Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Biochem Biophys Res Commun ; 674: 83-89, 2023 09 24.
Artigo em Inglês | MEDLINE | ID: mdl-37413709

RESUMO

The transcriptional regulators that drive regulatory T (Treg) cell development and function remain partially understood. Helios (Ikzf2) and Eos (Ikzf4) are closely-related members of the Ikaros family of transcription factors. They are highly expressed in CD4+ Treg cells and functionally important for Treg cell biology, as mice deficient for either Helios or Eos are susceptible to autoimmune diseases. However, it remains unknown if these factors exhibit specific or partially redundant functions in Treg cells. Here we show that mice with germline deletions of both Ikzf2 and Ikzf4 are not very different from animals with single Ikzf2 or Ikzf4 deletions. Double knockout Treg cells differentiate normally, and efficiently suppress effector T cell proliferation in vitro. Both Helios and Eos are required for optimal Foxp3 protein expression. Surprisingly, Helios and Eos regulate different, largely non-overlapping, sets of genes. Only Helios is required for proper Treg cell aging, as Helios deficiency results in reduced Treg cell frequencies in the spleen of older animals. These results indicate that Helios and Eos are required for distinct aspects of Treg cell function.


Assuntos
Fator de Transcrição Ikaros , Linfócitos T Reguladores , Animais , Camundongos , Doenças Autoimunes/genética , Suscetibilidade a Doenças/metabolismo , Fatores de Transcrição Forkhead/metabolismo , Fator de Transcrição Ikaros/genética , Fator de Transcrição Ikaros/metabolismo , Fatores de Transcrição/metabolismo
2.
J Exp Med ; 218(10)2021 10 04.
Artigo em Inglês | MEDLINE | ID: mdl-34459852

RESUMO

Our understanding of cell fate decisions in hematopoietic stem cells is incomplete. Here, we show that the transcription factor Helios is highly expressed in murine hematopoietic stem and progenitor cells (HSPCs), where it is required to suppress the separation of the platelet/megakaryocyte lineage from the HSPC pool. Helios acts mainly in quiescent cells, where it directly represses the megakaryocyte gene expression program in cells as early as the stem cell stage. Helios binding promotes chromatin compaction, notably at the regulatory regions of platelet-specific genes recognized by the Gata2 and Runx1 transcriptional activators, implicated in megakaryocyte priming. Helios null HSPCs are biased toward the megakaryocyte lineage at the expense of the lymphoid and partially resemble cells of aging animals. We propose that Helios acts as a guardian of HSPC pluripotency by continuously repressing the megakaryocyte fate, which in turn allows downstream lymphoid priming to take place. These results highlight the importance of negative and positive priming events in lineage commitment.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Células-Tronco Hematopoéticas/fisiologia , Megacariócitos/fisiologia , Fatores de Transcrição/metabolismo , Animais , Diferenciação Celular , Proteínas de Ligação a DNA/genética , Feminino , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Células-Tronco Hematopoéticas/citologia , Linfócitos/citologia , Linfócitos/fisiologia , Masculino , Megacariócitos/citologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Linfócitos T/citologia , Linfócitos T/fisiologia , Fatores de Transcrição/genética
3.
J Immunol ; 207(2): 421-435, 2021 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-34233909

RESUMO

Intracellular ion fluxes emerge as critical actors of immunoregulation but still remain poorly explored. In this study, we investigated the role of the redundant cation channels TMEM176A and TMEM176B (TMEM176A/B) in retinoic acid-related orphan receptor γt+ cells and conventional dendritic cells (DCs) using germline and conditional double knockout mice. Although Tmem176a/b appeared surprisingly dispensable for the protective function of Th17 and group 3 innate lymphoid cells in the intestinal mucosa, we found that they were required in conventional DCs for optimal Ag processing and presentation to CD4+ T cells. Using a real-time imaging method, we show that TMEM176A/B accumulate in dynamic post-Golgi vesicles preferentially linked to the late endolysosomal system and strongly colocalize with HLA-DM. Taken together, our results suggest that TMEM176A/B ion channels play a direct role in the MHC class II compartment of DCs for the fine regulation of Ag presentation and naive CD4+ T cell priming.


Assuntos
Apresentação de Antígeno/imunologia , Linfócitos T CD4-Positivos/imunologia , Células Dendríticas/imunologia , Antígenos de Histocompatibilidade Classe II/imunologia , Proteínas de Membrana/imunologia , Animais , Endossomos/imunologia , Feminino , Genes MHC da Classe II/imunologia , Complexo de Golgi/imunologia , Imunidade Inata/imunologia , Mucosa Intestinal/imunologia , Canais Iônicos/imunologia , Linfócitos/imunologia , Lisossomos/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células Th17/imunologia , Tretinoína/imunologia
4.
J Allergy Clin Immunol ; 143(2): 712-725.e5, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-29800647

RESUMO

BACKGROUND: Autosomal dominant gain-of-function mutations in human stimulator of interferon genes (STING) lead to a severe autoinflammatory disease called STING-associated vasculopathy with onset in infancy that is associated with enhanced expression of interferon-stimulated gene transcripts. OBJECTIVE: The goal of this study was to analyze the phenotype of a new mouse model of STING hyperactivation and the role of type I interferons in this system. METHODS: We generated a knock-in model carrying an amino acid substitution (V154M) in mouse STING, corresponding to a recurrent mutation seen in human patients with STING-associated vasculopathy with onset in infancy. Hematopoietic development and tissue histology were analyzed. Lymphocyte activation and proliferation were assessed in vitro. STING V154M/wild-type (WT) mice were crossed to IFN-α/ß receptor (IFNAR) knockout mice to evaluate the type I interferon dependence of the mutant Sting phenotype recorded. RESULTS: In STING V154M/WT mice we detected variable expression of inflammatory infiltrates in the lungs and kidneys. These mice showed a marked decrease in survival and developed a severe combined immunodeficiency disease (SCID) affecting B, T, and natural killer cells, with an almost complete lack of antibodies and a significant expansion of monocytes and granulocytes. The blockade in B- and T-cell development was present from early immature stages in bone marrow and thymus. In addition, in vitro experiments revealed an intrinsic proliferative defect of mature T cells. Although the V154M/WT mutant demonstrated increased expression of interferon-stimulated genes, the SCID phenotype was not reversed in STING V154M/WT IFNAR knockout mice. However, the antiproliferative defect in T cells was rescued partially by IFNAR deficiency. CONCLUSIONS: STING gain-of-function mice developed an interferon-independent SCID phenotype with a T-cell, B-cell, and natural killer cell developmental defect and hypogammaglobulinemia that is associated with signs of inflammation in lungs and kidneys. Only the intrinsic proliferative defect of T cells was partially interferon dependent.


Assuntos
Linfócitos B/fisiologia , Inflamação/genética , Células Matadoras Naturais/imunologia , Proteínas de Membrana/genética , Mutação/genética , Imunodeficiência Combinada Severa/genética , Linfócitos T/fisiologia , Agamaglobulinemia , Animais , Diferenciação Celular/genética , Modelos Animais de Doenças , Humanos , Interferon Tipo I/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptor de Interferon alfa e beta/genética
5.
PLoS Genet ; 14(7): e1007485, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-30001316

RESUMO

Plasmacytoid and conventional dendritic cells (pDCs and cDCs) arise from monocyte and dendritic progenitors (MDPs) and common dendritic progenitors (CDPs) through gene expression changes that remain partially understood. Here we show that the Ikaros transcription factor is required for DC development at multiple stages. Ikaros cooperates with Notch pathway activation to maintain the homeostasis of MDPs and CDPs. Ikaros then antagonizes TGFß function to promote pDC differentiation from CDPs. Strikingly, Ikaros-deficient CDPs and pDCs express a cDC-like transcriptional signature that is correlated with TGFß activation, suggesting that Ikaros is an upstream negative regulator of the TGFß pathway and a repressor of cDC-lineage genes in pDCs. Almost all of these phenotypes can be rescued by short-term in vitro treatment with γ-secretase inhibitors, which affects both TGFß-dependent and -independent pathways, but is Notch-independent. We conclude that Ikaros is a crucial differentiation factor in early dendritic progenitors that is required for pDC identity.


Assuntos
Diferenciação Celular/genética , Células Dendríticas/fisiologia , Fator de Transcrição Ikaros/metabolismo , Receptores Notch/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Secretases da Proteína Precursora do Amiloide/metabolismo , Animais , Transplante de Medula Óssea , Linhagem Celular , Regulação para Baixo , Células-Tronco Hematopoéticas/fisiologia , Fator de Transcrição Ikaros/genética , Camundongos , Camundongos Transgênicos , Monócitos/fisiologia , Mutação , Transdução de Sinais/genética , Regulação para Cima
6.
Eur J Immunol ; 48(5): 861-873, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29315532

RESUMO

Basophils have been recently recognized to play important roles in type 2 immune responses during allergies and parasitic infection, largely due to the development of novel tools for the in vivo study of these cells. As such, the genetically-engineered MCPT8DTR mouse line has been used to specifically deplete basophils following treatment with diphtheria toxin (DT). In this study, we showed that DT-injected MCPT8DTR mice exhibited a striking decrease of eosinophils and neutrophils in skin when subjected to a hapten fluorescein isothiocyanate (FITC)-induced allergic contact dermatitis (ACD) experimental protocol. Unexpectedly, we found that loss of skin eosinophils and neutrophils was not due to a lack of basophil-mediated recruitment, as DT injection caused a systemic reduction of eosinophils and neutrophils in MCPT8DTR mice in a time-dependent manner. Furthermore, we found that hematopoietic stem-cell-derived granulocyte-macrophage progenitors (GMPs) expressed MCPT8 gene, and that these cells were depleted upon DT injection. Finally, we optimized a protocol in which a low-dose DT achieved a better specificity for depleting basophils, but not GMPs, in MCPT8DTR mice, and demonstrate that basophils do not play a major role in recruiting eosinophils and neutrophils to ACD skin. These data provide new and valuable information about functional studies of basophils.


Assuntos
Basófilos/imunologia , Dermatite Alérgica de Contato/imunologia , Toxina Diftérica/toxicidade , Eosinófilos/imunologia , Células Progenitoras de Granulócitos e Macrófagos/citologia , Neutrófilos/imunologia , Triptases/metabolismo , Animais , Basófilos/citologia , Eosinófilos/citologia , Feminino , Células Progenitoras de Granulócitos e Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Transgênicos , Neutrófilos/citologia , Triptases/genética
7.
Arthritis Rheumatol ; 69(11): 2124-2135, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28777892

RESUMO

OBJECTIVE: The role of plasmacytoid dendritic cells (PDCs) and type I interferons (IFNs) in rheumatoid arthritis (RA) remains a subject of controversy. This study was undertaken to explore the contribution of PDCs and type I IFNs to RA pathogenesis using various animal models of PDC depletion and to monitor the effect of localized PDC recruitment and activation on joint inflammation and bone damage. METHODS: Mice with K/BxN serum-induced arthritis, collagen-induced arthritis, and human tumor necrosis factor transgene insertion were studied. Symptoms were evaluated by visual scoring, quantification of paw swelling, determination of cytokine levels by enzyme-linked immunosorbent assay, and histologic analysis. Imiquimod-dependent therapeutic effects were monitored by transcriptome analysis (using quantitative reverse transcriptase-polymerase chain reaction) and flow cytometric analysis of the periarticular tissue. RESULTS: PDC-deficient mice showed exacerbation of inflammatory and arthritis symptoms after arthritogenic serum transfer. In contrast, enhancing PDC recruitment and activation to arthritic joints by topical application of the Toll-like receptor 7 (TLR-7) agonist imiquimod significantly ameliorated arthritis in various mouse models. Imiquimod induced an IFN signature and led to reduced infiltration of inflammatory cells. CONCLUSION: The therapeutic effects of imiquimod on joint inflammation and bone destruction are dependent on TLR-7 sensing by PDCs and type I IFN signaling. Our findings indicate that local recruitment and activation of PDCs represents an attractive therapeutic opportunity for RA patients.


Assuntos
Adjuvantes Imunológicos/farmacologia , Aminoquinolinas/farmacologia , Artrite Experimental/imunologia , Artrite Reumatoide/imunologia , Células Dendríticas/efeitos dos fármacos , Interferon Tipo I/efeitos dos fármacos , Animais , Artrite Experimental/genética , Artrite Reumatoide/genética , Citocinas/efeitos dos fármacos , Citocinas/imunologia , Células Dendríticas/imunologia , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Perfilação da Expressão Gênica , Humanos , Fator de Transcrição Ikaros/genética , Imiquimode , Interferon Tipo I/imunologia , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Receptor 7 Toll-Like/genética , Fator de Necrose Tumoral alfa/genética
8.
PLoS One ; 11(6): e0157767, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27315244

RESUMO

The Ikaros transcription factor is a tumor suppressor that is also important for lymphocyte development. How post-translational modifications influence Ikaros function remains partially understood. We show that Ikaros undergoes sumoylation in developing T cells that correspond to mono-, bi- or poly-sumoylation by SUMO1 and/or SUMO2/3 on three lysine residues (K58, K240 and K425). Sumoylation occurs in the nucleus and requires DNA binding by Ikaros. Sumoylated Ikaros is less effective than unsumoylated forms at inhibiting the expansion of murine leukemic cells, and Ikaros sumoylation is abundant in human B-cell acute lymphoblastic leukemic cells, but not in healthy peripheral blood leukocytes. Our results suggest that sumoylation may be important in modulating the tumor suppressor function of Ikaros.


Assuntos
Proteínas de Ligação a DNA/genética , Fator de Transcrição Ikaros/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Transcrição Gênica , Animais , Linfócitos B/patologia , Linhagem Celular Tumoral , Proteínas de Ligação a DNA/biossíntese , Regulação Neoplásica da Expressão Gênica , Humanos , Fator de Transcrição Ikaros/biossíntese , Linfócitos/patologia , Camundongos , Leucemia-Linfoma Linfoblástico de Células Precursoras/patologia , Processamento de Proteína Pós-Traducional/genética , Proteína SUMO-1/genética , Sumoilação/genética , Proteínas Supressoras de Tumor
9.
Blood ; 122(5): 694-704, 2013 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-23801632

RESUMO

Wnt signaling is important for T-cell differentiation at the early CD4(-)CD8(-) stage and is subsequently downregulated with maturation. To assess the importance of this downregulation, we generated a mouse line (R26-ßcat) in which high levels of active ß-catenin are maintained throughout T-cell development. Young R26-ßcat mice show a differentiation block at the CD4(+)CD8(+) double-positive (DP) stage. These DP cells exhibit impaired apoptosis upon irradiation or dexamethasone treatment. All R26-ßcat mice develop T-cell leukemias at 5 to 6 months of age. R26-ßcat leukemias remain dependent on ß-catenin function but lack Notch pathway activation. They exhibit recurrent secondary genomic rearrangements that lead to Myc overexpression and loss of Pten activity. Because ß-catenin activation and Myc translocations were previously found in murine T-cell acute lymphoblastic leukemias (T-ALLs) deficient for Pten, our results suggest that activation of the canonical Wnt pathway is associated with a subtype of Notch-independent T-ALLs that bear Myc gene rearrangements and Pten mutations.


Assuntos
Genes myc/genética , PTEN Fosfo-Hidrolase/genética , Leucemia-Linfoma Linfoblástico de Células T Precursoras/genética , Receptores Notch/fisiologia , beta Catenina/fisiologia , Animais , Diferenciação Celular/genética , Deleção de Genes , Regulação Leucêmica da Expressão Gênica , Camundongos , Camundongos Transgênicos , Mutação/fisiologia , Leucemia-Linfoma Linfoblástico de Células T Precursoras/metabolismo , Leucemia-Linfoma Linfoblástico de Células T Precursoras/mortalidade , Receptores Notch/genética , Linfócitos T/metabolismo , Linfócitos T/fisiologia , Regulação para Cima/genética , Via de Sinalização Wnt/genética , Via de Sinalização Wnt/fisiologia , beta Catenina/agonistas , beta Catenina/genética , beta Catenina/metabolismo
10.
Proc Natl Acad Sci U S A ; 106(41): 17475-80, 2009 Oct 13.
Artigo em Inglês | MEDLINE | ID: mdl-19805133

RESUMO

Macrophages play an essential role in the resolution of tissue damage through removal of necrotic cells, thus paving the way for tissue regeneration. Macrophages also directly support the formation of new tissue to replace the injury, through their acquisition of an anti-inflammatory, or M2, phenotype, characterized by a gene expression program that includes IL-10, the IL-13 receptor, and arginase 1. We report that deletion of two CREB-binding sites from the Cebpb promoter abrogates Cebpb induction upon macrophage activation. This blocks the downstream induction of M2-specific Msr1, Il10, II13ra, and Arg-1 genes, whereas the inflammatory (M1) genes Il1, Il6, Tnfa, and Il12 are not affected. Mice carrying the mutated Cebpb promoter (betaDeltaCre) remove necrotic tissue from injured muscle, but exhibit severe defects in muscle fiber regeneration. Conditional deletion of the Cebpb gene in muscle cells does not affect regeneration, showing that the C/EBPbeta cascade leading to muscle repair is muscle-extrinsic. While betaDeltaCre macrophages efficiently infiltrate injured muscle they fail to upregulate Cebpb, leading to decreased Arg-1 expression. CREB-mediated induction of Cebpb expression is therefore required in infiltrating macrophages for upregulation of M2-specific genes and muscle regeneration, providing a direct genetic link between these two processes.


Assuntos
Proteína beta Intensificadora de Ligação a CCAAT/fisiologia , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/fisiologia , Regulação da Expressão Gênica , Macrófagos/fisiologia , Músculo Esquelético/fisiologia , Animais , Linfócitos B/fisiologia , Sítios de Ligação , Células da Medula Óssea/fisiologia , Proteína beta Intensificadora de Ligação a CCAAT/genética , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/genética , Camundongos , Regiões Promotoras Genéticas , Regeneração , Fatores de Transcrição/metabolismo
11.
Cancer Cell ; 13(4): 299-310, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18394553

RESUMO

Mutations in the CEBPA gene are present in 7%-10% of human patients with acute myeloid leukemia (AML). However, no genetic models exist that demonstrate their etiological relevance. To mimic the most common mutations affecting CEBPA-that is, those leading to loss of the 42 kDa C/EBPalpha isoform (p42) while retaining the 30kDa isoform (p30)-we modified the mouse Cebpa locus to express only p30. p30 supported the formation of granulocyte-macrophage progenitors. However, p42 was required for control of myeloid progenitor proliferation, and p42-deficient mice developed AML with complete penetrance. p42-deficient leukemia could be transferred by a Mac1+c-Kit+ population that gave rise only to myeloid cells in recipient mice. Expression profiling of this population against normal Mac1+c-Kit+ progenitors revealed a signature shared with MLL-AF9-transformed AML.


Assuntos
Proteína alfa Estimuladora de Ligação a CCAAT/metabolismo , Regulação Leucêmica da Expressão Gênica , Leucemia Mielomonocítica Aguda/genética , Leucemia Mielomonocítica Aguda/patologia , Modelos Biológicos , Proteínas Mutantes/metabolismo , Células-Tronco Neoplásicas/patologia , Animais , Proteína alfa Estimuladora de Ligação a CCAAT/deficiência , Proteína alfa Estimuladora de Ligação a CCAAT/genética , Diferenciação Celular , Progressão da Doença , Perfilação da Expressão Gênica , Granulócitos/citologia , Antígeno de Macrófago 1/metabolismo , Camundongos , Camundongos Knockout , Células Progenitoras Mieloides/patologia , Transplante de Neoplasias , Células-Tronco Neoplásicas/metabolismo , Fenótipo , Isoformas de Proteínas/metabolismo , Proteínas Proto-Oncogênicas c-kit/metabolismo
12.
Nat Immunol ; 7(10): 1048-56, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16951689

RESUMO

Wnt signaling increases hematopoietic stem cell self-renewal and is activated in both myeloid and lymphoid malignancies, indicating involvement in both normal and malignant hematopoiesis. We report here activated canonical Wnt signaling in the hematopoietic system through conditional expression of a stable form of beta-catenin. This enforced expression led to hematopoietic failure associated with loss of myeloid lineage commitment at the granulocyte-macrophage progenitor stage; blocked erythrocyte differentiation; disruption of lymphoid development; and loss of repopulating stem cell activity. Loss of hematopoietic stem cell function was associated with decreased expression of Cdkn1a (encoding the cell cycle inhibitor p21(cdk)), Sfpi1, Hoxb4 and Bmi1 (encoding the transcription factors PU.1, HoxB4 and Bmi-1, respectively) and altered integrin expression in Lin(-)Sca-1(+)c-Kit(+) cells, whereas PU.1 was upregulated in erythroid progenitors. Constitutive activation of canonical Wnt signaling therefore causes multilineage differentiation block and compromised hematopoietic stem cell maintenance.


Assuntos
Regulação da Expressão Gênica , Hematopoese/genética , Células-Tronco Hematopoéticas/fisiologia , Proteínas Wnt/metabolismo , beta Catenina/metabolismo , Animais , Diferenciação Celular , Linhagem da Célula/genética , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Expressão Gênica , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Proteínas de Homeodomínio/metabolismo , Integrinas/genética , Camundongos , Camundongos Mutantes , Proteínas Nucleares/metabolismo , Complexo Repressor Polycomb 1 , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Repressoras/metabolismo , Transdução de Sinais , Transativadores/metabolismo , Fatores de Transcrição/metabolismo , Proteínas Wnt/genética , beta Catenina/genética
13.
Mol Cell Biol ; 26(3): 1028-37, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16428455

RESUMO

The C/EBPalpha transcription factor regulates growth and differentiation of several tissues during embryonic development. Several hypotheses as to how C/EBPalpha inhibits cellular growth in vivo have been derived, mainly from studies of tissue culture cells. In fetal liver it has been proposed that a short, centrally located, 15-amino-acid proline-histidine-rich region (PHR) of C/EBPalpha is responsible for the growth-inhibitory function of the protein through its ability to interact with CDK2 and CDK4, thereby inhibiting their activities. Homozygous Cebpa(DeltaPHR/DeltaPHR) (DeltaPHR) mice, carrying a modified cebpa allele lacking amino acids 180 to 194, were born at the Mendelian ratio, reached adulthood, and displayed no apparent adverse phenotypes. When fetal livers from the DeltaPHR mice were analyzed for their expression of cell cycle markers, bromodeoxyuridine incorporation, cyclin-dependent kinase 2 kinase activity, and global gene expression, we failed to detect any cell cycle or developmental differences between the DeltaPHR mice and their control littermates. These in vivo data demonstrate that any C/EBPalpha-mediated growth repression via the PHR as well as the basic region is dispensable for proper embryonic development of, and cell cycle control in, the liver. Surprisingly, control experiments performed in C/EBPalpha null fetal livers yielded similar results.


Assuntos
Proteína alfa Estimuladora de Ligação a CCAAT/metabolismo , Histidina/química , Fígado/embriologia , Prolina/química , Adipócitos/citologia , Sequência de Aminoácidos , Animais , Proteína alfa Estimuladora de Ligação a CCAAT/química , Proteína alfa Estimuladora de Ligação a CCAAT/genética , Diferenciação Celular , Quinase 2 Dependente de Ciclina/metabolismo , Quinase 4 Dependente de Ciclina/metabolismo , Embrião de Mamíferos/metabolismo , Desenvolvimento Embrionário/genética , Histidina/genética , Humanos , Fígado/metabolismo , Camundongos , Camundongos Mutantes , Dados de Sequência Molecular , Prolina/genética , Estrutura Terciária de Proteína , Ratos , Deleção de Sequência
14.
Mol Cell Biol ; 26(1): 209-20, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16354692

RESUMO

The Ikaros transcription factor is both a key regulator of lymphocyte differentiation and a tumor suppressor in T lymphocytes. Mice carrying a hypomorphic mutation (Ik(L/L)) in the Ikaros gene all develop thymic lymphomas. Ik(L/L) tumors always exhibit strong activation of the Notch pathway, which is required for tumor cell proliferation in vitro. Notch activation occurs early in tumorigenesis and may precede transformation, as ectopic expression of the Notch targets Hes-1 and Deltex-1 is detected in thymocytes from young Ik(L/L) mice with no overt signs of transformation. Notch activation is further amplified by secondary mutations that lead to C-terminal truncations of Notch 1. Strikingly, restoration of Ikaros activity in tumor cells leads to a rapid and specific downregulation of Notch target gene expression and proliferation arrest. Furthermore, Ikaros binds to the Notch-responsive element in the Hes-1 promoter and represses Notch-dependent transcription from this promoter. Thus, Ikaros-mediated repression of Notch target gene expression may play a critical role in defining the tumor suppressor function of this factor.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Proteínas de Homeodomínio/genética , Fator de Transcrição Ikaros/deficiência , Linfoma de Células T/genética , Receptor Notch1/metabolismo , Elementos de Resposta , Sequência de Aminoácidos , Animais , Proliferação de Células , Fator de Transcrição Ikaros/genética , Camundongos , Dados de Sequência Molecular , Mutação , Regiões Promotoras Genéticas , Receptor Notch1/genética , Transdução de Sinais , Timo/metabolismo , Timo/patologia , Fatores de Transcrição HES-1
15.
Blood ; 101(6): 2219-26, 2003 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-12406904

RESUMO

The Ikaros gene encodes a zinc finger transcription factor that is selectively expressed by all hematopoietic cells. Although Ikaros is required for lymphocyte differentiation, its role in the myeloid lineage is unclear. We show here that Ikaros expression is temporally regulated during neutrophil differentiation: Ikaros is primarily expressed at immature stages and significantly less so in mature neutrophils. Furthermore Ik(L/L) mice, harboring a hypomorphic mutation at the Ikaros locus, exhibit several defects during neutrophil differentiation. (1) Ik(L/L) fetal livers contain high numbers of neutrophil lineage cells, and this increase is reflected in the number of GM-CSF-dependent progenitor cells. (2) The migratory potential and survival of neutrophil progenitors is altered in vitro. (3) Expression of the Gr-1 marker is delayed and repressed. In contrast, neutrophil function appears normal. These data demonstrate that Ikaros regulates early neutrophil differentiation but is dispensable in mature neutrophils.


Assuntos
Diferenciação Celular , Proteínas de Ligação a DNA , Neutrófilos/citologia , Fatores de Transcrição/fisiologia , Animais , Antígenos de Diferenciação Mielomonocítica/análise , Apoptose , Células da Medula Óssea/citologia , Células Cultivadas , Quimiotaxia de Leucócito , Citometria de Fluxo , Imunofluorescência , Expressão Gênica , Fator Estimulador de Colônias de Granulócitos/farmacologia , Heterozigoto , Homozigoto , Fator de Transcrição Ikaros , Fígado/citologia , Fígado/embriologia , Antígeno de Macrófago 1/análise , Camundongos , Mutação , Neutrófilos/fisiologia , Fagocitose , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Baço/citologia , Fatores de Transcrição/análise , Fatores de Transcrição/genética , beta-Galactosidase/genética
16.
Eur J Immunol ; 32(3): 720-30, 2002 03.
Artigo em Inglês | MEDLINE | ID: mdl-11870616

RESUMO

The Ikaros gene encodes a zinc-finger transcription factor required during early B cell development, as B-lineage cells are absent in mice lacking Ikaros. Here we describe a novel Ikaros-targeted mouse line carrying a beta-galactosidase reporter in which low amounts of Ikaros proteins remain expressed. In homozygote animals, B cells are absent during fetal development, but develop postnatally from a reduced pool of precursors. In vitro, the proliferation and differentiation of B-lineage progenitors are severely impaired. These defects are attenuated in vivo, but bone marrow B cells display an unusual pattern of cell surface marker expression and show decreased transcript levels for TdT, Rag-1, Rag-2 and lambda 5. These abnormalities suggest a partial block at the proB cell stage of differentiation. In the periphery, mature B cells exhibit a lower activation threshold but form fewer germinal centers in response to antigenic stimulation. Our results show that Ikaros controls multiple aspects of B cell differentiation and function.


Assuntos
Linfócitos B/citologia , Fatores de Transcrição/fisiologia , Animais , Formação de Anticorpos , Antígenos de Diferenciação de Linfócitos B/biossíntese , Antígenos de Diferenciação de Linfócitos B/genética , Linfócitos B/imunologia , Células da Medula Óssea/imunologia , Diferenciação Celular , Linhagem da Célula , DNA Nucleotidilexotransferase/biossíntese , DNA Nucleotidilexotransferase/genética , Proteínas de Ligação a DNA , Feto/imunologia , Regulação da Expressão Gênica no Desenvolvimento , Marcação de Genes , Genes RAG-1 , Genes Reporter , Genótipo , Proteínas de Homeodomínio/biossíntese , Fator de Transcrição Ikaros , Isotipos de Imunoglobulinas/biossíntese , Imunoglobulina M/biossíntese , Síndromes de Imunodeficiência/embriologia , Síndromes de Imunodeficiência/etiologia , Síndromes de Imunodeficiência/genética , Óperon Lac , Camundongos , Camundongos Transgênicos , Fenótipo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Transcrição/genética , Transcrição Gênica , beta-Galactosidase/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...