Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Nat Commun ; 13(1): 3710, 2022 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-35764638

RESUMEN

X-linked Severe Combined Immunodeficiency (SCID-X1) due to IL2RG mutations is potentially fatal in infancy where 'emergency' life-saving stem cell transplant may only achieve incomplete immune reconstitution following transplant. Salvage therapy SCID-X1 patients over 2 years old (NCT01306019) is a non-randomized, open-label, phase I/II clinical trial for administration of lentiviral-transduced autologous hematopoietic stem cells following busulfan (6 mg/kg total) conditioning. The primary and secondary objectives assess efficacy in restoring immunity and safety by vector insertion site analysis (VISA). In this ongoing study (19 patients treated), we report VISA in blood lineages from first eight treated patients with longer follow up found a > 60-fold increase in frequency of forward-orientated VIS within intron 3 of the High Mobility Group AT-hook 2 gene. All eight patients demonstrated emergence of dominant HMGA2 VIS clones in progenitor and myeloid lineages, but without disturbance of hematopoiesis. Our molecular analysis demonstrated a cryptic splice site within the chicken ß-globin hypersensitivity 4 insulator element in the vector generating truncated mRNA transcripts from many transcriptionally active gene containing forward-oriented intronic vector insert. A two base-pair change at the splice site within the lentiviral vector eliminated splicing activity while retaining vector functional capability. This highlights the importance of functional analysis of lentivectors for cryptic splicing for preclinical safety assessment and a redesign of clinical vectors to improve safety.


Asunto(s)
Enfermedades por Inmunodeficiencia Combinada Ligada al Cromosoma X , Antígenos CD34/genética , Células Clonales , Terapia Genética , Vectores Genéticos/genética , Humanos , Lentivirus/genética , Enfermedades por Inmunodeficiencia Combinada Ligada al Cromosoma X/genética , Enfermedades por Inmunodeficiencia Combinada Ligada al Cromosoma X/terapia
2.
Blood ; 116(8): 1263-71, 2010 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-20489056

RESUMEN

Destructive midline granulomatous disease characterized by necrotizing granulomas of the head and neck is most commonly caused by Wegener granulomatosis, natural killer/T-cell lymphomas, cocaine abuse, or infections. An adolescent patient with myasthenia gravis treated with thymectomy subsequently developed extensive granulomatous destruction of midface structures, palate, nasal septum, airways, and epiglottis. His lymphocyte numbers, total immunoglobulin G level, and T-cell receptor (TCR) repertoire appeared normal. Sequencing of Recombination activating gene-1 (Rag1) showed compound heterozygous Rag1 mutations; a novel deletion with no recombinase activity and a missense mutation resulting in 50% Rag activity. His thymus was dysplastic and, although not depleted of T cells, showed a notable absence of autoimmune regulator (AIRE) and Foxp3(+) regulatory T cells. This distinct Rag-deficient phenotype characterized by immune dysregulation with granulomatous hyperinflammation and autoimmunity, with relatively normal T and B lymphocyte numbers and a diverse TCR repertoire expands the spectrum of presentation in Rag deficiency. This study was registered at www.clinicaltrials.gov as #NCT00128973.


Asunto(s)
Enfermedad Granulomatosa Crónica/etiología , Enfermedad Granulomatosa Crónica/patología , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/fisiología , Mutación Missense/genética , Inmunodeficiencia Combinada Grave/etiología , Inmunodeficiencia Combinada Grave/patología , Adolescente , Animales , Células Cultivadas , Factores de Transcripción Forkhead , Reordenamiento Génico , Genes de Inmunoglobulinas , Enfermedad Granulomatosa Crónica/cirugía , Humanos , Inmunoglobulina G/inmunología , Inmunoglobulina G/metabolismo , Inmunofenotipificación , Linfocitos/inmunología , Linfocitos/metabolismo , Masculino , Ratones , Recombinasas/metabolismo , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Linfocitos T Reguladores/patología , Timectomía , Factores de Transcripción , Transgenes/fisiología , Proteína AIRE
3.
Stem Cells Dev ; 16(3): 361-70, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17610366

RESUMEN

Hematopoietic stem cell (HSC) graft cell dose impacts significantly on allogeneic transplant. Similarly, HSC gene therapy outcome is affected by loss of repopulating cells during culture required for ex vivo retrovirus transduction. Stromal cell-derived factor-1 (SDF-1) and its receptor CXCR4 play a central role in marrow trafficking of HSCs, and maneuvers that enhance CXCR4 activation might positively impact outcome in settings of limiting graft dose. CD26/dipeptidyl peptidase IV (DPP-IV) is an ectoenzyme protease that cleaves SDF-1, thus reducing CXCR4 activation. We show that injection of irradiated nonobese diabetic/severe combined immunodeficiency (NOD/SCID) mice with >or=2 micromol Diprotin A (a tripeptide specific inhibitor of CD26 protease activity) at the time of transplant of human granulocyte colony-stimulating factor (G-CSF) mobilized CD34(+) peripheral blood cells (CD34(+) PBCs) results in a >3.4-fold enhancement of engraftment of human cells. We also show that CD26 on residual stromal cells in the irradiated recipient marrow milieu, and not any CD26 activity in the human CD34(+) PBC graft itself, plays the critical role in regulating receptivity of this environment for the incoming graft. Human marrow stromal cells also express CD26, raising the possibility that Diprotin A treatment could significantly enhance engraftment of HSCs in humans in settings of limiting graft dose just as we observed in the NOD/SCID mouse human xenograft model.


Asunto(s)
Antígenos CD34/metabolismo , Células Sanguíneas/metabolismo , Trasplante de Médula Ósea , Oligopéptidos/metabolismo , Animales , Células Sanguíneas/citología , Células Sanguíneas/efectos de la radiación , Movimiento Celular , Factor Estimulante de Colonias de Granulocitos/metabolismo , Humanos , Ratones , Ratones Endogámicos NOD , Ratones SCID , Oligopéptidos/administración & dosificación , Trasplante Heterólogo
4.
Stem Cells ; 25(7): 1807-13, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17464090

RESUMEN

We used the nonobese diabetic/severe combined immunodeficient (NOD/SCID) mouse model to assess the repopulation potential of subpopulations of mobilized human CD34+ peripheral blood progenitor cells (PBPC). First, PBPC were transduced with gamma-retrovirus vector RD114-MFGS-CFP, which requires cell division for successful transduction, at 24 hours, 48 hours, and 72 hours to achieve 96% cyan fluorescent protein (CFP)-positive cells. Cells were sorted 12 hours after the last transduction into CFP-positive (divided cells) and CFP-negative populations. CFP-positive cells were transplanted postsort, whereas the CFP-negative cells were retransduced and injected at 120 hours. The CFP-negative sorted and retransduced cells contained markedly fewer vector copies and resulted in a 32-fold higher overall engraftment and in a 13-fold higher number of engrafted transgene positive cells. To assess cell proliferation as an underlying cause for the different engraftment levels, carboxyfluorescein succinimidyl ester-labeling of untransduced PBPC was performed to track the number of cell divisions. At 72 hours after initiation of culture, when 95% of all cells have divided, PBPC were sorted into nondivided and divided fractions and transplanted into NOD/SCID mice. Nondivided cells demonstrated 45-fold higher engraftment than divided cells. Late dividing PBPC in ex vivo culture retain high expression of the stem cell marker CD133, whereas rapidly proliferating cells lose CD133 in correlation to the number of cell divisions. Our studies demonstrate that late dividing progenitors transduced with gamma-retroviral vectors contribute most to NOD/SCID engraftment and transgene marking. Confining the gamma-retroviral transduction to CD133-positive cells on days 3 and 4 could greatly reduce the number of transplanted vector copies, limiting the risk of leukemia from insertional mutagenesis. Disclosure of potential conflicts of interest is found at the end of this article.


Asunto(s)
Diabetes Mellitus/metabolismo , Gammaretrovirus/genética , Vectores Genéticos , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/citología , Transducción Genética , Antígeno AC133 , Adulto , Animales , Antígenos CD/metabolismo , División Celular , Movimiento Celular , Células Cultivadas , Citometría de Flujo , Gammaretrovirus/fisiología , Glicoproteínas/metabolismo , Proteínas Fluorescentes Verdes/metabolismo , Células Madre Hematopoyéticas/metabolismo , Células Madre Hematopoyéticas/virología , Humanos , Ratones , Ratones SCID , Péptidos/metabolismo , Fenotipo , Factores de Tiempo
5.
Blood ; 110(1): 67-73, 2007 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-17369490

RESUMEN

Retroviral gene therapy can restore immunity to infants with X-linked severe combined immunodeficiency (XSCID) caused by mutations in the IL2RG gene encoding the common gamma chain (gammac) of receptors for interleukins 2 (IL-2), -4, -7, -9, -15, and -21. We investigated the safety and efficacy of gene therapy as salvage treatment for older XSCID children with inadequate immune reconstitution despite prior bone marrow transplant from a parent. Subjects received retrovirus-transduced autologous peripherally mobilized CD34(+) hematopoietic cells. T-cell function significantly improved in the youngest subject (age 10 years), and multilineage retroviral marking occurred in all 3 children.


Asunto(s)
Terapia Genética/métodos , Inmunidad/efectos de los fármacos , Receptores de Interleucina-2/administración & dosificación , Enfermedades por Inmunodeficiencia Combinada Ligada al Cromosoma X/inmunología , Enfermedades por Inmunodeficiencia Combinada Ligada al Cromosoma X/terapia , Trasplante de Células Madre Hematopoyéticas/métodos , Humanos , Lactante , Mutación , Receptores de Interleucina-2/genética , Retroviridae/genética , Linfocitos T/inmunología , Transducción Genética , Trasplante Autólogo , Enfermedades por Inmunodeficiencia Combinada Ligada al Cromosoma X/genética
6.
Blood ; 109(1): 78-84, 2007 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-16946301

RESUMEN

WHIM(warts, hypogammaglobulinemia, recurrent bacterial infection, and myelokathexis) syndrome is a rare immunodeficiency caused in many cases by autosomal dominant C-terminal truncation mutations in the chemokine receptor CXCR4. A prominent and unexplained feature of WHIM is myelokathexis (hypercellularity with apoptosis of mature myeloid cells in bone marrow and neutropenia). We transduced healthy human CD34(+) peripheral blood-mobilized stem cells (PBSCs) with retrovirus vector encoding wild-type (wt) CXCR4 or WHIM-type mutated CXCR4 and studied these cells ex vivo in culture and after engraftment in a nonobese diabetic/severe combined immunodeficiency (NOD/SCID) mouse xenograft model. Neither wt CXCR4 nor mutated CXCR4 transgene expression itself enhanced apoptosis of neutrophils arising in transduced PBSC cultures even with stimulation by a CXCR4 agonist, stromal cell-derived factor-1 (SDF-1 [CXCL12]). Excess wt CXCR4 expression by transduced human PBSCs enhanced marrow engraftment, but did not affect bone marrow (BM) apoptosis or the release of transduced leukocytes into PB. However, mutated CXCR4 transgene expression further enhanced BM engraftment, but was associated with a significant increase in apoptosis of transduced cells in BM and reduced release of transduced leukocytes into PB. We conclude that increased apoptosis of mature myeloid cells in WHIM is secondary to a failure of marrow release and progression to normal myeloid cell senescence, and not a direct effect of activation of mutated CXCR4.


Asunto(s)
Agammaglobulinemia/genética , Infecciones Bacterianas/etiología , Células de la Médula Ósea/patología , Síndromes de Inmunodeficiencia/genética , Neutropenia/genética , Receptores CXCR4/fisiología , Verrugas/genética , Sustitución de Aminoácidos , Animales , Apoptosis , Células de la Médula Ósea/metabolismo , Señalización del Calcio , Movimiento Celular , Quimiocina CXCL12 , Quimiocinas CXC/fisiología , Ensayo de Unidades Formadoras de Colonias , Susceptibilidad a Enfermedades , Dosificación de Gen , Supervivencia de Injerto , Humanos , Ratones , Ratones Endogámicos NOD , Ratones SCID , Mutación Missense , Trasplante de Células Madre de Sangre Periférica , Mutación Puntual , Quimera por Radiación , Receptores CXCR4/genética , Proteínas Recombinantes de Fusión/fisiología , Recurrencia , Transgenes , Trasplante Heterólogo
7.
Mol Ther ; 14(2): 202-11, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16600688

RESUMEN

We have recently reported that the RD114-pseudotyped MFGS-gp91phox vector achieves unprecedented levels of correction of the NADPH-oxidase gp91phox (approved gene symbol CYBB) defect in CD34(+) cells from patients with X-linked chronic granulomatous disease in the NOD/SCID mouse model. Considering clinical use of this vector, we transplanted autologous mobilized peripheral blood CD34(+) progenitor cells, transduced with the RD114-MFGS-gp91phox vector, into two healthy rhesus macaques following nonmyeloablative conditioning. The moderately high levels of in vivo marking seen in the first months following transduction decreased and stabilized at about 8 months posttransplant. Marking for both healthy animals after 15 months was 0.3 to 1.3 vector copies per 100 cells in lymphocytes, neutrophils, and monocytes. Vector insertion analyses performed by linear amplification-mediated PCR and sequencing identified 32 and 45 separate insertion sites in the animals. Identical insertion sites were found in myeloid cells and lymphocytes, demonstrating the successful transduction of lymphomyeloid progenitors. Some inserts landed in the vicinity of genes controlling cell cycle and proliferation. Statistical analyses of insertion sites 1 year posttransplant suggest a high diversity of insertion sites despite low marking.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas , Glicoproteínas de Membrana/genética , NADPH Oxidasas/genética , Transducción Genética , Animales , Antígenos CD34/análisis , Gammaretrovirus/genética , Dosificación de Gen , Vectores Genéticos , Macaca mulatta , Masculino , NADPH Oxidasa 2 , Transgenes , Acondicionamiento Pretrasplante , Trasplante Autólogo , Integración Viral , Irradiación Corporal Total
8.
Blood ; 107(8): 3091-7, 2006 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-16384923

RESUMEN

X-linked severe combined immunodeficiency (XSCID) is characterized by profound immunodeficiency and early mortality, the only potential cure being hematopoietic stem cell (HSC) transplantation or gene therapy. Current clinical gene therapy protocols targeting HSCs are based upon ex vivo gene transfer, potentially limited by the adequacy of HSC harvest, transduction efficiencies of repopulating HSCs, and the potential loss of their engraftment potential during ex vivo culture. We demonstrate an important proof of principle by showing achievement of durable immune reconstitution in XSCID dogs following intravenous injection of concentrated RD114-pseudotyped retrovirus vector encoding the corrective gene, the interleukin-2 receptor gamma chain (gamma c). In 3 of 4 dogs treated, normalization of numbers and function of T cells were observed. Two long-term-surviving animals (16 and 18 months) showed significant marking of B lymphocytes and myeloid cells, normalization of IgG levels, and protective humoral immune response to immunization. There were no adverse effects from in vivo gene therapy, and in one dog that reached sexual maturity, sparing of gonadal tissue from gene transfer was demonstrated. This is the first demonstration that in vivo gene therapy targeting HSCs can restore both cellular and humoral immunity in a large-animal model of a fatal immunodeficiency.


Asunto(s)
Terapia Genética , Vectores Genéticos/administración & dosificación , Receptores de Interleucina-2/genética , Recuperación de la Función/genética , Inmunodeficiencia Combinada Grave/terapia , Transducción Genética , Animales , Formación de Anticuerpos/genética , Formación de Anticuerpos/inmunología , Linfocitos B/inmunología , Perros , Terapia Genética/métodos , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/inmunología , Inmunización , Receptores de Interleucina-2/inmunología , Recuperación de la Función/inmunología , Retroviridae , Inmunodeficiencia Combinada Grave/genética , Inmunodeficiencia Combinada Grave/inmunología , Linfocitos T/inmunología , Transducción Genética/métodos , Trasplante Autólogo
9.
Exp Hematol ; 33(4): 460-8, 2005 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15781337

RESUMEN

OBJECTIVE: WHIM (warts, hypogammaglobulinemia, recurrent bacterial infection, myelokathexis) syndrome is an autosomal dominant immune deficiency with severe chronic neutropenia and marrow neutrophil apoptosis. Carboxy-termini truncating mutations in the chemokine receptor CXCR4 have been identified in WHIM patients. We created a retrovirus encoding mutated CXCR4 (truncating point mutation 1000C-->T [R334X] inherited heterozygously in several WHIM patients) in order to transducer healthy human CD34 stem cells and K562 to overexpress mutated CXCR4 and determined its effect on receptor responses to stromal-derived factor-1 (SDF1). METHODS: Retrovirus vector was engineered to coexpress WHIM-associated R334X mutated CXCR4 together with green fluorescent protein (GFP). Control vectors included similar constructs with wild-type CXCR4 (WT-CXCR4) or only GFP. CD34+ cells and K562 were transduced with these vectors. Populations of 100% transduced K562 were established by sorting GFP+ cells by flow cytometry. We performed migration and calcium flux assays of transduced CD34+ cells and transduced/sorted K562. We also examined receptor recycling in response to SDF1. RESULTS: Healthy human CD34+ cells and/or human erythroleukemia K562 cells transduced to express mutated CXCR4, WT-CXCR4, or GFP alone demonstrated that mutated CXCR4 was associated with enhanced calcium flux and enhanced migration. There was also decreased receptor internalization and enhanced recovery of surface mutated CXCR4 in response to SDF1 compared with WT-CXCR4. CONCLUSION: We propose that decreased internalization of WHIM-associated mutated CXCR4 leads to prolongation/enhancement of signaling in response to SDF1 and that this may provide the biochemical basis for the autosomal dominant abnormalities of cell trafficking and function associated with WHIM syndrome.


Asunto(s)
Endocitosis , Enfermedades Genéticas Congénitas/etiología , Receptores CXCR4/genética , Receptores CXCR4/metabolismo , Agammaglobulinemia , Infecciones Bacterianas , Línea Celular , Quimiocina CXCL12 , Quimiocinas CXC/farmacología , Relación Dosis-Respuesta a Droga , Humanos , Cinética , Neutropenia , Transducción de Señal/efectos de los fármacos , Síndrome , Transducción Genética , Verrugas
10.
Stem Cells ; 22(7): 1128-33, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15579633

RESUMEN

Hematopoietic stem cells (HSCs) lose marrow reconstitution potential during ex vivo culture. HSC migration to stromal cell-derived factor (SDF)-1 (CXCL12) correlates with CXC chemokine receptor 4 (CXCR4) expression and marrow engraftment. We demonstrate that mobilized human CD34+ peripheral blood stem cells (CD34+ PBSCs) lose CXCR4 expression during prolonged culture. We transduced CD34+ PBSCs with retrovirus vector encoding human CXCR4 and achieved 18-fold more CXCR4 expression in over 87% of CD34+ cells. CXCR4-transduced cells yielded increased calcium flux and up to a 10-fold increase in migration to SDF-1. Six-day cultured CXCR4-transduced cells demonstrated significant engraftment in nonobese diabetic/severe combined immunodeficient mice under conditions in which control transduced cells resulted in low or no engraftment. We conclude that transduction-mediated overexpression of CXCR4 significantly improves marrow engraftment of cultured PBSCs.


Asunto(s)
Células de la Médula Ósea/citología , Técnicas de Transferencia de Gen , Trasplante de Células Madre Hematopoyéticas/métodos , Células Madre Hematopoyéticas/citología , Receptores CXCR4/biosíntesis , Receptores CXCR4/genética , Transgenes , Animales , Antígenos CD34/biosíntesis , Calcio/metabolismo , Movimiento Celular , Citometría de Flujo , Supervivencia de Injerto , Humanos , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones SCID , NADPH Oxidasa 2 , NADPH Oxidasas/metabolismo , Transducción de Señal , Factores de Tiempo
11.
Exp Hematol ; 32(8): 709-19, 2004 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15308322

RESUMEN

OBJECTIVE: We characterized a novel in vivo selectable fusion protein, green fluorescence protein-O6-benzylguanine (BG)-resistant O6-methylguanine-methyltransferase (GFP-MGMT* [*refers to mutant MGMT]) used to delineate optimum selection regimens for transduced hematopoietic stem cells (HSC) ex vivo and in vivo. MATERIALS AND METHODS: We transduced human or mouse HSC with retrovirus vector encoding GFP-MGMT* where BG-resistant forms of human P140K-hMGMT* and mouse P144K-mMGMT* were studied. We evaluated selection of transduced HSC ex vivo and in vivo using either BG/1,3-bis(2-chloroethyl)-1-nitrosourea (BCNU) or BG/temozolomide (TMZ) combinations, evaluating transduction marking by flow cytometry and real-time TaqMan PCR. RESULTS: GFP-MGMT* transduction confers nuclear-localized GFP fluorescence and BG resistance. Optimum selection ex vivo of GFP-MGMT*-transduced HSC occurred with BG (2.5-10 microM)/BCNU (5-10 microM) or TMZ (100-200 microM), which increases marking while preserving maximum viable transduced cells. Starting at low levels (0.1%) or high levels (>30%) of in vivo bone marrow gene making in mice, in vivo selection with BG/BCNU (20/6 mg/kg) (weeks 4 and 5) or BG/TMZ (20/60 mg/kg) (daily x 5 at week 4) increased bone marrow marking to 8.58% +/- 3.52% or 82.0% +/- 3.4% GFP+ cells, respectively, in the low- or high-level initial marking mice. CONCLUSIONS: GFP-MGMT* is an informative tool to explore optimization of in vivo selection regimens using BG/BCNU or BG/TMZ to increase gene marking of HSC. Both timing and dosing of selection regimens and the starting level of marking may all be important to the level of selective increase of in vivo marking achieved.


Asunto(s)
Núcleo Celular/metabolismo , Guanina/análogos & derivados , Guanina/farmacología , Células Madre Hematopoyéticas/metabolismo , Proteínas Luminiscentes/metabolismo , O(6)-Metilguanina-ADN Metiltransferasa/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Animales , Células Cultivadas , Vectores Genéticos , Proteínas Fluorescentes Verdes , Ratones , Ratones Endogámicos C57BL , Transducción Genética
12.
Blood ; 102(8): 2789-97, 2003 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-12829597

RESUMEN

In previous studies amphotropic MFGS-gp91phox (murine onco-retrovirus vector) was used in a clinical trial of X-linked chronic granulomatous disease (X-CGD) gene therapy to achieve transient correction of oxidase activity in 0.1% of neutrophils. We later showed that transduced CD34+ peripheral blood stem cells (CD34+ PBSCs) from this trial transplanted into nonobese diabetic/severe combined immunodeficient (NOD/SCID) mice resulted in correction of only 2.5% of human neutrophils. However, higher rates of transduction into stem cells are required. In the current study we demonstrate that the same vector (MFGS-gp91phox) pseudo-typed with RD114 envelope in a 4-day culture/transduction regimen results in a 7-fold increase in correction of NOD/SCID mouse repopulating X-CGD CD34+ PBSCs (14%-22% corrected human neutrophils; human cell engraftment 13%-67%). This increase may result from high expression of receptor for RD114 that we demonstrate on CD34+CD38- stem cells. Using RD114-MFGS encoding cyan fluorescent protein to allow similar studies of normal CD34+ PBSCs, we show that progressively higher levels of gene marking of human neutrophils (67%-77%) can be achieved by prolongation of culture/transduction to 6 days, but with lower rates of human cell engraftment. Our data demonstrate the highest reported level of functional correction of any inherited metabolic disorder in human cells in vivo with the NOD/SCID mouse system using onco-retrovirus vector.


Asunto(s)
Enfermedad Granulomatosa Crónica/genética , Glicoproteínas de Membrana/genética , NADPH Oxidasas , Oxidorreductasas/genética , Animales , Antígenos CD34/biosíntesis , Citometría de Flujo , Técnicas de Transferencia de Gen , Terapia Genética/métodos , Vectores Genéticos , Enfermedad Granulomatosa Crónica/terapia , Proteínas Fluorescentes Verdes , Células Madre Hematopoyéticas , Humanos , Proteínas Luminiscentes/metabolismo , Ratones , Ratones Endogámicos NOD , Ratones SCID , NADP/metabolismo , NADPH Oxidasa 2 , Neutrófilos/metabolismo , ARN Mensajero/metabolismo , Retroviridae/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Madre/metabolismo , Factores de Tiempo , Transgenes , Ultracentrifugación , Microglobulina beta-2/genética
13.
Blood ; 100(13): 4381-90, 2002 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-12393624

RESUMEN

HIV-1-derived lentivectors are promising for gene transfer into hematopoietic stem cells but require preclinical in vivo evaluation relevant to specific human diseases. Nonobese diabetic/severe combined immunodeficient (NOD/SCID) mice accept human hematopoietic stem cell grafts, providing a unique opportunity for in vivo evaluation of therapies targeting human hematopoietic diseases. We demonstrate for the first time that hematopoietic stem cells from patients with X-linked chronic granulomatous disease (X-CGD) give rise to X-CGD-phenotype neutrophils in the NOD/SCID model that can be corrected using VSV-G-pseudotyped, 3rd-generation, self-inactivating (SIN) lentivector encoding gp91(phox). We transduced X-CGD patient-mobilized CD34(+) peripheral blood stem cells (CD34(+)PBSCs) with lentivector-gp91(phox) or amphotropic oncoretrovirus MFGS-gp91(phox) and evaluated correction ex vivo and in vivo in NOD/SCID mice. Only lentivector transduced CD34(+)PBSCs under ex vivo conditions nonpermissive for cell division, but both vectors performed best under conditions permissive for proliferation (multiple growth factors). Under the latter conditions, lentivector and MFGS achieved significant ex vivo correction of X-CGD CD34(+)PBSCs (18% and 54% of cells expressing gp91(phox), associated with 53% and 163% of normal superoxide production, respectively). However, lentivector, but not MFGS, achieved significant correction of human X-CGD neutrophils arising in vivo in NOD/SCID mice that underwent transplantation (20% and 2.4%, respectively). Thus, 3rd-generation SIN lentivector-gp91(phox) performs well as assessed in human X-CGD neutrophils differentiating in vivo, and our studies suggest that the NOD/SCID model is generally applicable for in vivo study of therapies evaluated in human blood cells expressing a specific disease phenotype.


Asunto(s)
Virus Defectuosos/genética , Terapia Genética , Vectores Genéticos/uso terapéutico , Enfermedad Granulomatosa Crónica/terapia , VIH-1/genética , Glicoproteínas de Membrana/genética , NADPH Oxidasas/deficiencia , Trasplante de Células Madre de Sangre Periférica , Animales , Antígenos CD34/análisis , División Celular , Prueba de Complementación Genética , Factor Estimulante de Colonias de Granulocitos/farmacología , Enfermedad Granulomatosa Crónica/enzimología , Enfermedad Granulomatosa Crónica/genética , Movilización de Célula Madre Hematopoyética , Células Madre Hematopoyéticas/enzimología , Humanos , Células K562 , Glicoproteínas de Membrana/deficiencia , Ratones , Ratones Endogámicos NOD , Ratones SCID , NADPH Oxidasa 2 , NADPH Oxidasas/genética , Fosfoproteínas/genética , Retroviridae/genética , Trasplante Heterólogo , Virus de la Estomatitis Vesicular Indiana/fisiología , Cromosoma X/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA