Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Immunity ; 45(3): 610-625, 2016 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-27612641

RESUMEN

The nature of gut intraepithelial lymphocytes (IELs) lacking antigen receptors remains controversial. Herein we showed that, in humans and in mice, innate intestinal IELs expressing intracellular CD3 (iCD3(+)) differentiate along an Id2 transcription factor (TF)-independent pathway in response to TF NOTCH1, interleukin-15 (IL-15), and Granzyme B signals. In NOTCH1-activated human hematopoietic precursors, IL-15 induced Granzyme B, which cleaved NOTCH1 into a peptide lacking transcriptional activity. As a result, NOTCH1 target genes indispensable for T cell differentiation were silenced and precursors were reprogrammed into innate cells with T cell marks including intracellular CD3 and T cell rearrangements. In the intraepithelial lymphoma complicating celiac disease, iCD3(+) innate IELs acquired gain-of-function mutations in Janus kinase 1 or Signal transducer and activator of transcription 3, which enhanced their response to IL-15. Overall we characterized gut T cell-like innate IELs, deciphered their pathway of differentiation and showed their malignant transformation in celiac disease.


Asunto(s)
Enfermedad Celíaca/inmunología , Interleucina-15/inmunología , Intestinos/inmunología , Linfoma/inmunología , Subgrupos de Linfocitos T/inmunología , Animales , Complejo CD3/inmunología , Diferenciación Celular/inmunología , Células Cultivadas , Granzimas/inmunología , Humanos , Proteína 2 Inhibidora de la Diferenciación/inmunología , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Receptor Notch1/inmunología , Factor de Transcripción STAT3/inmunología , Transducción de Señal/inmunología , Transcripción Genética/inmunología
2.
J Allergy Clin Immunol ; 143(2): 712-725.e5, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-29800647

RESUMEN

BACKGROUND: Autosomal dominant gain-of-function mutations in human stimulator of interferon genes (STING) lead to a severe autoinflammatory disease called STING-associated vasculopathy with onset in infancy that is associated with enhanced expression of interferon-stimulated gene transcripts. OBJECTIVE: The goal of this study was to analyze the phenotype of a new mouse model of STING hyperactivation and the role of type I interferons in this system. METHODS: We generated a knock-in model carrying an amino acid substitution (V154M) in mouse STING, corresponding to a recurrent mutation seen in human patients with STING-associated vasculopathy with onset in infancy. Hematopoietic development and tissue histology were analyzed. Lymphocyte activation and proliferation were assessed in vitro. STING V154M/wild-type (WT) mice were crossed to IFN-α/ß receptor (IFNAR) knockout mice to evaluate the type I interferon dependence of the mutant Sting phenotype recorded. RESULTS: In STING V154M/WT mice we detected variable expression of inflammatory infiltrates in the lungs and kidneys. These mice showed a marked decrease in survival and developed a severe combined immunodeficiency disease (SCID) affecting B, T, and natural killer cells, with an almost complete lack of antibodies and a significant expansion of monocytes and granulocytes. The blockade in B- and T-cell development was present from early immature stages in bone marrow and thymus. In addition, in vitro experiments revealed an intrinsic proliferative defect of mature T cells. Although the V154M/WT mutant demonstrated increased expression of interferon-stimulated genes, the SCID phenotype was not reversed in STING V154M/WT IFNAR knockout mice. However, the antiproliferative defect in T cells was rescued partially by IFNAR deficiency. CONCLUSIONS: STING gain-of-function mice developed an interferon-independent SCID phenotype with a T-cell, B-cell, and natural killer cell developmental defect and hypogammaglobulinemia that is associated with signs of inflammation in lungs and kidneys. Only the intrinsic proliferative defect of T cells was partially interferon dependent.


Asunto(s)
Linfocitos B/fisiología , Inflamación/genética , Células Asesinas Naturales/inmunología , Proteínas de la Membrana/genética , Mutación/genética , Inmunodeficiencia Combinada Grave/genética , Linfocitos T/fisiología , Agammaglobulinemia , Animales , Diferenciación Celular/genética , Modelos Animales de Enfermedad , Humanos , Interferón Tipo I/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptor de Interferón alfa y beta/genética
3.
Haematologica ; 103(5): 778-786, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29472357

RESUMEN

Sickle cell disease is characterized by chronic anemia and vaso-occlusive crises, which eventually lead to multi-organ damage and premature death. Hematopoietic stem cell transplantation is the only curative treatment but it is limited by toxicity and poor availability of HLA-compatible donors. A gene therapy approach based on the autologous transplantation of lentiviral-corrected hematopoietic stem and progenitor cells was shown to be efficacious in one patient. However, alterations of the bone marrow environment and properties of the red blood cells hamper the harvesting and immunoselection of patients' stem cells from bone marrow. The use of Filgrastim to mobilize large numbers of hematopoietic stem and progenitor cells into the circulation has been associated with severe adverse events in sickle cell patients. Thus, broader application of the gene therapy approach requires the development of alternative mobilization methods. We set up a phase I/II clinical trial whose primary objective was to assess the safety of a single injection of Plerixafor in sickle cell patients undergoing red blood cell exchange to decrease the hemoglobin S level to below 30%. The secondary objective was to measure the efficiency of mobilization and isolation of hematopoietic stem and progenitor cells. No adverse events were observed. Large numbers of CD34+ cells were mobilized extremely quickly. Importantly, the mobilized cells contained high numbers of hematopoietic stem cells, expressed high levels of stemness genes, and engrafted very efficiently in immunodeficient mice. Thus, Plerixafor can be safely used to mobilize hematopoietic stem cells in sickle cell patients; this finding opens up new avenues for treatment approaches based on gene addition and genome editing. Clinicaltrials.gov identifier: NCT02212535.


Asunto(s)
Anemia de Células Falciformes/terapia , Transfusión Sanguínea , Movilización de Célula Madre Hematopoyética , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/efectos de los fármacos , Células Madre Hematopoyéticas/metabolismo , Compuestos Heterocíclicos/administración & dosificación , Anemia de Células Falciformes/metabolismo , Anemia de Células Falciformes/patología , Fármacos Anti-VIH/administración & dosificación , Antígenos CD34/metabolismo , Antidrepanocíticos/administración & dosificación , Bencilaminas , Estudios de Casos y Controles , Células Cultivadas , Estudios de Cohortes , Ciclamas , Células Madre Hematopoyéticas/citología , Humanos , Hidroxiurea/administración & dosificación
4.
J Clin Immunol ; 37(7): 715-726, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28842786

RESUMEN

BACKGROUND: Common variable immunodeficiency (CVID) is characterized by infections and hypogammaglobulinemia. Neutropenia is rare during CVID. METHODS: The French DEFI study enrolled patients with primary hypogammaglobulinemia. Patients with CVID and neutropenia were retrospectively analyzed. RESULTS: Among 473 patients with CVID, 16 patients displayed neutropenia (lowest count [0-1400]*106/L). Sex ratio (M/F) was 10/6. Five patients died during the follow-up (11 years) with an increased percentage of deaths compared to the whole DEFI group (31.3 vs 3.4%, P < 0.05). Neutropenia was diagnosed for 10 patients before 22 years old. The most frequent symptoms, except infections, were autoimmune cytopenia, i.e., thrombopenia or anemia (11/16). Ten patients were affected with lymphoproliferative diseases. Two patients were in the infection only group and the others belonged to one or several other CVID groups. The median level of IgG was 2.6 g/L [0.35-4.4]. Most patients presented increased numbers of CD21low CD38low B cell, as already described in CVID autoimmune cytopenia group. Neutropenia was considered autoimmune in 11 cases. NGS for 52 genes of interest was performed on 8 patients. No deleterious mutations were found in LRBA, CTLA4, and PIK3. More than one potentially damaging variant in other genes associated with CVID were present in most patients arguing for a multigene process. CONCLUSION: Neutropenia is generally associated with another cytopenia and presumably of autoimmune origin during CVID. In the DEFI study, neutropenia is coupled with more severe clinical outcomes. It appears as an "alarm bell" considering patients' presentation and the high rate of deaths. Whole exome sequencing diagnosis should improve management.


Asunto(s)
Inmunodeficiencia Variable Común/epidemiología , Neutropenia/epidemiología , Adolescente , Adulto , Niño , Preescolar , Inmunodeficiencia Variable Común/sangre , Inmunodeficiencia Variable Común/genética , Inmunodeficiencia Variable Común/inmunología , Comorbilidad , Femenino , Francia/epidemiología , Humanos , Inmunoglobulinas/sangre , Lactante , Recién Nacido , Recuento de Leucocitos , Masculino , Persona de Mediana Edad , Neutropenia/sangre , Neutropenia/genética , Neutropenia/inmunología , Secuenciación del Exoma , Adulto Joven
5.
J Allergy Clin Immunol ; 138(6): 1681-1689.e8, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27405666

RESUMEN

BACKGROUND: We investigated 7 male patients (from 5 different families) presenting with profound lymphopenia, hypogammaglobulinemia, fluctuating monocytopenia and neutropenia, a poor immune response to vaccine antigens, and increased susceptibility to bacterial and varicella zoster virus infections. OBJECTIVE: We sought to characterize the genetic defect involved in a new form of X-linked immunodeficiency. METHODS: We performed genetic analyses and an exhaustive phenotypic and functional characterization of the lymphocyte compartment. RESULTS: We observed hemizygous mutations in the moesin (MSN) gene (located on the X chromosome and coding for MSN) in all 7 patients. Six of the latter had the same missense mutation, which led to an amino acid substitution (R171W) in the MSN four-point-one, ezrin, radixin, moesin domain. The seventh patient had a nonsense mutation leading to a premature stop codon mutation (R533X). The naive T-cell counts were particularly low for age, and most CD8+ T cells expressed the senescence marker CD57. This phenotype was associated with impaired T-cell proliferation, which was rescued by expression of wild-type MSN. MSN-deficient T cells also displayed poor chemokine receptor expression, increased adhesion molecule expression, and altered migration and adhesion capacities. CONCLUSION: Our observations establish a causal link between an ezrin-radixin-moesin protein mutation and a primary immunodeficiency that could be referred to as X-linked moesin-associated immunodeficiency.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Cromosomas Humanos X/genética , Síndromes de Inmunodeficiencia/genética , Infecciones/genética , Proteínas de Microfilamentos/genética , Mutación/genética , Adolescente , Adulto , Anciano , Adhesión Celular , Movimiento Celular , Niño , Preescolar , Estudios de Asociación Genética , Humanos , Recuento de Linfocitos , Masculino , Linaje
6.
J Allergy Clin Immunol ; 136(6): 1619-1626.e5, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26220525

RESUMEN

BACKGROUND: Myb-Like, SWIRM, and MPN domains 1 (MYSM1) is a metalloprotease that deubiquitinates the K119-monoubiquitinated form of histone 2A (H2A), a chromatin marker associated with gene transcription silencing. Likewise, it has been reported that murine Mysm1 participates in transcription derepression of genes, among which are transcription factors involved in hematopoietic stem cell homeostasis, hematopoiesis, and lymphocyte differentiation. However, whether MYSM1 has a similar function in human subjects remains unclear. Here we describe a patient presenting with a complete lack of B lymphocytes, T-cell lymphopenia, defective hematopoiesis, and developmental abnormalities. OBJECTIVES: We sought to characterize the underlying genetic cause of this syndrome. METHODS: We performed genome-wide homozygosity mapping, followed by whole-exome sequencing. RESULTS: Genetic analysis revealed that this novel disorder is caused by a homozygous MYSM1 missense mutation affecting the catalytic site within the deubiquitinase JAB1/MPN/Mov34 (JAMM)/MPN domain. Remarkably, during the course of our study, the patient recovered a normal immunohematologic phenotype. Genetic analysis indicated that this improvement originated from a spontaneous genetic reversion of the MYSM1 mutation in a hematopoietic stem cell. CONCLUSIONS: We here define a novel human immunodeficiency and provide evidence that MYSM1 is essential for proper immunohematopoietic development in human subjects. In addition, we describe one of the few examples of spontaneous in vivo genetic cure of a human immunodeficiency.


Asunto(s)
Proteínas de Unión al ADN/genética , Síndromes de Inmunodeficiencia/genética , Factores de Transcripción/genética , Linfocitos B/citología , Diferenciación Celular , Hematopoyesis/genética , Humanos , Lactante , Linfopenia/genética , Masculino , Mutación , Linfocitos T/citología , Transactivadores , Proteasas Ubiquitina-Específicas
7.
Curr Opin Pediatr ; 27(2): 233-9, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25635584

RESUMEN

PURPOSE OF REVIEW: Here we present fetal genotoxicity and mitochondrial toxicity, induced by nucleoside reverse transcriptase inhibitors (NRTIs), in HIV-1-infected pregnant women treated to prevent mother-to-child HIV-1 transmission, and in virus-free pregnant patas monkeys. RECENT FINDINGS: In the offspring of pregnant patas monkeys given human-equivalent NRTI protocols, aneuploidy was found in cultured bone marrow cells taken at birth, 1, and 3 years of age. In some newborn human infants, the offspring of HIV-1-infected mothers given zidovudine (AZT) therapy, aneuploidy, mitochondrial DNA (mtDNA) depletion, morphologically damaged mitochondria, and reduction in cardiac left ventricular muscle were observed. NRTI-exposed human and patas umbilical cords had similar levels of mtDNA depletion and mitochondrial morphological damage. NRTI-exposed patas offspring showed a compensatory increase in heart mtDNA, and a 50% loss of brain mtDNA at 1 year of age. Mitochondrial morphological damage and mtDNA loss were persistent in blood cells of NRTI-exposed infants up to 2 years of age, and in heart and brain from NRTI-exposed patas up to 3 years of age (human equivalent of 15 years). SUMMARY: Whereas use of NRTIs in human pregnancy protects many thousands of children worldwide, some HIV-1-uninfected infants born to HIV-1-infected mothers receiving antiretroviral drug therapy sustain toxicities that may have adverse consequences later in life.


Asunto(s)
ADN Mitocondrial/efectos de los fármacos , Infecciones por VIH/tratamiento farmacológico , Transmisión Vertical de Enfermedad Infecciosa/prevención & control , Complicaciones Infecciosas del Embarazo/tratamiento farmacológico , Inhibidores de la Transcriptasa Inversa/efectos adversos , Zidovudina/efectos adversos , Aneuploidia , Animales , Animales Recién Nacidos , ADN Mitocondrial/fisiología , Modelos Animales de Enfermedad , Erythrocebus patas , Femenino , Infecciones por VIH/prevención & control , Humanos , Lactante , Recién Nacido , Intercambio Materno-Fetal , Embarazo , Inhibidores de la Transcriptasa Inversa/farmacología , Inhibidores de la Transcriptasa Inversa/toxicidad , Zidovudina/farmacología , Zidovudina/toxicidad
8.
J Allergy Clin Immunol ; 134(1): 145-54, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24582315

RESUMEN

BACKGROUND: Expression of the pre-B-cell receptor (pre-BCR) by pre-BII cells constitutes a crucial checkpoint in B-cell differentiation. Mutations that affect the pre-B-cell receptor result in early B-cell differentiation blockades that lead to primary B-cell immunodeficiencies. BLNK adaptor protein has a key role in the pre-B-cell receptor signaling cascade, as illustrated by the abnormal B-cell development in the 4 patients with BLNK gene defects reported to date. However, the BLNK protein's precise function in human B-cell differentiation has not been completely specified. METHODS: B-cell development, including IgVH and Vk chain repertoires analysis, was studied in the bone marrow of a new case of BLNK deficiency in vitro and in vivo. RESULTS: Here, we report on a patient with agammaglobulinemia, with a total absence of circulating B cells. We detected a homozygous mutation in BLNK, which leads to the complete abrogation of BLNK protein expression. In the bone marrow, we identified a severe differentiation blockade at the pre-BI- to pre-BII-cell transition. IgVH gene rearrangements and selection of the IgH repertoire were normal, whereas the patient's pre-BI cells showed very restricted usage of the IgVκ repertoire. Complementation of bone marrow progenitors from the patient with the BLNK gene and transplantation into NOD/SCID/γcko mice allowed the complete restoration of B-cell differentiation and a normal usage of the IgVκ genes.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/inmunología , Agammaglobulinemia/inmunología , Linfocitos B/inmunología , Proteínas Adaptadoras Transductoras de Señales/genética , Agammaglobulinemia/genética , Agammaglobulinemia/patología , Animales , Linfocitos B/patología , Linfocitos B/trasplante , Médula Ósea/inmunología , Médula Ósea/patología , Diferenciación Celular , Expresión Génica , Prueba de Complementación Genética , Humanos , Cadenas kappa de Inmunoglobulina/genética , Cadenas kappa de Inmunoglobulina/inmunología , Ratones , Ratones Endogámicos NOD , Ratones SCID , Mutación , Precursores de Proteínas/genética , Precursores de Proteínas/inmunología , Receptores de Antígenos de Linfocitos B/genética , Receptores de Antígenos de Linfocitos B/inmunología , Anticuerpos de Dominio Único/genética , Anticuerpos de Dominio Único/inmunología , Trasplante Heterólogo
9.
Blood ; 119(15): 3458-68, 2012 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-22174160

RESUMEN

The molecular mechanisms that underlie T-cell quiescence are poorly understood. In the present study, we report a primary immunodeficiency phenotype associated with MST1 deficiency and primarily characterized by a progressive loss of naive T cells. The in vivo consequences include recurrent bacterial and viral infections and autoimmune manifestations. MST1-deficient T cells poorly expressed the transcription factor FOXO1, the IL-7 receptor, and BCL2. Conversely, FAS expression and the FAS-mediating apoptotic pathway were up-regulated. These abnormalities suggest that increased cell death of naive and proliferating T cells is the main mechanism underlying this novel immunodeficiency. Our results characterize a new mechanism in primary T-cell immunodeficiencies and highlight a role of the MST1/FOXO1 pathway in controlling the death of human naive T cells.


Asunto(s)
Síndromes de Inmunodeficiencia/genética , Síndromes de Inmunodeficiencia/inmunología , Mutación , Proteínas Serina-Treonina Quinasas/genética , Linfocitos T/fisiología , Adolescente , Supervivencia Celular/genética , Células Cultivadas , Niño , Preescolar , Familia , Femenino , Genes Recesivos/fisiología , Humanos , Síndromes de Inmunodeficiencia/sangre , Péptidos y Proteínas de Señalización Intracelular , Masculino , Mutación/fisiología , Linaje , Linfocitos T/inmunología
11.
J Infect Dis ; 208(2): 235-43, 2013 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-23559464

RESUMEN

BACKGROUND: The genotoxicity of zidovudine has been established in experimental models. The objective of the study was to identify genotoxicity markers in cord blood cells from newborns exposed in utero to antiretroviral (ARV) combinations containing zidovudine. METHODS: Cells were investigated by karyotyping and gene expression analysis of the CD34(+) hematopoietic stem/progenitor cell (HPC) compartment. RESULTS: Karyotyping of the cord blood cells from 15 ARV-exposed newborns and 12 controls revealed a higher proportion of aneuploid cells in the exposed group (median, 18.8% [interquartile range, 10.0%-26.7%] vs 6.6% [interquartile range, 3.1%-11.7%]; P < .001). All chromosomes were involved, with a random distribution of these alterations. Gene expression profiling of CD34(+) HPCs from 7 ARV-exposed and 6 control newborns revealed that >300 genes were significantly upregulated or downregulated by at least 1.5-fold in the exposed group (P < .05 for all comparisons). Significant alterations of genes involved in cell cycle control, mitotic checkpoints, and DNA repair were identified. Although this study does not allow discrimination between the roles of each of the 3 drugs, both cytogenetic and transcriptional findings are similar to those in cellular experiments that used zidovudine alone. CONCLUSIONS: The cord blood cells, including hematopoietic stem cells, from newborns exposed in utero to a zidovudine-based ARV combination present cytogenetic and transcriptional abnormalities compatible with DNA damage.


Asunto(s)
Fármacos Anti-VIH/efectos adversos , Sangre Fetal/efectos de los fármacos , Células Madre Hematopoyéticas/efectos de los fármacos , Zidovudina/efectos adversos , Adulto , Antígenos CD34/genética , Antígenos CD34/metabolismo , Ciclo Celular/genética , Reparación del ADN/genética , Combinación de Medicamentos , Femenino , Sangre Fetal/citología , Sangre Fetal/fisiología , Perfilación de la Expresión Génica/métodos , Infecciones por VIH/tratamiento farmacológico , Infecciones por VIH/genética , Infecciones por VIH/metabolismo , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Humanos , Recién Nacido , Transmisión Vertical de Enfermedad Infecciosa , Cariotipificación/métodos , Intercambio Materno-Fetal/fisiología , Embarazo , Complicaciones Infecciosas del Embarazo/tratamiento farmacológico , Complicaciones Infecciosas del Embarazo/metabolismo , Complicaciones Infecciosas del Embarazo/virología , Efectos Tardíos de la Exposición Prenatal , Células Madre/metabolismo , Transcriptoma/genética , Adulto Joven , Zidovudina/farmacocinética
12.
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
14.
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
15.
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
17.
Blood Cells Mol Dis ; 47(1): 72-8, 2011 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-21531153

RESUMEN

An important proof of principle has been achieved with the development of an in vitro T-cell differentiation assay based on the coculture of hematopoietic progenitors with the OP9-Delta1 stromal cell line. The original murine T cell differentiation assay has since been adapted for human T-cell differentiation, however with lower efficiency. The choice of both medium and cytokines is crucial in this assay, therefore our work has been focused on these two factors. The use of freshly reconstituted medium, the optimization of interleukine-7 (IL-7) concentration, and the addition of stem cell factor (SCF) have allowed to improve the proliferation of progenitors and T-cell precursors as well as the yield of double positive CD4+CD8+ T cells, and mature γδ and αß T cells. These optimizations make the OP9-Delta1 system sensitive enough to perform both quantitative and qualitative assays with various type of progenitors, including those transduced by a retroviral vector. The improved OP9-Delta1 assay therefore constitutes an extremely useful test for basic research purposes and for translational medicine.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Medios de Cultivo Condicionados/farmacología , Citocinas/farmacología , Linfocitos T/citología , Linfocitos T/efectos de los fármacos , Adyuvantes Inmunológicos/farmacología , Células Cultivadas , Técnicas de Cocultivo , Citocinas/inmunología , Células Madre Hematopoyéticas/inmunología , Humanos , Interleucina-7/farmacología , Factor de Células Madre/farmacología , Células del Estroma/inmunología
18.
Blood ; 113(17): 3953-60, 2009 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-19221036

RESUMEN

We have isolated c-Kit(+)Lin(-) cells from both human and murine amniotic fluid (AF) and investigated their hematopoietic potential. In vitro, the c-Kit(+)Lin(-) population in both species displayed a multilineage hematopoietic potential, as demonstrated by the generation of erythroid, myeloid, and lymphoid cells. In vivo, cells belonging to all 3 hematopoietic lineages were found after primary and secondary transplantation of murine c-Kit(+)Lin(-) cells into immunocompromised hosts, thus demonstrating the ability of these cells to self-renew. Gene expression analysis of c-Kit(+) cells isolated from murine AF confirmed these results. The presence of cells with similar characteristics in the surrounding amnion indicates the possible origin of AF c-Kit(+)Lin(-) cells. This is the first report showing that cells isolated from the AF do have hematopoietic potential; our results support the idea that AF may be a new source of stem cells for therapeutic applications.


Asunto(s)
Líquido Amniótico/metabolismo , Linaje de la Célula , Hematopoyesis , Proteínas Proto-Oncogénicas c-kit/metabolismo , Animales , Femenino , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Cinética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo
19.
Blood ; 112(7): 2722-9, 2008 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-18625888

RESUMEN

To address the role of chromatin structure in the establishment of hematopoietic stem cell (HSC) multilineage potential and commitment to the lymphoid lineage, we have analyzed histone modifications at a panel of lymphoid- and myeloid-affiliated genes in multipotent and lineage-committed hematopoietic cells isolated from human cord blood. Our results show that many B- and T-lymphoid genes, although silent in HSCs, are associated with acetylated histones H3 and H4. We also detected histone H3 lysine 4 methylation but not repressive lysine 9 or 27 methylation marks at these loci, indicative of an open chromatin structure. Interestingly, the relative level of H3 lysine 4 dimethylation to trimethylation at B-specific loci was high in multipotent CD34(+)CD38(lo) progenitors and decreased as they become actively transcribed in B-lineage cells. In vitro differentiation of CD34(+) cells toward the erythroid, granulocyte, and T-cell lineages resulted in a loss of histone acetylation at nonlineage-associated genes. This study provides evidence that histone modifications involved in chromatin decondensation are already in place at lymphoid-specific genes in primary human HSCs, supporting the idea that these genes are "primed" for expression before lineage commitment. This permissive chromatin structure is progressively lost as the stem cell differentiates.


Asunto(s)
Genes , Células Madre Hematopoyéticas/metabolismo , Histonas/metabolismo , Linfocitos/metabolismo , ADP-Ribosil Ciclasa 1/metabolismo , Acetilación , Antígenos CD19/metabolismo , Antígenos CD34/metabolismo , Diferenciación Celular , Linaje de la Célula , Sangre Fetal/citología , Células Madre Hematopoyéticas/citología , Humanos , Linfocitos/citología , Células Mieloides/citología , Células Mieloides/metabolismo , Células Satélite del Músculo Esquelético/citología , Células Satélite del Músculo Esquelético/metabolismo
20.
Sci Adv ; 6(7)2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-32917636

RESUMEN

Sickle cell disease (SCD) is caused by a single amino acid change in the adult hemoglobin (Hb) ß chain that causes Hb polymerization and red blood cell (RBC) sickling. The co-inheritance of mutations causing fetal γ-globin production in adult life hereditary persistence of fetal Hb (HPFH) reduces the clinical severity of SCD. HPFH mutations in the HBG γ-globin promoters disrupt binding sites for the repressors BCL11A and LRF. We used CRISPR-Cas9 to mimic HPFH mutations in the HBG promoters by generating insertions and deletions, leading to disruption of known and putative repressor binding sites. Editing of the LRF-binding site in patient-derived hematopoietic stem/progenitor cells (HSPCs) resulted in γ-globin derepression and correction of the sickling phenotype. Xenotransplantation of HSPCs treated with gRNAs targeting the LRF-binding site showed a high editing efficiency in repopulating HSPCs. This study identifies the LRF-binding site as a potent target for genome-editing treatment of SCD.


Asunto(s)
Anemia de Células Falciformes , Talasemia beta , Anemia de Células Falciformes/genética , Anemia de Células Falciformes/terapia , Sitios de Unión , Sistemas CRISPR-Cas , Hemoglobina Fetal/genética , Hemoglobina Fetal/metabolismo , Edición Génica/métodos , Humanos , Fenotipo , Globinas beta/genética , Globinas beta/metabolismo , Talasemia beta/genética , Talasemia beta/metabolismo , Talasemia beta/terapia , gamma-Globinas/genética , gamma-Globinas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA