Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
Blood ; 139(13): 1939-1953, 2022 03 31.
Artículo en Inglés | MEDLINE | ID: mdl-35015835

RESUMEN

Understanding the biological and clinical impact of copy number aberrations (CNAs) on the development of precision therapies in cancer remains an unmet challenge. Genetic amplification of chromosome 1q (chr1q-amp) is a major CNA conferring an adverse prognosis in several types of cancer, including in the blood cancer multiple myeloma (MM). Although several genes across chromosome 1 (chr1q) portend high-risk MM disease, the underpinning molecular etiology remains elusive. Here, with reference to the 3-dimensional (3D) chromatin structure, we integrate multi-omics data sets from patients with MM with genetic variables to obtain an associated clinical risk map across chr1q and to identify 103 adverse prognosis genes in chr1q-amp MM. Prominent among these genes, the transcription factor PBX1 is ectopically expressed by genetic amplification and epigenetic activation of its own preserved 3D regulatory domain. By binding to reprogrammed superenhancers, PBX1 directly regulates critical oncogenic pathways and a FOXM1-dependent transcriptional program. Together, PBX1 and FOXM1 activate a proliferative gene signature that predicts adverse prognosis across multiple types of cancer. Notably, pharmacological disruption of the PBX1-FOXM1 axis with existing agents (thiostrepton) and a novel PBX1 small molecule inhibitor (T417) is selectively toxic against chr1q-amp myeloma and solid tumor cells. Overall, our systems medicine approach successfully identifies CNA-driven oncogenic circuitries, links them to clinical phenotypes, and proposes novel CNA-targeted therapy strategies in MM and other types of cancer.


Asunto(s)
Mieloma Múltiple , Cromosomas Humanos Par 1/metabolismo , Proteína Forkhead Box M1/genética , Humanos , Mieloma Múltiple/tratamiento farmacológico , Mieloma Múltiple/genética , Mieloma Múltiple/metabolismo , Factor de Transcripción 1 de la Leucemia de Células Pre-B/genética , Pronóstico , Análisis de Sistemas , Factores de Transcripción/genética
2.
Nat Commun ; 12(1): 5450, 2021 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-34521827

RESUMEN

Multiple myeloma is a genetically heterogeneous cancer of the bone marrow plasma cells (PC). Distinct myeloma transcriptome profiles are primarily driven by myeloma initiating events (MIE) and converge into a mutually exclusive overexpression of the CCND1 and CCND2 oncogenes. Here, with reference to their normal counterparts, we find that myeloma PC enhanced chromatin accessibility combined with paired transcriptome profiling can classify MIE-defined genetic subgroups. Across and within different MM genetic subgroups, we ascribe regulation of genes and pathways critical for myeloma biology to unique or shared, developmentally activated or de novo formed candidate enhancers. Such enhancers co-opt recruitment of existing transcription factors, which although not transcriptionally deregulated per se, organise aberrant gene regulatory networks that help identify myeloma cell dependencies with prognostic impact. Finally, we identify and validate the critical super-enhancer that regulates ectopic expression of CCND2 in a subset of patients with MM and in chronic lymphocytic leukemia.


Asunto(s)
Carcinogénesis/genética , Ciclina D1/genética , Ciclina D2/genética , Regulación Neoplásica de la Expresión Génica , Leucemia Linfocítica Crónica de Células B/genética , Mieloma Múltiple/genética , Transcriptoma , Células de la Médula Ósea/metabolismo , Células de la Médula Ósea/patología , Carcinogénesis/metabolismo , Carcinogénesis/patología , Estudios de Casos y Controles , Línea Celular Tumoral , Cromatina/química , Cromatina/metabolismo , Ciclina D1/metabolismo , Ciclina D2/metabolismo , Elementos de Facilitación Genéticos , Perfilación de la Expresión Génica , Redes Reguladoras de Genes , N-Metiltransferasa de Histona-Lisina/genética , N-Metiltransferasa de Histona-Lisina/metabolismo , Humanos , Leucemia Linfocítica Crónica de Células B/metabolismo , Leucemia Linfocítica Crónica de Células B/mortalidad , Leucemia Linfocítica Crónica de Células B/patología , Mieloma Múltiple/metabolismo , Mieloma Múltiple/mortalidad , Mieloma Múltiple/patología , Células Plasmáticas/metabolismo , Células Plasmáticas/patología , Proteínas Proto-Oncogénicas c-maf/genética , Proteínas Proto-Oncogénicas c-maf/metabolismo , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Análisis de Supervivencia
3.
Clin Immunol ; 183: 8-16, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28645875

RESUMEN

The ontogeny of the natural, public IgM repertoire remains incompletely explored. Here, high-resolution immunogenetic analysis of B cells from (unrelated) fetal, child, and adult samples, shows that although fetal liver (FL) and bone marrow (FBM) IgM repertoires are equally diversified, FL is the main source of IgM natural immunity during the 2nd trimester. Strikingly, 0.25% of all prenatal clonotypes, comprising 18.7% of the expressed repertoire, are shared with the postnatal samples, consistent with persisting fetal IgM+ B cells being a source of natural IgM repertoire in adult life. Further, the origins of specific stereotypic IgM+ B cell receptors associated with chronic lymphocytic leukemia, can be traced back to fetal B cell lymphopoiesis, suggesting that persisting fetal B cells can be subject to malignant transformation late in life. Overall, these novel data provide unique insights into the ontogeny of physiological and malignant B lymphopoiesis that spans the human lifetime.


Asunto(s)
Linfocitos B/inmunología , Médula Ósea/inmunología , Feto/inmunología , Cadenas Pesadas de Inmunoglobulina/genética , Inmunoglobulina M/genética , Leucemia Linfocítica Crónica de Células B/genética , Hígado/inmunología , Linfopoyesis/genética , Receptores de Antígenos de Linfocitos B/genética , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Cadenas Pesadas de Inmunoglobulina/inmunología , Inmunoglobulina M/inmunología , Leucemia Linfocítica Crónica de Células B/inmunología , Linfopoyesis/inmunología , Receptores de Antígenos de Linfocitos B/inmunología , Análisis de Secuencia de ADN
4.
Blood ; 124(20): 3151-4, 2014 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-25293775

RESUMEN

A rare point mutation in the core promoter -270GC-rich box of PIGM, a housekeeping gene, disrupts binding of the generic transcription factor (TF) Sp1 and causes inherited glycosylphosphatidylinositol (GPI) deficiency (IGD). We show that whereas PIGM messenger RNA levels and surface GPI expression in IGD B cells are low, GPI expression is near normal in IGD erythroid cells. This divergent phenotype results from differential promoter chromatin accessibility and binding of Sp1. Specifically, whereas PIGM transcription in B cells is dependent on Sp1 binding to the -270GC-rich box and is associated with lower promoter accessibility, in erythroid cells, Sp1 activates PIGM transcription by binding upstream of (but not to) the -270GC-rich box. These findings explain intact PIGM transcription in IGD erythroid cells and the lack of clinically significant intravascular hemolysis in patients with IGD. Furthermore, they provide novel insights into tissue-specific transcriptional control of a housekeeping gene by a generic TF.


Asunto(s)
Glicosilfosfatidilinositoles/deficiencia , Hemoglobinuria Paroxística/genética , Manosiltransferasas/genética , Activación Transcripcional , Linfocitos B/metabolismo , Linfocitos B/patología , Eritrocitos/metabolismo , Eritrocitos/patología , Glicosilfosfatidilinositoles/genética , Glicosilfosfatidilinositoles/metabolismo , Hemoglobinuria Paroxística/metabolismo , Hemoglobinuria Paroxística/patología , Humanos , Mutación , Fenotipo , Regiones Promotoras Genéticas , Convulsiones , Factor de Transcripción Sp1/metabolismo
5.
J Gene Med ; 16(1-2): 1-10, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24590875

RESUMEN

BACKGROUND: Mutations in the ß-globin gene (HBB) cause haemoglobinopathies where current treatments have serious limitations. Gene correction by homologous recombination (HR) is an attractive approach to gene therapy for such diseases and is stimulated by gene-specific endonucleases, including zinc finger nucleases (ZFNs). Customised nucleases targeting HBB have previously been shown to promote HR-mediated HBB modification in 0.3­60% of drug-selected cells, although frequencies among unselected cells, more relevant to the goal of correcting HBB in primary stem cells, have not been reported. METHODS: ZFNs targeting HBB were tested for HBB binding (two-hybrid assay) or HBB cleavage followed by inaccurate end joining (surveyor assay)in bacteria or human cancer cell lines, respectively. ZFN-stimulated HR was measured in cell lines by a modified fluorescence-based reporter assay or by targeted insertion of a drug-resistance marker into endogenous HBB confirmed by Southern analyses. RESULTS: Although the ZFNs that we assembled in-house showed limited potential, a commercially commissioned nuclease (ZFN4) enhanced HR mediated HBB modification in up to 95% of drug-selected cells. Among unselected cells, however, this frequency was less than 0.2%. Furthermore, ZFN4 cleaved HBB at an efficiency of 1­2% (surveyor assay) and enhanced the HR reporter assay 20-fold less efficiently than a control endonuclease. CONCLUSIONS: With ZFN4, we achieved higher efficiencies of HR-mediated HBB modification than previously reported for drug-selected cells. Our measurements of ZFN4-induced HR in unselected cells, however, suggest that improved nucleases must be developed if therapeutic HBB correction is to be achievable in primary stem cells.


Asunto(s)
Endodesoxirribonucleasas/genética , Factores de Transcripción/genética , Dedos de Zinc/genética , Globinas beta/genética , Anemia de Células Falciformes/genética , Anemia de Células Falciformes/terapia , Línea Celular Tumoral , Reparación del ADN por Unión de Extremidades , Endodesoxirribonucleasas/biosíntesis , Endodesoxirribonucleasas/metabolismo , Marcación de Gen , Terapia Genética , Recombinación Homóloga , Humanos , Mutación , Factores de Transcripción/biosíntesis , Factores de Transcripción/metabolismo , Talasemia beta/genética , Talasemia beta/terapia
6.
J Immunol ; 186(5): 2910-7, 2011 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-21278350

RESUMEN

Invariant NKT (iNKT) cells modulate innate and adaptive immune responses through activation of myeloid dendritic cells and macrophages and via enhanced clonogenicity, differentiation, and egress of their shared myeloid progenitors. Because these same progenitors give rise to osteoclasts (OCs), which also mediate the egress of hematopoietic progenitors and orchestrate bone remodeling, we hypothesized that iNKT cells would extend their myeloid cell regulatory role to the development and function of OCs. In this study, we report that selective activation of iNKT cells by α-galactosylceramide causes myeloid cell egress, enhances OC progenitor and precursor development, modifies the intramedullary kinetics of mature OCs, and enhances their resorptive activity. OC progenitor activity is positively regulated by TNF-α and negatively regulated by IFN-γ, but is IL-4 and IL-17 independent. These data demonstrate a novel role of iNKT cells that couples osteoclastogenesis with myeloid cell egress in conditions of immune activation.


Asunto(s)
Diferenciación Celular/inmunología , Activación de Linfocitos/inmunología , Células T Asesinas Naturales/citología , Células T Asesinas Naturales/inmunología , Osteoclastos/inmunología , Osteoclastos/metabolismo , Animales , Movimiento Celular/inmunología , Células Dendríticas/citología , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Regulación hacia Abajo/inmunología , Interferón gamma/fisiología , Factor Estimulante de Colonias de Macrófagos/fisiología , Macrófagos/citología , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Células Mieloides/citología , Células Mieloides/inmunología , Células Mieloides/metabolismo , Células T Asesinas Naturales/metabolismo , Osteoclastos/citología , Ligando RANK/fisiología , Células Madre/citología , Células Madre/inmunología , Células Madre/metabolismo , Factor de Necrosis Tumoral alfa/fisiología , Regulación hacia Arriba/inmunología
7.
Biol Blood Marrow Transplant ; 16(2): 263-72, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19822218

RESUMEN

We conducted a prospective, multicenter investigation of human-leukocyte antigen (HLA) identical sibling bone marrow transplantation (BMT) in children with severe sickle cell disease (SCD) between 1991 and 2000. To determine if children were protected from complications of SCD after successful BMT, we extended our initial study of BMT for SCD to conduct assessments of the central nervous system (CNS) and of pulmonary function 2 or more years after transplantation. In addition, the impact on gonadal function was studied. After BMT, patients with stroke who had stable engraftment of donor cells experienced no subsequent stroke events after BMT, and brain magnetic resonance imaging (MRI) exams demonstrated stable or improved appearance. However, 2 patients with graft rejection had a second stroke after BMT. After transplantation, most patients also had unchanged or improved pulmonary function. Among the 11 patients who had restrictive lung changes at baseline, 5 were improved and 6 had persistent restrictive disease after BMT. Of the 2 patients who had obstructive changes at baseline, 1 improved and 1 had worsened obstructive disease after BMT. There was, however, significant gonadal toxicity after BMT, particularly among female recipients. In summary, individuals who had stable donor engraftment did not experience sickle-related complications after BMT, and were protected from progressive CNS and pulmonary disease.


Asunto(s)
Anemia de Células Falciformes/terapia , Trasplante de Médula Ósea/efectos adversos , Enfermedades del Sistema Nervioso Central/etiología , Trastornos Gonadales/etiología , Estado de Salud , Enfermedades Pulmonares Obstructivas/etiología , Adolescente , Anemia de Células Falciformes/complicaciones , Anemia de Células Falciformes/fisiopatología , Enfermedades del Sistema Nervioso Central/fisiopatología , Niño , Selección de Donante , Femenino , Estudios de Seguimiento , Trastornos Gonadales/fisiopatología , Supervivencia de Injerto , Enfermedad Injerto contra Huésped/tratamiento farmacológico , Enfermedad Injerto contra Huésped/etiología , Enfermedad Injerto contra Huésped/mortalidad , Antígenos HLA/inmunología , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Prueba de Histocompatibilidad , Humanos , Enfermedades Pulmonares Obstructivas/fisiopatología , Masculino , Hermanos , Análisis de Supervivencia , Quimera por Trasplante , Resultado del Tratamiento
8.
J Immunol ; 181(5): 3268-76, 2008 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-18713998

RESUMEN

Invariant NKT (iNKT) cells are a subset of highly conserved immunoregulatory T cells that modify a variety of immune responses, including alloreactivity. Central to their function is the interaction of the invariant TCR with glycosphingolipid (GSL) ligands presented by the nonpolymorphic MHC class I molecule CD1d and their ability to secrete rapidly large amounts of immunomodulatory cytokines when activated. Whether iNKT cells, like NK and conventional T cells, can directly display alloreactivity is not known. We show in this study that human iNKT cells and APC can establish a direct cross-talk leading to preferential maturation of allogeneic APC and a considerably higher reactivity of iNKT cells cultured with allogeneic rather that autologous APC. Although the allogeneic activation of iNKT cells is invariant TCR-CD1d interaction-dependent, GSL profiling suggests it does not involve the recognition of disparate CD1d/GSL complexes. Instead, we show that contrary to previous reports, iNKT cells, like NK and T cells, express killer Ig receptors at a frequency similar to that of conventional T cells and that iNKT cell allogeneic activation requires up-regulation and function of activating killer Ig receptors. Thus, iNKT cells can display alloreactivity, for which they use mechanisms characteristic of both NK and conventional T cells.


Asunto(s)
Antígenos CD1/metabolismo , Isoantígenos/inmunología , Células Asesinas Naturales/inmunología , Receptores de Antígenos de Linfocitos T/metabolismo , Receptores Inmunológicos/inmunología , Presentación de Antígeno , Células Presentadoras de Antígenos , Antígenos CD1d , Glicoesfingolípidos/metabolismo , Humanos , Activación de Linfocitos , Unión Proteica , Linfocitos T
9.
Early Hum Dev ; 84(8): 515-23, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18621490

RESUMEN

The diagnosis, acute management and follow-up of neonates with haemolytic disease of the newborn (HDN) still represents a significant area of activity for maternity/neonatal services. ABO incompatability is now the single largest cause of HDN in the western world. However, with increasing knowledge at the molecular level, and closer liaison between neonatal paediatricians and haematologists, the diagnosis of non-immune causes of HDN is increasing. As these conditions have an inherited basis and therefore have implications for other family members (or future children), it remains a high priority for all neonatal paediatricians to achieve an accurate diagnosis in all cases of HDN. As the efficacy of phototherapy increases the role of exchange transfusion in acute management is rapidly decreasing. This makes gauging the appropriate time to intervene with exchange transfusion a difficult clinical decision, and guidelines appropriate to the spectrum of contemporary disease are required. In the future intravenous immunoglobulin and/or intramuscular metalloporphyrins may further reduce the need for exchange transfusion and continue to change the spectrum of HDN faced by neonatal paediatricians.


Asunto(s)
Eritroblastosis Fetal/terapia , Eritroblastosis Fetal/sangre , Eritroblastosis Fetal/inmunología , Recambio Total de Sangre/métodos , Recambio Total de Sangre/tendencias , Femenino , Humanos , Inmunoglobulinas Intravenosas/uso terapéutico , Recién Nacido/inmunología , Fototerapia/métodos , Fototerapia/normas , Fototerapia/tendencias , Embarazo/inmunología
10.
N Engl J Med ; 356(16): 1641-7, 2007 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-17442906

RESUMEN

Disrupted binding of the transcription factor Sp1 to the mutated promoter region of the mannosyl transferase-encoding gene PIGM causes inherited glycosylphosphatidylinositol (GPI) deficiency characterized by splanchnic vein thrombosis and epilepsy. We show that this results in histone hypoacetylation at the promoter of PIGM. The histone deacetylase inhibitor butyrate increases PIGM transcription and surface GPI expression in vitro as well as in vivo through enhanced histone acetylation in an Sp1-dependent manner. More important, the drug caused complete cessation of intractable seizures in a child with inherited GPI deficiency.


Asunto(s)
Butiratos/uso terapéutico , Glicosilfosfatidilinositoles/deficiencia , Inhibidores de Histona Desacetilasas , Manosiltransferasas/genética , Errores Innatos del Metabolismo/tratamiento farmacológico , Factor de Transcripción Sp1/metabolismo , Transcripción Genética/efectos de los fármacos , Acetilación/efectos de los fármacos , Adolescente , Butiratos/farmacología , Epilepsia Tipo Ausencia/tratamiento farmacológico , Epilepsia Tipo Ausencia/etiología , Femenino , Glicosilfosfatidilinositoles/biosíntesis , Humanos , Manosiltransferasas/metabolismo , Errores Innatos del Metabolismo/complicaciones , Errores Innatos del Metabolismo/genética , Mutación , Regiones Promotoras Genéticas/fisiología , Trombosis/tratamiento farmacológico , Trombosis/etiología
12.
Curr Hematol Rep ; 5(1): 55-63, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16537047

RESUMEN

Neonatal thrombocytopenia is a common clinical problem. The majority of episodes are early-onset thrombocytopenias due to impaired fetal megakaryocytopoiesis associated with placental insufficiency; the commonest causes of severe early-onset thrombocytopenia are immune thrombocytopenias, congenital infections, and asphyxia. By contrast, about 90% of cases of severe thrombocytopenia presenting after the first few days of life are due to late-onset bacterial sepsis, necrotizing enterocolitis, or both. Although clinically stable neonates tolerate relatively low platelet counts without significant risk of hemorrhage, ill or clinically unstable neonates with profound thrombocytopenia often have a poor outcome. Currently, the only therapy is platelet transfusion. Despite many published guidelines for platelet transfusion in the newborn, however, there have been no randomized trials to define the safe lower limit for platelet counts in sick neonates. The platelet threshold at which the benefits of transfusion outweigh the risks in neonates remains unclear. Well-designed trials are urgently needed.


Asunto(s)
Trombocitopenia/etiología , Edad de Inicio , Transfusión Sanguínea , Humanos , Recién Nacido , Recuento de Plaquetas , Trombocitopenia/diagnóstico , Trombocitopenia/terapia
13.
Blood ; 107(8): 3138-44, 2006 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-16373666

RESUMEN

Invariant natural killer T cells (iNKT cells) are a small subset of immunoregulatory T cells highly conserved in humans and mice. On activation by glycolipids presented by the MHC-like molecule CD1d, iNKT cells promptly secrete T helper 1 and 2 (Th1/2) cytokines but also cytokines with hematopoietic potential such as GM-CSF. Here, we show that the myeloid clonogenic potential of human hematopoietic progenitors is increased in the presence of glycolipid-activated, GM-CSF-secreting NKT cells; conversely, short- and long-term progenitor activity is decreased in the absence of NKT cells, implying regulation of hematopoiesis in both the presence and the absence of immune activation. In accordance with these findings, iNKT-cell-deficient mice display impaired hematopoiesis characterized by peripheral-blood cytopenias, reduced marrow cellularity, lower frequency of hematopoietic stem cells (HSCs), and reduced early and late hematopoietic progenitors. We also show that CD1d is expressed on human HSCs. CD1d-expressing HSCs display short- and long-term clonogenic potential and can present the glycolipid alpha-galactosylceramide to iNKT cells. Thus, iNKT cells emerge as the first subset of regulatory T cells that are required for effective hematopoiesis in both steady-state conditions and under conditions of immune activation.


Asunto(s)
Presentación de Antígeno/inmunología , Galactosilceramidas/inmunología , Hematopoyesis/inmunología , Células Madre Hematopoyéticas/inmunología , Células Asesinas Naturales/inmunología , Activación de Linfocitos/fisiología , Animales , Presentación de Antígeno/efectos de los fármacos , Femenino , Galactosilceramidas/administración & dosificación , Factor Estimulante de Colonias de Granulocitos y Macrófagos/inmunología , Hematopoyesis/efectos de los fármacos , Células Madre Hematopoyéticas/citología , Humanos , Células Asesinas Naturales/citología , Activación de Linfocitos/efectos de los fármacos , Ratones , Ratones Noqueados , Células TH1/citología , Células TH1/inmunología , Células Th2/citología , Células Th2/inmunología
14.
J Immunol ; 175(8): 5087-94, 2005 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-16210612

RESUMEN

NKT cells are a small subset of regulatory T cells conserved in humans and mice. In humans they express the Valpha24Jalpha18 invariant chain (hence invariant NKT (iNKT) cells) and are restricted by the glycolipid-presenting molecule CD1d. In mice, iNKT cells may enhance or inhibit anti-infectious and antitumor T cell responses but suppress autoimmune and alloreactive responses. We postulated that iNKT cells might also modulate human alloreactive responses. Using MLR assays we demonstrate that in the presence of the CD1d-presented glycolipid alpha-galactosylceramide (alphaGC) alloreactivity is enhanced (37 +/- 12%; p < 0.001) in an iNKT cell-dependent manner. iNKT cells are activated early during the course of the MLR, presumably by natural ligands. In MLR performed without exogenous ligands, depletion of iNKT cells significantly diminished the alloresponse in terms of proliferation (58.8 +/- 24%; p < 0.001) and IFN-gamma secretion (43.2 +/- 15.2%; p < 0.001). Importantly, adding back fresh iNKT cells restored the reactivity of iNKT cell-depleted MLR to near baseline levels. CD1d-blocking mAbs equally reduced the reactivity of the iNKT cell-replete and -depleted MLR compared with IgG control, indicating that the effect of iNKT cells in the in vitro alloresponse is CD1d-dependent. These findings suggest that human iNKT cells, although not essential for its development, can enhance the alloreactive response.


Asunto(s)
Células Asesinas Naturales/inmunología , Activación de Linfocitos/inmunología , Linfocitos T/inmunología , Antígenos CD1/fisiología , Antígenos CD1d , Complejo CD3/metabolismo , Proliferación Celular , Células Cultivadas , Citometría de Flujo , Galactosilceramidas/fisiología , Humanos , Células Asesinas Naturales/citología , Células Asesinas Naturales/metabolismo , Ligandos , Prueba de Cultivo Mixto de Linfocitos , Linfocitos T/citología , Linfocitos T/metabolismo
15.
PLoS Med ; 2(2): e44, 2005 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15737000

RESUMEN

BACKGROUND TO THE DEBATE: Umbilical cord blood--the blood that remains in the placenta after birth--can be collected and stored frozen for years. A well-accepted use of cord blood is as an alternative to bone marrow as a source of hematopoietic stem cells for allogeneic transplantation to siblings or to unrelated recipients; women can donate cord blood for unrelated recipients to public banks. However, private banks are now open that offer expectant parents the option to pay a fee for the chance to store cord blood for possible future use by that same child (autologous transplantation).


Asunto(s)
Bancos de Sangre/economía , Ética Médica , Sangre Fetal , Sector Privado , Trasplante de Células Madre de Sangre del Cordón Umbilical/etnología , Humanos , Manejo de Especímenes , Trasplante Autólogo
16.
Stem Cells ; 23(1): 93-102, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-15625126

RESUMEN

First-trimester fetal blood contains a readily expandable population of stem cells, human fetal mesenchymal stem cells (hfMSCs), which might be exploited for autologous intrauterine gene therapy. We investigated the self-renewal and differentiation of hfMSCs after transduction with onco-retroviral and lentiviral vectors. After transduction with either a MoMuLV retrovirus or an HIV-1-based lentiviral vector carrying the ss-galactosidase and green fluorescent reporter gene, respectively, transgene expression, self-renewal, and differentiation capabilities were assessed 2 and 14 weeks later. Transduction with the lentiviral vector resulted in higher efficiencies than with the MoMuLV-based vector (mean, 97.7 +/- 1.4% versus 80.2 +/- 5.4%; p = .02). Transgene expression was maintained with lentiviral-transduced cells (94.6 +/- 2.6%) but decreased over 14 weeks in culture with onco-retroviral-transduced cells (48.3 +/- 3.9%). The self-renewal capability of these cells and their ability to undergo osteogenic, adipogenic, and myogenic differentiation was unimpaired after transduction with either vector. Finally, clonal expansion of lentivirally modified cells was expanded over 20 population doublings with maintenance of multiline age differentiation capacity. These results suggest that hfMSCs may be suitable targets for ex vivo genetic manipulation with onco-retroviral or lentiviral vectors without affecting their stem cell properties.


Asunto(s)
Sangre Fetal/citología , Técnicas de Transferencia de Gen , Vectores Genéticos , Células Madre Mesenquimatosas/citología , Transducción Genética , Diferenciación Celular/fisiología , Clonación Molecular , Sangre Fetal/metabolismo , Sangre Fetal/virología , Expresión Génica , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Lentivirus/genética , Lentivirus/metabolismo , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/virología , Retroviridae/genética , Retroviridae/metabolismo , Células Madre/metabolismo
17.
Lancet ; 364(9429): 179-82, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15246731

RESUMEN

Fetal cells enter maternal blood during pregnancy and persist in women with autoimmune disease. The frequency of subsequent fetomaternal microchimerism in healthy women and its cell type is unknown. To test the hypothesis that fetal mesenchymal stem cells persist in maternal organs, we studied female bone marrow and ribs. Male cells were identified by XY fluorescence in-situ hybridisation in marrow-derived mesenchymal stem cells and in rib sections from all women with male pregnancies, but not in controls (9/9 vs 0/5, p=0.0005). We conclude that fetal stem cells transferred into maternal blood engraft in marrow, where they remain throughout life. This finding has implications for normal pregnancy, for obstetric complications that increase fetomaternal trafficking, and for graft survival after transplantation.


Asunto(s)
Células de la Médula Ósea/citología , Quimera , Feto/citología , Intercambio Materno-Fetal , Mesodermo/citología , Células Madre/citología , Anciano , Anciano de 80 o más Años , Huesos/citología , Cromosomas Humanos X , Cromosomas Humanos Y , Femenino , Humanos , Hibridación Fluorescente in Situ , Masculino , Persona de Mediana Edad , Embarazo , Costillas , Factores de Tiempo
18.
Blood ; 102(4): 1224-31, 2003 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-12714525

RESUMEN

Hematopoietic stem cell transplantation (SCT) is the only proven cure for chronic myeloid leukemia (CML), a rare disease in childhood. We report outcomes of 314 children with Philadelphia-chromosome-positive (Ph+) CML undergoing SCT from HLA-matched siblings (n = 182) or volunteer-unrelated donors (VUD; n = 132). Three-year overall survival (OS) and leukemia-free survival (LFS) rates were 66% and 55% (n = 314). For 156 children in first chronic phase (CP1) who underwent transplantation from HLA-identical siblings, OS and LFS rates were 75% and 63%. For 97 children who underwent SCT in CP1 from VUD, 3-year OS and LFS rates were 65% and 56%, reflecting higher transplantation-related mortality (TRM) after VUD SCT (35% vs 20%; multivariate hazard ratio [HR], 1.9; 95% confidence interval [CI], 1.0-3.5; P =.05). In a multivariate model for OS and LFS, outcomes were superior in CP1 than in advanced phase (AP/CP1) (OS HR, 2.0; 95% CI, 1.3-3; P =.001; LFS HR, 1.8; 95% CI, 1.2-2.6; P =.003). For relapse, donor source (VUD/sibling) (HR, 0.38; 95% CI, 0.19-0.76; P =.006) and disease stage (AP/CP1) (HR, 2.4; 95% CI, 1.36-4.3; P =.003) were significant. This is the first large series to show that SCT confers long-term LFS in most children with CML and helps assess alternative therapy, including tyrosine kinase inhibitors.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas , Leucemia Mielógena Crónica BCR-ABL Positiva/terapia , Adolescente , Adulto , Niño , Preescolar , Citomegalovirus , Infecciones por Citomegalovirus/sangre , Supervivencia sin Enfermedad , Femenino , Enfermedad Injerto contra Huésped/etiología , Enfermedad Injerto contra Huésped/prevención & control , Humanos , Lactante , Interferón-alfa/uso terapéutico , Leucemia Mielógena Crónica BCR-ABL Positiva/virología , Masculino , Análisis Multivariante , Modelos de Riesgos Proporcionales , Hermanos , Tasa de Supervivencia , Donantes de Tejidos , Resultado del Tratamiento
19.
Am J Obstet Gynecol ; 188(4): 1002-10, 2003 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-12712101

RESUMEN

OBJECTIVE: The purpose of this study was to investigate whether purified CD34(+) cells from first-trimester fetal blood are a source of primitive and committed hemopoietic progenitors. STUDY DESIGN: CD34(+) cells from first-trimester fetal blood and term cord blood were assayed for committed hemopoietic progenitor cells, high proliferative potential colony-forming cells, and long-term culture-initiating cells. RESULTS: First-trimester CD34(+) cells that were compared with cells at term generated fewer hemopoietic progenitor cells and fewer high proliferative potential colony-forming cells with lower recloning efficiency(P <.001). First-trimester CD34(+) cells tended to contain more long-term culture-initiating cells, both in bulk cultures and by limiting dilution analysis. The ratio between committed and primitive progenitors was 3 in the first-trimester and 20 in the term cord blood, respectively. CONCLUSION: First-trimester fetal blood is enriched in primitive (compared with committed) hemopoietic progenitors and may be an advantageous source of stem cells for prenatal therapy.


Asunto(s)
Antígenos CD34/análisis , Células Sanguíneas/citología , Células Sanguíneas/inmunología , Sangre Fetal , Células Madre Hematopoyéticas/citología , Células Cultivadas , Ensayo de Unidades Formadoras de Colonias , Femenino , Humanos , Embarazo , Primer Trimestre del Embarazo , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...