Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
2.
N Engl J Med ; 386(24): 2295-2302, 2022 06 16.
Artículo en Inglés | MEDLINE | ID: mdl-35704481

RESUMEN

Lifelong immunosuppression is required for allograft survival after kidney transplantation but may not ultimately prevent allograft loss resulting from chronic rejection. We developed an approach that attempts to abrogate immune rejection and the need for post-transplantation immunosuppression in three patients with Schimke immuno-osseous dysplasia who had both T-cell immunodeficiency and renal failure. Each patient received sequential transplants of αß T-cell-depleted and CD19 B-cell-depleted haploidentical hematopoietic stem cells and a kidney from the same donor. Full donor hematopoietic chimerism and functional ex vivo T-cell tolerance was achieved, and the patients continued to have normal renal function without immunosuppression at 22 to 34 months after kidney transplantation. (Funded by the Kruzn for a Kure Foundation.).


Asunto(s)
Trasplante de Células Madre Hematopoyéticas , Síndromes de Inmunodeficiencia , Trasplante de Riñón , Síndrome Nefrótico , Osteocondrodisplasias , Enfermedades de Inmunodeficiencia Primaria , Arteriosclerosis/genética , Arteriosclerosis/terapia , Rechazo de Injerto/prevención & control , Humanos , Síndromes de Inmunodeficiencia/terapia , Riñón/fisiología , Trasplante de Riñón/efectos adversos , Síndrome Nefrótico/genética , Síndrome Nefrótico/terapia , Osteocondrodisplasias/genética , Osteocondrodisplasias/terapia , Enfermedades de Inmunodeficiencia Primaria/genética , Enfermedades de Inmunodeficiencia Primaria/terapia , Embolia Pulmonar/genética , Embolia Pulmonar/terapia , Acondicionamiento Pretrasplante/métodos
3.
J Allergy Clin Immunol ; 145(4): 1262-1271.e13, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31874182

RESUMEN

BACKGROUND: Immune-dysregulation, polyendocrinopathy, enteropathy, X-linked (IPEX) syndrome is a lethal disease caused by mutations in a transcription factor critical for the function of thymus-derived regulatory T (Treg) cells (ie, FOXP3), resulting in impaired Treg function and autoimmunity. At present, hematopoietic stem cell transplantation is the therapy of choice for patients with IPEX syndrome. If not available, multiple immunosuppressive regimens have been used with poor disease-free survival at long-term follow-up. Rapamycin has been shown to suppress peripheral T cells while sparing Treg cells expressing wild-type FOXP3, thereby proving beneficial in the clinical setting of immune dysregulation. However, the mechanisms of immunosuppression selective to Treg cells in patients with IPEX syndrome are unclear. OBJECTIVE: We sought to determine the cellular and molecular basis of the clinical benefit observed under rapamycin treatment in 6 patients with IPEX syndrome with different FOXP3 mutations. METHODS: Phenotype and function of FOXP3-mutated Treg cells from rapamycin-treated patients with IPEX syndrome were tested by flow cytometry and in vitro suppression assays, and the gene expression profile of rapamycin-conditioned Treg cells by droplet-digital PCR. RESULTS: Clinical and histologic improvements in patients correlated with partially restored Treg function, independent of FOXP3 expression or Treg frequency. Expression of TNF-receptor-superfamily-member 18 (TNFRSF18, glucocorticoid-induced TNF-receptor-related) and EBV-induced-3 (EBI3, an IL-35 subunit) in patients' Treg cells increased during treatment as compared with that of Treg cells from untreated healthy subjects. Furthermore inhibition of glucocorticoid-induced TNF-receptor-related and Ebi3 partially reverted in vitro suppression by in vivo rapamycin-conditioned Treg cells. CONCLUSIONS: Rapamycin is able to affect Treg suppressive function via a FOXP3-independent mechanism, thus sustaining the clinical improvement observed in patients with IPEX syndrome under rapamycin treatment.


Asunto(s)
Diabetes Mellitus Tipo 1/congénito , Diarrea/inmunología , Factores de Transcripción Forkhead/genética , Enfermedades Genéticas Ligadas al Cromosoma X/inmunología , Enfermedades del Sistema Inmune/congénito , Inmunosupresores/uso terapéutico , Mutación/genética , Sirolimus/uso terapéutico , Linfocitos T Reguladores/inmunología , Movimiento Celular , Células Cultivadas , Niño , Diabetes Mellitus Tipo 1/tratamiento farmacológico , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/inmunología , Diarrea/tratamiento farmacológico , Diarrea/genética , Regulación de la Expresión Génica , Enfermedades Genéticas Ligadas al Cromosoma X/tratamiento farmacológico , Enfermedades Genéticas Ligadas al Cromosoma X/genética , Proteína Relacionada con TNFR Inducida por Glucocorticoide/metabolismo , Humanos , Enfermedades del Sistema Inmune/tratamiento farmacológico , Enfermedades del Sistema Inmune/genética , Enfermedades del Sistema Inmune/inmunología , Tolerancia Inmunológica , Interleucinas/genética , Interleucinas/metabolismo , Activación de Linfocitos , Masculino , Antígenos de Histocompatibilidad Menor/genética , Antígenos de Histocompatibilidad Menor/metabolismo
5.
Mol Ther ; 21(1): 175-84, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22371846

RESUMEN

Gene therapy with ex vivo-transduced hematopoietic stem/progenitor cells may represent a valid therapeutic option for monogenic immunohematological disorders such as Wiskott-Aldrich syndrome (WAS), a primary immunodeficiency associated with thrombocytopenia. We evaluated the preclinical safety and efficacy of human CD34(+) cells transduced with lentiviral vectors (LV) encoding WAS protein (WASp). We first set up and validated a transduction protocol for CD34(+) cells derived from bone marrow (BM) or mobilized peripheral blood (MPB) using a clinical grade, highly purified LV. Robust transduction of progenitor cells was obtained in normal donors and WAS patients' cells, without evidence of toxicity. To study biodistribution of human cells and exclude vector release in vivo, LV-transduced CD34(+) cells were transplanted in immunodeficient mice, showing a normal engraftment and differentiation ability towards transduced lymphoid and myeloid cells in hematopoietic tissues. Vector mobilization to host cells and transmission to germline cells of the LV were excluded by different molecular assays. Analysis of vector integrations showed polyclonal integration patterns in vitro and in human engrafted cells in vivo. In summary, this work establishes the preclinical safety and efficacy of human CD34(+) cells gene therapy for the treatment of WAS.


Asunto(s)
Antígenos CD34/inmunología , Células de la Médula Ósea/citología , Trasplante de Médula Ósea , Vectores Genéticos , Lentivirus/genética , Transducción Genética , Síndrome de Wiskott-Aldrich/terapia , Animales , Células de la Médula Ósea/inmunología , Ratones , Ratones Noqueados
7.
Eur J Immunol ; 41(4): 1120-31, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21400500

RESUMEN

Mutations of forkhead box p3 (FOXP3), the master gene for naturally occurring regulatory T cells (nTregs), are responsible for the impaired function of nTregs, resulting in an autoimmune disease known as the immune dysregulation, polyendocrinopathy, enteropathy, X-linked (IPEX) syndrome. The relevance of other peripheral tolerance mechanisms, such as the presence and function of type 1 regulatory T (Tr1) cells, the major adaptive IL-10-producing Treg subset, in patients with IPEX syndrome remains to be clarified. FOXP3(mutated) Tr1-polarized cells, differentiated in vitro from CD4(+) T cells of four IPEX patients, were enriched in IL-10(+) IL-4(-) IFN-γ(+) T cells, a cytokine production profile specific for Tr1 cells, and expressed low levels of FOXP3 and high levels of Granzyme-B. IPEX Tr1 cells were hypoproliferative and suppressive, thus indicating that FOXP3 mutations did not impair their function. Furthermore, we isolated Tr1 cell clones from the peripheral blood of one FOXP3(null) patient, demonstrating that Tr1 cells are present in vivo and they can be expanded in vitro in the absence of WT FOXP3. Overall, our results (i) show that functional Tr1 cells differentiate independently of FOXP3, (ii) confirm that human Tr1 and nTregs are distinct T-cell lineages, and (iii) suggest that under favorable conditions Tr1 cells could exert regulatory functions in IPEX patients.


Asunto(s)
Diferenciación Celular , Enteritis/inmunología , Factores de Transcripción Forkhead/genética , Enfermedades Genéticas Ligadas al Cromosoma X/inmunología , Mutación , Poliendocrinopatías Autoinmunes/inmunología , Linfocitos T Reguladores/inmunología , Linaje de la Célula , Células Cultivadas , Enteritis/genética , Factores de Transcripción Forkhead/inmunología , Enfermedades Genéticas Ligadas al Cromosoma X/genética , Humanos , Inmunidad Innata , Subunidad alfa del Receptor de Interleucina-2/inmunología , Poliendocrinopatías Autoinmunes/genética , Síndrome , Linfocitos T Reguladores/citología
8.
Pediatr Transplant ; 14(3): 393-401, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20070557

RESUMEN

Immune-mediated refractoriness to platelet transfusion is a major problem in patients undergoing HSCT. In a cohort of 50 pediatric patients affected by beta thalassemia coming from Middle East countries, we experienced a high incidence of refractoriness because of anti-HLA antibodies during post-HSCT aplasia. In a risk factors analysis, factors predicting a negative transfusion outcome were presence of spleen and the number of anti-HLA antibodies. We adopted a policy to select platelet donors by avoiding HLA antigens against which the patient had specific antibodies. Transfusion of dedicated units resulted in 26% refractoriness compared to 74% to random units (p < 0.0001). When dedicated transfusions were used, the presence of spleen did not influence transfusion outcome. Analyzing transfusion outcome depending on the degree of HLA match and ABO compatibility, 76% successful transfusions were obtained with HLA-matched- ABO compatible followed by 67% in HLA-1mismatch- ABO compatible or HLA-matched- ABO incompatible and by 46% in HLA-1mismatch- ABO incompatible. In conclusion, we provide evidence that the selection of platelet donors according to patient characteristics, anti-HLA antibodies and ABO matching, is successful in reducing platelet refractoriness in heavily alloimmunized thalassemia patients undergoing transplantation.


Asunto(s)
Transfusión de Plaquetas/efectos adversos , Talasemia beta/inmunología , Talasemia beta/terapia , Sistema del Grupo Sanguíneo ABO/inmunología , Adolescente , Tipificación y Pruebas Cruzadas Sanguíneas/métodos , Distribución de Chi-Cuadrado , Niño , Preescolar , Femenino , Antígenos HLA/inmunología , Humanos , Terapia de Inmunosupresión/métodos , Lactante , Modelos Lineales , Masculino , Factores de Riesgo , Trasplante de Células Madre , Acondicionamiento Pretrasplante/métodos
9.
Br J Haematol ; 147(4): 554-60, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19747363

RESUMEN

Hepatic veno-occlusive disease (VOD) is a common complication of haematopoietic stem cell transplantation (HSCT), with reported incidences of 5-40% in children. Recently, defibrotide (DF) has been successfully used as prophylaxis and treatment of VOD. This study reports data on 63 human leucocyte antigen-matched HSCT performed in 57 children affected by beta thalassemia at very high risk for developing VOD (liver fibrosis, iron overload, hepatitis C virus infections, busulphan-based conditioning, methotraexate + ciclosporine). All patients received a busulphan-based conditioning regimen, either orally (four HSCT) or intravenously (59 HSCT). All patients received oral DF (40 mg/kg per day, final dose) as VOD prophylaxis from median day -9 to median day +29. In order to overcome the lack of oral paediatric formulations, a galenic formulation was administered. DF was well tolerated. Only one patient fulfilled Seattle Criteria for VOD diagnosis. This patient had discontinued DF 6 d prior to VOD onset, due to high risk of haemorrhage. We concluded that oral defibrotide prophylaxis and i.v. busulphan safely abated VOD incidence in high-risk patients who had undergone HSCT. A galenic preparation of oral DF also permits this treatment in low-weight patients. Costs of DF prophylaxis are acceptable considering the reduced incidence of VOD.


Asunto(s)
Busulfano/uso terapéutico , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Enfermedad Veno-Oclusiva Hepática/prevención & control , Inhibidores de Agregación Plaquetaria/uso terapéutico , Polidesoxirribonucleótidos/uso terapéutico , Talasemia beta/terapia , Adolescente , Niño , Preescolar , Evaluación de Medicamentos , Femenino , Enfermedad Injerto contra Huésped/prevención & control , Trasplante de Células Madre Hematopoyéticas/métodos , Enfermedad Veno-Oclusiva Hepática/etiología , Humanos , Inmunosupresores/uso terapéutico , Masculino , Estudios Retrospectivos , Acondicionamiento Pretrasplante/métodos
10.
Int Immunol ; 20(3): 421-31, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18270368

RESUMEN

Forkhead box P3 (FOXP3) is considered a specific marker for CD4(+)CD25(+) regulatory T (Treg) cells, but increasing evidence suggests that human CD4(+)CD25(-) effector T (Teff) cells can transiently express FOXP3 upon activation. We demonstrate that the signal transducer and activator of transcription 5 (STAT5)-signaling cytokines, IL-2, IL-15 and to a lesser extent IL-7, induce FOXP3 up-regulation in vitro in activated human Teff cells. In contrast, cytokines which do not activate STAT5, such as IL-4 or transforming growth factor-beta alone, do not directly induce FOXP3 expression in activated Teff cells. Moreover, expression of a constitutively active form of STAT5a is sufficient to induce FOXP3 expression in Teff cells. Expression of FOXP3 in activated Teff cells requires both TCR-mediated activation and endogenous IL-2, but is not dependent on cell division and does not induce suppressive function. The presence of STAT5-activating cytokines is also required to maintain high FOXP3 expression and suppressive activity of Treg cells in vitro. These data indicate that activation of STAT5 sustains FOXP3 expression in both Treg and Teff cells and contribute to our understanding of how cytokines affect the expression of FOXP3.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Factores de Transcripción Forkhead/inmunología , Regulación de la Expresión Génica , Factor de Transcripción STAT5/inmunología , Linfocitos T Reguladores/inmunología , Linfocitos T CD4-Positivos/efectos de los fármacos , Línea Celular , Células Cultivadas , Inhibidores Enzimáticos/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Inmunosupresores/farmacología , Interleucina-2/farmacología , Subunidad alfa del Receptor de Interleucina-2/inmunología , Transducción de Señal , Sirolimus/farmacología , Linfocitos T Reguladores/efectos de los fármacos
11.
Int Immunol ; 19(4): 345-54, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17329235

RESUMEN

Forkhead box P3 (FOXP3) is currently thought to be the most specific marker for naturally occurring CD4(+)CD25(+) T regulatory cells (nTregs). In mice, expression of FoxP3 is strictly correlated with regulatory activity, whereas increasing evidence suggests that in humans, activated T effector cells (Teffs) may also express FOXP3. In order to better define the role of FOXP3 in human Teff cells, we investigated the intensity and kinetics of expression in ex vivo Teff cells, suppressed Teff cells and Teff cell lines. We found that all dividing Teff cells expressed FOXP3, but only transiently, and at levels that were significantly lower than those in suppressive nTregs. This temporary expression in Teff cells was insufficient to suppress expression of reported targets of FOXP3 repressor activity, including CD127, IL-2 and IFN-gamma, and was not correlated with induction of a nTreg phenotype. Thus expression of FOXP3 is a normal consequence of CD4(+) T cell activation and, in humans, it can no longer be used as an exclusive marker of nTregs. These data indicate that our current understanding of how FOXP3 contributes to immune tolerance in humans needs to be re-evaluated.


Asunto(s)
Proliferación Celular , Citocinas/metabolismo , Factores de Transcripción Forkhead/metabolismo , Subgrupos de Linfocitos T/metabolismo , Anticuerpos/farmacología , Antígenos CD/metabolismo , Antígenos de Diferenciación/metabolismo , Antígenos de Diferenciación de Linfocitos T/metabolismo , Antígenos CD28/inmunología , Complejo CD3/inmunología , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Antígeno CTLA-4 , Línea Celular , Anergia Clonal , Técnicas de Cocultivo , Cisteína/análogos & derivados , Cisteína/farmacología , Flagelina/farmacología , Proteína Relacionada con TNFR Inducida por Glucocorticoide , Humanos , Interferón gamma/metabolismo , Interleucina-2/metabolismo , Interleucina-2/farmacología , Subunidad alfa del Receptor de Interleucina-2/metabolismo , Subunidad alfa del Receptor de Interleucina-7/metabolismo , Lectinas Tipo C , Lipoproteínas/farmacología , Activación de Linfocitos/inmunología , Receptores de Factor de Crecimiento Nervioso/metabolismo , Receptores del Factor de Necrosis Tumoral/metabolismo , Subgrupos de Linfocitos T/efectos de los fármacos , Subgrupos de Linfocitos T/inmunología , Linfocitos T Reguladores/efectos de los fármacos , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Receptores Toll-Like/inmunología
12.
Blood ; 101(12): 5076-83, 2003 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-12609834

RESUMEN

We previously reported that interleukin-10 (IL-10) and transforming growth factor (TGF)-beta treatment of primary mixed lymphocyte reaction (MLR) cultures resulted in secondary alloantigen-specific hyporesponsiveness and protection from graft-versus-host disease (GVHD) lethality. Here, we report that CD4+ T cells recovered from the IL-10- and TGF-beta-treated primary MLR cultures have immunoregulatory function. Tolerized cells significantly inhibited proliferation of naive alloreactive CD4+ T cells in a primary MLR. Inhibition of the naive alloresponse was observed with as few as 1 tolerized cell to 10 naive responder cells. Tolerized cells were able to significantly reduce GVHD lethality when injected with naive alloreactive CD4+ T cells into major histocombatibility class (MHC) II disparate recipients. Rigorous CD25 depletion of the primary MLR had no effect on generation of a regulatory capacity, suggesting that the regulatory cells likely originated from CD4+CD25- T cells. Immune suppression was mediated independently of IL-10 and TGF-beta production, as neutralizing antibodies for IL-10, IL-10R, and TGF-beta were unable to revert suppression, and IL-10- deficient CD4+ T cells were able to mediate in vitro and in vivo suppression. The generation of immunoregulatory cells from a CD4+CD25- population during tolerization with IL-10 and TGF-beta provides an additional mechanism to prevent GVHD lethality by T cells that may escape full tolerance induction.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Tolerancia Inmunológica , Interleucina-10/farmacología , Isoantígenos/inmunología , Receptores de Interleucina-2/análisis , Factor de Crecimiento Transformador beta/farmacología , Animales , Anticuerpos/farmacología , Citometría de Flujo , Enfermedad Injerto contra Huésped/mortalidad , Enfermedad Injerto contra Huésped/prevención & control , Interleucina-10/inmunología , Prueba de Cultivo Mixto de Linfocitos , Ratones , Ratones Endogámicos C57BL , Receptores de Interleucina/inmunología , Receptores de Interleucina-10 , Factor de Crecimiento Transformador beta/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...