Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
Proc Natl Acad Sci U S A ; 120(32): e2206860120, 2023 08 08.
Artículo en Inglés | MEDLINE | ID: mdl-37523546

RESUMEN

Mbtd1 (mbt domain containing 1) encodes a nuclear protein containing a zinc finger domain and four malignant brain tumor (MBT) repeats. We previously generated Mbtd1-deficient mice and found that MBTD1 is highly expressed in fetal hematopoietic stem cells (HSCs) and sustains the number and function of fetal HSCs. However, since Mbtd1-deficient mice die soon after birth possibly due to skeletal abnormalities, its role in adult hematopoiesis remains unclear. To address this issue, we generated Mbtd1 conditional knockout mice and analyzed adult hematopoietic tissues deficient in Mbtd1. We observed that the numbers of HSCs and progenitors increased and Mbtd1-deficient HSCs exhibited hyperactive cell cycle, resulting in a defective response to exogenous stresses. Mechanistically, we found that MBTD1 directly binds to the promoter region of FoxO3a, encoding a forkhead protein essential for HSC quiescence, and interacts with components of TIP60 chromatin remodeling complex and other proteins involved in HSC and other stem cell functions. Restoration of FOXO3a activity in Mbtd1-deficient HSCs in vivo rescued cell cycle and pool size abnormalities. These findings indicate that MBTD1 is a critical regulator for HSC pool size and function, mainly through the maintenance of cell cycle quiescence by FOXO3a.


Asunto(s)
Médula Ósea , Células Madre Hematopoyéticas , Animales , Ratones , Ciclo Celular/genética , Hematopoyesis/genética , Células Madre Hematopoyéticas/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Factores de Transcripción/metabolismo
2.
Blood ; 137(7): 908-922, 2021 02 18.
Artículo en Inglés | MEDLINE | ID: mdl-33174606

RESUMEN

Epigenetic regulation is essential for the maintenance of the hematopoietic system, and its deregulation is implicated in hematopoietic disorders. In this study, UTX, a demethylase for lysine 27 on histone H3 (H3K27) and a component of COMPASS-like and SWI/SNF complexes, played an essential role in the hematopoietic system by globally regulating aging-associated genes. Utx-deficient (UtxΔ/Δ) mice exhibited myeloid skewing with dysplasia, extramedullary hematopoiesis, impaired hematopoietic reconstituting ability, and increased susceptibility to leukemia, which are the hallmarks of hematopoietic aging. RNA-sequencing (RNA-seq) analysis revealed that Utx deficiency converted the gene expression profiles of young hematopoietic stem-progenitor cells (HSPCs) to those of aged HSPCs. Utx expression in hematopoietic stem cells declined with age, and UtxΔ/Δ HSPCs exhibited increased expression of an aging-associated marker, accumulation of reactive oxygen species, and impaired repair of DNA double-strand breaks. Pathway and chromatin immunoprecipitation analyses coupled with RNA-seq data indicated that UTX contributed to hematopoietic homeostasis mainly by maintaining the expression of genes downregulated with aging via demethylase-dependent and -independent epigenetic programming. Of note, comparison of pathway changes in UtxΔ/Δ HSPCs, aged muscle stem cells, aged fibroblasts, and aged induced neurons showed substantial overlap, strongly suggesting common aging mechanisms among different tissue stem cells.


Asunto(s)
Envejecimiento/genética , Regulación de la Expresión Génica/genética , Hematopoyesis/genética , Sistema Hematopoyético/fisiología , Código de Histonas/genética , Histona Demetilasas/fisiología , Animales , Senescencia Celular/genética , Roturas del ADN de Doble Cadena , Reparación del ADN , Femenino , Predisposición Genética a la Enfermedad , Hematopoyesis Extramedular , Histona Demetilasas/deficiencia , Histona Demetilasas/genética , Reconstitución Inmune , Histona Demetilasas con Dominio de Jumonji/metabolismo , Leucemia Experimental/genética , Leucemia Experimental/virología , Masculino , Ratones , Ratones Noqueados , Virus de la Leucemia Murina de Moloney/fisiología , Células Mieloides/patología , Quimera por Radiación , Especies Reactivas de Oxígeno/metabolismo , Proteínas Recombinantes/metabolismo , Factores de Transcripción/metabolismo , Integración Viral
3.
Blood ; 129(15): 2148-2160, 2017 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-28209720

RESUMEN

Chronic myelomonocytic leukemia (CMML) is a hematological malignancy characterized by uncontrolled proliferation of dysplastic myelomonocytes and frequent progression to acute myeloid leukemia (AML). We identified mutations in the Cbl gene, which encodes a negative regulator of cytokine signaling, in a subset of CMML patients. To investigate the contribution of mutant Cbl in CMML pathogenesis, we generated conditional knockin mice for Cbl that express wild-type Cbl in a steady state and inducibly express CblQ367P , a CMML-associated Cbl mutant. CblQ367P mice exhibited sustained proliferation of myelomonocytes, multilineage dysplasia, and splenomegaly, which are the hallmarks of CMML. The phosphatidylinositol 3-kinase (PI3K)-AKT and JAK-STAT pathways were constitutively activated in CblQ367P hematopoietic stem cells, which promoted cell cycle progression and enhanced chemokine-chemokine receptor activity. Gem, a gene encoding a GTPase that is upregulated by CblQ367P , enhanced hematopoietic stem cell activity and induced myeloid cell proliferation. In addition, Evi1, a gene encoding a transcription factor, was found to cooperate with CblQ367P and progress CMML to AML. Furthermore, targeted inhibition for the PI3K-AKT and JAK-STAT pathways efficiently suppressed the proliferative activity of CblQ367P -bearing CMML cells. Our findings provide insights into the molecular mechanisms underlying mutant Cbl-induced CMML and propose a possible molecular targeting therapy for mutant Cbl-carrying CMML patients.


Asunto(s)
Ciclo Celular , Células Madre Hematopoyéticas , Leucemia Mielógena Crónica BCR-ABL Positiva , Mutación Missense , Mielopoyesis , Proteínas Proto-Oncogénicas c-cbl , Regulación hacia Arriba , Sustitución de Aminoácidos , Animales , Regulación Enzimológica de la Expresión Génica , Células Madre Hematopoyéticas/metabolismo , Células Madre Hematopoyéticas/patología , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Leucemia Mielógena Crónica BCR-ABL Positiva/metabolismo , Leucemia Mielógena Crónica BCR-ABL Positiva/patología , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patología , Ratones , Ratones Transgénicos , Monocitos/metabolismo , Monocitos/patología , Proteínas de Unión al GTP Monoméricas/biosíntesis , Proteínas de Unión al GTP Monoméricas/genética , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas c-cbl/biosíntesis , Proteínas Proto-Oncogénicas c-cbl/genética , Transducción de Señal
4.
Proc Natl Acad Sci U S A ; 113(37): 10370-5, 2016 09 13.
Artículo en Inglés | MEDLINE | ID: mdl-27578866

RESUMEN

Polycomb repressive complex 2 (PRC2) catalyzes the monomethylation, dimethylation, and trimethylation of histone H3 Lys27 (H3K27) and acts as a central epigenetic regulator that marks the repressive chromatin domain. Embryonic ectoderm development (EED), an essential component of PRC2, interacts with trimethylated H3K27 (H3K27me3) through the aromatic cage structure composed of its three aromatic amino acids, Phe97, Trp364, and Tyr365. This interaction allosterically activates the histone methyltransferase activity of PRC2 and thereby propagates repressive histone marks. In this study, we report the analysis of knock-in mice harboring the myeloid disorder-associated EED Ile363Met (I363M) mutation, analogous to the EED aromatic cage mutants. The I363M homozygotes displayed a remarkable and preferential reduction of H3K27me3 and died at midgestation. The heterozygotes increased the clonogenic capacity and bone marrow repopulating activity of hematopoietic stem/progenitor cells (HSPCs) and were susceptible to leukemia. Lgals3, a PRC2 target gene encoding a multifunctional galactose-binding lectin, was derepressed in I363M heterozygotes, which enhanced the stemness of HSPCs. Thus, our work provides in vivo evidence that the structural integrity of EED to H3K27me3 propagation is critical, especially for embryonic development and hematopoietic homeostasis, and that its perturbation increases the predisposition to hematologic malignancies.


Asunto(s)
Galectina 3/genética , Leucemia/genética , Complejo Represivo Polycomb 2/química , Animales , Desarrollo Embrionario/genética , Epigénesis Genética/genética , Galectina 3/química , Predisposición Genética a la Enfermedad , Células Madre Hematopoyéticas/química , Células Madre Hematopoyéticas/metabolismo , N-Metiltransferasa de Histona-Lisina/química , N-Metiltransferasa de Histona-Lisina/genética , Humanos , Ratones , Complejo Represivo Polycomb 2/genética
5.
Blood ; 125(22): 3437-46, 2015 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-25872778

RESUMEN

We previously reported that deficiency for Samd9L, which was cloned as a candidate gene for -7/7q- syndrome, accelerated leukemia cooperatively with enhanced expression of a histone demethylase: F-box and leucine-rich repeat protein 10 (Fbxl10, also known as Jhdm1b, Kdm2b, and Ndy1). To further investigate the role of Fbxl10 in leukemogenesis, we generated transgenic (Tg) mice that overexpress Fbxl10 in hematopoietic stem cells (HSCs). Interestingly, Fbxl10 Tg mice developed myeloid or B-lymphoid leukemia with complete penetrance. HSCs from the Tg mice exhibited an accelerated G0/G1-to-S transition with a normal G0 to G1 entry, resulting in pleiotropic progenitor cell expansion. Fbxl10 Tg HSCs displayed enhanced expression of neuron-specific gene family member 2 (Nsg2), and forced expression of Nsg2 in primary bone marrow cells resulted in expansion of immature cells. In addition, the genes involved in mitochondrial oxidative phosphorylation were markedly enriched in Fbxl10 Tg HSCs, coupled with increased cellular adenosine 5'-triphosphate levels. Moreover, chromatin immunoprecipitation followed by sequencing analysis demonstrated that Fbxl10 directly binds to the regulatory regions of Nsg2 and oxidative phosphorylation genes. These findings define Fbxl10 as a bona fide oncogene, whose deregulated expression contributes to the development of leukemia involving metabolic proliferative advantage and Nsg2-mediated impaired differentiation.


Asunto(s)
Proteínas Portadoras/metabolismo , Proteínas F-Box/genética , Células Madre Hematopoyéticas/metabolismo , Histona Demetilasas con Dominio de Jumonji/genética , Leucemia/genética , Leucemia/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Animales , Linfocitos B/patología , Proteínas Portadoras/genética , Diferenciación Celular/genética , Proliferación Celular/genética , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Proteínas F-Box/metabolismo , Histona Demetilasas con Dominio de Jumonji/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Células Mieloides/patología , Proteínas del Tejido Nervioso/genética , Oncogenes , Regulación hacia Arriba/genética
6.
Proc Natl Acad Sci U S A ; 108(6): 2468-73, 2011 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-21252303

RESUMEN

To clarify the molecular pathways governing hematopoietic stem cell (HSC) development, we screened a fetal liver (FL) HSC cDNA library and identified a unique gene, hematopoietic expressed mammalian polycomb (hemp), encoding a protein with a zinc-finger domain and four malignant brain tumor (mbt) repeats. To investigate its biological role, we generated mice lacking Hemp (hemp(-/-)). Hemp(-/-) mice exhibited a variety of skeletal malformations and died soon after birth. In the FL, hemp was preferentially expressed in the HSC and early progenitor cell fractions, and analyses of fetal hematopoiesis revealed that the number of FL mononuclear cells, including HSCs, was reduced markedly in hemp(-/-) embryos, especially during early development. In addition, colony-forming and competitive repopulation assays demonstrated that the proliferative and reconstitution abilities of hemp(-/-) FL HSCs were significantly impaired. Microarray analysis revealed alterations in the expression levels of several genes implicated in hematopoietic development and differentiation in hemp(-/-) FL HSCs. These results demonstrate that Hemp, an mbt-containing protein, plays essential roles in HSC function and skeletal formation. It is also hypothesized that Hemp might be involved in certain congenital diseases, such as Klippel-Feil anomaly.


Asunto(s)
Desarrollo Óseo/fisiología , Huesos/embriología , Proteínas Cromosómicas no Histona/metabolismo , Embrión de Mamíferos/metabolismo , Regulación del Desarrollo de la Expresión Génica/fisiología , Células Madre Hematopoyéticas/metabolismo , Proteínas Represoras/metabolismo , Animales , Diferenciación Celular/fisiología , Proteínas Cromosómicas no Histona/genética , Embrión de Mamíferos/citología , Perfilación de la Expresión Génica , Hematopoyesis/fisiología , Síndrome de Klippel-Feil/genética , Síndrome de Klippel-Feil/metabolismo , Ratones , Ratones Noqueados , Análisis de Secuencia por Matrices de Oligonucleótidos , Proteínas Represoras/genética
7.
BMC Immunol ; 13: 72, 2012 Dec 26.
Artículo en Inglés | MEDLINE | ID: mdl-23268649

RESUMEN

BACKGROUND: CD72 is an inhibitory co-receptor expressed on B cells. We previously demonstrated significant association of the polymorphism of the CD72 gene with susceptibility to human systemic lupus erythematosus (SLE) in individuals carrying a SLE-susceptible FCGR2B genotype (FCGR2B-232Thr/Thr). The human CD72 locus generates a splicing isoform that lacks exon 8 (CD72Δex8) as well as full-length CD72 (CD72fl), and the CD72 polymorphism regulates exon 8 skipping. RESULTS: Here we demonstrated that individuals carrying the disease-protective CD72 genotype exhibit significantly lower serum immunoglobulin levels than do individuals carrying other CD72 genotypes (P < 0.05). Although expression level of CD72fl in the peripheral blood B cells was similar regardless of CD72 genotype, the protein level of CD72Δex8 was increased in individuals carrying the disease-protective CD72 genotype, suggesting a crucial role of CD72Δex8 in regulation of antibody production. By expressing these human CD72 isoforms in mouse cell lines, we further demonstrated that CD72Δex8 is accumulated in endoplasmic reticulum (ER) and fails to regulate BCR signaling whereas human CD72fl is efficiently transported to the cell surface and inhibits signaling through the B cell antigen receptor (BCR), as is the case for mouse CD72. CONCLUSION: Human CD72 polymorphism appears to regulate antibody production as well as susceptibility to SLE by regulating expression of ER-localizing CD72Δex8.


Asunto(s)
Antígenos CD/metabolismo , Antígenos de Diferenciación de Linfocitos B/metabolismo , Linfocitos B/inmunología , Retículo Endoplásmico/metabolismo , Lupus Eritematoso Sistémico/inmunología , Proteínas Mutantes/metabolismo , Isoformas de Proteínas/metabolismo , Animales , Formación de Anticuerpos/genética , Antígenos CD/genética , Antígenos CD/inmunología , Antígenos de Diferenciación de Linfocitos B/genética , Antígenos de Diferenciación de Linfocitos B/inmunología , Línea Celular , Predisposición Genética a la Enfermedad , Humanos , Inmunoglobulinas/sangre , Ratones , Proteínas Mutantes/genética , Proteínas Mutantes/inmunología , Polimorfismo Genético , Isoformas de Proteínas/genética , Isoformas de Proteínas/inmunología , Empalme de Proteína , Transporte de Proteínas , Transgenes/genética
8.
J Immunol ; 185(12): 7330-9, 2010 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-21057086

RESUMEN

Naturally occurring regulatory T (Treg) cells play a central role in the maintenance of immune homeostasis and in restraining the development of spontaneous inflammatory responses. However, the underlying mechanisms of Treg homeostasis remain incompletely understood. Of particular note, the IL-2Rα (CD25) is crucial for the homeostasis of Treg cells and the prevention of lymphoproliferative autoimmune disease. In this paper, we report that the basic helix-loop-helix transcription factor Dec1 is involved in the homeostasis of Treg cells and plays a role in their survival or expansion after adoptive transfer to lymphopenic recipients. Hence, it is crucial for the suppression of effector T cell-mediated inflammatory responses. Enforced expression of Dec1 upregulates CD25 expression during thymocyte development and increases the number of Treg cells in the periphery. Dec1 binds the transcription factor Runx1 and colocalizes with Runx1 in Treg cells. Specifically, we demonstrate that in Treg cells the Dec1/Runx1 complex binds to regulatory elements present in the Il-2rα locus. Collectively, these data show how Dec1 mechanistically acts in Treg cells.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/inmunología , Proteínas de Homeodominio/inmunología , Homeostasis/fisiología , Subunidad alfa del Receptor de Interleucina-2/inmunología , Regulación hacia Arriba/fisiología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Supervivencia Celular/fisiología , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Subunidad alfa 2 del Factor de Unión al Sitio Principal/inmunología , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Subunidad alfa del Receptor de Interleucina-2/genética , Subunidad alfa del Receptor de Interleucina-2/metabolismo , Ratones , Ratones Transgénicos , Unión Proteica/genética , Unión Proteica/inmunología , Linfocitos T Reguladores
9.
Dev Biol ; 345(2): 226-36, 2010 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-20647008

RESUMEN

Although internal ribosome entry site (IRES)-mediated translation is considered important for proper cellular function, its precise biological role is not fully understood. Runx1 gene, which encodes a transcription factor implicated in hematopoiesis, angiogenesis, and leukemogenesis, contains IRES sequences in the 5' untranslated region. To clarify the roles of the IRES element in Runx1 function, we generated knock-in mice for either wild-type Runx1 or Runx1/Evi1, a Runx1 fusion protein identified in human leukemia. In both cases, native promoter-dependent transcription was retained, whereas IRES-mediated translation was eliminated. Interestingly, homozygotes expressing wild-type Runx1 deleted for the IRES element (Runx1(Delta IRES/Delta IRES)) died in utero with prominent dilatation of peripheral blood vessels due to impaired pericyte development. In addition, hematopoietic cells in the Runx1(Delta IRES/Delta IRES) fetal liver were significantly decreased, and exhibited an altered differentiation pattern, a reduced proliferative activity, and an impaired reconstitution ability. On the other hand, heterozygotes expressing Runx1/Evi1 deleted for the IRES element (Runx1(+/RE Delta IRES)) were born normally and did not show any hematological abnormalities, in contrast that conventional Runx1/Evi1 heterozygotes die in utero with central nervous system hemorrhage and Runx1/Evi1 chimeric mice develop acute leukemia. The findings reported here demonstrate the essential roles of the IRES element in Runx1 function under physiological and pathological conditions.


Asunto(s)
Regiones no Traducidas 5' , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Hematopoyesis/genética , Leucemia/genética , Neovascularización Fisiológica/genética , Animales , Diferenciación Celular , Regulación Leucémica de la Expresión Génica , Técnicas de Sustitución del Gen , Heterocigoto , Hígado/patología , Ratones , Modelos Animales , Ribosomas/metabolismo
10.
Hepatology ; 52(3): 1089-99, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20623582

RESUMEN

UNLABELLED: p130Cas, Crk-associated substrate (Cas), is an adaptor/scaffold protein that plays a central role in actin cytoskeletal reorganization. We previously showed that mice in which Cas was deleted (Cas(-/-)) died in utero because of early cardiovascular maldevelopment. To further investigate the in vivo roles of Cas, we generated mice with a hypomorphic Cas allele lacking the exon 2-derived region (Cas(Deltaex2/Deltaex2)), which encodes Src homology domain 3 (SH3) of Cas. Cas(Deltaex2/Deltaex2) mice again died as embryos, but they particularly showed progressive liver degeneration with hepatocyte apoptosis. Because Cas expression in the liver is preferentially detected in sinusoidal endothelial cells (SECs), the observed hepatocyte apoptosis was most likely ascribable to impaired function of SECs. To address this possibility, we stably introduced a Cas mutant lacking the SH3 domain (Cas DeltaSH3) into an SEC line (NP31). Intriguingly, the introduction of Cas DeltaSH3 induced a loss of fenestrae, the characteristic cell-penetrating pores in SECs that serve as a critical route for supplying oxygen and nutrients to hepatocytes. The disappearance of fenestrae in Cas DeltaSH3-expressing cells was associated with an attenuation of actin stress fiber formation, a marked reduction in tyrosine phosphorylation of Cas, and defective binding of Cas to CrkII. CONCLUSION: Cas plays pivotal roles in liver development through the reorganization of the actin cytoskeleton and formation of fenestrae in SECs.


Asunto(s)
Extensiones de la Superficie Celular/fisiología , Proteína Sustrato Asociada a CrK/fisiología , Endotelio/fisiología , Hígado/embriología , Hígado/fisiología , Actinas/fisiología , Animales , Apoptosis/fisiología , Línea Celular , Extensiones de la Superficie Celular/ultraestructura , Proteína Sustrato Asociada a CrK/genética , Citoesqueleto/fisiología , Citoesqueleto/ultraestructura , Endotelio/citología , Endotelio/ultraestructura , Exones/genética , Femenino , Hepatocitos/citología , Hepatocitos/fisiología , Hepatocitos/ultraestructura , Hígado/citología , Masculino , Ratones , Ratones Endogámicos C57BL , Modelos Animales , Fosforilación/fisiología , Ratas
11.
Blood ; 113(19): 4702-10, 2009 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-19234145

RESUMEN

Chronic myelogenous leukemia (CML) is a hematopoietic disorder originating from p210BCR/ABL-transformed stem cells, which begins as indolent chronic phase (CP) but progresses into fatal blast crisis (BC). To investigate molecular mechanism(s) underlying disease evolution, CML-exhibiting p210BCR/ABL transgenic mice were crossed with BXH2 mice that transmit a replication-competent retrovirus. Whereas nontransgenic mice in the BXH2 background exclusively developed acute myeloid leukemia, p210BCR/ABL transgenic littermates developed nonmyeloid leukemias, in which inverse polymerase chain reaction detected 2 common viral integration sites (CISs). Interestingly, one CIS was transgene's own promoter, which up-regulated p210BCR/ABL expression. The other was the 5' noncoding region of a transcription factor, Zfp423, which induced aberrant Zfp423 expression. The cooperative activities of Zfp423 and p210BCR/ABL were demonstrated as follows: (1) introduction of Zfp423 in p210BCR/ABL transgenic bone marrow (BM) cells increased colony-forming ability, (2) suppression of ZNF423 (human homologue of Zfp423) in ZNF423-expressing, p210BCR/ABL-positive hematopoietic cells retarded cell growth, (3) mice that received a transplant of BM cells transduced with Zfp423 and p210BCR/ABL developed acute leukemia, and (4) expression of ZNF423 was found in human BCR/ABL-positive cell lines and CML BC samples. These results demonstrate that enhanced expression of p210BCR/ABL and deregulated expression of Zfp423/ZNF423 contribute to CML BC.


Asunto(s)
Linfocitos B/patología , Crisis Blástica/genética , Proteínas de Unión al ADN/fisiología , Proteínas de Fusión bcr-abl/fisiología , Regulación Leucémica de la Expresión Génica , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Factores de Transcripción/fisiología , Animales , Northern Blotting , Southern Blotting , Western Blotting , Trasplante de Médula Ósea , Proliferación Celular , Ensayo de Unidades Formadoras de Colonias , Proteínas de Unión al ADN/antagonistas & inhibidores , Femenino , Citometría de Flujo , Reordenamiento Génico , Humanos , Leucemia Mielógena Crónica BCR-ABL Positiva/metabolismo , Leucemia Mielógena Crónica BCR-ABL Positiva/patología , Masculino , Ratones , Ratones Noqueados , Fenotipo , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño/farmacología , Retroviridae , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Tasa de Supervivencia , Factores de Transcripción/antagonistas & inhibidores , Dedos de Zinc
12.
Genes Cells ; 14(12): 1383-94, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19930468

RESUMEN

The activation mechanisms of Src family kinases (SFKs) involve the dissociation of the intramolecular interaction between the Src homology (SH) 3 and kinase domain. This process is mediated by the intermolecular attack of outer ligands to the SH3 domain. By using a yeast two-hybrid screen, we isolated a relevant ligand involved in the activation mechanisms of SFKs. This molecule was found to be identical to a recently recognized kinetochore protein--designated as centromere protein (CENP)-V--which is required for the progression of mitosis. We show here that human CENP-V plays further roles in cell dynamics; the proline-rich region of human CENP-V associates with the SH3 domains of SFKs and potently activates SFKs, whereas another domain of CENP-V that possesses a highly conserved cysteine array confers the ability to associate with stabilized microtubules (MTs). Human CENP-V distributes to the cell protrusion and to the leading edge of migrating cells in response to external stimuli, and depletion of CENP-V by RNA interference significantly attenuates closure of a scratch wound. These findings indicate that human CENP-V is involved in directional cell motility as well as in the progression of mitosis, as a scaffolding molecule that links MTs and SFKs.


Asunto(s)
Centrómero/metabolismo , Proteínas de Unión al ADN/metabolismo , Microtúbulos/metabolismo , Dominios Homologos src/fisiología , Familia-src Quinasas/metabolismo , Secuencia de Aminoácidos , Western Blotting , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Movimiento Celular , Proteínas Cromosómicas no Histona , Proteínas de Unión al ADN/antagonistas & inhibidores , Proteínas de Unión al ADN/genética , Activación Enzimática , Femenino , Técnica del Anticuerpo Fluorescente , Humanos , Inmunoprecipitación , Cinetocoros , Datos de Secuencia Molecular , ARN Interferente Pequeño/farmacología , Homología de Secuencia de Aminoácido , Distribución Tisular , Células Tumorales Cultivadas , Técnicas del Sistema de Dos Híbridos , Ubiquitinación , Cicatrización de Heridas
13.
Clin Cancer Res ; 26(8): 2065-2079, 2020 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-32047002

RESUMEN

PURPOSE: Epigenetic deregulation is deeply implicated in the pathogenesis of bladder cancer. KDM6A (Lysine (K)-specific demethylase 6A) is a histone modifier frequently mutated in bladder cancer. However, the molecular mechanisms of how KDM6A deficiency contributes to bladder cancer development remains largely unknown. We hypothesized that clarification of the pathogenic mechanisms underlying KDM6A-mutated bladder cancer can help in designing new anticancer therapies. EXPERIMENTAL DESIGN: We generated mice lacking Kdm6a in the urothelium and crossed them with mice heterozygous for p53, whose mutation/deletion significantly overlaps with the KDM6A mutation in muscle-invasive bladder cancer (MIBC). In addition, BBN (N-butyl-N-(4-hydroxybutyl) nitrosamine), a cigarette smoke-like mutagen, was used as a tumor-promoting agent. Isolated urothelia were subjected to phenotypic, pathologic, molecular, and cellular analyses. The clinical relevance of our findings was further analyzed using genomic and clinical data of patients with MIBC. RESULTS: We found that Kdm6a deficiency activated cytokine and chemokine pathways, promoted M2 macrophage polarization, increased cancer stem cells and caused bladder cancer in cooperation with p53 haploinsufficiency. We also found that BBN treatment significantly enhanced the expression of proinflammatory molecules and accelerated disease development. Human bladder cancer samples with decreased KDM6A expression also showed activated proinflammatory pathways. Notably, dual inhibition of IL6 and chemokine (C-C motif) ligand 2, upregulated in response to Kdm6a deficiency, efficiently suppressed Kdm6a-deficient bladder cancer cell growth. CONCLUSIONS: Our findings provide insights into multistep carcinogenic processes of bladder cancer and suggest molecular targeted therapeutic approaches for patients with bladder cancer with KDM6A dysfunction.


Asunto(s)
Carcinogénesis/patología , Histona Demetilasas/fisiología , Inflamación/patología , Macrófagos/inmunología , Proteína p53 Supresora de Tumor/fisiología , Neoplasias de la Vejiga Urinaria/patología , Urotelio/patología , Animales , Carcinogénesis/genética , Carcinogénesis/inmunología , Bases de Datos Genéticas/estadística & datos numéricos , Modelos Animales de Enfermedad , Humanos , Inflamación/genética , Inflamación/inmunología , Inflamación/metabolismo , Ratones , Ratones Endogámicos C3H , Ratones Noqueados , Neoplasias de la Vejiga Urinaria/genética , Neoplasias de la Vejiga Urinaria/inmunología , Neoplasias de la Vejiga Urinaria/metabolismo , Urotelio/inmunología
14.
Cancer Sci ; 100(7): 1219-26, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19432895

RESUMEN

Chronic myelogenous leukemia (CML) is a hematological malignancy that begins as indolent chronic phase (CP) but inevitably progresses to fatal blast crisis (BC). p210BCR/ABL, a chimeric protein with enhanced kinase activity, initiates CML CP, and additional genetic alterations account for progression to BC, but the precise mechanisms underlying disease evolution are not fully understood. In the present study, we investigated the possible contribution of dysfunction of Bcl11b, a zinc-finger protein required for thymocyte differentiation, and of H2AX, a histone protein involved in DNA repair, to the transition from CML CP to BC. For this purpose, we crossed CML CP-exhibiting p210BCR/ABL transgenic (BA(tg/-)) mice with Bcl11b heterozygous (Bcl11b(+/-)) mice and H2AX heterozygous (H2AX(+/-)) mice. Interestingly, p210BCR/ABL transgenic, Bcl11b heterozygous (BA(tg/-)Bcl11b(+/-)) mice and p210BCR/ABL transgenic, H2AX heterozygous (BA(tg/-)H2AX(+/-)) mice frequently developed CML BC with T-cell phenotype and died in a short period. In addition, whereas p210BCR/ABL was expressed in all of the leukemic tissues, the expression of Bcl11b and H2AX was undetectable in several tumors, which was attributed to the loss of the residual normal allele or the lack of mRNA expression. These results indicate that Bcl11b and H2AX function as tumor suppressor and that haploinsufficiency and acquired loss of these gene products cooperate with p210BCR/ABL to develop CML BC.


Asunto(s)
Crisis Blástica/genética , Histonas/genética , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Leucemia Mielógena Crónica BCR-ABL Positiva/patología , Proteínas Represoras/genética , Proteínas Supresoras de Tumor/genética , Animales , Aberraciones Cromosómicas , Proteínas de Fusión bcr-abl/genética , Proteínas de Fusión bcr-abl/metabolismo , Haplotipos/genética , Heterocigoto , Histonas/metabolismo , Humanos , Ratones , Ratones Transgénicos , Proteínas Represoras/metabolismo , Proteínas Supresoras de Tumor/metabolismo
15.
Genes Cells ; 13(2): 145-57, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18233957

RESUMEN

p130Cas (Cas, Crk-associated substrate) is an adaptor molecule composed of a Src homology 3 (SH3) domain, a substrate domain (SD) and a Src binding domain (SBD). The SH3 domain of Cas associates with focal adhesion kinase (FAK), but its role in cellular function has not fully been understood. To address this issue, we established and analyzed primary fibroblasts derived from mice expressing a truncated Cas lacking exon 2, which encodes the SH3 domain (Cas Deltaexon 2). In comparison to wild-type cells, Cas exon 2(Delta/Delta) cells showed reduced motility, which could be due to impaired tyrosine-phosphorylation of FAK and Cas, reduced FAK/Cas/Src/CrkII binding, and also impaired localization of Cas Deltaexon 2 to focal adhesions on fibronectin. In addition, to analyze downstream signaling pathways regulated by Cas exon 2, we performed microarray analyses. Interestingly, we found that a deficiency of Cas exon 2 up-regulated expression of CXC Chemokine Receptor-4 and CC Chemokine Receptor-5, which may be regulated by IkappaBalpha phosphorylation. These results indicate that the SH3-encoding exon of Cas participates in cell motility, tyrosine-phosphorylation of FAK and Cas, FAK/Cas/Src/CrkII complex formation, recruitment of Cas to focal adhesions and regulation of cell motility-associated gene expression in primary fibroblasts.


Asunto(s)
Proteína Sustrato Asociada a CrK/genética , Animales , Movimiento Celular , Células Cultivadas , Proteína Sustrato Asociada a CrK/química , Proteína Sustrato Asociada a CrK/metabolismo , Exones , Fibroblastos/metabolismo , Quinasa 1 de Adhesión Focal/química , Quinasa 1 de Adhesión Focal/metabolismo , Adhesiones Focales/metabolismo , Expresión Génica , Proteínas I-kappa B/metabolismo , Ratones , Ratones Noqueados , Modelos Biológicos , Complejos Multiproteicos , Inhibidor NF-kappaB alfa , Proteínas Proto-Oncogénicas c-crk/química , Proteínas Proto-Oncogénicas c-crk/metabolismo , Receptores CCR5/genética , Receptores CXCR4/genética , Eliminación de Secuencia , Transducción de Señal , Trombospondinas/genética , Cicatrización de Heridas , Dominios Homologos src
17.
Sci Rep ; 6: 29454, 2016 07 19.
Artículo en Inglés | MEDLINE | ID: mdl-27432459

RESUMEN

Polycomb repressive complex 2 (PRC2) participates in transcriptional repression through methylation of histone H3K27. The WD-repeat protein embryonic ectoderm development (EED) is a non-catalytic but an essential component of PRC2 and its mutations were identified in hematopoietic malignancies. To clarify the role(s) of EED in adult hematopoiesis and leukemogenesis, we generated Eed conditional knockout (Eed(Δ/Δ)) mice. Eed(Δ/Δ) mice died in a short period with rapid decrease of hematopoietic cells. Hematopoietic stem/progenitor cells (HSPCs) were markedly decreased with impaired bone marrow (BM) repopulation ability. Cell cycle analysis of HSPCs demonstrated increased S-phase fraction coupled with suppressed G0/G1 entry. Genes encoding cell adhesion molecules are significantly enriched in Eed(Δ/Δ) HSPCs, and consistently, Eed(Δ/Δ) HSPCs exhibited increased attachment to a major extracellular matrix component, fibronectin. Thus, EED deficiency increases proliferation on one side but promotes quiescence possibly by enhanced adhesion to the hematopoietic niche on the other, and these conflicting events would lead to abnormal differentiation and functional defect of Eed(Δ/Δ) HSPCs. In addition, Eed haploinsufficiency induced hematopoietic dysplasia, and Eed heterozygous mice were susceptible to malignant transformation and developed leukemia in cooperation with Evi1 overexpression. Our results demonstrated differentiation stage-specific and dose-dependent roles of EED in normal hematopoiesis and leukemogenesis.


Asunto(s)
Haploinsuficiencia , Hematopoyesis , Leucemia/metabolismo , Complejo Represivo Polycomb 2/genética , Complejo Represivo Polycomb 2/fisiología , Animales , Antígenos CD34/metabolismo , Adhesión Celular , Ciclo Celular , Diferenciación Celular , Proliferación Celular , Transformación Celular Neoplásica , Matriz Extracelular/metabolismo , Femenino , Sangre Fetal/citología , Fibronectinas/química , Fibronectinas/metabolismo , Citometría de Flujo , Técnicas de Transferencia de Gen , Neoplasias Hematológicas/genética , Neoplasias Hematológicas/metabolismo , Células Madre Hematopoyéticas/citología , Heterocigoto , Histonas , Leucemia/genética , Masculino , Ratones , Ratones Endogámicos C57BL , ARN Interferente Pequeño/metabolismo
18.
J Biochem ; 131(6): 773-9, 2002 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12038971

RESUMEN

Platelet-activating factor (PAF) is a pro-inflammatory lipid mediator possessing a unique 1-O-alkyl glycerophospholipid (GPC) backbone (1-O-alkyl-2-acetyl-sn-glycero-3-phosphocholin). Cloned PAF receptor, which belongs to the G protein-coupled receptor superfamily, transduces pleiotropic functions including cell motility, smooth muscle contraction, and synthesis and release of mediators and cytokines via multiple heterotrimeric G proteins. Pharmacological studies have suggested that PAF functions in a variety of settings including allergy, inflammation, neural functions, reproduction, and atherosclerosis. Establishment of PAFR(-/-) mice confirmed that the PAF receptor is responsible for pro-inflammatory responses, but that its roles in other settings remain to be clarified.


Asunto(s)
Factor de Activación Plaquetaria/metabolismo , Glicoproteínas de Membrana Plaquetaria/fisiología , Receptores de Superficie Celular/fisiología , Receptores Acoplados a Proteínas G , Transducción de Señal/fisiología , Secuencia de Aminoácidos , Animales , Humanos , Proteínas de la Membrana/química , Proteínas de la Membrana/fisiología , Glicoproteínas de Membrana Plaquetaria/química , Conformación Proteica , Receptores de Superficie Celular/química
19.
PLoS One ; 9(1): e87425, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24498102

RESUMEN

A20 is a negative regulator of NF-κB, and mutational loss of A20 expression is involved in the pathogenesis of autoimmune diseases and B-cell lymphomas. To clarify the role of A20 in adult hematopoiesis, we generated conditional A20 knockout mice (A20(flox/flox) ) and crossed them with Mx-1Cre (MxCre (+)) and ERT2Cre (ERT2Cre (+)) transgenic mice in which Cre is inducibly activated by endogenous interferon and exogenous tamoxifen, respectively. A20(flox/flox) MxCre (+) (A20Mx) mice spontaneously exhibited myeloid proliferation, B cell apoptosis, and anemia with overproduction of pro-inflammatory cytokines. Bone marrow transplantation demonstrated that these changes were caused by hematopoietic cells. NF-κB was constitutively activated in A20Mx hematopoietic stem cells (HSCs), which caused enhanced cell cycle entry and impaired repopulating ability. Tamoxifen stimulation of A20(flox/flox) ERT2Cre (+) (A20ERT2) mice induced fulminant apoptosis and subsequent myeloproliferation, lymphocytopenia, and progressive anemia with excessive production of pro-inflammatory cytokines, as observed in A20Mx mice. These results demonstrate that A20 plays essential roles in the homeostasis of adult hematopoiesis by preventing apoptosis and inflammation. Our findings provide insights into the mechanism underlying A20 dysfunction and human diseases in which A20 expression is impaired.


Asunto(s)
Apoptosis/fisiología , Proliferación Celular , Proteínas de Unión al ADN/metabolismo , Hematopoyesis/fisiología , Células Madre Hematopoyéticas/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Células Mieloides/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Cisteína Endopeptidasas , Citocinas/metabolismo , Proteínas de Unión al ADN/genética , Células Madre Hematopoyéticas/citología , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Ratones , Ratones Noqueados , Células Mieloides/citología , FN-kappa B/metabolismo , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa , Ubiquitina-Proteína Ligasas/genética
20.
FEBS Lett ; 587(10): 1529-35, 2013 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-23583447

RESUMEN

CIZ1 is a nuclear protein involved in DNA replication and is also implicated in human diseases including cancers. To gain an insight into its function in vivo, we generated mice lacking Ciz1. Ciz1-deficient (Ciz1(-/-)) mice grew without any obvious abnormalities, and Ciz1(-/-) mouse embryonic fibroblasts (MEFs) did not show any defects in cell cycle status, cell growth, and DNA damage response. However, Ciz1(-/-) MEFs were sensitive to hydroxyurea-mediated replication stress and susceptible to oncogene-induced cellular transformation. In addition, Ciz1(-/-) mice developed various types of leukemias by retroviral insertional mutagenesis. These results indicate that CIZ1 functions as a tumor suppressor in vivo.


Asunto(s)
Proteínas Nucleares/fisiología , Proteínas Supresoras de Tumor , Animales , Proteínas de la Ataxia Telangiectasia Mutada , Ciclo Celular/genética , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Proliferación Celular , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Células Cultivadas , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Embrión de Mamíferos/citología , Fibroblastos/metabolismo , Fibroblastos/fisiología , Ratones , Ratones Noqueados , Modelos Biológicos , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Unión Proteica , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo , Proteínas Supresoras de Tumor/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA