Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 68
Filtrar
1.
Int J Mol Sci ; 22(3)2021 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-33499314

RESUMEN

The Ikaros zing-finger family transcription factors (IKZF TFs) are important regulators of lymphocyte development and differentiation and are also highly expressed in B cell malignancies, including Multiple Myeloma (MM), where they are required for cancer cell growth and survival. Moreover, IKZF TFs negatively control the functional properties of many immune cells. Thus, the targeting of these proteins has relevant therapeutic implications in cancer. Indeed, accumulating evidence demonstrated that downregulation of Ikaros and Aiolos, two members of the IKZF family, in malignant plasma cells as well as in adaptative and innate lymphocytes, is key for the anti-myeloma activity of Immunomodulatory drugs (IMiDs). This review is focused on IKZF TF-related pathways in MM. In particular, we will address how the depletion of IKZF TFs exerts cytotoxic effects on MM cells, by reducing their survival and proliferation, and concomitantly potentiates the antitumor immune response, thus contributing to therapeutic efficacy of IMiDs, a cornerstone in the treatment of this neoplasia.


Asunto(s)
Factor de Transcripción Ikaros/fisiología , Mieloma Múltiple/tratamiento farmacológico , Animales , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular , Supervivencia Celular , Humanos , Factor de Transcripción Ikaros/genética , Inmunidad/efectos de los fármacos , Inmunomodulación/efectos de los fármacos , Lenalidomida/farmacología , Linfocitos/citología , Ratones , Mieloma Múltiple/inmunología , Talidomida/farmacología
2.
Blood ; 132(6): 577-586, 2018 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-29954751

RESUMEN

Primary effusion lymphoma (PEL) is an aggressive cancer with few treatment options. The immunomodulatory drugs (IMiDs) lenalidomide and pomalidomide have recently been shown to kill PEL cell lines, and lenalidomide is in clinical trials against PEL. IMiDs bind to the CRL4CRBN E3 ubiquitin ligase complex, leading to the acquisition of the Ikaros family zinc finger proteins 1 and 3 (IKZF1 and IKZF3), casein kinase 1 α (CK1α), and zinc finger protein 91 (ZFP91) as neosubstrates. IMiDs are effective against multiple myeloma because of degradation of IKZF1 and IKZF3 and the consequent loss of interferon regulatory factor 4 (IRF4) and MYC expression. Lenalidomide is also effective in chromosome 5q deletion-associated myelodysplastic syndrome as a result of degradation of CK1α. An essential IKZF1-IRF4-MYC axis has recently been proposed to underlie the toxicity of IMiDs in PEL. Here, we further investigate IMiD effectors in PEL cell lines, based on genome-wide CRISPR/Cas9 screens for essential human genes. These screens and extensive validation experiments show that, of the 4 neosubstrates, only CK1α is essential for the survival of PEL cell lines. In contrast, IKZF1 and IKZF3 are dispensable, individually or in combination. IRF4 was critical in all 8 PEL cell lines tested, and surprisingly, IMiDs triggered downregulation of IRF4 expression independently of both IKZF1 and IKZF3. Reexpression of CK1α and/or IRF4 partially rescued PEL cell lines from IMiD-mediated toxicity. In conclusion, IMiD toxicity in PEL cell lines is independent of IKZF1 and IKZF3 but proceeds through degradation of the neosubstrate CK1α and downregulation of IRF4.


Asunto(s)
Caseína Quinasa Ialfa/fisiología , Factores Inmunológicos/farmacología , Factores Reguladores del Interferón/fisiología , Lenalidomida/farmacología , Linfoma de Efusión Primaria/tratamiento farmacológico , Proteínas de Neoplasias/fisiología , Talidomida/análogos & derivados , Sistemas CRISPR-Cas , Caseína Quinasa Ialfa/genética , Línea Celular Tumoral , Regulación hacia Abajo/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Técnicas de Inactivación de Genes , Humanos , Factor de Transcripción Ikaros/fisiología , Factores Inmunológicos/uso terapéutico , Factores Reguladores del Interferón/biosíntesis , Factores Reguladores del Interferón/genética , Lenalidomida/uso terapéutico , Linfoma de Efusión Primaria/genética , Linfoma de Efusión Primaria/metabolismo , Terapia Molecular Dirigida , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/genética , Interferencia de ARN , ARN Interferente Pequeño/genética , Transducción de Señal , Talidomida/farmacología , Talidomida/uso terapéutico , Ubiquitina-Proteína Ligasas/fisiología
3.
Biochem Biophys Res Commun ; 510(1): 128-134, 2019 02 26.
Artículo en Inglés | MEDLINE | ID: mdl-30683310

RESUMEN

Unveiling the mechanism of the relapse of acute lymphoblastic leukemia (ALL) is the key to improve the prognosis of ALL and remains a huge challenge. Glycan-based interactions play a vital role in immune surveillance, cell-cell adhesion and cell-matrix interaction, contributing to treatment failure in tumor. However, the glycan essential for leukemia development and its upstream regulatory mechanism by oncogenic drivers were rarely reported. Here, we demonstrated that LeX, a well-characterized cancer-related glycan epitope, strengthened the cell-matrix interaction via glycosylating α5ß1 integrin under the control of the driver oncogenic Ikaros isoform (IK6) in ALL. By analyzing the expression profile of Ikaros and the level of FUT4/LeX in clinical samples, we found that FUT4/LeX was positively correlated with dysfunctional Ikaros isoforms. IK1 (Full length Ikaros) regulates the level of FUT4 as a transcription repressor, while IK6 abolished the wild-type Ikaros mediated transcriptional repression and resulted in higher level of FUT4 expression. Moreover, we demonstrated that FUT4 could activate α5ß1-mediated sequential signal transduction and accelerate adhesion and invasion between integrin α5ß1 in leukemia cells and fibronectin in extracellular matrix (ECM) via increasing glycosylation. Together, our study provides a new insight into the mechanisms by which Ikaros mutation induced ALL cells invasion and a potential strategy for drug-resistance ALL by blocking LeX in combination with common chemotherapy.


Asunto(s)
Empalme Alternativo/fisiología , Factor de Transcripción Ikaros/fisiología , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , Células Cultivadas , Quinasa 1 de Adhesión Focal/metabolismo , Fucosiltransferasas/metabolismo , Glicosilación , Humanos , Factor de Transcripción Ikaros/genética , Integrina alfa5beta1/metabolismo , Antígeno Lewis X/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Transducción de Señal
4.
Trends Genet ; 31(9): 500-8, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26049627

RESUMEN

Transcription factors are important determinants of lineage specification during hematopoiesis. They favor recruitment of cofactors involved in epigenetic regulation, thereby defining patterns of gene expression in a development- and lineage-specific manner. Additionally, transcription factors can facilitate transcription preinitiation complex (PIC) formation and assembly on chromatin. Interestingly, a few lineage-specific transcription factors, including IKAROS, also regulate transcription elongation. IKAROS is a tumor suppressor frequently inactivated in leukemia and associated with a poor prognosis. It forms a complex with the nucleosome remodeling and deacetylase (NuRD) complex and the positive transcription elongation factor b (P-TEFb), which is required for productive transcription elongation. It has also been reported that IKAROS interacts with factors involved in transcription termination. Here we review these and other recent findings that establish IKAROS as the first transcription factor found to act as a multifunctional regulator of the transcription cycle in hematopoietic cells.


Asunto(s)
Ensamble y Desensamble de Cromatina , ADN Polimerasa II/fisiología , Factor de Transcripción Ikaros/fisiología , Transcripción Genética , Animales , Regulación de la Expresión Génica , Humanos
5.
Haematologica ; 103(4): 565-574, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29519871

RESUMEN

Transcription factor IKZF1 (IKAROS) acts as a critical regulator of lymphoid differentiation and is frequently deleted or mutated in B-cell precursor acute lymphoblastic leukemia. IKZF1 gene defects are associated with inferior treatment outcome in both childhood and adult B-cell precursor acute lymphoblastic leukemia and occur in more than 70% of BCR-ABL1-positive and BCR-ABL1-like cases of acute lymphoblastic leukemia. Over the past few years, much has been learned about the tumor suppressive function of IKZF1 during leukemia development and the molecular pathways that relate to its impact on treatment outcome. In this review, we provide a concise overview on the role of IKZF1 during normal lymphopoiesis and the pathways that contribute to leukemia pathogenesis as a consequence of altered IKZF1 function. Furthermore, we discuss different mechanisms by which IKZF1 alterations impose therapy resistance on leukemic cells, including enhanced cell adhesion and modulation of glucocorticoid response.


Asunto(s)
Factor de Transcripción Ikaros/genética , Factor de Transcripción Ikaros/fisiología , Adulto , Adhesión Celular/genética , Niño , Resistencia a Medicamentos/genética , Eliminación de Gen , Humanos , Mutación
6.
Nucleic Acids Res ; 44(22): 10644-10661, 2016 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-27604872

RESUMEN

Aberrant stem cell-like gene regulatory networks are a feature of leukaemogenesis. The ETS-related gene (ERG), an important regulator of normal haematopoiesis, is also highly expressed in T-ALL and acute myeloid leukaemia (AML). However, the transcriptional regulation of ERG in leukaemic cells remains poorly understood. In order to discover transcriptional regulators of ERG, we employed a quantitative mass spectrometry-based method to identify factors binding the 321 bp ERG +85 stem cell enhancer region in MOLT-4 T-ALL and KG-1 AML cells. Using this approach, we identified a number of known binders of the +85 enhancer in leukaemic cells along with previously unknown binders, including ETV6 and IKZF1. We confirmed that ETV6 and IKZF1 were also bound at the +85 enhancer in both leukaemic cells and in healthy human CD34+ haematopoietic stem and progenitor cells. Knockdown experiments confirmed that ETV6 and IKZF1 are transcriptional regulators not just of ERG, but also of a number of genes regulated by a densely interconnected network of seven transcription factors. At last, we show that ETV6 and IKZF1 expression levels are positively correlated with expression of a number of heptad genes in AML and high expression of all nine genes confers poorer overall prognosis.


Asunto(s)
Factor de Transcripción Ikaros/fisiología , Proteínas Proto-Oncogénicas c-ets/fisiología , Proteínas Represoras/fisiología , Transcripción Genética , Secuencia de Bases , Sitios de Unión , Línea Celular Tumoral , Secuencia de Consenso , Elementos de Facilitación Genéticos , Regulación Leucémica de la Expresión Génica , Redes Reguladoras de Genes , Humanos , Estimación de Kaplan-Meier , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/mortalidad , Pronóstico , Modelos de Riesgos Proporcionales , Unión Proteica , Proteoma , Proteómica , Regulador Transcripcional ERG/fisiología , Proteína ETS de Variante de Translocación 6
7.
Proc Natl Acad Sci U S A ; 110(8): E716-25, 2013 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-23382203

RESUMEN

During cerebral cortex development, a series of projection neuron types is generated in a fixed temporal order. In Drosophila neuroblasts, the transcription factor hunchback encodes first-born identity within neural lineages. One of its mammalian homologs, Ikaros, was recently reported to play an equivalent role in retinal progenitor cells, raising the question as to whether Ikaros/Hunchback proteins could be general factors regulating the development of early-born fates throughout the nervous system. Ikaros is also expressed in progenitor cells of the mouse cerebral cortex, and this expression is highest during the early stages of neurogenesis and thereafter decreases over time. Transgenic mice with sustained Ikaros expression in cortical progenitor cells and neurons have developmental defects, including displaced progenitor cells within the cortical plate, increased early neural differentiation, and disrupted cortical lamination. Sustained Ikaros expression results in a prolonged period of generation of deep layer neurons into the stages when, normally, only late-born upper layer neurons are generated, as well as a delayed production of late-born neurons. Consequently, more early-born and fewer late-born neurons are present in the cortex of these mice at birth. This phenotype was observed in all parts of the cortex, including those with minimal structural defects, demonstrating that it is not secondary to abnormalities in cortical morphogenesis. These data suggest that Ikaros plays a similar role in regulating early temporal fates in the mammalian cerebral cortex as Ikaros/Hunchback proteins do in the Drosophila nerve cord.


Asunto(s)
Corteza Cerebral/citología , Factor de Transcripción Ikaros/fisiología , Neuronas/citología , Animales , Animales Modificados Genéticamente , Linaje de la Célula , Electroporación , Hibridación in Situ , Reacción en Cadena de la Polimerasa , Ratas , Ratas Sprague-Dawley
8.
Blood ; 121(13): 2440-51, 2013 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-23335373

RESUMEN

The transcription factor Ikaros regulates the development of hematopoietic cells. Ikaros-deficient animals fail to develop B cells and display a T-cell malignancy, which is correlated with altered Notch signaling. Recently, loss of Ikaros was associated with progression of myeloproliferative neoplasms to acute myeloid leukemia and increasing evidence shows that Ikaros is also critical for the regulation of myeloid development. Previous studies showed that Ikaros-deficient mice have increased megakaryopoiesis, but the molecular mechanism of this phenomenon remains unknown. Here, we show that Ikaros overexpression decreases NOTCH-induced megakaryocytic specification, and represses expression of several megakaryocytic genes including GATA-1 to block differentiation and terminal maturation. We also demonstrate that Ikaros expression is differentially regulated by GATA-2 and GATA-1 during megakaryocytic differentiation and reveal that the combined loss of Ikzf1 and Gata1 leads to synthetic lethality in vivo associated with prominent defects in erythroid cells and an expansion of megakaryocyte progenitors. Taken together, our observations demonstrate an important functional interplay between Ikaros, GATA factors, and the NOTCH signaling pathway in specification and homeostasis of the megakaryocyte lineage.


Asunto(s)
Factor de Transcripción GATA1/metabolismo , Factor de Transcripción Ikaros/fisiología , Receptores Notch/metabolismo , Trombopoyesis/genética , Animales , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Proliferación Celular , Células Cultivadas , Regulación hacia Abajo/genética , Embrión de Mamíferos , Regulación del Desarrollo de la Expresión Génica , Factor de Transcripción Ikaros/genética , Factor de Transcripción Ikaros/metabolismo , Megacariocitos/metabolismo , Megacariocitos/fisiología , Ratones , Ratones Noqueados , Modelos Biológicos , Unión Proteica/genética , Unión Proteica/fisiología , Transducción de Señal/genética , Transducción de Señal/fisiología
9.
Blood ; 122(15): 2572-81, 2013 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-23990620

RESUMEN

The Ikaros gene (Ikzf1) encodes a family of zinc-finger transcription factors implicated in hematopoietic cell differentiation. Here we show that Ikaros suppresses the development of basophils, which are proinflammatory cells of the myeloid lineage. In the absence of extrinsic basophil-inducing signals, Ikaros(-/-) (Ik(-/-)) mice exhibit increases in basophil numbers in blood and bone marrow and in their direct precursors in bone marrow and the spleen, as well as decreased numbers of intestinal mast cells. In vitro culture of Ik(-/-) bone marrow under mast cell differentiation conditions also results in predominance of basophils. Basophil expansion is associated with an increase in basophil progenitors, increased expression of Cebpa and decreased expression of mast cell-specifying genes Hes1 and microphthalmia-associated transcription factor (Mitf). Ikaros directly associates with regulatory sites within Cebpa and Hes1 and regulates the acquisition of permissive H3K4 tri-methylation marks at the Cebpa locus and reduces H3K4 tri-methylation at the Hes1 promoter. Ikaros blockade in cultured cells or transfer of Ik(-/-) bone marrow into irradiated Ik(+/+) recipients also results in increased basophils confirming a cell-intrinsic effect of Ikaros on basophil development. We conclude that Ikaros is a suppressor of basophil differentiation under steady-state conditions and that it acts by regulating permissive chromatin modifications of Cebpa.


Asunto(s)
Basófilos/citología , Basófilos/fisiología , Proteínas Potenciadoras de Unión a CCAAT/genética , Factor de Transcripción Ikaros/genética , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/fisiología , Células de la Médula Ósea/citología , Células de la Médula Ósea/fisiología , Proteínas Potenciadoras de Unión a CCAAT/fisiología , Diferenciación Celular/genética , Linaje de la Célula/genética , Células Cultivadas , Epigénesis Genética/genética , Femenino , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/fisiología , Factor de Transcripción Ikaros/fisiología , Masculino , Mastocitos/citología , Mastocitos/fisiología , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Factor de Transcripción HES-1
10.
Nucleic Acids Res ; 40(15): 7303-18, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22638570

RESUMEN

The sarcomeric myosin gene, Myh7b, encodes an intronic microRNA, miR-499, which regulates cardiac and skeletal muscle biology, yet little is known about its transcriptional regulation. To identify the transcription factors involved in regulating Myh7b/miR-499 gene expression, we have mapped the transcriptional start sites and identified an upstream 6.2 kb region of the mouse Myh7b gene whose activity mimics the expression pattern of the endogenous Myh7b gene both in vitro and in vivo. Through promoter deletion analysis, we have mapped a distal E-box element and a proximal Ikaros site that are essential for Myh7b promoter activity in muscle cells. We show that the myogenic regulatory factors, MyoD, Myf5 and Myogenin, bind to the E-box, while a lymphoid transcription factor, Ikaros 4 (Eos), binds to the Ikaros motif. Further, we show that through physical interaction, MyoD and Eos form an active transcriptional complex on the chromatin to regulate the expression of the endogenous Myh7b/miR-499 gene in muscle cells. We also provide the first evidence that Eos can regulate expression of additional myosin genes (Myosin 1 and ß-Myosin) via the miR-499/Sox6 pathway. Therefore, our results indicate a novel role for Eos in the regulation of the myofiber gene program.


Asunto(s)
Proteínas Portadoras/metabolismo , Regulación de la Expresión Génica , Factor de Transcripción Ikaros/metabolismo , MicroARNs/genética , Factores Reguladores Miogénicos/metabolismo , Cadenas Pesadas de Miosina/genética , Miosina Tipo II/genética , Proteínas del Tejido Nervioso/metabolismo , Transcripción Genética , Animales , Secuencia de Bases , Proteínas Portadoras/química , Proteínas Portadoras/fisiología , Células Cultivadas , Proteínas de Unión al ADN , Elementos E-Box , Humanos , Factor de Transcripción Ikaros/fisiología , Ratones , MicroARNs/biosíntesis , Datos de Secuencia Molecular , Músculo Esquelético/metabolismo , Proteína MioD/metabolismo , Miocardio/metabolismo , Factores Reguladores Miogénicos/fisiología , Cadenas Pesadas de Miosina/biosíntesis , Miosina Tipo II/biosíntesis , Proteínas del Tejido Nervioso/química , Proteínas del Tejido Nervioso/fisiología , Regiones Promotoras Genéticas , Dominios y Motivos de Interacción de Proteínas , Sitio de Iniciación de la Transcripción
11.
Blood ; 116(25): 5443-54, 2010 Dec 16.
Artículo en Inglés | MEDLINE | ID: mdl-20829372

RESUMEN

The Notch pathway is frequently activated in T-cell acute lymphoblastic leukemias (T-ALLs). Of the Notch receptors, Notch1 is a recurrent target of gain-of-function mutations and Notch3 is expressed in all T-ALLs, but it is currently unclear how these receptors contribute to T-cell transformation in vivo. We investigated the role of Notch1 and Notch3 in T-ALL progression by a genetic approach, in mice bearing a knockdown mutation in the Ikaros gene that spontaneously develop Notch-dependent T-ALL. While deletion of Notch3 has little effect, T cell-specific deletion of floxed Notch1 promoter/exon 1 sequences significantly accelerates leukemogenesis. Notch1-deleted tumors lack surface Notch1 but express γ-secretase-cleaved intracellular Notch1 proteins. In addition, these tumors accumulate high levels of truncated Notch1 transcripts that are caused by aberrant transcription from cryptic initiation sites in the 3' part of the gene. Deletion of the floxed sequences directly reprograms the Notch1 locus to begin transcription from these 3' promoters and is accompanied by an epigenetic reorganization of the Notch1 locus that is consistent with transcriptional activation. Further, spontaneous deletion of 5' Notch1 sequences occurs in approximately 75% of Ikaros-deficient T-ALLs. These results reveal a novel mechanism for the oncogenic activation of the Notch1 gene after deletion of its main promoter.


Asunto(s)
Factor de Transcripción Ikaros/fisiología , Leucemia-Linfoma Linfoblástico de Células T Precursoras/genética , Regiones Promotoras Genéticas/genética , Receptor Notch1/genética , Activación Transcripcional/fisiología , Animales , Northern Blotting , Western Blotting , Transformación Celular Neoplásica , Cartilla de ADN/química , Cartilla de ADN/genética , Citometría de Flujo , Regulación Neoplásica de la Expresión Génica , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/fisiología , Ratones , Ratones Noqueados , Mutación/genética , Leucemia-Linfoma Linfoblástico de Células T Precursoras/metabolismo , Leucemia-Linfoma Linfoblástico de Células T Precursoras/patología , ARN Mensajero/genética , Receptor Notch3 , Receptores Notch/fisiología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Eliminación de Secuencia , Tasa de Supervivencia
12.
Blood ; 116(25): 5455-64, 2010 Dec 16.
Artículo en Inglés | MEDLINE | ID: mdl-20852131

RESUMEN

Point mutations that trigger ligand-independent proteolysis of the Notch1 ectodomain occur frequently in human T-cell acute lymphoblastic leukemia (T-ALL) but are rare in murine T-ALL, suggesting that other mechanisms account for Notch1 activation in murine tumors. Here we show that most murine T-ALLs harbor Notch1 deletions that fall into 2 types, both leading to ligand-independent Notch1 activation. Type 1 deletions remove exon 1 and the proximal promoter, appear to be RAG-mediated, and are associated with mRNA transcripts that initiate from 3' regions of Notch1. In line with the RAG dependency of these rearrangements, RAG2 binds to the 5' end of Notch1 in normal thymocytes near the deletion breakpoints. Type 2 deletions remove sequences between exon 1 and exons 26 to 28 of Notch1, appear to be RAG-independent, and are associated with transcripts in which exon 1 is spliced out of frame to 3' Notch1 exons. Translation of both types of transcripts initiates at a conserved methionine residue, M1727, which lies within the Notch1 transmembrane domain. Polypeptides initiating at M1727 insert into membranes and are subject to constitutive cleavage by γ-secretase. Thus, like human T-ALL, murine T-ALL is often associated with acquired mutations that cause ligand-independent Notch1 activation.


Asunto(s)
Proteínas de Homeodominio/fisiología , Iniciación de la Cadena Peptídica Traduccional/genética , Leucemia-Linfoma Linfoblástico de Células T Precursoras/genética , Regiones Promotoras Genéticas/genética , Receptor Notch1/genética , Activación Transcripcional/fisiología , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Northern Blotting , Southern Blotting , Western Blotting , Neoplasias Óseas/genética , Neoplasias Óseas/metabolismo , Neoplasias Óseas/patología , Regulación Neoplásica de la Expresión Génica , Humanos , Factor de Transcripción Ikaros/fisiología , Ratones , Ratones Noqueados , Datos de Secuencia Molecular , Mutación/genética , Osteosarcoma/genética , Osteosarcoma/metabolismo , Osteosarcoma/patología , Leucemia-Linfoma Linfoblástico de Células T Precursoras/metabolismo , Leucemia-Linfoma Linfoblástico de Células T Precursoras/patología , ARN Mensajero/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Eliminación de Secuencia , Homología de Secuencia de Ácido Nucleico , Células Tumorales Cultivadas
13.
J Immunol ; 185(1): 410-7, 2010 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-20511547

RESUMEN

Ikaros and Notch are transcriptional regulators essential for normal T cell development. Aberrant activation of Notch target genes is observed in Ikaros-deficient thymocytes as well as leukemia cell lines. However, it is not known whether Notch deregulation plays a preferential or obligatory role in the leukemia that arise in Ikaros null (Ik(-/-)) mice. To answer this question, the expression of the DNA-binding Notch target gene activator RBP-Jkappa was abrogated in Ik(-/-) double-positive thymocytes. This was accomplished through conditional inactivation using CD4-Cre transgenic mice containing floxed RBP-Jkappa alleles (RBPJ(fl/fl)). Ik(-/-) x RBPJ(fl/fl) x CD4-Cre(+) transgenic mice develop clonal T cell populations in the thymus that escape to the periphery, with similar kinetics and penetrance as their CD4-Cre(-) counterparts. The clonal populations do not display increased RBP-Jkappa expression compared with nontransformed thymocytes, suggesting there is no selection for clones that have not fully deleted RBP-Jkappa. However, RBPJ-deficient clonal populations do not expand as aggressively as their RBPJ-sufficient counterparts, suggesting a qualitative role for deregulated Notch target gene activation in the leukemogenic process. Finally, these studies show that RBP-Jkappa plays no role in Notch target gene repression in double-positive thymocytes but rather that it is Ikaros that is required for the repression of these genes at this critical stage of T cell development.


Asunto(s)
Marcación de Gen , Factor de Transcripción Ikaros/deficiencia , Factor de Transcripción Ikaros/genética , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/genética , Inmunofenotipificación , Leucemia Experimental/genética , Receptores Notch/antagonistas & inhibidores , Receptores Notch/genética , Animales , Línea Celular , Línea Celular Tumoral , Silenciador del Gen/inmunología , Factor de Transcripción Ikaros/fisiología , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/antagonistas & inhibidores , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/metabolismo , Leucemia Experimental/inmunología , Leucemia Experimental/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Receptores Notch/metabolismo , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Subgrupos de Linfocitos T/patología
14.
Dev Biol ; 339(1): 51-64, 2010 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-20026024

RESUMEN

Here we provide evidence for a C2H2 zinc finger gene family with similarity to Ikaros and hunchback. The founding member of this family is Caenorhabditis elegans ehn-3, which has important and poorly understood functions in somatic gonad development. We examined the expression and function of four additional hunchback/Ikaros-like (HIL) genes in C. elegans reproductive system development. Two genes, ehn-3 and R08E3.4, are expressed in somatic gonadal precursors (SGPs) and have overlapping functions in their development. In ehn-3; R08E3.4 double mutants, we find defects in the generation of distal tip cells, anchor cells, and spermatheca; three of the five tissues derived from the SGPs. We provide in vivo evidence that C. elegans HIL proteins have functionally distinct zinc finger domains, with specificity residing in the N-terminal set of four zinc fingers and a likely protein-protein interaction domain provided by the C-terminal pair of zinc fingers. In addition, we find that a chimeric human Ikaros protein containing the N-terminal zinc fingers of EHN-3 functions in C. elegans. Together, these results lend support to the idea that the C. elegans HIL genes and Ikaros have similar functional domains. We propose that hunchback, Ikaros, and the HIL genes arose from a common ancestor that was present prior to the divergence of protostomes and deuterostomes.


Asunto(s)
Proteínas de Caenorhabditis elegans/fisiología , Caenorhabditis elegans/embriología , Genitales/embriología , Factor de Transcripción Ikaros/fisiología , Dedos de Zinc , Alelos , Animales , Interferencia de ARN , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
15.
J Immunol ; 183(9): 5518-25, 2009 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-19828627

RESUMEN

IL-10 is a regulatory cytokine critical for controlling inflammatory responses. Here we show that Ikaros, a zinc finger DNA-binding protein, plays an important role in the regulation of Il10 in murine CD4(+) T cells. Upon initial stimulation of the TCR, T cells deficient in Ikaros express significantly lower levels of IL-10 compared with wild-type T cells. In addition, under Th2 skewing conditions, which induce IL-10 production by wild-type T cells, Ikaros null T cells are unable to properly differentiate, producing only low levels of IL-10. Expression of a dominant-negative isoform of Ikaros in wild-type Th2 cells represses IL-10 production but does not significantly alter expression levels of the genes encoding the transcription factors GATA-3 and T-bet. Furthermore, expression of Ikaros in Ikaros null T cells restores expression of the Th2 cytokines IL-10 and IL-4 while reducing production of the Th1 cytokine, IFN-gamma. Coexpression of Ikaros and GATA-3 further increases IL-10 production, showing that these two factors have an additive effect on activating Il10 expression. Finally, we show that Ikaros binds to conserved regulatory regions of the Il10 gene locus in Th2 cells, supporting a direct role for Ikaros in Il10 expression. Thus, we provide evidence for Ikaros as a regulator of Il10 and Ifng gene expression and suggest a role for Ikaros in directing lineage-specific cytokine gene activation and repression.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Factor de Transcripción Ikaros/fisiología , Interleucina-10/biosíntesis , Animales , Células Cultivadas , Citocinas/biosíntesis , Citocinas/deficiencia , Citocinas/genética , Regulación de la Expresión Génica/inmunología , Humanos , Factor de Transcripción Ikaros/deficiencia , Factor de Transcripción Ikaros/genética , Interferón gamma/biosíntesis , Interferón gamma/genética , Interleucina-10/genética , Activación de Linfocitos/genética , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Ratas
16.
J Immunol ; 183(4): 2303-11, 2009 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-19620299

RESUMEN

Helios is a member of the Ikaros family of zinc finger transcription factors. It is expressed mainly in T cells, where it associates with Ikaros-containing complexes and has been proposed to act as a rate-limiting factor for Ikaros function. Overexpression of wild-type or dominant-negative Helios isoforms profoundly alters alphabeta T cell differentiation and activation, and endogenous Helios is expressed at strikingly high levels in regulatory T cells. Helios has also been implicated as a tumor suppressor in human T cell acute lymphoblastic leukemias. These studies suggest a central role for Helios in T cell development and homeostasis, but whether this protein is physiologically required in T cells is unclear. We report herein that inactivation of the Helios gene by homologous recombination does not impair the differentiation and effector cell function of alphabeta and gammadelta T cells, NKT cells, and regulatory T cells. These results suggest that Helios is not essential for T cells, and that its function can be compensated for by other members of the Ikaros family.


Asunto(s)
Diferenciación Celular/genética , Diferenciación Celular/inmunología , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/genética , Subgrupos de Linfocitos T/citología , Subgrupos de Linfocitos T/inmunología , Factores de Transcripción/deficiencia , Factores de Transcripción/genética , Animales , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Proliferación Celular , Células Cultivadas , Células Clonales , Proteínas de Unión al ADN/fisiología , Silenciador del Gen/inmunología , Factor de Transcripción Ikaros/genética , Factor de Transcripción Ikaros/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Familia de Multigenes/inmunología , Receptores de Antígenos de Linfocitos T/fisiología , Subgrupos de Linfocitos T/metabolismo , Factores de Transcripción/fisiología
17.
J Immunol ; 182(2): 741-5, 2009 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-19124715

RESUMEN

Ikaros, a hematopoietic transcription factor, has well defined effects on early lymphocyte development in the bone marrow and thymus. In this study we demonstrate that Ikaros is a positive regulator of Th2 cytokine gene expression in peripheral T cells. CD4+ T cells from naive Ikaros(null) mice cultured under Th2-skewing conditions express the Th1 cytokine IFN-gamma and have reduced IL-4, IL-5, and IL-13 expression. Ikaros directly associates with several Th2 locus regulatory regions in naive CD4+ T cells. The decreased ability to express Th2 cytokines in Ikaros(null)T cells corresponds with histone 3 hypoacetylation across the Th2 cytokine locus as well as decreased GATA3 and cMaf and increased T-bet and STAT1 expression. These data support a model whereby Ikaros directly activates Th2 gene expression by promoting local chromatin accessibility during CD4+ T cell differentiation and also acts indirectly to regulate expression of Th2- and Th1-specific transcription factors.


Asunto(s)
Diferenciación Celular/inmunología , Factor de Transcripción Ikaros/fisiología , Células Th2/citología , Células Th2/inmunología , Regulación hacia Arriba/inmunología , Animales , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Células Cultivadas , Cromatina/genética , Cromatina/metabolismo , Regulación de la Expresión Génica/inmunología , Hematopoyesis/genética , Hematopoyesis/inmunología , Factor de Transcripción Ikaros/deficiencia , Factor de Transcripción Ikaros/genética , Factor de Transcripción Ikaros/metabolismo , Interferón gamma/biosíntesis , Interferón gamma/genética , Interferón gamma/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Inmunológicos , Unión Proteica/genética , Unión Proteica/inmunología , Células TH1/inmunología , Células TH1/metabolismo , Células Th2/metabolismo , Activación Transcripcional/inmunología , Regulación hacia Arriba/genética
18.
Leukemia ; 35(8): 2205-2219, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33483615

RESUMEN

The majority of cases of T-cell acute lymphoblastic leukemia (T-ALL) contain chromosomal abnormalities that drive overexpression of oncogenic transcription factors. However, whether these initiating oncogenes are required for leukemia maintenance is poorly understood. To address this, we developed a tetracycline-regulated mouse model of T-ALL driven by the oncogenic transcription factor Lmo2. This revealed that whilst thymus-resident pre-Leukemic Stem Cells (pre-LSCs) required continuous Lmo2 expression, the majority of leukemias relapsed despite Lmo2 withdrawal. Relapse was associated with a mature phenotype and frequent mutation or loss of tumor suppressor genes including Ikzf1 (Ikaros), with targeted deletion Ikzf1 being sufficient to transform Lmo2-dependent leukemias to Lmo2-independence. Moreover, we found that the related transcription factor TAL1 was dispensable in several human T-ALL cell lines that contain SIL-TAL1 chromosomal deletions driving its overexpression, indicating that evolution to oncogene independence can also occur in human T-ALL. Together these results indicate an evolution of oncogene addiction in murine and human T-ALL and show that loss of Ikaros is a mechanism that can promote self-renewal of T-ALL lymphoblasts in the absence of an initiating oncogenic transcription factor.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/fisiología , Regulación Leucémica de la Expresión Génica , Factor de Transcripción Ikaros/fisiología , Proteínas con Dominio LIM/fisiología , Leucemia-Linfoma Linfoblástico de Células T Precursoras/patología , Animales , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Oncogenes , Leucemia-Linfoma Linfoblástico de Células T Precursoras/etiología , Leucemia-Linfoma Linfoblástico de Células T Precursoras/metabolismo
19.
Curr Opin Immunol ; 19(2): 116-22, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17307348

RESUMEN

Hematopoiesis is the developmental process by which all blood and immune cells are generated. A decade-old scheme has supported an early and complete separation of the erythro-myeloid from the lymphoid lineages. Recent advances have re-drawn this map, separating lymphoid and myeloid from erythroid programs early in development. Subsequently, the fate restriction of both the lympho-myeloid and the erythro-megakaryocyte progenitors is dependent on Ikaros and its associated chromatin regulators. Genetic studies of this family of nuclear factors are now providing unique insight into the functional molecular signatures that bestow plasticity to the hematopoietic stem cell and its early progeny.


Asunto(s)
Cromatina/genética , Células Precursoras Eritroides/metabolismo , Regulación del Desarrollo de la Expresión Génica , Hematopoyesis/genética , Factor de Transcripción Ikaros/fisiología , Células Madre Multipotentes/metabolismo , Animales , Factor de Transcripción Ikaros/genética , Ratones
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda