Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 76
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Nat Immunol ; 17(8): 956-65, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27376470

RESUMEN

During T cell development, multipotent progenitors relinquish competence for other fates and commit to the T cell lineage by turning on Bcl11b, which encodes a transcription factor. To clarify lineage commitment mechanisms, we followed developing T cells at the single-cell level using Bcl11b knock-in fluorescent reporter mice. Notch signaling and Notch-activated transcription factors collaborate to activate Bcl11b expression irrespectively of Notch-dependent proliferation. These inputs work via three distinct, asynchronous mechanisms: an early locus 'poising' function dependent on TCF-1 and GATA-3, a stochastic-permissivity function dependent on Notch signaling, and a separate amplitude-control function dependent on Runx1, a factor already present in multipotent progenitors. Despite their necessity for Bcl11b expression, these inputs act in a stage-specific manner, providing a multitiered mechanism for developmental gene regulation.


Asunto(s)
Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Factor de Transcripción GATA3/metabolismo , Regulación del Desarrollo de la Expresión Génica , Factor Nuclear 1-alfa del Hepatocito/metabolismo , Linfopoyesis/genética , Receptores Notch/metabolismo , Proteínas Represoras/metabolismo , Linfocitos T/fisiología , Proteínas Supresoras de Tumor/metabolismo , Animales , Diferenciación Celular/genética , Linaje de la Célula/genética , Rastreo Celular , Células Cultivadas , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Factor de Transcripción GATA3/genética , Factor Nuclear 1-alfa del Hepatocito/genética , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas Represoras/genética , Transducción de Señal , Análisis de la Célula Individual , Proteínas Supresoras de Tumor/genética
2.
Blood ; 140(21): 2261-2275, 2022 11 24.
Artículo en Inglés | MEDLINE | ID: mdl-35605191

RESUMEN

Adoptive transfer of T cells expressing chimeric antigen receptors (CAR-T) effectively treats refractory hematologic malignancies in a subset of patients but can be limited by poor T-cell expansion and persistence in vivo. Less differentiated T-cell states correlate with the capacity of CAR-T to proliferate and mediate antitumor responses, and interventions that limit tumor-specific T-cell differentiation during ex vivo manufacturing enhance efficacy. NOTCH signaling is involved in fate decisions across diverse cell lineages and in memory CD8+ T cells was reported to upregulate the transcription factor FOXM1, attenuate differentiation, and enhance proliferation and antitumor efficacy in vivo. Here, we used a cell-free culture system to provide an agonistic NOTCH1 signal during naïve CD4+ T-cell activation and CAR-T production and studied the effects on differentiation, transcription factor expression, cytokine production, and responses to tumor. NOTCH1 agonism efficiently induced a stem cell memory phenotype in CAR-T derived from naïve but not memory CD4+ T cells and upregulated expression of AhR and c-MAF, driving heightened production of interleukin-22, interleukin-10, and granzyme B. NOTCH1-agonized CD4+ CAR-T demonstrated enhanced antigen responsiveness and proliferated to strikingly higher frequencies in mice bearing human lymphoma xenografts. NOTCH1-agonized CD4+ CAR-T also provided superior help to cotransferred CD8+ CAR-T, driving improved expansion and curative antitumor responses in vivo at low CAR-T doses. Our data expand the mechanisms by which NOTCH can shape CD4+ T-cell behavior and demonstrate that activating NOTCH1 signaling during genetic modification ex vivo is a potential strategy for enhancing the function of T cells engineered with tumor-targeting receptors.


Asunto(s)
Linfoma , Receptores Quiméricos de Antígenos , Humanos , Ratones , Animales , Inmunoterapia Adoptiva , Linfocitos T CD4-Positivos , Factores de Transcripción , Linfoma/tratamiento farmacológico , Receptores de Antígenos de Linfocitos T , Receptor Notch1/genética
3.
Blood ; 130(22): 2373-2376, 2017 11 30.
Artículo en Inglés | MEDLINE | ID: mdl-29021230

RESUMEN

On 1 September 2017, the US Food and Drug Administration (FDA) approved gemtuzumab ozogamicin (GO) for the treatment of adults with newly diagnosed CD33+ acute myeloid leukemia and for patients aged ≥2 years with CD33+ acute myeloid leukemia who have experienced a relapse or who have not responded to initial treatment. This signals a new chapter in the long and unusual story of GO, which was the first antibody-drug conjugate approved for human use by the FDA.


Asunto(s)
Aminoglicósidos/uso terapéutico , Anticuerpos Monoclonales Humanizados/uso terapéutico , Antineoplásicos Inmunológicos/uso terapéutico , Leucemia Mieloide Aguda/tratamiento farmacológico , Aminoglicósidos/administración & dosificación , Aminoglicósidos/efectos adversos , Anticuerpos Monoclonales Humanizados/administración & dosificación , Anticuerpos Monoclonales Humanizados/efectos adversos , Antineoplásicos Inmunológicos/administración & dosificación , Antineoplásicos Inmunológicos/efectos adversos , Ensayos Clínicos como Asunto , Gemtuzumab , Humanos , Metaanálisis como Asunto , Lectina 3 Similar a Ig de Unión al Ácido Siálico/análisis
4.
Development ; 140(18): 3799-808, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23924634

RESUMEN

Genetic regulation of the cell fate transition from lateral plate mesoderm to the specification of cardiomyocytes requires suppression of Wnt/ß-catenin signaling, but the mechanism for this is not well understood. By analyzing gene expression and chromatin dynamics during directed differentiation of human embryonic stem cells (hESCs), we identified a suppressor of Wnt/ß-catenin signaling, transmembrane protein 88 (TMEM88), as a potential regulator of cardiovascular progenitor cell (CVP) specification. During the transition from mesoderm to the CVP, TMEM88 has a chromatin signature of genes that mediate cell fate decisions, and its expression is highly upregulated in advance of key cardiac transcription factors in vitro and in vivo. In early zebrafish embryos, tmem88a is expressed broadly in the lateral plate mesoderm, including the bilateral heart fields. Short hairpin RNA targeting of TMEM88 during hESC cardiac differentiation increases Wnt/ß-catenin signaling, confirming its role as a suppressor of this pathway. TMEM88 knockdown has no effect on NKX2.5 or GATA4 expression, but 80% of genes most highly induced during CVP development have reduced expression, suggesting adoption of a new cell fate. In support of this, analysis of later stage cell differentiation showed that TMEM88 knockdown inhibits cardiomyocyte differentiation and promotes endothelial differentiation. Taken together, TMEM88 is crucial for heart development and acts downstream of GATA factors in the pre-cardiac mesoderm to specify lineage commitment of cardiomyocyte development through inhibition of Wnt/ß-catenin signaling.


Asunto(s)
Proteínas de la Membrana/metabolismo , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Proteínas Wnt/metabolismo , Proteínas de Pez Cebra/metabolismo , Pez Cebra/embriología , Animales , Linaje de la Célula/genética , Regulación hacia Abajo/genética , Embrión no Mamífero/citología , Embrión no Mamífero/metabolismo , Células Endoteliales/citología , Células Endoteliales/metabolismo , Regulación del Desarrollo de la Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Proteínas de la Membrana/genética , Ratones , Modelos Biológicos , Transducción de Señal/genética , Células Madre/citología , Células Madre/metabolismo , Regulación hacia Arriba/genética , Pez Cebra/genética , Proteínas de Pez Cebra/genética , beta Catenina/metabolismo
5.
Blood ; 123(5): 650-8, 2014 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-24243972

RESUMEN

Increasing evidence supports the importance of cell extrinsic regulation in stem cell fate control. Hematopoietic stem cells (HSC) are responsive to local signals from their niche and to systemic feedback from progenitors and mature cells. The Notch ligand Delta-1 (DL1), a key component of the stem cell niche, regulates human hematopoietic lineage development in a dose-dependent manner and has been used clinically for primitive progenitor expansion. How DL1 acts to regulate HSC fate and whether these actions are related to its lineage skewing effects are poorly understood. Here we demonstrate that, although DL1 activates signal transducer and activator of transcription 3 signaling similarly to the gp130-activating cytokine interleukin-6 (IL-6), it has opposite effects on myeloid cell production. Mechanistically, these different outcomes are attributable to a DL1-mediated reduction in membrane (m)-bound IL-6 receptor (R) expression, converting progenitor cells from being directly IL-6 responsive to requiring both IL-6 and soluble (s) IL-6R for activation. Concomitant reduction of both mIL-6R (by DL1 supplementation) and sIL-6R (using dynamically fed cultures) reduced myeloid cell production and led to enhanced outputs of human HSCs. This work describes a new mode of cytokine action in which DL1 changes cytokine receptor distributions on hematopoietic cells, altering feedback networks and their impact on stem cell fate.


Asunto(s)
Células Madre Hematopoyéticas/citología , Interleucina-6/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas de la Membrana/metabolismo , Comunicación Paracrina , Animales , Células Cultivadas , Femenino , Hematopoyesis , Células Madre Hematopoyéticas/metabolismo , Humanos , Quinasas Janus/metabolismo , Ratones , Ratones SCID , Factor de Transcripción STAT3/metabolismo
6.
Blood ; 121(5): 717-8, 2013 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-23372149

RESUMEN

In this issue of Blood, Rafii et al present an elegant study of human embryonic stem cell (ESC)­derived hematopoiesis incorporating live imaging at the single-cell level to track hematopoietic lineage potential during the endothelial to hematopoietic transition.


Asunto(s)
Diferenciación Celular , Células Madre Embrionarias/metabolismo , Células Endoteliales/metabolismo , Células Madre Hematopoyéticas/metabolismo , Transducción Genética , Humanos
7.
Blood ; 122(7): 1162-73, 2013 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-23733337

RESUMEN

Notch signaling regulates several cellular processes including cell fate decisions and proliferation in both invertebrates and mice. However, comparatively less is known about the role of Notch during early human development. Here, we examined the function of Notch signaling during hematopoietic lineage specification from human pluripotent stem cells of both embryonic and adult fibroblast origin. Using immobilized Notch ligands and small interfering RNA to Notch receptors we have demonstrated that Notch1, but not Notch2, activation induced hairy and enhancer of split 1 (HES1) expression and generation of committed hematopoietic progenitors. Using gain- and loss-of-function approaches, this was shown to be attributed to Notch-signaling regulation through HES1, which dictated cell fate decisions from bipotent precursors either to the endothelial or hematopoietic lineages at the clonal level. Our study reveals a previously unappreciated role for the Notch pathway during early human hematopoiesis, whereby Notch signaling via HES1 represents a toggle switch of hematopoietic vs endothelial fate specification.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Células Madre Embrionarias/citología , Endotelio Vascular/citología , Hematopoyesis/fisiología , Células Madre Hematopoyéticas/citología , Proteínas de Homeodominio/metabolismo , Células Madre Pluripotentes Inducidas/citología , Receptor Notch1/metabolismo , Apoptosis , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/antagonistas & inhibidores , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Biomarcadores/metabolismo , Western Blotting , Diferenciación Celular , Movimiento Celular , Proliferación Celular , Células Cultivadas , Dermis/citología , Dermis/metabolismo , Células Madre Embrionarias/metabolismo , Endotelio Vascular/metabolismo , Fibroblastos/citología , Fibroblastos/metabolismo , Citometría de Flujo , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Células Madre Hematopoyéticas/metabolismo , Proteínas de Homeodominio/antagonistas & inhibidores , Proteínas de Homeodominio/genética , Humanos , Técnicas para Inmunoenzimas , Células Madre Pluripotentes Inducidas/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , ARN Interferente Pequeño/genética , Receptor Notch1/antagonistas & inhibidores , Receptor Notch1/genética , Receptores Notch/metabolismo , Transducción de Señal , Factor de Transcripción HES-1
8.
Blood ; 122(8): 1455-63, 2013 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-23770776

RESUMEN

Outcomes in acute myeloid leukemia (AML) remain unsatisfactory, and novel treatments are urgently needed. One strategy explores antibodies and their drug conjugates, particularly those targeting CD33. Emerging data with gemtuzumab ozogamicin (GO) demonstrate target validity and activity in some patients with AML, but efficacy is limited by heterogeneous drug conjugation, linker instability, and a high incidence of multidrug resistance. We describe here the development of SGN-CD33A, a humanized anti-CD33 antibody with engineered cysteines conjugated to a highly potent, synthetic DNA cross-linking pyrrolobenzodiazepine dimer via a protease-cleavable linker. The use of engineered cysteine residues at the sites of drug linker attachment results in a drug loading of approximately 2 pyrrolobenzodiazepine dimers per antibody. In preclinical testing, SGN-CD33A is more potent than GO against a panel of AML cell lines and primary AML cells in vitro and in xenotransplantation studies in mice. Unlike GO, antileukemic activity is observed with SGN-CD33A in AML models with the multidrug-resistant phenotype. Mechanistic studies indicate that the cytotoxic effects of SGN-CD33A involve DNA damage with ensuing cell cycle arrest and apoptotic cell death. Together, these data suggest that SGN-CD33A has CD33-directed antitumor activity and support clinical testing of this novel therapeutic in patients with AML.


Asunto(s)
Anticuerpos Monoclonales Humanizados/química , Benzodiazepinas/química , Resistencia a Antineoplásicos , Inmunoconjugados/química , Leucemia Mieloide Aguda/tratamiento farmacológico , Lectina 3 Similar a Ig de Unión al Ácido Siálico/química , Animales , Apoptosis , Ciclo Celular , Reactivos de Enlaces Cruzados/química , Reactivos de Enlaces Cruzados/farmacología , Cisteína/genética , Dimerización , Diseño de Fármacos , Células HEK293 , Células HL-60 , Humanos , Leucemia Mieloide Aguda/inmunología , Ratones
9.
Cytotherapy ; 17(5): 579-92, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25559145

RESUMEN

BACKGROUND AIMS: Human cord blood (CB) is enriched in circulating endothelial colony forming cells (ECFCs) that display high proliferative potential and in vivo vessel forming ability. Because Notch signaling is critical for embryonic blood vessel formation in utero, we hypothesized that Notch pathway activation may enhance cultured ECFC vasculogenic properties in vivo. METHODS: In vitro ECFC stimulation with an immobilized chimeric Notch ligand (Delta-like1(ext-IgG)) led to significant increases in the mRNA and protein levels of Notch regulated Hey2 and EphrinB2 that were blocked by treatment with γ-secretase inhibitor addition. However, Notch stimulated preconditioning in vitro failed to enhance ECFC vasculogenesis in vivo. In contrast, in vivo co-implantation of ECFCs with OP9-Delta-like 1 stromal cells that constitutively expressed the Notch ligand delta-like 1 resulted in enhanced Notch activated ECFC-derived increased vessel density and enlarged vessel area in vivo, an effect not induced by OP9 control stromal implantation. RESULTS: This Notch activation was associated with diminished apoptosis in the exposed ECFC. CONCLUSIONS: We conclude that Notch pathway activation in ECFC in vivo via co-implanted stromal cells expressing delta-like 1 promotes vasculogenesis and augments blood vessel formation via diminishing apoptosis of the implanted ECFC.


Asunto(s)
Células Endoteliales/citología , Sangre Fetal/citología , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Proteínas de la Membrana/metabolismo , Neovascularización Fisiológica , Receptores Notch/metabolismo , Animales , Proteínas de Unión al Calcio , Células Cultivadas , Colágeno/farmacología , Ensayo de Unidades Formadoras de Colonias , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Humanos , Recién Nacido , Ligandos , Ratones Endogámicos NOD , Ratones SCID , Neovascularización Fisiológica/efectos de los fármacos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Células del Estroma/citología , Células del Estroma/metabolismo
10.
Blood ; 119(26): 6198-208, 2012 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-22286199

RESUMEN

Although the identification of cancer stem cells as therapeutic targets is now actively being pursued in many human malignancies, the leukemic stem cells in acute myeloid leukemia (AML) are a paradigm of such a strategy. Heterogeneity of these cells was suggested by clonal analyses indicating the existence of both leukemias resulting from transformed multipotent CD33(-) stem cells as well others arising from, or predominantly involving, committed CD33(+) myeloid precursors. The latter leukemias, which may be associated with an intrinsically better prognosis, offer a particularly attractive target for stem cell-directed therapies. Targeting the CD33 differentiation antigen with gemtuzumab ozogamicin was the first attempt of such an approach. Emerging clinical data indicate that gemtuzumab ozogamicin is efficacious not only for acute promyelocytic leukemia but, in combination with conventional chemotherapy, also for other favorable- and intermediate-risk AMLs, providing the first proof-of-principle evidence for the validity of this strategy. Herein, we review studies on the nature of stem cells in AML, discuss clinical data on the effectiveness of CD33-directed therapy, and consider the mechanistic basis for success and failure in various AML subsets.


Asunto(s)
Antígenos CD/inmunología , Antígenos de Diferenciación Mielomonocítica/inmunología , Inmunoterapia/métodos , Leucemia Mieloide Aguda/terapia , Células Madre Neoplásicas/inmunología , Antígenos CD/metabolismo , Antígenos de Diferenciación Mielomonocítica/metabolismo , Ensayos Clínicos como Asunto/estadística & datos numéricos , Humanos , Leucemia Mieloide Aguda/patología , Modelos Biológicos , Terapia Molecular Dirigida/métodos , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Lectina 3 Similar a Ig de Unión al Ácido Siálico , Resultado del Tratamiento
11.
Blood ; 119(8): 1856-60, 2012 Feb 23.
Artículo en Inglés | MEDLINE | ID: mdl-22219225

RESUMEN

SIRT1 is an NAD(+)-dependent histone deacetylase implicated in the establishment of the primitive hematopoietic system during mouse embryonic development. However, investigation of the role of SIRT1 in adult hematopoiesis has been complicated by the high perinatal mortality of SIRT1-deficient mice (SIRT1(-/-)). We performed a comprehensive in vivo study of the hematopoietic stem cell (HSC) compartment in adult SIRT1(-/-) mice and show that, apart from anemia and leukocytosis in older mice, the production of mature blood cells, lineage distribution within hematopoietic organs, and frequencies of the most primitive HSC populations are comparable to those of wild-type littermate controls. Furthermore, we show that SIRT1-deficient BM cells confer stable long-term reconstitution in competitive repopulation and serial transplantation experiments. The results of the present study rule out an essential physiologic role for cell-autonomous SIRT1 signaling in the maintenance of the adult HSC compartment in mice.


Asunto(s)
Hematopoyesis/fisiología , Células Madre Hematopoyéticas/fisiología , Sirtuina 1/fisiología , Factores de Edad , Animales , Antígenos CD/metabolismo , Antígenos Ly/metabolismo , Recuento de Células Sanguíneas , Células de la Médula Ósea/metabolismo , Trasplante de Médula Ósea , Femenino , Citometría de Flujo , Hematopoyesis/genética , Células Madre Hematopoyéticas/metabolismo , Inmunofenotipificación , Masculino , Proteínas de la Membrana/metabolismo , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Proto-Oncogénicas c-kit/metabolismo , Receptores de Superficie Celular/metabolismo , Miembro 1 de la Familia de Moléculas Señalizadoras de la Activación Linfocitaria , Sirtuina 1/deficiencia , Sirtuina 1/genética , Factores de Tiempo
12.
Blood ; 119(16): 3705-11, 2012 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-22378848

RESUMEN

CD33 is expressed on the majority of acute myeloid leukemia (AML) leukemic blasts and is the target for gemtuzumab ozogamicin (GO), a toxin-conjugated anti-CD33 mAb. In the present study, we quantified the CD33 mean fluorescent intensity of leukemic blasts prospectively in 619 de novo pediatric AML patients enrolled in Children's Oncology Group GO-containing clinical trials and determined its correlation with disease characteristics and clinical outcome. CD33 expression varied more than 2-log fold; a median mean fluorescent intensity of 129 (range, 3-1550.07) was observed. Patients were divided into 4 quartiles, quartiles 1-4 (Q1-4) based on expression and disease characteristics and clinical response defined across quartiles. High CD33 expression was associated with high-risk FLT3/ITD mutations (P < .001) and was inversely associated with low-risk disease (P < .001). Complete remission (CR) rates were similar, but patients in Q4 had significantly lower overall survival (57% ± 16% vs 77% ± 7%, P = .002) and disease-free survival from CR (44% ± 16% vs 62% ± 8%, P = .022). In a multivariate model, high CD33 expression remained a significant predictor of overall survival (P = .011) and disease-free survival (P = .038) from CR. Our findings suggest that CD33 expression is heterogeneous within de novo pediatric AML. High expression is associated with adverse disease features and is an independent predictor of inferior outcome. The correlation between CD33 expression and GO response is under investigation. These studies are registered at www.clinicaltrials.gov as NCT00070174 and NCT00372593.


Asunto(s)
Aminoglicósidos/administración & dosificación , Anticuerpos Monoclonales Humanizados/administración & dosificación , Antígenos CD/metabolismo , Antígenos de Diferenciación Mielomonocítica/metabolismo , Antineoplásicos/administración & dosificación , Leucemia Mieloide Aguda/tratamiento farmacológico , Antígenos CD/inmunología , Antígenos de Diferenciación Mielomonocítica/inmunología , Niño , Preescolar , Monitoreo de Drogas/métodos , Femenino , Gemtuzumab , Humanos , Lactante , Leucemia Mieloide Aguda/epidemiología , Leucemia Mieloide Aguda/inmunología , Masculino , Modelos de Riesgos Proporcionales , Estudios Prospectivos , Factores de Riesgo , Lectina 3 Similar a Ig de Unión al Ácido Siálico , Resultado del Tratamiento , Adulto Joven
13.
Blood ; 117(23): 6083-90, 2011 Jun 09.
Artículo en Inglés | MEDLINE | ID: mdl-21436068

RESUMEN

Despite progress in our understanding of the growth factors that support the progressive maturation of the various cell lineages of the hematopoietic system, less is known about factors that govern the self-renewal of hematopoietic stem and progenitor cells (HSPCs), and our ability to expand human HSPC numbers ex vivo remains limited. Interest in stem cell expansion has been heightened by the increasing importance of HSCs in the treatment of both malignant and nonmalignant diseases, as well as their use in gene therapy. To date, most attempts to ex vivo expand HSPCs have used hematopoietic growth factors but have not achieved clinically relevant effects. More recent approaches, including our studies in which activation of the Notch signaling pathway has enabled a clinically relevant ex vivo expansion of HSPCs, have led to renewed interest in this arena. Here we briefly review early attempts at ex vivo expansion by cytokine stimulation followed by an examination of our studies investigating the role of Notch signaling in HSPC self-renewal. We will also review other recently developed approaches for ex vivo expansion, primarily focused on the more extensively studied cord blood-derived stem cell. Finally, we discuss some of the challenges still facing this field.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Citocinas/farmacología , Sangre Fetal/citología , Células Madre Hematopoyéticas/citología , Sangre Fetal/metabolismo , Células Madre Hematopoyéticas/metabolismo , Humanos , Receptores Notch/metabolismo , Transducción de Señal/efectos de los fármacos
14.
Blood ; 118(7): 1784-96, 2011 Aug 18.
Artículo en Inglés | MEDLINE | ID: mdl-21715312

RESUMEN

The mechanisms regulating the emergence of BM prothymocytes remain poorly characterized. Genome-wide transcriptome analyses looking for genes expressed in human prothymocytes led to the identification of AF1q/MLLT11 as a candidate gene conceivably involved in this process. Analysis of AF1q protein subcellular localization and intracellular trafficking showed that despite pronounced karyophily, it was subjected to constitutive nuclear export followed by ubiquitin-mediated degradation in the centrosomal area. Using in vitro assays based on either forced expression or shRNA-mediated silencing of AF1q, we provide evidence that the protein promotes T- over B-cell differentiation in multipotent hematopoietic progenitors. At the molecular level, AF1q confers to multipotent progenitors an increased susceptibility to Delta-like/Notch-mediated signaling. Consistent with these findings, enforced AF1q expression in humanized mice fosters the emergence of BM CD34(+)CD7(+) prothymocytes, enhances subsequent thymus colonization, and accelerates intrathymic T-cell development. In contrast, AF1q silencing provokes a global shift of BM lymphopoiesis toward the B-cell lineage, hinders prothymocyte development, inhibits thymus colonization, and leads to intrathymic accumulation of B cells. Our results indicate that AF1q cooperates with the Notch signaling pathway to foster the emergence of BM prothymocytes and drive subsequent intrathymic specification toward the T-cell lineage.


Asunto(s)
Células Madre Hematopoyéticas/citología , Linfopoyesis , Proteínas de Neoplasias/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Receptores Notch/metabolismo , Linfocitos T/citología , Transporte Activo de Núcleo Celular , Secuencia de Aminoácidos , Animales , Linfocitos B/citología , Linfocitos B/metabolismo , Células Cultivadas , Silenciador del Gen , Células HeLa , Células Madre Hematopoyéticas/metabolismo , Humanos , Ratones , Ratones SCID , Datos de Secuencia Molecular , Proteínas de Neoplasias/genética , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteínas Proto-Oncogénicas/genética , Alineación de Secuencia , Transducción de Señal , Linfocitos T/metabolismo
15.
Blood ; 117(17): 4449-59, 2011 Apr 28.
Artículo en Inglés | MEDLINE | ID: mdl-21372153

RESUMEN

Notch signaling critically mediates various hematopoietic lineage decisions and is induced in mammals by Notch ligands that are classified into 2 families, Delta-like (Delta-like-1, -3 and -4) and Jagged (Jagged1 and Jagged2), based on structural homology with both Drosophila ligands Delta and Serrate, respectively. Because the functional differences between mammalian Notch ligands were still unclear, we have investigated their influence on early human hematopoiesis and show that Jagged2 affects hematopoietic lineage decisions very similarly as Delta-like-1 and -4, but very different from Jagged1. OP9 coculture experiments revealed that Jagged2, like Delta-like ligands, induces T-lineage differentiation and inhibits B-cell and myeloid development. However, dose-dependent Notch activation studies, gene expression analysis, and promoter activation assays indicated that Jagged2 is a weaker Notch1-activator compared with the Delta-like ligands, revealing a Notch1 specific signal strength hierarchy for mammalian Notch ligands. Strikingly, Lunatic-Fringe- mediated glycosylation of Notch1 potentiated Notch signaling through Delta-like ligands and also Jagged2, in contrast to Jagged1. Thus, our results reveal a unique role for Jagged1 in preventing the induction of T-lineage differentiation in hematopoietic stem cells and show an unexpected functional similarity between Jagged2 and the Delta-like ligands.


Asunto(s)
Hematopoyesis/fisiología , Células Madre Hematopoyéticas/citología , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Animales , Linfocitos B/citología , Linfocitos B/metabolismo , Proteínas de Unión al Calcio/metabolismo , Diferenciación Celular/fisiología , Células Cultivadas , Proteínas de Drosophila , Glicosilación , Glicosiltransferasas/metabolismo , Células Madre Hematopoyéticas/metabolismo , Humanos , Proteína Jagged-1 , Proteína Jagged-2 , Receptor Notch1/metabolismo , Proteínas Serrate-Jagged , Transducción de Señal/fisiología , Linfocitos T/citología , Linfocitos T/metabolismo
16.
Blood ; 118(17): 4561-6, 2011 Oct 27.
Artículo en Inglés | MEDLINE | ID: mdl-21873548

RESUMEN

IDH1 SNP rs11554137 was recently reported in association with poor prognosis in normal karyotype adult acute myeloid leukemia (AML). We aimed to determine the prevalence, clinical associations, and prognostic significance of SNP rs11554137 in unselected pediatric and adult AML patients. Diagnostic marrow specimens from 527 AML patients treated on the pediatric trial Children's Oncology Group-AAML03P1 (N = 253) or adult SWOG trials (N = 274) were analyzed for the presence of the SNP. SNP rs11554137 was present in 11% of all patients. SNP status had no prognostic impact on survival in pediatric patients. In adult AML, overall survival for SNP-positive patients was 10% versus 18% for SNP-negative patients (P = .44). Among the 142 adults who achieved complete remission, 5-year relapse-free survival was significantly worse for SNP-positive patients (0% vs 25%, P = .0014). However, among adults with normal cytogenetics, FLT3/ITD was present in 90% of SNP-positive patients versus 59% of SNP-negative patients (P = .0053). In multivariate analysis, adjusting for the effects of age, cytogenetics, and FLT3/ITD, the independent prognostic effect of SNP positivity was not statistically significant (hazard ratio = 1.72, P = .18). The clinical profile of SNP-positive patients suggests that SNP rs11554137 may have biologic effects that bear further investigation. The clinical trials in this study are registered at http://www.clinicaltrials.gov as #NCT000707174 and #NCT00899171.


Asunto(s)
Isocitrato Deshidrogenasa/genética , Leucemia Mieloide Aguda/diagnóstico , Polimorfismo de Nucleótido Simple/fisiología , Adolescente , Adulto , Edad de Inicio , Niño , Preescolar , Ensayos Clínicos como Asunto , Femenino , Humanos , Lactante , Recién Nacido , Isocitrato Deshidrogenasa/fisiología , Leucemia Mieloide Aguda/epidemiología , Leucemia Mieloide Aguda/genética , Masculino , Oncología Médica/organización & administración , Persona de Mediana Edad , Mutación Missense/fisiología , Pronóstico , Sociedades Médicas , Adulto Joven
17.
Stem Cells ; 30(10): 2212-20, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22865615

RESUMEN

Transplantation of myogenic stem cells possesses great potential for long-term repair of dystrophic muscle. However, a single donor muscle biopsy is unlikely to provide enough cells to effectively transplant the muscle mass of a patient affected by muscular dystrophy. Expansion of cells ex vivo using traditional culture techniques significantly reduces engraftment potential. We hypothesized that activation of Notch signaling during ex vivo expansion would maintain donor cell engraftment potential. In this study, we expanded freshly isolated canine muscle-derived cells on tissue culture plates coated with Delta-1(ext) -IgG to activate Notch signaling or with human IgG as a control. A model of canine-to-murine xenotransplantation was used to quantitatively compare canine muscle cell engraftment and determine whether engrafted donor cells could function as satellite cells in vivo. We show that Delta-1(ext) -IgG inhibited differentiation of canine muscle-derived cells and increased the level of genes normally expressed in myogenic precursors. Moreover, cells expanded on Delta-1(ext) -IgG resulted in a significant increase in the number of donor-derived fibers, as compared to cells expanded on human IgG, reaching engraftment levels similar to freshly isolated cells. Importantly, cells expanded on Delta-1(ext) -IgG engrafted to the recipient satellite cell niche and contributed to further regeneration. A similar strategy of expanding human muscle-derived cells on Notch ligand might facilitate engraftment and muscle regeneration for patients affected with muscular dystrophy.


Asunto(s)
Supervivencia de Injerto , Células Musculares/metabolismo , Músculo Esquelético/metabolismo , Receptores Notch/metabolismo , Células Madre/metabolismo , Animales , Comunicación Celular , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Perros , Humanos , Inmunoglobulina G/farmacología , Ratones , Ratones SCID , Células Musculares/citología , Células Musculares/trasplante , Músculo Esquelético/citología , Regeneración , Transducción de Señal , Especificidad de la Especie , Trasplante de Células Madre , Células Madre/citología , Trasplante Heterólogo
18.
bioRxiv ; 2023 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-37333272

RESUMEN

Decoding the gene regulatory mechanisms mediating self-renewal of hematopoietic stem cells (HSCs) during their amplification in the fetal liver (FL) is relevant for advancing therapeutic applications aiming to expand transplantable HSCs, a long-standing challenge. Here, to explore intrinsic and extrinsic regulation of self-renewal in FL-HSCs at the single cell level, we engineered a culture platform designed to recapitulate the FL endothelial niche, which supports the amplification of serially engraftable HSCs ex vivo. Leveraging this platform in combination with single cell index flow cytometry, serial transplantation assays, and single cell RNA-sequencing, we elucidated previously unrecognized heterogeneity in immunophenotypically defined FL-HSCs and demonstrated that differentiation latency and transcriptional signatures of biosynthetic dormancy are distinguishing properties of self-renewing FL-HSCs with capacity for serial, long-term multilineage hematopoietic reconstitution. Altogether, our findings provide key insights into HSC expansion and generate a novel resource for future exploration of the intrinsic and niche-derived signaling pathways that support FL-HSC self-renewal.

19.
Blood ; 116(26): 5859-66, 2010 Dec 23.
Artículo en Inglés | MEDLINE | ID: mdl-20921340

RESUMEN

Umbilical cord blood (UCB) is an attractive cell source for hematopoietic cell transplantation (HCT). Here we examine whether the combination of homeobox B4 (HOXB4) and Delta-1 ligand (DL) synergize when used together. Monkey and human UCB CD34(+) cells were transduced with a HOXB4-expressing gammaretroviral vector and cultured with DL. Individual and combined effects of HOXB4 and DL were assessed by colony-forming unit assays, flow cytometry, and nonobese diabetic/severe combined immune deficienct mouse transplantation. The presence of DL yielded higher percentage of CD34(+) and CD7(+) cells and lower percentages of CD14(+) cells than non-DL cultures. Furthermore, HOXB4 yielded higher percentages of CD34(+) and CD14(+) cells than non-HOXB4 cultures. Interestingly, coculture with DL-expressing OP9 cells resulted in better maintenance of HOXB4 than culture in DL-conditioned medium. Culture of HOXB4-transduced human cells in the presence of DL yielded enhanced generation of repopulating cells with higher levels of engraftment of human CD45(+), CD34(+), CD3(+), CD20(+), and CD41(+) cells compared with either factor individually. Our results demonstrate enhanced generation of hematopoietic progenitors by combining HOXB4 and DL; addition of DL further enhances expansion of multipotent cells capable of repopulating lymphoid and megakaryocyte lineages, which is not observed with HOXB4 alone.


Asunto(s)
Sangre Fetal/citología , Sangre Fetal/trasplante , Proteínas de Homeodominio/fisiología , Proteínas de la Membrana/fisiología , Factores de Transcripción/fisiología , Animales , Células Cultivadas , Ensayo de Unidades Formadoras de Colonias , Medios de Cultivo Condicionados/farmacología , Sangre Fetal/metabolismo , Citometría de Flujo , Humanos , Péptidos y Proteínas de Señalización Intracelular , Linfocitos/metabolismo , Macaca nemestrina , Ratones , Ratones Endogámicos NOD , Ratones SCID , Células Mieloides/metabolismo , Fenotipo
20.
Blood ; 115(4): 757-65, 2010 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-19822900

RESUMEN

We investigated the potential role of an immune reaction in mediating the dominant engraftment of 1 cord blood unit in 14 patients who received a double-unit cord blood transplantation (CBT). In 10 patients, dominant engraftment of a single donor unit emerged by day 28 after CBT. In 9 of these 10 patients, a significant subset of CD8(+) CD45RO(+/-)CCR7(-) T cells, present in peripheral blood mononuclear cells and derived from the engrafting cord blood unit, produced interferon-gamma (IFN-gamma) in response to the nonengrafting unit. No significant population of IFN-gamma-secreting cells was detectable when posttransplantation peripheral blood mononuclear cells were stimulated against cells from the engrafted unit (P < .001) or from a random human leukocyte antigen disparate third party (P = .003). Three patients maintained persistent mixed chimerism after CBT, and no significant IFN-gamma-secreting cells were detected after similar stimulations in these patients (P < .005). Our data provide the first direct evidence in human double-unit CBT recipients that immune rejection mediated by effector CD8(+) T cells developing after CBT from naive precursors is responsible for the failure of 1 unit to engraft. Future investigations based on these findings may result in strategies to predict a dominant unit and enhance graft-versus-leukemia effect.


Asunto(s)
Linfocitos T CD8-positivos/citología , Trasplante de Células Madre de Sangre del Cordón Umbilical/métodos , Rechazo de Injerto/inmunología , Efecto Injerto vs Leucemia/inmunología , Células Madre Hematopoyéticas/citología , Leucemia/terapia , Adolescente , Adulto , Anciano , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Diferenciación Celular/inmunología , Citometría de Flujo , Hematopoyesis , Humanos , Interferón gamma/metabolismo , Leucemia/inmunología , Leucemia Mielógena Crónica BCR-ABL Positiva/inmunología , Leucemia Mielógena Crónica BCR-ABL Positiva/terapia , Leucemia Mieloide Aguda/inmunología , Leucemia Mieloide Aguda/terapia , Persona de Mediana Edad , Leucemia-Linfoma Linfoblástico de Células Precursoras/inmunología , Leucemia-Linfoma Linfoblástico de Células Precursoras/terapia , Quimera por Trasplante/inmunología , Acondicionamiento Pretrasplante , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA