Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Blood Cancer Discov ; 4(3): 180-207, 2023 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-36763002

RESUMEN

Acute myeloid leukemia (AML) is fueled by leukemic stem cells (LSC) whose determinants are challenging to discern from hematopoietic stem cells (HSC) or uncover by approaches focused on general cell properties. We have identified a set of RNA-binding proteins (RBP) selectively enriched in human AML LSCs. Using an in vivo two-step CRISPR-Cas9 screen to assay stem cell functionality, we found 32 RBPs essential for LSCs in MLL-AF9;NrasG12D AML. Loss-of-function approaches targeting key hit RBP ELAVL1 compromised LSC-driven in vivo leukemic reconstitution, and selectively depleted primitive malignant versus healthy cells. Integrative multiomics revealed differentiation, splicing, and mitochondrial metabolism as key features defining the leukemic ELAVL1-mRNA interactome with mitochondrial import protein, TOMM34, being a direct ELAVL1-stabilized target whose repression impairs AML propagation. Altogether, using a stem cell-adapted in vivo CRISPR screen, this work demonstrates pervasive reliance on RBPs as regulators of LSCs and highlights their potential as therapeutic targets in AML. SIGNIFICANCE: LSC-targeted therapies remain a significant unmet need in AML. We developed a stem-cell-adapted in vivo CRISPR screen to identify key LSC drivers. We uncover widespread RNA-binding protein dependencies in LSCs, including ELAVL1, which we identify as a novel therapeutic vulnerability through its regulation of mitochondrial metabolism. This article is highlighted in the In This Issue feature, p. 171.


Asunto(s)
Leucemia Mieloide Aguda , Humanos , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/tratamiento farmacológico , Diferenciación Celular , Células Madre Hematopoyéticas/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Proteínas de Unión al ARN/uso terapéutico , Proteínas del Complejo de Importación de Proteínas Precursoras Mitocondriales , Proteína 1 Similar a ELAV/genética , Proteína 1 Similar a ELAV/metabolismo
3.
Nat Immunol ; 22(6): 723-734, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33958784

RESUMEN

Continuous supply of immune cells throughout life relies on the delicate balance in the hematopoietic stem cell (HSC) pool between long-term maintenance and meeting the demands of both normal blood production and unexpected stress conditions. Here we identified distinct subsets of human long-term (LT)-HSCs that responded differently to regeneration-mediated stress: an immune checkpoint ligand CD112lo subset that exhibited a transient engraftment restraint (termed latency) before contributing to hematopoietic reconstitution and a primed CD112hi subset that responded rapidly. This functional heterogeneity and CD112 expression are regulated by INKA1 through direct interaction with PAK4 and SIRT1, inducing epigenetic changes and defining an alternative state of LT-HSC quiescence that serves to preserve self-renewal and regenerative capacity upon regeneration-mediated stress. Collectively, our data uncovered the molecular intricacies underlying HSC heterogeneity and self-renewal regulation and point to latency as an orchestrated physiological response that balances blood cell demands with preserving a stem cell reservoir.


Asunto(s)
Autorrenovación de las Células/inmunología , Células Madre Hematopoyéticas/fisiología , Reconstitución Inmune , Células Madre Multipotentes/fisiología , Estrés Fisiológico/inmunología , Adulto , Animales , Autorrenovación de las Células/genética , Células Cultivadas , Epigénesis Genética/inmunología , Femenino , Sangre Fetal/citología , Citometría de Flujo , Técnicas de Silenciamiento del Gen , Hematopoyesis , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Humanos , Separación Inmunomagnética , Recién Nacido , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Masculino , Ratones , Persona de Mediana Edad , Nectinas/metabolismo , Cultivo Primario de Células , RNA-Seq , Análisis de la Célula Individual , Sirtuina 1/metabolismo , Estrés Fisiológico/genética , Trasplante Heterólogo , Quinasas p21 Activadas/genética , Quinasas p21 Activadas/metabolismo
4.
Cell Stem Cell ; 28(3): 488-501.e10, 2021 03 04.
Artículo en Inglés | MEDLINE | ID: mdl-33242413

RESUMEN

Lifelong blood production requires long-term hematopoietic stem cells (LT-HSCs), marked by stemness states involving quiescence and self-renewal, to transition into activated short-term HSCs (ST-HSCs) with reduced stemness. As few transcriptional changes underlie this transition, we used single-cell and bulk assay for transposase-accessible chromatin sequencing (ATAC-seq) on human HSCs and hematopoietic stem and progenitor cell (HSPC) subsets to uncover chromatin accessibility signatures, one including LT-HSCs (LT/HSPC signature) and another excluding LT-HSCs (activated HSPC [Act/HSPC] signature). These signatures inversely correlated during early hematopoietic commitment and differentiation. The Act/HSPC signature contains CCCTC-binding factor (CTCF) binding sites mediating 351 chromatin interactions engaged in ST-HSCs, but not LT-HSCs, enclosing multiple stemness pathway genes active in LT-HSCs and repressed in ST-HSCs. CTCF silencing derepressed stemness genes, restraining quiescent LT-HSCs from transitioning to activated ST-HSCs. Hence, 3D chromatin interactions centrally mediated by CTCF endow a gatekeeper function that governs the earliest fate transitions HSCs make by coordinating disparate stemness pathways linked to quiescence and self-renewal.


Asunto(s)
Cromatina , Células Madre Hematopoyéticas , Diferenciación Celular , División Celular , Hematopoyesis , Humanos
5.
JCI Insight ; 5(4)2020 02 27.
Artículo en Inglés | MEDLINE | ID: mdl-31990679

RESUMEN

Inherited bone marrow failure syndromes, such as Fanconi anemia (FA) and Shwachman-Diamond syndrome (SDS), feature progressive cytopenia and a risk of acute myeloid leukemia (AML). Using deep phenotypic analysis of early progenitors in FA/SDS bone marrow samples, we revealed selective survival of progenitors that phenotypically resembled granulocyte-monocyte progenitors (GMP). Whole-exome and targeted sequencing of GMP-like cells in leukemia-free patients revealed a higher mutation load than in healthy controls and molecular changes that are characteristic of AML: increased G>A/C>T variants, decreased A>G/T>C variants, increased trinucleotide mutations at Xp(C>T)pT, and decreased mutation rates at Xp(C>T)pG sites compared with other Xp(C>T)pX sites and enrichment for Cancer Signature 1 (X indicates any nucleotide). Potential preleukemic targets in the GMP-like cells from patients with FA/SDS included SYNE1, DST, HUWE1, LRP2, NOTCH2, and TP53. Serial analysis of GMPs from an SDS patient who progressed to leukemia revealed a gradual increase in mutational burden, enrichment of G>A/C>T signature, and emergence of new clones. Interestingly, the molecular signature of marrow cells from 2 FA/SDS patients with leukemia was similar to that of FA/SDS patients without transformation. The predicted founding clones in SDS-derived AML harbored mutations in several genes, including TP53, while in FA-derived AML the mutated genes included ARID1B and SFPQ. We describe an architectural change in the hematopoietic hierarchy of FA/SDS with remarkable preservation of GMP-like populations harboring unique mutation signatures. GMP-like cells might represent a cellular reservoir for clonal evolution.


Asunto(s)
Trastornos de Fallo de la Médula Ósea/patología , Células Madre Hematopoyéticas/patología , Modelos Genéticos , Trastornos de Fallo de la Médula Ósea/genética , Evolución Clonal , Humanos , Leucemia Mieloide Aguda/genética , Mutación , Síndromes Mielodisplásicos/genética
6.
Blood ; 133(20): 2198-2211, 2019 05 16.
Artículo en Inglés | MEDLINE | ID: mdl-30796022

RESUMEN

There is a growing body of evidence that the molecular properties of leukemia stem cells (LSCs) are associated with clinical outcomes in acute myeloid leukemia (AML), and LSCs have been linked to therapy failure and relapse. Thus, a better understanding of the molecular mechanisms that contribute to the persistence and regenerative potential of LSCs is expected to result in the development of more effective therapies. We therefore interrogated functionally validated data sets of LSC-specific genes together with their known protein interactors and selected 64 candidates for a competitive in vivo gain-of-function screen to identify genes that enhanced stemness in human cord blood hematopoietic stem and progenitor cells. A consistent effect observed for the top hits was the ability to restrain early repopulation kinetics while preserving regenerative potential. Overexpression (OE) of the most promising candidate, the orphan gene C3orf54/INKA1, in a patient-derived AML model (8227) promoted the retention of LSCs in a primitive state manifested by relative expansion of CD34+ cells, accumulation of cells in G0, and reduced output of differentiated progeny. Despite delayed early repopulation, at later times, INKA1-OE resulted in the expansion of self-renewing LSCs. In contrast, INKA1 silencing in primary AML reduced regenerative potential. Mechanistically, our multidimensional confocal analysis found that INKA1 regulates G0 exit by interfering with nuclear localization of its target PAK4, with concomitant reduction of global H4K16ac levels. These data identify INKA1 as a novel regulator of LSC latency and reveal a link between the regulation of stem cell kinetics and pool size during regeneration.


Asunto(s)
Regulación Leucémica de la Expresión Génica , Péptidos y Proteínas de Señalización Intracelular/genética , Leucemia Mieloide Aguda/genética , Células Madre Neoplásicas/metabolismo , Animales , Puntos de Control del Ciclo Celular , Línea Celular Tumoral , Femenino , Humanos , Leucemia Mieloide Aguda/patología , Masculino , Ratones Endogámicos NOD , Células Madre Neoplásicas/citología , Células Madre Neoplásicas/patología , Regulación hacia Arriba , Quinasas p21 Activadas/análisis
8.
Cancer Cell ; 29(2): 186-200, 2016 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-26859458

RESUMEN

Chromosomal rearrangements are a hallmark of acute lymphoblastic leukemia (ALL) and are important ALL initiating events. We describe four different rearrangements of the erythropoietin receptor gene EPOR in Philadelphia chromosome-like (Ph-like) ALL. All of these rearrangements result in truncation of the cytoplasmic tail of EPOR at residues similar to those mutated in primary familial congenital polycythemia, with preservation of the proximal tyrosine essential for receptor activation and loss of distal regulatory residues. This resulted in deregulated EPOR expression, hypersensitivity to erythropoietin stimulation, and heightened JAK-STAT activation. Expression of truncated EPOR in mouse B cell progenitors induced ALL in vivo. Human leukemic cells with EPOR rearrangements were sensitive to JAK-STAT inhibition, suggesting a therapeutic option in high-risk ALL.


Asunto(s)
Orden Génico , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Receptores de Eritropoyetina/genética , Secuencia de Aminoácidos , Antineoplásicos/uso terapéutico , Secuencia de Bases , Humanos , Datos de Secuencia Molecular , Mutación , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico
9.
Science ; 351(6269): aab2116, 2016 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-26541609

RESUMEN

In a classical view of hematopoiesis, the various blood cell lineages arise via a hierarchical scheme starting with multipotent stem cells that become increasingly restricted in their differentiation potential through oligopotent and then unipotent progenitors. We developed a cell-sorting scheme to resolve myeloid (My), erythroid (Er), and megakaryocytic (Mk) fates from single CD34(+) cells and then mapped the progenitor hierarchy across human development. Fetal liver contained large numbers of distinct oligopotent progenitors with intermingled My, Er, and Mk fates. However, few oligopotent progenitor intermediates were present in the adult bone marrow. Instead, only two progenitor classes predominate, multipotent and unipotent, with Er-Mk lineages emerging from multipotent cells. The developmental shift to an adult "two-tier" hierarchy challenges current dogma and provides a revised framework to understand normal and disease states of human hematopoiesis.


Asunto(s)
Linaje de la Célula/fisiología , Células Eritroides/citología , Hematopoyesis/fisiología , Células Progenitoras de Megacariocitos/citología , Megacariocitos/citología , Células Mieloides/citología , Adulto , Antígenos CD34/análisis , Linaje de la Célula/genética , Separación Celular , Células Cultivadas , Sangre Fetal/citología , Perfilación de la Expresión Génica , Hematopoyesis/genética , Humanos , Hígado/citología , Hígado/embriología , Células Madre Multipotentes/citología , Transcripción Genética
10.
Int J Hematol ; 102(5): 513-22, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26440972

RESUMEN

Chronological human aging is associated with a number of changes in the hematopoietic system, occurring at many levels from stem to mature cells, and the marrow microenvironment as well. This review will focus mainly on the aging of hematopoietic stem and progenitor cells (HSPCs), and on the associated increases in the incidence of hematological malignancies. HSPCs manifest reduced function and acquire molecular changes with chronological aging. Furthermore, while for many years it has been known that the human hematopoietic system becomes increasingly clonal with chronological aging (clonal hematopoiesis), only in the last few years has it become clear that clonal hematopoiesis may result from the accumulation of preleukemic mutations in HSPCs. Such mutations confer a selective advantage that leads to clonal hematopoiesis, and that may occasionally result in the development of leukemia, and define the existence of both preleukemic stem cells, and of 'preleukemia' as a clinical entity. While it is well appreciated that clonal hematopoiesis is very common in the elderly, several questions remain unanswered: why and how does clonal hematopoiesis develop? How is clonal hematopoiesis related to the age-related changes observed in the hematopoietic system? And why do only some individuals with clonal hematopoiesis develop leukemia?


Asunto(s)
Envejecimiento , Hematopoyesis , Células Madre Hematopoyéticas , Leucemia , Mutación , Lesiones Precancerosas , Envejecimiento/metabolismo , Envejecimiento/patología , Células Madre Hematopoyéticas/metabolismo , Células Madre Hematopoyéticas/patología , Humanos , Leucemia/genética , Leucemia/metabolismo , Leucemia/patología , Lesiones Precancerosas/genética , Lesiones Precancerosas/metabolismo
11.
Cancer Cell ; 28(3): 343-56, 2015 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-26321221

RESUMEN

Alterations of IKZF1, encoding the lymphoid transcription factor IKAROS, are a hallmark of high-risk acute lymphoblastic leukemia (ALL), however the role of IKZF1 alterations in ALL pathogenesis is poorly understood. Here, we show that in mouse models of BCR-ABL1 leukemia, Ikzf1 and Arf alterations synergistically promote the development of an aggressive lymphoid leukemia. Ikzf1 alterations result in acquisition of stem cell-like features, including self-renewal and increased bone marrow stromal adhesion. Retinoid receptor agonists reversed this phenotype, partly by inducing expression of IKZF1, resulting in abrogation of adhesion and self-renewal, cell cycle arrest, and attenuation of proliferation without direct cytotoxicity. Retinoids potentiated the activity of dasatinib in mouse and human BCR-ABL1 ALL, providing an additional therapeutic option in IKZF1-mutated ALL.


Asunto(s)
Proteínas de Fusión bcr-abl/genética , Factor de Transcripción Ikaros/genética , Mutación/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Retinoides/metabolismo , Animales , Puntos de Control del Ciclo Celular/genética , Humanos , Ratones , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , Receptores de Ácido Retinoico/metabolismo
12.
Cell Stem Cell ; 16(3): 302-13, 2015 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-25704240

RESUMEN

Regulated blood production is achieved through the hierarchical organization of dormant hematopoietic stem cell (HSC) subsets that differ in self-renewal potential and division frequency, with long-term (LT)-HSCs dividing the least. The molecular mechanisms underlying this variability in HSC division kinetics are unknown. We report here that quiescence exit kinetics are differentially regulated within human HSC subsets through the expression level of CDK6. LT-HSCs lack CDK6 protein. Short-term (ST)-HSCs are also quiescent but contain high CDK6 protein levels that permit rapid cell cycle entry upon mitogenic stimulation. Enforced CDK6 expression in LT-HSCs shortens quiescence exit and confers competitive advantage without impacting function. Computational modeling suggests that this independent control of quiescence exit kinetics inherently limits LT-HSC divisions and preserves the HSC pool to ensure lifelong hematopoiesis. Thus, differential expression of CDK6 underlies heterogeneity in stem cell quiescence states that functionally regulates this highly regenerative system.


Asunto(s)
División Celular/fisiología , Simulación por Computador , Quinasa 6 Dependiente de la Ciclina/biosíntesis , Regulación Enzimológica de la Expresión Génica/fisiología , Células Madre Hematopoyéticas/enzimología , Modelos Biológicos , Hematopoyesis/fisiología , Células Madre Hematopoyéticas/citología , Humanos
13.
Nature ; 506(7488): 328-33, 2014 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-24522528

RESUMEN

In acute myeloid leukaemia (AML), the cell of origin, nature and biological consequences of initiating lesions, and order of subsequent mutations remain poorly understood, as AML is typically diagnosed without observation of a pre-leukaemic phase. Here, highly purified haematopoietic stem cells (HSCs), progenitor and mature cell fractions from the blood of AML patients were found to contain recurrent DNMT3A mutations (DNMT3A(mut)) at high allele frequency, but without coincident NPM1 mutations (NPM1c) present in AML blasts. DNMT3A(mut)-bearing HSCs showed a multilineage repopulation advantage over non-mutated HSCs in xenografts, establishing their identity as pre-leukaemic HSCs. Pre-leukaemic HSCs were found in remission samples, indicating that they survive chemotherapy. Therefore DNMT3A(mut) arises early in AML evolution, probably in HSCs, leading to a clonally expanded pool of pre-leukaemic HSCs from which AML evolves. Our findings provide a paradigm for the detection and treatment of pre-leukaemic clones before the acquisition of additional genetic lesions engenders greater therapeutic resistance.


Asunto(s)
Células Madre Hematopoyéticas/citología , Leucemia Mieloide Aguda/patología , Células Madre Neoplásicas/citología , Animales , Diferenciación Celular , División Celular , Linaje de la Célula , Células Clonales/citología , Células Clonales/metabolismo , Células Clonales/patología , ADN (Citosina-5-)-Metiltransferasas/genética , ADN (Citosina-5-)-Metiltransferasas/metabolismo , ADN Metiltransferasa 3A , Resistencia a Antineoplásicos/efectos de los fármacos , Femenino , Hematopoyesis , Células Madre Hematopoyéticas/efectos de los fármacos , Células Madre Hematopoyéticas/metabolismo , Células Madre Hematopoyéticas/patología , Xenoinjertos , Humanos , Isocitrato Deshidrogenasa/genética , Leucemia Mieloide Aguda/diagnóstico , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/genética , Ratones , Ratones Endogámicos NOD , Ratones SCID , Mutación/genética , Trasplante de Neoplasias , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Proteínas Nucleares/genética , Nucleofosmina , Inducción de Remisión , Linfocitos T/metabolismo , Linfocitos T/patología
14.
Blood ; 123(6): 863-74, 2014 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-24345756

RESUMEN

Constitutive heterozygous GATA2 mutation is associated with deafness, lymphedema, mononuclear cytopenias, infection, myelodysplasia (MDS), and acute myeloid leukemia. In this study, we describe a cross-sectional analysis of 24 patients and 6 relatives with 14 different frameshift or substitution mutations of GATA2. A pattern of dendritic cell, monocyte, B, and natural killer (NK) lymphoid deficiency (DCML deficiency) with elevated Fms-like tyrosine kinase 3 ligand (Flt3L) was observed in all 20 patients phenotyped, including patients with Emberger syndrome, monocytopenia with Mycobacterium avium complex (MonoMAC), and MDS. Four unaffected relatives had a normal phenotype indicating that cellular deficiency may evolve over time or is incompletely penetrant, while 2 developed subclinical cytopenias or elevated Flt3L. Patients with GATA2 mutation maintained higher hemoglobin, neutrophils, and platelets and were younger than controls with acquired MDS and wild-type GATA2. Frameshift mutations were associated with earlier age of clinical presentation than substitution mutations. Elevated Flt3L, loss of bone marrow progenitors, and clonal myelopoiesis were early signs of disease evolution. Clinical progression was associated with increasingly elevated Flt3L, depletion of transitional B cells, CD56(bright) NK cells, naïve T cells, and accumulation of terminally differentiated NK and CD8(+) memory T cells. These studies provide a framework for clinical and laboratory monitoring of patients with GATA2 mutation and may inform therapeutic decision-making.


Asunto(s)
Linfocitos B/patología , Células Dendríticas/patología , Factor de Transcripción GATA2/genética , Células Asesinas Naturales/patología , Monocitos/patología , Mutación/genética , Síndromes Mielodisplásicos/patología , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Linfocitos B/inmunología , Linfocitos B/metabolismo , Biomarcadores , Estudios de Casos y Controles , Niño , Preescolar , Evolución Clonal , Estudios Transversales , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Ensayo de Inmunoadsorción Enzimática , Femenino , Estudios de Seguimiento , Estudios de Asociación Genética , Humanos , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Masculino , Persona de Mediana Edad , Monocitos/inmunología , Monocitos/metabolismo , Síndromes Mielodisplásicos/sangre , Síndromes Mielodisplásicos/genética , Linaje , Pronóstico , Adulto Joven , Tirosina Quinasa 3 Similar a fms/metabolismo
15.
Int J Cancer ; 134(10): 2330-41, 2014 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-24154973

RESUMEN

A locus on human chromosome 11q23 tagged by marker rs3802842 was associated with colorectal cancer (CRC) in a genome-wide association study; this finding has been replicated in case-control studies worldwide. In order to identify biologic factors at this locus that are related to the etiopathology of CRC, we used microarray-based target selection methods, coupled to next-generation sequencing, to study 103 kb at the 11q23 locus. We genotyped 369 putative variants from 1,030 patients with CRC (cases) and 1,061 individuals without CRC (controls) from the Ontario Familial Colorectal Cancer Registry. Two previously uncharacterized genes, COLCA1 and COLCA2, were found to be co-regulated genes that are transcribed from opposite strands. Expression levels of COLCA1 and COLCA2 transcripts correlate with rs3802842 genotypes. In colon tissues, COLCA1 co-localizes with crystalloid granules of eosinophils and granular organelles of mast cells, neutrophils, macrophages, dendritic cells and differentiated myeloid-derived cell lines. COLCA2 is present in the cytoplasm of normal epithelial, immune and other cell lineages, as well as tumor cells. Tissue microarray analysis demonstrates the association of rs3802842 with lymphocyte density in the lamina propria (p = 0.014) and levels of COLCA1 in the lamina propria (p = 0.00016) and COLCA2 (tumor cells, p = 0.0041 and lamina propria, p = 6 × 10(-5)). In conclusion, genetic, expression and immunohistochemical data implicate COLCA1 and COLCA2 in the pathogenesis of colon cancer. Histologic analyses indicate the involvement of immune pathways.


Asunto(s)
Colon/metabolismo , Neoplasias Colorrectales/genética , Regulación Neoplásica de la Expresión Génica , Predisposición Genética a la Enfermedad/genética , Sistema Inmunológico/metabolismo , Polimorfismo de Nucleótido Simple , Secuencia de Aminoácidos , Western Blotting , Células CACO-2 , Línea Celular Tumoral , Colon/patología , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Predisposición Genética a la Enfermedad/clasificación , Células HCT116 , Células HEK293 , Células HL-60 , Células HT29 , Células HeLa , Humanos , Sistema Inmunológico/patología , Inmunohistoquímica , Células Jurkat , Células K562 , Células MCF-7 , Datos de Secuencia Molecular , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Filogenia , ARN , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Homología de Secuencia de Aminoácido , Células U937
16.
Nat Immunol ; 14(7): 756-63, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23708252

RESUMEN

Understanding how differentiation programs originate from the gene-expression 'landscape' of hematopoietic stem cells (HSCs) is crucial for the development of new clinical therapies. We mapped the transcriptional dynamics underlying the first steps of commitment by tracking transcriptome changes in human HSCs and eight early progenitor populations. We found that transcriptional programs were extensively shared, extended across lineage-potential boundaries and were not strictly lineage affiliated. Elements of stem, lymphoid and myeloid programs were retained in multilymphoid progenitors (MLPs), which reflected a hybrid transcriptional state. By functional single cell analysis, we found that the transcription factors Bcl-11A, Sox4 and TEAD1 (TEF1) governed transcriptional networks in MLPs, which led to B cell specification. Overall, we found that integrated transcriptome approaches can be used to identify previously unknown regulators of multipotency and show additional complexity in lymphoid commitment.


Asunto(s)
Linfocitos B/citología , Redes Reguladoras de Genes , Hematopoyesis/fisiología , Células Madre Hematopoyéticas/citología , Diferenciación Celular/genética , Linaje de la Célula , Biología Computacional , Perfilación de la Expresión Génica/métodos , Humanos , ARN Mensajero/química , ARN Mensajero/genética , Factores de Transcripción/genética
17.
Proc Natl Acad Sci U S A ; 109(39): 15871-6, 2012 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-23019372

RESUMEN

To better understand the process of B-lymphocyte lineage restriction, we have investigated molecular and functional properties in early B-lineage cells from Pax-5-deficient animals crossed to a B-lineage-restricted reporter mouse, allowing us to identify B-lineage-specified progenitors independently of conventional surface markers. Pax-5 deficiency resulted in a dramatic increase in the frequency of specified progenitor B-cells marked by expression of a λ5 (Igll1) promoter-controlled reporter gene. Gene expression analysis of ex vivo isolated progenitor cells revealed that Pax-5 deficiency has a minor impact on B-cell specification. However, single-cell in vitro differentiation analysis of ex vivo isolated cells revealed that specified B-lineage progenitors still displayed a high degree of plasticity for development into NK or T lineage cells. In contrast, we were unable to detect any major changes in myeloid lineage potential in specified Pax-5-deficient cells. By comparison of gene expression patterns in ex vivo isolated Pax-5- and Ebf-1-deficient progenitors, it was possible to identify a set of B-cell-restricted genes dependent on Ebf-1 but not Pax-5, supporting the idea that B-cell specification and commitment is controlled by distinct regulatory networks.


Asunto(s)
Linfocitos B/inmunología , Diferenciación Celular/fisiología , Regulación de la Expresión Génica/inmunología , Animales , Linfocitos B/citología , Humanos , Células Asesinas Naturales/citología , Células Asesinas Naturales/inmunología , Ratones , Ratones Transgénicos , Factor de Transcripción PAX5/genética , Factor de Transcripción PAX5/inmunología , Linfocitos T/citología , Linfocitos T/inmunología , Transactivadores/genética , Transactivadores/inmunología
18.
Mol Med ; 18: 1169-82, 2012 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-22777388

RESUMEN

Human immunodeficiency virus type 1 (HIV-1) infection enhances the expression of inhibitory molecules on T cells, leading to T-cell impairment. The signaling pathways underlying the regulation of inhibitory molecules and subsequent onset of T-cell impairment remain elusive. We showed that both autologous and allogeneic T cells exposed to HIV-pulsed dendritic cells (DCs) upregulated cytotoxic T-lymphocyte antigen (CTLA-4), tumor-necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL), lymphocyte-activation gene-3 (LAG3), T-cell immunoglobulin mucin-3 (TIM-3), CD160 and certain suppression-associated transcription factors, such as B-lymphocyte induced maturation protein-1 (BLIMP-1), deltex homolog 1 protein (DTX1) and forkhead box P3 (FOXP3), leading to T-cell suppression. This induction was regulated by p38 mitogen-activated protein kinase/signal transducer and activator of transcription-3 (P38MAPK/STAT3) pathways, because their blockade significantly abrogated expression of all the inhibitory molecules studied and a subsequent recovery in T-cell proliferation. Neither interleukin-6 (IL-6) nor IL-10 nor growth factors known to activate STAT3 signaling events were responsible for STAT3 activation. Involvement of the P38MAPK/STAT3 pathways was evident because these proteins had a higher level of phosphorylation in the HIV-1-primed cells. Furthermore, blockade of viral CD4 binding and fusion significantly reduced the negative effects DCs imposed on primed T cells. In conclusion, HIV-1 interaction with DCs modulated their functionality, causing them to trigger the activation of the P38MAPK/STAT3 pathway in T cells, which was responsible for the upregulation of inhibitory molecules.


Asunto(s)
Células Dendríticas/virología , VIH-1/inmunología , Activación de Linfocitos/inmunología , Factor de Transcripción STAT3/metabolismo , Transducción de Señal/inmunología , Linfocitos T/inmunología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Proliferación Celular/efectos de los fármacos , Citosol/efectos de los fármacos , Citosol/metabolismo , Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Células Dendríticas/patología , Enterotoxinas/farmacología , VIH-1/efectos de los fármacos , Humanos , Memoria Inmunológica/efectos de los fármacos , Interleucina-10/metabolismo , Interleucina-6/metabolismo , Activación de Linfocitos/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Pruebas de Neutralización , Transducción de Señal/efectos de los fármacos , Linfocitos T/efectos de los fármacos , Linfocitos T/enzimología , Linfocitos T/patología
19.
Immunity ; 36(6): 921-32, 2012 Jun 29.
Artículo en Inglés | MEDLINE | ID: mdl-22608498

RESUMEN

Multiple transcription factors guide the development of mature functional natural killer (NK) cells, yet little is known about their function. We used global gene expression and genome-wide binding analyses combined with developmental and functional studies to unveil three roles for the ETS1 transcription factor in NK cells. ETS1 functions at the earliest stages of NK cell development to promote expression of critical transcriptional regulators including T-BET and ID2, NK cell receptors (NKRs) including NKp46, Ly49H, and Ly49D, and signaling molecules essential for NKR function. As a consequence, Ets1(-/-) NK cells fail to degranulate after stimulation through activating NKRs. Nonetheless, these cells are hyperresponsive to cytokines and have characteristics of chronic stimulation including increased expression of inhibitory NKRs and multiple activation-associated genes. Therefore, ETS1 regulates a broad gene expression program in NK cells that promotes target cell recognition while limiting cytokine-driven activation.


Asunto(s)
Células Asesinas Naturales/inmunología , Proteína Proto-Oncogénica c-ets-1/deficiencia , Secuencias de Aminoácidos , Animales , Sitios de Unión , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Células Cultivadas/efectos de los fármacos , Células Cultivadas/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/inmunología , Proteína 2 Inhibidora de la Diferenciación/biosíntesis , Proteína 2 Inhibidora de la Diferenciación/genética , Interleucina-15/farmacología , Interleucina-15/fisiología , Péptidos y Proteínas de Señalización Intracelular/genética , Células Asesinas Naturales/efectos de los fármacos , Células Asesinas Naturales/metabolismo , Activación de Linfocitos/efectos de los fármacos , Activación de Linfocitos/genética , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Datos de Secuencia Molecular , Proteína Proto-Oncogénica c-ets-1/genética , Proteína Proto-Oncogénica c-ets-1/fisiología , Quimera por Radiación , Receptores de Células Asesinas Naturales/biosíntesis , Receptores de Células Asesinas Naturales/genética , Transducción de Señal/genética , Transducción de Señal/inmunología , Proteínas de Dominio T Box/biosíntesis , Proteínas de Dominio T Box/genética , Factores de Transcripción/biosíntesis , Factores de Transcripción/genética , Transcripción Genética/efectos de los fármacos , Transcripción Genética/inmunología
20.
Blood ; 118(5): 1283-90, 2011 Aug 04.
Artículo en Inglés | MEDLINE | ID: mdl-21652681

RESUMEN

Deficiencies in the IL-7 signaling pathway result in severe disruptions of lymphoid development in adult mice. To understand more about how IL-7 deficiency impacts early lymphoid development, we have investigated lineage restriction events within the common lymphoid progenitor (CLP) compartment in IL-7 knockout mice. This revealed that although IL-7 deficiency had a minor impact on the development of LY6D(-) multipotent CLPs, the formation of the lineage restricted LY6D(+) CLP population was dramatically reduced. This was reflected in a low-level transcription of B-lineage genes as well as in a loss of functional B-cell commitment. The few Ly6D(+) CLPs developed in the absence of IL-7 displayed increased lineage plasticity and low expression of Ebf-1. Absence of Ebf-1 could be linked to increased plasticity because even though Ly6D(+) cells develop in Ebf-1-deficient mice, these cells retain both natural killer and dendritic cell potential. This reveals that IL-7 is essential for normal development of Ly6D(+) CLPs and that Ebf-1 is crucial for lineage restriction in early lymphoid progenitors.


Asunto(s)
Linaje de la Célula/genética , Interleucina-7/fisiología , Células Progenitoras Linfoides/fisiología , Transactivadores/fisiología , Animales , Antígenos Ly/metabolismo , Linfocitos B/metabolismo , Linfocitos B/fisiología , Diferenciación Celular/genética , Células Cultivadas , Proteínas Ligadas a GPI/metabolismo , Perfilación de la Expresión Génica , Interleucina-7/genética , Células Asesinas Naturales/metabolismo , Células Asesinas Naturales/fisiología , Células Progenitoras Linfoides/citología , Células Progenitoras Linfoides/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Análisis por Micromatrices , Linfocitos T/metabolismo , Linfocitos T/fisiología , Transactivadores/genética , Transactivadores/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...