an insulated γ-globin lentiviral vector can improve the pathology of SCD in mice, thereby illustrating a potentially safe and effective strategy for gene therapy in humans.

9.
Exp Hematol ; 43(5): 346-351, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25681747

RESUMEN

Sickle cell disease (SCD) can be cured by allogeneic hematopoietic stem cell transplant. However, this is only possible when a matched donor is available, making the development of gene therapy using autologous hematopoietic stem cells a highly desirable alternative. We used a culture model of human erythropoiesis to directly compare two insulated, self-inactivating, and erythroid-specific lentiviral vectors, encoding for γ-globin (V5m3-400) or a modified ß-globin (ßAS3-FB) for production of antisickling hemoglobin (Hb) and correction of red cell deformability after deoxygenation. Bone marrow CD34+ cells from three SCD patients were transduced using V5m3-400 or ßAS3-FB and compared with mock-transduced SCD or healthy donor CD34+ cells. Lentiviral transduction did not impair cell growth or differentiation, as gauged by proliferation and acquisition of erythroid markers. Vector copy number averaged approximately one copy per cell, and corrective globin mRNA levels were increased more than sevenfold over mock-transduced controls. Erythroblasts derived from healthy donor and mock-transduced SCD cells produced a low level of fetal Hb that was increased to 23.6 ± 4.1% per vector copy for cells transduced with V5m3-400. Equivalent levels of modified normal adult Hb of 17.6 ± 3.8% per vector copy were detected for SCD cells transduced with ßAS3-FB. These levels of antisickling Hb production were sufficient to reduce sickling of terminal-stage red blood cells upon deoxygenation. We concluded that the achieved levels of fetal Hb and modified normal adult Hb would likely prove therapeutic to SCD patients who lack matched donors.


Asunto(s)
Células de la Médula Ósea/metabolismo , Lentivirus/genética , Globinas beta/genética , gamma-Globinas/genética , Anemia de Células Falciformes/genética , Anemia de Células Falciformes/terapia , Antígenos CD34/metabolismo , Hemoglobina Fetal/genética , Citometría de Flujo , Expresión Génica , Técnicas de Transferencia de Gen , Terapia Genética/métodos , Vectores Genéticos/genética , Hemoglobinas/genética , Humanos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Globinas beta/metabolismo , gamma-Globinas/metabolismo
10.
J Pediatr Surg ; 42(7): 1172-9, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17618876

RESUMEN

BACKGROUND: We evaluated the potential of bioluminescence imaging (BLI) for early tumor detection, demonstrating occult sites of disseminated disease and assessing disease progression in a murine model of neuroblastoma. METHODS: Neuroblastoma cells engineered to express the enzyme firefly luciferase were used to establish localized tumors and disseminated disease in SCID mice. Bioluminescent signal intensity was measured at serial time points, and compared with traditional methods of evaluating tumor growth. RESULTS: Bioluminescence imaging detected subcutaneous and retroperitoneal tumors weeks before they were palpable or appreciable by ultrasound. Bioluminescent signal intensity at both sites then paralleled tumor growth. After intravenous administration of tumor cells, BLI revealed disseminated disease in the liver, lungs, and bone marrow, again weeks before any gross disease was present. The presence of tumor within these sites at early time points was confirmed by reverse transcriptase-polymerase chain reaction. Finally, BLI permitted a real-time, noninvasive, quantitative method for following response to therapy in a model of minimal residual disease. CONCLUSION: Bioluminescence imaging detects tumor much earlier than traditional methods. In addition, it can detect, quantify, and follow micrometastasis in real-time during disease progression. This methodology is extremely valuable for studying tumor tissue tropism, mechanisms of metastasis, and response to therapy in murine tumor models.


Asunto(s)
Mediciones Luminiscentes/métodos , Neuroblastoma/patología , Animales , Southern Blotting , Progresión de la Enfermedad , Diagnóstico Precoz , Técnicas para Inmunoenzimas , Luciferasas de Luciérnaga , Ratones , Ratones SCID , Análisis de Regresión , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sensibilidad y Especificidad
11.
Blood ; 101(6): 2175-83, 2003 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-12411297

RESUMEN

Increased fetal hemoglobin (HbF) levels diminish the clinical severity of beta-thalassemia and sickle cell anemia. A treatment strategy using autologous stem cell-targeted gene transfer of a gamma-globin gene may therefore have therapeutic potential. We evaluated oncoretroviral- and lentiviral-based gamma-globin vectors for expression in transduced erythroid cell lines. Compared with gamma-globin, oncoretroviral vectors containing either a beta-spectrin or beta-globin promoter and the alpha-globin HS40 element, a gamma-globin lentiviral vector utilizing the beta-globin promoter and elements from the beta-globin locus control region demonstrated a higher probability of expression. This lentiviral vector design was evaluated in lethally irradiated mice that received transplants of transduced bone marrow cells. Long-term, stable erythroid expression of human gamma-globin was observed with levels of vector-encoded gamma-globin mRNA ranging from 9% to 19% of total murine alpha-globin mRNA. The therapeutic efficacy of the vector was subsequently evaluated in a murine model of beta-thalassemia intermedia. The majority of mice that underwent transplantation expressed significant levels of chimeric m(alpha)(2)h(gamma)(2) molecules (termed HbF), the amount of which correlated with the degree of phenotypic improvement. A group of animals with a mean HbF level of 21% displayed a 2.5 g/dL (25 g/L) improvement in Hb concentration and normalization of erythrocyte morphology relative to control animals. gamma-Globin expression and phenotypic improvement was variably lower in other animals due to differences in vector copy number and chromosomal position effects. These data establish the potential of using a gamma-globin lentiviral vector for gene therapy of beta-thalassemia.


Asunto(s)
Globinas/genética , Lentivirus/genética , Fenotipo , Transfección , Talasemia beta/terapia , Animales , Células de la Médula Ósea/metabolismo , Trasplante de Médula Ósea , Eritrocitos/metabolismo , Hemoglobina Fetal/análisis , Expresión Génica , Terapia Genética , Vectores Genéticos , Globinas/deficiencia , Humanos , Leucemia Eritroblástica Aguda , Ratones , Ratones Noqueados , ARN Mensajero/análisis , Retroviridae/genética , Bazo/citología , Células Tumorales Cultivadas , Talasemia beta/genética
12.
Blood ; 104(8): 2281-90, 2004 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-15198957

RESUMEN

Since increased fetal hemoglobin diminishes the severity of beta-thalassemia and sickle cell anemia, a strategy using autologous, stem cell-targeted gene transfer of a gamma-globin gene may be therapeutically useful. We previously found that a gamma-globin lentiviral vector utilizing the beta-globin promoter and elements from the beta-globin locus control region (LCR) totaling 1.7 kb could correct murine beta-thalassemia. However, therapeutic consistency was compromised by chromosomal position effects on vector expression. In contrast, we show here that the majority of animals that received transplants of beta-thalassemic stem cells transduced with a new vector containing 3.2 kb of LCR sequences expressed high levels of fetal hemoglobin (17%-33%), with an average vector copy number of 1.3. This led to a mean 26 g/L (2.6 g/dL) increase in hemoglobin concentration and enhanced amelioration of other hematologic parameters. Analysis of clonal erythroid cells of secondary spleen colonies from mice that underwent transplantation demonstrated an increased resistance of the larger LCR vector to stable and variegating position effects. This trend was also observed for vector insertion sites located inside genes, where vector expression was often compromised, in contrast to intergenic sites, where higher levels of expression were observed. These data emphasize the importance of overcoming detrimental position effects for consistent therapeutic globin vector expression.


Asunto(s)
Globinas/genética , Globinas/uso terapéutico , Lentivirus/genética , Región de Control de Posición/genética , Talasemia beta/genética , Talasemia beta/terapia , Anemia de Células Falciformes/genética , Anemia de Células Falciformes/terapia , Animales , Células Cultivadas , ADN Intergénico/genética , Células Eritroides/metabolismo , Citometría de Flujo , Expresión Génica , Terapia Genética , Vectores Genéticos/genética , Genoma , Genoma Viral , Globinas/metabolismo , VIH-1/genética , Ratones , ARN Viral/genética
13.
Blood ; 102(2): 506-13, 2003 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-12663444

RESUMEN

Successful gene therapy of beta-thalassemia will require replacement of the abnormal erythroid compartment with erythropoiesis derived from genetically corrected, autologous hematopoietic stem cells (HSCs). However, currently attainable gene transfer efficiencies into human HSCs are unlikely to yield sufficient numbers of corrected cells for a clinical benefit. Here, using a murine model of beta-thalassemia, we demonstrate for the first time that selective enrichment in vivo of transplanted, drug-resistant HSCs can be used therapeutically and may therefore be a useful approach to overcome limiting gene transfer. We used an oncoretroviral vector to transfer a methylguanine methyltransferase (MGMT) drug-resistance gene into normal bone marrow cells. These cells were transplanted into beta-thalassemic mice given nonmyeloablative pretransplantation conditioning with temozolomide (TMZ) and O6-benzylguanine (BG). A majority of mice receiving 2 additional courses of TMZ/BG demonstrated in vivo selection of the drug-resistant cells and amelioration of anemia, compared with untreated control animals. These results were extended using a novel gamma-globin/MGMT dual gene lentiviral vector. Following drug treatment, normal mice that received transduced cells had an average 67-fold increase in gamma-globin expressing red cells. These studies demonstrate that MGMT-based in vivo selection may be useful to increase genetically corrected cells to therapeutic levels in patients with beta-thalassemia.


Asunto(s)
Dacarbazina/análogos & derivados , Resistencia a Medicamentos/genética , Globinas/genética , Guanina/análogos & derivados , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/efectos de los fármacos , O(6)-Metilguanina-ADN Metiltransferasa/genética , Talasemia beta/terapia , Animales , Cruzamientos Genéticos , Femenino , Vectores Genéticos/genética , Guanina/farmacología , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Lentivirus/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , O(6)-Metilguanina-ADN Metiltransferasa/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Selección Genética , Temozolomida , Quimera por Trasplante , Acondicionamiento Pretrasplante , Talasemia beta/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA