Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 122
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
2.
Nature ; 467(7313): 318-22, 2010 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-20844535

RESUMEN

The ß-haemoglobinopathies are the most prevalent inherited disorders worldwide. Gene therapy of ß-thalassaemia is particularly challenging given the requirement for massive haemoglobin production in a lineage-specific manner and the lack of selective advantage for corrected haematopoietic stem cells. Compound ß(E)/ß(0)-thalassaemia is the most common form of severe thalassaemia in southeast Asian countries and their diasporas. The ß(E)-globin allele bears a point mutation that causes alternative splicing. The abnormally spliced form is non-coding, whereas the correctly spliced messenger RNA expresses a mutated ß(E)-globin with partial instability. When this is compounded with a non-functional ß(0) allele, a profound decrease in ß-globin synthesis results, and approximately half of ß(E)/ß(0)-thalassaemia patients are transfusion-dependent. The only available curative therapy is allogeneic haematopoietic stem cell transplantation, although most patients do not have a human-leukocyte-antigen-matched, geno-identical donor, and those who do still risk rejection or graft-versus-host disease. Here we show that, 33 months after lentiviral ß-globin gene transfer, an adult patient with severe ß(E)/ß(0)-thalassaemia dependent on monthly transfusions since early childhood has become transfusion independent for the past 21 months. Blood haemoglobin is maintained between 9 and 10 g dl(-1), of which one-third contains vector-encoded ß-globin. Most of the therapeutic benefit results from a dominant, myeloid-biased cell clone, in which the integrated vector causes transcriptional activation of HMGA2 in erythroid cells with further increased expression of a truncated HMGA2 mRNA insensitive to degradation by let-7 microRNAs. The clonal dominance that accompanies therapeutic efficacy may be coincidental and stochastic or result from a hitherto benign cell expansion caused by dysregulation of the HMGA2 gene in stem/progenitor cells.


Asunto(s)
Transfusión Sanguínea , Terapia Genética , Proteína HMGA2/metabolismo , Globinas beta/genética , Globinas beta/metabolismo , Talasemia beta/genética , Talasemia beta/terapia , Adolescente , Células Sanguíneas/citología , Células Sanguíneas/metabolismo , Células de la Médula Ósea/citología , Células de la Médula Ósea/metabolismo , Preescolar , Células Clonales/metabolismo , Expresión Génica , Vectores Genéticos/genética , Proteína HMGA2/genética , Homeostasis , Humanos , Lentivirus/genética , Masculino , MicroARNs/genética , Especificidad de Órganos , ARN Mensajero/análisis , ARN Mensajero/genética , Factores de Tiempo , Activación Transcripcional , Adulto Joven , Talasemia beta/metabolismo
3.
Blood ; 121(9): 1510-6, 2013 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-23264593

RESUMEN

On the basis of a nationwide database of 160 patients with Wiskott-Aldrich syndrome (WAS), we identified a subset of infants who were significantly more likely to be attributed with an Ochs score of 5 before the age of 2 (n = 26 of 47 [55%], P = 2.8 × 10(−7)). A retrospective analysis revealed that these patients often had severe refractory thrombocytopenia (n = 13), autoimmune hemolytic anemia (n = 15), and vasculitis (n = 6). One patient had developed 2 distinct cancers. Hemizygous mutations predictive of the absence of WAS protein were identified in 19 of the 24 tested patients, and the absence of WAS protein was confirmed in all 10 investigated cases. Allogeneic hematopoietic stem cell transplantation (HSCT) was found to be a curative treatment with a relatively good prognosis because it was successful in 17 of 22 patients. Nevertheless, 3 patients experienced significant disease sequelae and 4 patients died before HSCT. Therefore, the present study identifies a distinct subgroup of WAS patients with early-onset, life-threatening manifestations. We suggest that HSCT is a curative strategy in this subgroup of patients and should be performed as early in life as possible, even when a fully matched donor is lacking.


Asunto(s)
Síndrome de Wiskott-Aldrich/diagnóstico , Adolescente , Adulto , Edad de Inicio , Niño , Preescolar , Estudios de Cohortes , Humanos , Lactante , Recién Nacido , Pronóstico , Proyectos de Investigación , Índice de Severidad de la Enfermedad , Síndrome de Wiskott-Aldrich/epidemiología , Síndrome de Wiskott-Aldrich/patología , Adulto Joven
4.
Blood ; 121(19): 3981-7, 2013 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-23493783

RESUMEN

We report transplantation outcomes of 258 children with Hurler syndrome (HS) after a myeloablative conditioning regimen from 1995 to 2007. Median age at transplant was 16.7 months and median follow-up was 57 months. The cumulative incidence of neutrophil recovery at day 60 was 91%, acute graft-versus-host disease (GVHD) (grade II-IV) at day 100 was 25%, and chronic GVHD and 5 years was 16%. Overall survival and event-free survival (EFS) at 5 years were 74% and 63%, respectively. EFS after HLA-matched sibling donor (MSD) and 6/6 matched unrelated cord blood (CB) donor were similar at 81%, 66% after 10/10 HLA-matched unrelated donor (UD), and 68% after 5/6 matched CB donor. EFS was lower after transplantation in 4/6 matched unrelated CB (UCB) (57%; P = .031) and HLA-mismatched UD (41%; P = .007). Full-donor chimerism (P = .039) and normal enzyme levels (P = .007) were higher after CB transplantation (92% and 98%, respectively) compared with the other grafts sources (69% and 59%, respectively). In conclusion, results of allogeneic transplantation for HS are encouraging, with similar EFS rates after MSD, 6/6 matched UCB, 5/6 UCB, and 10/10 matched UD. The use of mismatched UD and 4/6 matched UCB was associated with lower EFS.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas/métodos , Células Madre Hematopoyéticas/citología , Mucopolisacaridosis I/terapia , Agonistas Mieloablativos/uso terapéutico , Acondicionamiento Pretrasplante/métodos , Adolescente , Niño , Preescolar , Femenino , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Prueba de Histocompatibilidad , Humanos , Lactante , Masculino , Mucopolisacaridosis I/epidemiología , Mucopolisacaridosis I/mortalidad , Agonistas Mieloablativos/efectos adversos , Estudios Retrospectivos , Donantes de Tejidos , Obtención de Tejidos y Órganos/métodos , Acondicionamiento Pretrasplante/efectos adversos , Resultado del Tratamiento , Adulto Joven
5.
Nat Rev Immunol ; 2(8): 615-21, 2002 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12154380

RESUMEN

The concept that the outcome of a devastating disease can be modified by inserting a transgene into abnormal cells is appealing. However, the gene-transfer technologies that are available at present have limited the success of gene therapy so far. Nevertheless, severe combined immunodeficiencies are a useful model, because gene transfer can confer a selective advantage to transduced cells. In this way, a proof of concept for gene therapy has been provided.


Asunto(s)
Terapia Genética , Inmunodeficiencia Combinada Grave/genética , Inmunodeficiencia Combinada Grave/terapia , Adenosina Desaminasa/genética , Animales , Grupo Citocromo b/genética , Humanos , Inmunodeficiencia Combinada Grave/inmunología , Linfocitos T
6.
Blood ; 120(10): 2144-54, 2012 Sep 06.
Artículo en Inglés | MEDLINE | ID: mdl-22730537

RESUMEN

Invariant natural killer T (iNKT) cells can experimentally dissociate GVL from graft-versus-host-disease (GVHD). Their role in human conventional allogeneic hematopoietic stem cell transplantation (HSCT) is unknown. Here, we analyzed the post-HSCT recovery of iNKT cells in 71 adult allografted patients. Results were compared with conventional T- and NK-cell recovery and correlated to the occurrence of GVHD, relapse, and survival. We observed that posttransplantation iNKT cells, likely of donor origin, recovered independently of T and NK cells in the first 90 days after HSCT and reached greater levels in recipient younger than 45 years (P = .003) and after a reduced-intensity conditioning regimen (P = .03). Low posttransplantation iNKT/T ratios (ie, < 10(-3)) were an independent factor associated with the occurrence of acute GVHD (aGVHD; P = .001). Inversely, reaching iNKT/T ratios > 10(-3) before day 90 was associated with reduced nonrelapse mortality (P = .009) without increased risk of relapse and appeared as an independent predictive factor of an improved overall survival (P = .028). Furthermore, an iNKT/T ratio on day 15 > 0.58 × 10(-3) was associated with a 94% risk reduction of aGVHD. These findings provide a proof of concept that early postallogeneic HSCT iNKT cell recovery can predict the occurrence of aGVHD and an improved overall survival.


Asunto(s)
Enfermedad Injerto contra Huésped/inmunología , Trasplante de Células Madre Hematopoyéticas , Células T Asesinas Naturales/inmunología , Acondicionamiento Pretrasplante/métodos , Enfermedad Aguda , Adolescente , Adulto , Factores de Edad , Femenino , Enfermedad Injerto contra Huésped/patología , Enfermedad Injerto contra Huésped/terapia , Efecto Injerto vs Leucemia/inmunología , Neoplasias Hematológicas/inmunología , Neoplasias Hematológicas/patología , Neoplasias Hematológicas/terapia , Humanos , Recuento de Linfocitos , Masculino , Persona de Mediana Edad , Células T Asesinas Naturales/patología , Recurrencia , Factores de Riesgo , Análisis de Supervivencia , Linfocitos T/inmunología , Linfocitos T/patología , Factores de Tiempo , Donantes de Tejidos , Trasplante Homólogo
7.
Blood ; 119(12): 2949-55, 2012 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-22308292

RESUMEN

Pediatric patients with SCID constitute medical emergencies. In the absence of an HLA-identical hematopoietic stem cell (HSC) donor, mismatched related-donor transplantation (MMRDT) or unrelated-donor umbilical cord blood transplantation (UCBT) are valuable treatment options. To help transplantation centers choose the best treatment option, we retrospectively compared outcomes after 175 MMRDTs and 74 UCBTs in patients with SCID or Omenn syndrome. Median follow-up time was 83 months and 58 months for UCBT and MMRDT, respectively. Most UCB recipients received a myeloablative conditioning regimen; most MMRDT recipients did not. UCB recipients presented a higher frequency of complete donor chimerism (P = .04) and faster total lymphocyte count recovery (P = .04) without any statistically significance with the preparative regimen they received. The MMRDT and UCBT groups did not differ in terms of T-cell engraftment, CD4(+) and CD3(+) cell recoveries, while Ig replacement therapy was discontinued sooner after UCBT (adjusted P = .02). There was a trend toward a greater incidence of grades II-IV acute GVHD (P = .06) and more chronic GVHD (P = .03) after UCBT. The estimated 5-year overall survival rates were 62% ± 4% after MMRDT and 57% ± 6% after UCBT. For children with SCID and no HLA-identical sibling donor, both UCBT and MMRDT represent available HSC sources for transplantation with quite similar outcomes.


Asunto(s)
Trasplante de Células Madre de Sangre del Cordón Umbilical , Enfermedad Injerto contra Huésped/epidemiología , Trasplante de Células Madre Hematopoyéticas , Inmunodeficiencia Combinada Grave/inmunología , Inmunodeficiencia Combinada Grave/cirugía , Preescolar , Trasplante de Células Madre de Sangre del Cordón Umbilical/efectos adversos , Trasplante de Células Madre de Sangre del Cordón Umbilical/mortalidad , Femenino , Enfermedad Injerto contra Huésped/mortalidad , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Trasplante de Células Madre Hematopoyéticas/mortalidad , Histocompatibilidad , Humanos , Incidencia , Lactante , Recién Nacido , Estimación de Kaplan-Meier , Masculino , Modelos de Riesgos Proporcionales , Estudios Retrospectivos , Inmunodeficiencia Combinada Grave/mortalidad , Acondicionamiento Pretrasplante/métodos , Resultado del Tratamiento
8.
Blood ; 120(17): 3615-24; quiz 3626, 2012 Oct 25.
Artículo en Inglés | MEDLINE | ID: mdl-22791287

RESUMEN

Deficiency of the purine salvage enzyme adenosine deaminase leads to SCID (ADA-SCID). Hematopoietic cell transplantation (HCT) can lead to a permanent cure of SCID; however, little data are available on outcome of HCT for ADA-SCID in particular. In this multicenter retrospective study, we analyzed outcome of HCT in 106 patients with ADA-SCID who received a total of 119 transplants. HCT from matched sibling and family donors (MSDs, MFDs) had significantly better overall survival (86% and 81%) in comparison with HCT from matched unrelated (66%; P < .05) and haploidentical donors (43%; P < .001). Superior overall survival was also seen in patients who received unconditioned transplants in comparison with myeloablative procedures (81% vs 54%; P < .003), although in unconditioned haploidentical donor HCT, nonengraftment was a major problem. Long-term immune recovery showed that regardless of transplant type, overall T-cell numbers were similar, although a faster rate of T-cell recovery was observed after MSD/MFD HCT. Humoral immunity and donor B-cell engraftment was achieved in nearly all evaluable surviving patients and was seen even after unconditioned HCT. These data detail for the first time the outcomes of HCT for ADA-SCID and show that, if patients survive HCT, long-term cellular and humoral immune recovery is achieved.


Asunto(s)
Agammaglobulinemia/tratamiento farmacológico , Trasplante de Células Madre Hematopoyéticas , Inmunodeficiencia Combinada Grave/tratamiento farmacológico , Acondicionamiento Pretrasplante , Adenosina Desaminasa/deficiencia , Adenosina Desaminasa/inmunología , Agammaglobulinemia/inmunología , Agammaglobulinemia/mortalidad , Agammaglobulinemia/patología , Niño , Preescolar , Femenino , Supervivencia de Injerto , Prueba de Histocompatibilidad , Humanos , Inmunidad Celular , Inmunidad Humoral , Lactante , Recién Nacido , Estimación de Kaplan-Meier , Recuento de Linfocitos , Masculino , Agonistas Mieloablativos/uso terapéutico , Estudios Retrospectivos , Inmunodeficiencia Combinada Grave/inmunología , Inmunodeficiencia Combinada Grave/mortalidad , Inmunodeficiencia Combinada Grave/patología , Hermanos , Linfocitos T/inmunología , Resultado del Tratamiento , Donante no Emparentado
9.
Hum Mutat ; 34(2): 374-84, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23111928

RESUMEN

Ataxia telangiectasia-mutated (ATM) and ataxia telangiectasia and Rad3-related (ATR) kinases are two key regulators of DNA-damage responses (DDR) that are mainly activated in response to DNA double-strand breaks and single-stranded DNA damages, respectively. Seckel syndrome, a rare genetic disorder characterized by a microcephaly and a markedly reduced body size, has been associated with defective ATR-dependent DNA damage signaling. However, the only human genetic ATR defect reported so far is a hypomorphic splicing mutation identified in five related individuals with Seckel syndrome. Here, we report the first case of primary microcephaly with compound heterozygous mutations in ATR: a 540 kb genomic deletion on one allele and a missense mutation leading to splice dysregulation on the other, which ultimately lead to a sharp decrease in ATR expression. DNA combing technology revealed a profound spontaneous alteration of several DNA replication parameters in patient's cells and FISH analyses highlighted the genomic instability caused by ATR deficiency. Collectively, our results emphasize the crucial role for ATR in the control of DNA replication, and reinforce the complementary and nonredundant contributions of ATM and ATR in human cells to face DNA damages and warrant genome integrity.


Asunto(s)
Proteínas de Ciclo Celular/genética , Replicación del ADN , ADN de Cadena Simple , Proteínas de Unión al ADN/genética , Inestabilidad Genómica , Microcefalia/genética , Proteínas Serina-Treonina Quinasas/genética , Proteínas Supresoras de Tumor/genética , Alelos , Animales , Proteínas de la Ataxia Telangiectasia Mutada , Secuencia de Bases , Western Blotting , Línea Celular , Niño , Hibridación Genómica Comparativa , Roturas del ADN de Doble Cadena , Femenino , Fibroblastos/citología , Fibroblastos/metabolismo , Técnica del Anticuerpo Fluorescente , Eliminación de Gen , Estudio de Asociación del Genoma Completo , Heterocigoto , Humanos , Hibridación Fluorescente in Situ , Masculino , Ratones , Datos de Secuencia Molecular , Mutación Missense , Empalme del ARN , Transducción de Señal
10.
Br J Haematol ; 160(2): 146-52, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23167301

RESUMEN

Severe combined immunodeficiencies (SCIDs) correspond to the most severe form of primary immunodeficiency. The extreme severity of the clinical presentation in SCID has legitimately led physicians to consider these conditions as medical emergencies. Hundreds of patients worldwide have undergone allogeneic haematopoietic stem cell transplantation (HCST) in the last 40 years. The complete absence of the T cell compartment in SCID prompted the development (starting in the early 1980s) of haploidentical, parental HSCT for the many patients who do not have a human leucocyte antigen (HLA)-identical sibling. Despite the undeniable progress made in this field over recent years, the long-lasting immunodeficiency that follows partially HLA-incompatible transplantation is still responsible for a mortality rate of 30% at one year post-transplantation. New approaches for reconstituting T cell compartments more rapidly are under intense preclinical development and are discussed herein.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas , Inmunodeficiencia Combinada Grave/cirugía , Trasplante de Células Madre de Sangre del Cordón Umbilical , Citocinas/inmunología , Europa (Continente)/epidemiología , Terapia Genética , Rechazo de Injerto/inmunología , Enfermedad Injerto contra Huésped/epidemiología , Enfermedad Injerto contra Huésped/prevención & control , Trasplante de Células Madre Hematopoyéticas/mortalidad , Histocompatibilidad , Humanos , Lactante , Recién Nacido , Subgrupos Linfocitarios/inmunología , Subgrupos Linfocitarios/patología , Subgrupos Linfocitarios/trasplante , Transfusión de Linfocitos , Linfopoyesis , Tamizaje Neonatal , Inmunodeficiencia Combinada Grave/clasificación , Inmunodeficiencia Combinada Grave/diagnóstico , Inmunodeficiencia Combinada Grave/epidemiología , Inmunodeficiencia Combinada Grave/genética , Inmunodeficiencia Combinada Grave/inmunología , Timo/embriología , Timo/inmunología , Timo/patología , Donantes de Tejidos , Trasplante Homólogo , Resultado del Tratamiento , Estados Unidos/epidemiología
11.
N Engl J Med ; 363(4): 355-64, 2010 Jul 22.
Artículo en Inglés | MEDLINE | ID: mdl-20660403

RESUMEN

BACKGROUND: The outcomes of gene therapy to correct congenital immunodeficiencies are unknown. We reviewed long-term outcomes after gene therapy in nine patients with X-linked severe combined immunodeficiency (SCID-X1), which is characterized by the absence of the cytokine receptor common gamma chain. METHODS: The nine patients, who lacked an HLA-identical donor, underwent ex vivo retrovirus-mediated transfer of gamma chain to autologous CD34+ bone marrow cells between 1999 and 2002. We assessed clinical events and immune function on long-term follow-up. RESULTS: Eight patients were alive after a median follow-up period of 9 years (range, 8 to 11). Gene therapy was initially successful at correcting immune dysfunction in eight of the nine patients. However, acute leukemia developed in four patients, and one died. Transduced T cells were detected for up to 10.7 years after gene therapy. Seven patients, including the three survivors of leukemia, had sustained immune reconstitution; three patients required immunoglobulin-replacement therapy. Sustained thymopoiesis was established by the persistent presence of naive T cells, even after chemotherapy in three patients. The T-cell-receptor repertoire was diverse in all patients. Transduced B cells were not detected. Correction of the immunodeficiency improved the patients' health. CONCLUSIONS: After nearly 10 years of follow-up, gene therapy was shown to have corrected the immunodeficiency associated with SCID-X1. Gene therapy may be an option for patients who do not have an HLA-identical donor for hematopoietic stem-cell transplantation and for whom the risks are deemed acceptable. This treatment is associated with a risk of acute leukemia. (Funded by INSERM and others.)


Asunto(s)
Terapia Genética , Subunidad gamma Común de Receptores de Interleucina/genética , Inmunodeficiencia Combinada Grave/terapia , Antígenos CD34 , Linfocitos B/inmunología , Estudios de Seguimiento , Terapia Genética/efectos adversos , Humanos , Inmunoglobulinas/sangre , Lactante , Subunidad gamma Común de Receptores de Interleucina/deficiencia , Células Asesinas Naturales/fisiología , Recuento de Linfocitos , Leucemia-Linfoma Linfoblástico de Células Precursoras/etiología , Inmunodeficiencia Combinada Grave/inmunología , Linfocitos T/inmunología
12.
Blood ; 117(17): 4420-4, 2011 Apr 28.
Artículo en Inglés | MEDLINE | ID: mdl-21304104

RESUMEN

The understanding of the hierarchical organization of the human hematopoietic system is of major biologic and clinical significance. The validity of the conventional model in which hematopoiesis is solely maintained by a pool of multipotent long-term hematopoietic stem cells (LT-HSCs) has been recently challenged by several mouse studies. These new data point to the existence of a heterogeneous stem cell population that consists of distinct subsets of LT-HSCs, which include stem cells biased toward lineage-specific differentiation programs. This review attempts to discuss the balanced versus biased patterns of lineage output of human LT-HSCs gathered in 3 different gene therapy trials on the basis of vector integration site analysis by deep sequencing. The distribution of integration sites observed tends to support the validity of the revised model.


Asunto(s)
Hematopoyesis/fisiología , Células Madre Hematopoyéticas/fisiología , Células Madre Multipotentes/fisiología , Animales , Humanos , Modelos Biológicos
13.
Blood ; 118(3): 544-53, 2011 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-21622646

RESUMEN

In human B-acute lymphoblastic leukemia (B-ALL), RAG1-induced genomic alterations are important for disease progression. However, given that biallelic loss of the RAG1 locus is observed in a subset of cases, RAG1's role in the development of B-ALL remains unclear. We chose a p19Arf(-/-)Rag1(-/-) mouse model to confirm the previously published results concerning the contribution of CDKN2A (p19ARF /INK4a) and RAG1 copy number alterations in precursor B cells to the initiation and/or progression to B-acute lymphoblastic leukemia (B-ALL). In this murine model, we identified a new, Rag1-independent leukemia-initiating mechanism originating from a Sca1(+)CD19(+) precursor cell population and showed that Notch1 expression accelerates the cells' self-renewal capacity in vitro. In human RAG1-deficient BM, a similar CD34(+)CD19(+) population expressed p19ARF. These findings suggest that combined loss of p19Arf and Rag1 results in B-cell precursor leukemia in mice and may contribute to the progression of precursor B-ALL in humans.


Asunto(s)
Linfocitos B/fisiología , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Células Madre Hematopoyéticas/fisiología , Proteínas de Homeodominio/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras B/genética , Animales , Antígenos CD19/metabolismo , Antígenos CD34/metabolismo , Antígenos Ly/metabolismo , Apoptosis/fisiología , Linfocitos B/metabolismo , Linfocitos B/patología , Células de la Médula Ósea/citología , Células de la Médula Ósea/fisiología , Células Cultivadas , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Regulación Leucémica de la Expresión Génica/fisiología , Células Madre Hematopoyéticas/metabolismo , Células Madre Hematopoyéticas/patología , Proteínas de Homeodominio/metabolismo , Humanos , Proteínas de la Membrana/metabolismo , Ratones , Ratones Mutantes , Trasplante de Neoplasias , Fenotipo , Leucemia-Linfoma Linfoblástico de Células Precursoras B/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras B/patología , Proteínas Proto-Oncogénicas c-kit/genética , Proteínas Proto-Oncogénicas c-kit/metabolismo , Receptor Notch1/genética , Transducción de Señal/fisiología , Células del Estroma/citología , Células del Estroma/fisiología
14.
Blood ; 118(6): 1675-84, 2011 Aug 11.
Artículo en Inglés | MEDLINE | ID: mdl-21659547

RESUMEN

In this retrospective collaborative study, we have analyzed long-term outcome and donor cell engraftment in 194 patients with Wiskott-Aldrich syndrome (WAS) who have been treated by hematopoietic cell transplantation (HCT) in the period 1980- 2009. Overall survival was 84.0% and was even higher (89.1% 5-year survival) for those who received HCT since the year 2000, reflecting recent improvement of outcome after transplantation from mismatched family donors and for patients who received HCT from an unrelated donor at older than 5 years. Patients who went to transplantation in better clinical conditions had a lower rate of post-HCT complications. Retrospective analysis of lineage-specific donor cell engraftment showed that stable full donor chimerism was attained by 72.3% of the patients who survived for at least 1 year after HCT. Mixed chimerism was associated with an increased risk of incomplete reconstitution of lymphocyte count and post-HCT autoimmunity, and myeloid donor cell chimerism < 50% was associated with persistent thrombocytopenia. These observations indicate continuous improvement of outcome after HCT for WAS and may have important implications for the development of novel protocols aiming to obtain full correction of the disease and reduce post-HCT complications.


Asunto(s)
Linaje de la Célula , Trasplante de Células Madre Hematopoyéticas/métodos , Quimera por Trasplante/sangre , Síndrome de Wiskott-Aldrich/cirugía , Autoinmunidad/inmunología , Donantes de Sangre , Niño , Preescolar , Estudios de Seguimiento , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Humanos , Mutación , Evaluación de Resultado en la Atención de Salud/estadística & datos numéricos , Complicaciones Posoperatorias/sangre , Complicaciones Posoperatorias/etiología , Complicaciones Posoperatorias/inmunología , Estudios Retrospectivos , Análisis de Supervivencia , Trombocitopenia/sangre , Trombocitopenia/etiología , Factores de Tiempo , Síndrome de Wiskott-Aldrich/sangre , Síndrome de Wiskott-Aldrich/genética
15.
Blood ; 117(1): 53-62, 2011 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-20926771

RESUMEN

X-linked lymphoproliferative disease (XLP1) is a rare immunodeficiency characterized by severe immune dysregulation and caused by mutations in the SH2D1A/SAP gene. Clinical manifestations are varied and include hemophagocytic lymphohistiocytosis (HLH), lymphoma and dysgammaglobulinemia, often triggered by Epstein-Barr virus infection. Historical data published before improved treatment regimens shows very poor outcome. We describe a large cohort of 91 genetically defined XLP1 patients collected from centers worldwide and report characteristics and outcome data for 43 patients receiving hematopoietic stem cell transplant (HSCT) and 48 untransplanted patients. The advent of better treatment strategies for HLH and malignancy has greatly reduced mortality for these patients, but HLH still remains the most severe feature of XLP1. Survival after allogeneic HSCT is 81.4% with good immune reconstitution in the large majority of patients and little evidence of posttransplant lymphoproliferative disease. However, survival falls to 50% in patients with HLH as a feature of disease. Untransplanted patients have an overall survival of 62.5% with the majority on immunoglobulin replacement therapy, but the outcome for those untransplanted after HLH is extremely poor (18.8%). HSCT should be undertaken in all patients with HLH, because outcome without transplant is extremely poor. The outcome of HSCT for other manifestations of XLP1 is very good, and if HSCT is not undertaken immediately, patients must be monitored closely for evidence of disease progression.


Asunto(s)
Antígenos CD/genética , Péptidos y Proteínas de Señalización Intracelular/genética , Linfohistiocitosis Hemofagocítica/genética , Trastornos Linfoproliferativos/genética , Mutación/genética , Receptores de Superficie Celular/genética , Adolescente , Adulto , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Niño , Preescolar , Infecciones por Virus de Epstein-Barr/genética , Infecciones por Virus de Epstein-Barr/patología , Infecciones por Virus de Epstein-Barr/virología , Femenino , Trasplante de Células Madre Hematopoyéticas , Herpesvirus Humano 4/genética , Humanos , Lactante , Recién Nacido , Linfohistiocitosis Hemofagocítica/patología , Linfohistiocitosis Hemofagocítica/terapia , Trastornos Linfoproliferativos/patología , Trastornos Linfoproliferativos/terapia , Masculino , Persona de Mediana Edad , Proteína Asociada a la Molécula de Señalización de la Activación Linfocitaria , Miembro 1 de la Familia de Moléculas Señalizadoras de la Activación Linfocitaria , Tasa de Supervivencia , Adulto Joven
16.
Stem Cells ; 30(8): 1771-80, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22689616

RESUMEN

Slow T-cell reconstitution is a major clinical concern after transplantation of cord blood (CB)-derived hematopoietic stem cells. Adoptive transfer of in vitro-generated T-cell progenitors has emerged as a promising strategy for promoting de novo thymopoiesis and thus accelerating T-cell reconstitution. Here, we describe the development of a new culture system based on the immobilized Notch ligand Delta-like-4 (DL-4). Culture of human CD34(+) CB cells in this new DL-4 system enabled the in vitro generation of large amounts of T-cell progenitor cells that (a) displayed the phenotypic and molecular signatures of early thymic progenitors and (b) had high T lymphopoietic potential. When transferred into NOD/SCID/γc(-/-) (NSG) mice, DL-4 primed T-cell progenitors migrated to the thymus and developed into functional, mature, polyclonal αß T cells that subsequently left the thymus and accelerated T-cell reconstitution. T-cell reconstitution was even faster and more robust when ex vivo-manipulated and nonmanipulated CB samples were simultaneously injected into NSG mice (i.e., a situation reminiscent of the double CB transplant setting). This work provides further evidence of the ability of in vitro-generated human T-cell progenitors to accelerate T-cell reconstitution and also introduces a feeder-cell-free culture technique with the potential for rapid, safe transfer to a clinical setting.


Asunto(s)
Células Madre Hematopoyéticas/citología , Péptidos y Proteínas de Señalización Intercelular/inmunología , Linfocitos T/citología , Proteínas Adaptadoras Transductoras de Señales , Animales , Proteínas de Unión al Calcio , Diferenciación Celular/fisiología , Trasplante de Células Madre Hematopoyéticas/métodos , Células Madre Hematopoyéticas/inmunología , Humanos , Inmunoterapia , Péptidos y Proteínas de Señalización Intercelular/genética , Linfopoyesis/fisiología , Ratones , Ratones Endogámicos NOD , Ratones SCID , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología , Linfocitos T/inmunología
17.
Stem Cells ; 30(8): 1675-84, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22644669

RESUMEN

Mutations in the survival of motor neuron gene (SMN1) are responsible for spinal muscular atrophy, a fatal neuromuscular disorder. Mice carrying a homozygous deletion of Smn exon 7 directed to skeletal muscle (HSA-Cre, Smn(F7/F7) mice) present clinical features of human muscular dystrophies for which new therapeutic approaches are highly warranted. Herein we demonstrate that tail vein transplantation of mouse amniotic fluid stem (AFS) cells enhances the muscle strength and improves the survival rate of the affected animals. Second, after cardiotoxin injury of the Tibialis Anterior, only AFS-transplanted mice efficiently regenerate. Most importantly, secondary transplants of satellite cells (SCs) derived from treated mice show that AFS cells integrate into the muscle stem cell compartment and have long-term muscle regeneration capacity indistinguishable from that of wild-type-derived SC. This is the first study demonstrating the functional and stable integration of AFS cells into the skeletal muscle, highlighting their value as cell source for the treatment of muscular dystrophies.


Asunto(s)
Líquido Amniótico/citología , Músculo Esquelético/citología , Atrofia Muscular Espinal/genética , Atrofia Muscular Espinal/cirugía , Nicho de Células Madre/fisiología , Trasplante de Células Madre/métodos , Células Madre/citología , Líquido Amniótico/metabolismo , Animales , Modelos Animales de Enfermedad , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Músculo Esquelético/fisiopatología , Atrofia Muscular Espinal/patología , Atrofia Muscular Espinal/fisiopatología , Distribución Aleatoria , Células Madre/metabolismo , Proteína 1 para la Supervivencia de la Neurona Motora/genética
18.
Transfusion ; 53(3): 564-9, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22725259

RESUMEN

BACKGROUND: This report describes the specific kinetics of the peripheral blood (PB) CD34+ cell concentration in a selected group of very poor stem cell mobilizer patients treated with granulocyte-colony-stimulating factor (G-CSF) and plerixafor and determines the kinetics' impact on apheresis. STUDY DESIGN AND METHODS: All patients had previously experienced at least two failures of mobilization (without use of plerixafor). The present salvage therapy consisted in the administration of 10 µg/kg/day G-CSF for 5 days added to a dose of plerixafor administered at between 5 a.m. and 6 a.m. on Day 5. The PB CD34+ cell counts were tested every 3 hours thereafter. Apheresis was initiated as soon as the PB CD34+ cell count reached 10 × 10(6) /L. RESULTS: A PB CD34+ cell count higher than 10 × 10(6) /L was observed as soon as 3 hours after plerixafor administration in 10 of the 11 patients who reached this threshold at some point in the monitoring process. Interestingly, all patients presented an early decrease in the PB CD34+ cell count 8 to 12 hours after plerixafor administration (below 10 × 10(6) /L for seven patients). CONCLUSION: Had such patients been tested for PB CD34+ cell mobilization according to conventional criteria (i.e., 11 hr after plerixafor administration), apheresis would not have been performed at the optimal timing. For very poor stem cell mobilizer patients, early monitoring of PB CD34+ cell count may be required for the optimal initiation of apheresis.


Asunto(s)
Antígenos CD34/metabolismo , Eliminación de Componentes Sanguíneos/métodos , Movilización de Célula Madre Hematopoyética/métodos , Compuestos Heterocíclicos/administración & dosificación , Adulto , Anciano , Antígenos CD34/sangre , Bencilaminas , Recuento de Células Sanguíneas , Células Sanguíneas/metabolismo , Ciclamas , Esquema de Medicación , Sinergismo Farmacológico , Femenino , Factor Estimulante de Colonias de Granulocitos/administración & dosificación , Factor Estimulante de Colonias de Granulocitos/farmacología , Compuestos Heterocíclicos/farmacología , Humanos , Masculino , Persona de Mediana Edad , Mieloma Múltiple/tratamiento farmacológico , Mieloma Múltiple/terapia , Trasplante de Células Madre de Sangre Periférica , Factores de Tiempo , Insuficiencia del Tratamiento
19.
Nucleic Acids Res ; 39(11): e72, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21415009

RESUMEN

Human genetic diseases have been successfully corrected by integration of functional copies of the defective genes into human cells, but in some cases integration of therapeutic vectors has activated proto-oncogenes and contributed to leukemia. For this reason, extensive efforts have focused on analyzing integration site populations from patient samples, but the most commonly used methods for recovering newly integrated DNA suffer from severe recovery biases. Here, we show that a new method based on phage Mu transposition in vitro allows convenient and consistent recovery of integration site sequences in a form that can be analyzed directly using DNA barcoding and pyrosequencing. The method also allows simple estimation of the relative abundance of gene-modified cells from human gene therapy subjects, which has previously been lacking but is crucial for detecting expansion of cell clones that may be a prelude to adverse events.


Asunto(s)
Marcación de Gen , Terapia Genética , Análisis de Secuencia de ADN/métodos , Bacteriófago mu/genética , Línea Celular , Humanos , Reacción en Cadena de la Polimerasa
20.
Rev Prat ; 63(4): 457-61, 2013 Apr.
Artículo en Francés | MEDLINE | ID: mdl-23682464

RESUMEN

Gene therapy is a form of molecular medicine based on the addition of a corrected copy of a gene to the somatic cells of an individual in order to cure or to alleviate the underlying disease. This approach provides new targeted therapies for an increasing number of acquired or inherited diseases. Since its very first beginning in the 90's essentially for cancer treatment, this therapeutical approach has met unpredicted obstacles as well as true success. Recently, significant clinical benefits have been reported for several inherited diseases of the hematopoietic system but also of the retina and some cancers, widening its application provided that severe adverse events can be efficiently and stably prevented.


Asunto(s)
Terapia Genética , Terapia Genética/métodos , Terapia Genética/tendencias , Vectores Genéticos , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA