Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 106
Filtrar
1.
EMBO Rep ; 25(6): 2662-2697, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38744970

RESUMO

The multifunctional RNA-binding protein hnRNPL is implicated in antibody class switching but its broader function in B cells is unknown. Here, we show that hnRNPL is essential for B cell activation, germinal center formation, and antibody responses. Upon activation, hnRNPL-deficient B cells show proliferation defects and increased apoptosis. Comparative analysis of RNA-seq data from activated B cells and another eight hnRNPL-depleted cell types reveals common effects on MYC and E2F transcriptional programs required for proliferation. Notably, while individual gene expression changes are cell type specific, several alternative splicing events affecting histone modifiers like KDM6A and SIRT1, are conserved across cell types. Moreover, hnRNPL-deficient B cells show global changes in H3K27me3 and H3K9ac. Epigenetic dysregulation after hnRNPL loss could underlie differential gene expression and upregulation of lncRNAs, and explain common and cell type-specific phenotypes, such as dysfunctional mitochondria and ROS overproduction in mouse B cells. Thus, hnRNPL is essential for the resting-to-activated B cell transition by regulating transcriptional programs and metabolism, at least in part through the alternative splicing of several histone modifiers.


Assuntos
Processamento Alternativo , Linfócitos B , Epigênese Genética , Ativação Linfocitária , Animais , Humanos , Camundongos , Apoptose/genética , Linfócitos B/metabolismo , Linfócitos B/imunologia , Proliferação de Células/genética , Regulação da Expressão Gênica , Centro Germinativo/imunologia , Centro Germinativo/metabolismo , Histonas/metabolismo , Ativação Linfocitária/genética
2.
Blood ; 142(25): 2175-2191, 2023 12 21.
Artigo em Inglês | MEDLINE | ID: mdl-37756525

RESUMO

ABSTRACT: Growth factor independence 1 (GFI1) is a DNA-binding transcription factor and a key regulator of hematopoiesis. GFI1-36N is a germ line variant, causing a change of serine (S) to asparagine (N) at position 36. We previously reported that the GFI1-36N allele has a prevalence of 10% to 15% among patients with acute myeloid leukemia (AML) and 5% to 7% among healthy Caucasians and promotes the development of this disease. Using a multiomics approach, we show here that GFI1-36N expression is associated with increased frequencies of chromosomal aberrations, mutational burden, and mutational signatures in both murine and human AML and impedes homologous recombination (HR)-directed DNA repair in leukemic cells. GFI1-36N exhibits impaired binding to N-Myc downstream-regulated gene 1 (Ndrg1) regulatory elements, causing decreased NDRG1 levels, which leads to a reduction of O6-methylguanine-DNA-methyltransferase (MGMT) expression levels, as illustrated by both transcriptome and proteome analyses. Targeting MGMT via temozolomide, a DNA alkylating drug, and HR via olaparib, a poly-ADP ribose polymerase 1 inhibitor, caused synthetic lethality in human and murine AML samples expressing GFI1-36N, whereas the effects were insignificant in nonmalignant GFI1-36S or GFI1-36N cells. In addition, mice that received transplantation with GFI1-36N leukemic cells treated with a combination of temozolomide and olaparib had significantly longer AML-free survival than mice that received transplantation with GFI1-36S leukemic cells. This suggests that reduced MGMT expression leaves GFI1-36N leukemic cells particularly vulnerable to DNA damage initiating chemotherapeutics. Our data provide critical insights into novel options to treat patients with AML carrying the GFI1-36N variant.


Assuntos
Proteínas de Ligação a DNA , Leucemia Mieloide Aguda , Humanos , Camundongos , Animais , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Temozolomida , Leucemia Mieloide Aguda/tratamento farmacológico , Leucemia Mieloide Aguda/genética , Dano ao DNA , Reparo do DNA , Células Germinativas/metabolismo , DNA , Fatores de Transcrição/genética
3.
J Immunol ; 207(6): 1599-1615, 2021 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-34408010

RESUMO

GFI1 is a DNA-binding transcription factor that regulates hematopoiesis by repressing target genes through its association with complexes containing histone demethylases such as KDM1A (LSD1) and histone deacetylases (HDACs). To study the consequences of the disruption of the complex between GFI1 and histone-modifying enzymes, we have used knock-in mice harboring a P2A mutation in GFI1 coding region that renders it unable to bind LSD1 and associated histone-modifying enzymes such as HDACs. GFI1P2A mice die prematurely and show increased numbers of memory effector and regulatory T cells in the spleen accompanied by a severe systemic inflammation with high serum levels of IL-6, TNF-α, and IL-1ß and overexpression of the gene encoding the cytokine oncostatin M (OSM). We identified lung alveolar macrophages, CD8 T cell from the spleen and thymic eosinophils, and monocytes as the sources of these cytokines in GFI1P2A mice. Chromatin immunoprecipitation showed that GFI1/LSD1 complexes occupy sites at the Osm promoter and an intragenic region of the Tnfα gene and that a GFI1P2A mutant still remains bound at these sites even without LSD1. Methylation and acetylation of histone H3 at these sites were enriched in cells from GFI1P2A mice, the H3K27 acetylation being the most significant. These data suggest that the histone modification facilitated by GFI1 is critical to control inflammatory pathways in different cell types, including monocytes and eosinophils, and that a disruption of GFI1-associated complexes can lead to systemic inflammation with fatal consequences.


Assuntos
Proteínas de Ligação a DNA/deficiência , Histona Desmetilases/metabolismo , Proteínas Mutantes/metabolismo , Transdução de Sinais/genética , Síndrome de Resposta Inflamatória Sistêmica/sangue , Fatores de Transcrição/deficiência , Animais , Transplante de Medula Óssea/métodos , Linfócitos T CD8-Positivos/imunologia , Citocinas/sangue , Citocinas/genética , Proteínas de Ligação a DNA/genética , Feminino , Expressão Gênica , Técnicas de Introdução de Genes , Histonas/metabolismo , Macrófagos/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mutação , Ligação Proteica , Síndrome de Resposta Inflamatória Sistêmica/genética , Síndrome de Resposta Inflamatória Sistêmica/imunologia , Fatores de Transcrição/genética
4.
Mol Cell ; 54(4): 651-62, 2014 May 22.
Artigo em Inglês | MEDLINE | ID: mdl-24837677

RESUMO

The circadian clock drives daily rhythms in gene expression to control metabolism, behavior, and physiology; while the underlying transcriptional feedback loops are well defined, the impact of alternative splicing on circadian biology remains poorly understood. Here we describe a robust circadian and light-inducible splicing switch that changes the reading frame of the mouse mRNA encoding U2-auxiliary-factor 26 (U2AF26). This results in translation far into the 3' UTR, generating a C terminus with homology to the Drosophila clock regulator TIMELESS. This new U2AF26 variant destabilizes PERIOD1 protein, and U2AF26-deficient mice show nearly arrhythmic PERIOD1 protein levels and broad defects in circadian mRNA expression in peripheral clocks. At the behavioral level, these mice display increased phase advance adaptation following experimental jet lag. These data suggest light-induced U2af26 alternative splicing to be a buffering mechanism that limits PERIOD1 induction, thus stabilizing the circadian clock against abnormal changes in light:dark conditions.


Assuntos
Processamento Alternativo , Relógios Circadianos , Ritmo Circadiano , Mutação da Fase de Leitura , Proteínas Circadianas Period/metabolismo , RNA Mensageiro/metabolismo , Ribonucleoproteínas/genética , Ribonucleoproteínas/metabolismo , Animais , Comportamento Animal/efeitos da radiação , Encéfalo/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular Tumoral , Relógios Circadianos/genética , Regulação da Expressão Gênica , Células HEK293 , Células HeLa , Humanos , Fígado/metabolismo , Camundongos , Camundongos Transgênicos , Células NIH 3T3 , Estabilidade Proteica , RNA Mensageiro/genética , Fator de Processamento U2AF
5.
Haematologica ; 105(10): 2457-2470, 2020 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-33054086

RESUMO

GFI1B-related thrombocytopenia (GFI1B-RT) is a rare bleeding disorder mainly caused by the presence of truncated GFI1B proteins with dominant-negative properties. The disease is characterized by low platelet counts, the presence of abnormal platelets, a megakaryocytic expansion and mild erythroid defects. However, no animal models faithfully reproducing the GFI1B-RT phenotype observed in patients exist. We had previously generated mice with floxed Gfi1b alleles that can be eliminated by Cre recombinase, but those animals developed a much more severe phenotype than GFI1B-RT patients and were of limited interest in assessing the disease. Using CRISPR/Cas9 technology, we have now established three independent mouse lines that carry mutated Gfi1b alleles producing proteins lacking DNA binding zinc fingers and thereby acting in a dominant negative (DN) manner. Mice heterozygous for these Gfi1b-DN alleles show reduced platelet counts and an expansion of megakaryocytes similar to features of human GFI1B-RT but lacking the distinctively large agranular platelets. In addition, Gfi1b-DN mice exhibit an expansion of erythroid precursors indicative of a mildly abnormal erythropoiesis but without noticeable red blood cell defects. When associated with megakaryocyte-specific ablation of the remaining allele, the Gfi1b-DN alleles triggered erythroid-specific deleterious defects. Gfi1b-DN mice also showed a delayed recovery from platelet depletion, indicating a defect in stress thrombopoiesis. However, injecting Gfi1b-DN mice with romiplostim, a thrombopoietin receptor super agonist, increased platelet numbers even beyond normal levels. Thus, our data support a causal link between DN mutations in GFI1B and thrombocytopenia and suggest that patients with GFI1B-RT could be treated successfully with thrombopoietin agonists.


Assuntos
Trombocitopenia , Trombopoese , Animais , Plaquetas , Eritropoese , Humanos , Megacariócitos , Camundongos , Mutação , Proteínas Proto-Oncogênicas/genética , Proteínas Repressoras/genética , Trombocitopenia/genética , Trombopoese/genética
6.
Immunity ; 33(6): 917-28, 2010 Dec 14.
Artigo em Inglês | MEDLINE | ID: mdl-21167753

RESUMO

B cell development requires the coordinated action of transcription factors and cytokines, in particular interleukin-7 (IL-7). We report that mice lacking the POZ (Poxvirus and zinc finger) domain of the transcription factor Miz-1 (Zbtb17(ΔPOZ/ΔPOZ)) almost entirely lacked follicular B cells, as shown by the fact that their progenitors failed to activate the Jak-Stat5 pathway and to upregulate the antiapoptotic gene Bcl2 upon IL-7 stimulation. We show that Miz-1 exerted a dual role in the interleukin-7 receptor (IL-7R) pathway by directly repressing the Janus kinase (Jak) inhibitor suppressor of cytokine signaling 1 (Socs1) and by activating Bcl2 expression. Zbtb17(ΔPOZ/ΔPOZ) (Miz-1-deficient) B cell progenitors had low expression of early B cell genes as transcription factor 3 (Tcf3) and early B cell factor 1 (Ebf1) and showed a propensity for apoptosis. Only the combined re-expression of Bcl2 and Ebf1 could reconstitute the ability of Miz-1-deficient precursors to develop into CD19(+) B cells.


Assuntos
Linfócitos B/metabolismo , Medula Óssea/patologia , Proteínas Nucleares/metabolismo , Proteínas Inibidoras de STAT Ativados/metabolismo , Receptores de Interleucina-7/metabolismo , Proteína de Morte Celular Associada a bcl/biossíntese , Animais , Linfócitos B/imunologia , Linfócitos B/patologia , Diferenciação Celular/genética , Diferenciação Celular/imunologia , Linhagem da Célula/genética , Linhagem da Célula/imunologia , Sobrevivência Celular/genética , Células Cultivadas , Camundongos , Camundongos Mutantes , Mutação/genética , Proteínas Nucleares/genética , Proteínas Nucleares/imunologia , Proteínas Inibidoras de STAT Ativados/genética , Proteínas Inibidoras de STAT Ativados/imunologia , Receptores de Interleucina-7/genética , Receptores de Interleucina-7/imunologia , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Ubiquitina-Proteína Ligases , Proteína de Morte Celular Associada a bcl/genética , Proteína de Morte Celular Associada a bcl/imunologia
7.
Genes Dev ; 24(12): 1281-94, 2010 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-20551174

RESUMO

The Myc protein suppresses the transcription of several cyclin-dependent kinase inhibitors (CKIs) via binding to Miz1; whether this interaction is important for Myc's ability to induce or maintain tumorigenesis is not known. Here we show that the oncogenic potential of a point mutant of Myc (MycV394D) that is selectively deficient in binding to Miz1 is greatly attenuated. Binding of Myc to Miz1 is continuously required to repress CKI expression and inhibit accumulation of trimethylated histone H3 at Lys 9 (H3K9triMe), a hallmark of cellular senescence, in T-cell lymphomas. Lymphomas that arise express high amounts of transforming growth factor beta-2 (TGFbeta-2) and TGFbeta-3. Upon Myc suppression, TGFbeta signaling is required to induce CKI expression and cellular senescence and suppress tumor recurrence. Binding of Myc to Miz1 is required to antagonize growth suppression and induction of senescence by TGFbeta. We demonstrate that, since lymphomas express high levels of TGFbeta, they are poised to elicit an autocrine program of senescence upon Myc inactivation, demonstrating that TGFbeta is a key factor that establishes oncogene addiction of T-cell lymphomas.


Assuntos
Comunicação Autócrina/fisiologia , Linfoma de Células T/fisiopatologia , Proteínas Nucleares/metabolismo , Proteínas Inibidoras de STAT Ativados/metabolismo , Proteínas Proto-Oncogênicas c-myc/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Animais , Doxiciclina/farmacologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Células HeLa , Humanos , Camundongos , Camundongos Transgênicos , Mutação/genética , Proteínas Proto-Oncogênicas c-myc/genética , Ubiquitina-Proteína Ligases
8.
Curr Opin Hematol ; 24(1): 32-37, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27820734

RESUMO

PURPOSE OF REVIEW: Growth factor independent 1 (Gfi1) is a transcriptional repressor that plays multiple roles during myeloid commitment and development. Gfi1-deficient mice lack granulocytes, accumulate aberrant monocytes and show a hyperactivity of macrophages toward bacterial cell wall components. Since these initial findings, numerous additional studies have confirmed the role of Gfi1 in myeloid differentiation from hematopoietic stem cells and multipotent progenitors to bipotential lymphoid/myeloid precursors and myeloid effector cells. This review will summarize the existing knowledge concerning the mechanisms through which Gfi1 exerts these actions and will highlight recent insights into its additional implication in myeloid malignancies. RECENT FINDINGS: Gfi1 has more recently been implicated in myeloid malignancies, in particular in myelodysplasia, myeloproliferative neoplasms and in acute myeloid leukemia, a fatal disease, which is essentially treated today the same way as 30 years ago. SUMMARY: Recent findings on the role of Gfi1 in myeloid malignancies together with the knowledge base built over many years on this molecule may help us to find new ways to predict the progression of acute myeloid leukemia and to design more efficient epigenetic drugs to treat this disease.


Assuntos
Diferenciação Celular , Proteínas de Ligação a DNA/metabolismo , Células Mieloides/citologia , Células Mieloides/metabolismo , Fatores de Transcrição/metabolismo , Animais , Diferenciação Celular/genética , Linhagem da Célula/genética , Proteínas de Ligação a DNA/genética , Suscetibilidade a Doenças , Granulócitos/metabolismo , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Humanos , Inflamação/imunologia , Inflamação/metabolismo , Inflamação/patologia , Linfopoese/genética , Mielopoese/genética , Transdução de Sinais , Fatores de Transcrição/genética
9.
Blood ; 126(24): 2561-9, 2015 Dec 10.
Artigo em Inglês | MEDLINE | ID: mdl-26447191

RESUMO

The DNA-binding zinc finger transcription factors Gfi1 and Gfi1b were discovered more than 20 years ago and are recognized today as major regulators of both early hematopoiesis and hematopoietic stem cells. Both proteins function as transcriptional repressors by recruiting histone-modifying enzymes to promoters and enhancers of target genes. The establishment of Gfi1 and Gfi1b reporter mice made it possible to visualize their cell type-specific expression and to understand their function in hematopoietic lineages. We now know that Gfi1 is primarily important in myeloid and lymphoid differentiation, whereas Gfi1b is crucial for the generation of red blood cells and platelets. Several rare hematologic diseases are associated with acquired or inheritable mutations in the GFI1 and GFI1B genes. Certain patients with severe congenital neutropenia carry mutations in the GFI1 gene that lead to the disruption of the C-terminal zinc finger domains. Other mutations have been found in the GFI1B gene in families with inherited bleeding disorders. In addition, the Gfi1 locus is frequently found to be a proviral integration site in retrovirus-induced lymphomagenesis, and new, emerging data suggest a role of Gfi1 in human leukemia and lymphoma, underlining the role of both factors not only in normal hematopoiesis, but also in a wide spectrum of human blood diseases.


Assuntos
Proteínas de Ligação a DNA/fisiologia , Hematopoese/fisiologia , Proteínas Proto-Oncogênicas/fisiologia , Proteínas Repressoras/fisiologia , Fatores de Transcrição/fisiologia , Animais , Células Sanguíneas/fisiologia , Síndrome Congênita de Insuficiência da Medula Óssea , Proteínas de Ligação a DNA/deficiência , Proteínas de Ligação a DNA/genética , Regulação da Expressão Gênica , Redes Reguladoras de Genes , Genes Reporter , Doenças Hematológicas/genética , Neoplasias Hematológicas/genética , Hematopoese/genética , Células-Tronco Hematopoéticas/fisiologia , Código das Histonas/fisiologia , Humanos , Vírus da Leucemia Murina/fisiologia , Camundongos , Camundongos Transgênicos , Modelos Moleculares , Neutropenia/congênito , Neutropenia/genética , Conformação Proteica , Proteínas Proto-Oncogênicas/genética , Proteínas Repressoras/genética , Fatores de Transcrição/deficiência , Fatores de Transcrição/genética , Integração Viral
10.
Haematologica ; 102(3): 484-497, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-28082345

RESUMO

Mutations in GFI1B are associated with inherited bleeding disorders called GFI1B-related thrombocytopenias. We show here that mice with a megakaryocyte-specific Gfi1b deletion exhibit a macrothrombocytopenic phenotype along a megakaryocytic dysplasia reminiscent of GFI1B-related thrombocytopenia. GFI1B deficiency increases megakaryocyte proliferation and affects their ploidy, but also abrogates their responsiveness towards integrin signaling and their ability to spread and reorganize their cytoskeleton. Gfi1b-null megakaryocytes are also unable to form proplatelets, a process independent of integrin signaling. GFI1B-deficient megakaryocytes exhibit aberrant expression of several components of both the actin and microtubule cytoskeleton, with a dramatic reduction of α-tubulin. Inhibition of FAK or ROCK, both important for actin cytoskeleton organization and integrin signaling, only partially restored their response to integrin ligands, but the inhibition of PAK, a regulator of the actin cytoskeleton, completely rescued the responsiveness of Gfi1b-null megakaryocytes to ligands, but not their ability to form proplatelets. We conclude that Gfi1b controls major functions of megakaryocytes such as integrin-dependent cytoskeleton organization, spreading and migration through the regulation of PAK activity whereas the proplatelet formation defect in GFI1B-deficient megakaryocytes is due, at least partially, to an insufficient α-tubulin content.


Assuntos
Citoesqueleto/metabolismo , Integrinas/metabolismo , Megacariócitos/metabolismo , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Transdução de Sinais , Actinas/química , Actinas/metabolismo , Animais , Diferenciação Celular/genética , Células Cultivadas , Perfilação da Expressão Gênica , Estudos de Associação Genética , Megacariócitos/efeitos dos fármacos , Megacariócitos/patologia , Megacariócitos/ultraestrutura , Camundongos , Camundongos Knockout , Microtúbulos/metabolismo , Fenótipo , Contagem de Plaquetas , Inibidores de Proteínas Quinases/farmacologia , Multimerização Proteica , Trombocitopenia/sangue , Trombocitopenia/diagnóstico , Trombocitopenia/genética , Trombocitopenia/metabolismo , Transcriptoma
11.
Proc Natl Acad Sci U S A ; 111(50): E5411-9, 2014 Dec 16.
Artigo em Inglês | MEDLINE | ID: mdl-25468973

RESUMO

To be effective, the adaptive immune response requires a large repertoire of antigen receptors, which are generated through V(D)J recombination in lymphoid precursors. These precursors must be protected from DNA damage-induced cell death, however, because V(D)J recombination generates double-strand breaks and may activate p53. Here we show that the BTB/POZ domain protein Miz-1 restricts p53-dependent induction of apoptosis in both pro-B and DN3a pre-T cells that actively rearrange antigen receptor genes. Miz-1 exerts this function by directly activating the gene for ribosomal protein L22 (Rpl22), which binds to p53 mRNA and negatively regulates its translation. This mechanism limits p53 expression levels and thus contains its apoptosis-inducing functions in lymphocytes, precisely at differentiation stages in which V(D)J recombination occurs.


Assuntos
Regulação da Expressão Gênica/fisiologia , Células Progenitoras Linfoides/fisiologia , Proteínas Nucleares/metabolismo , Biossíntese de Proteínas/fisiologia , Proteínas Inibidoras de STAT Ativados/metabolismo , Proteínas de Ligação a RNA/metabolismo , Proteínas Ribossômicas/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Recombinação V(D)J/genética , Análise de Variância , Animais , Morte Celular/fisiologia , Imunoprecipitação da Cromatina , Citometria de Fluxo , Regulação da Expressão Gênica/genética , Vetores Genéticos/genética , Immunoblotting , Imunoprecipitação , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Nucleares/genética , Biossíntese de Proteínas/genética , Proteínas Inibidoras de STAT Ativados/genética , Reação em Cadeia da Polimerase em Tempo Real , Ubiquitina-Proteína Ligases , Recombinação V(D)J/fisiologia
12.
PLoS Genet ; 9(9): e1003713, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24068942

RESUMO

Growth factor independent 1 (Gfi1) is a transcriptional repressor originally identified as a gene activated in T-cell leukemias induced by Moloney-murine-leukemia virus infection. Notch1 is a transmembrane receptor that is frequently mutated in human T-cell acute lymphoblastic leukemia (T-ALL). Gfi1 is an important factor in the initiation and maintenance of lymphoid leukemias and its deficiency significantly impedes Notch dependent initiation of T-ALL in animal models. Here, we show that immature hematopoietic cells require Gfi1 to competently integrate Notch-activated signaling. Notch1 activation coupled with Gfi1 deficiency early in T-lineage specification leads to a dramatic loss of T-cells, whereas activation in later stages leaves development unaffected. In Gfi1 deficient multipotent precursors, Notch activation induces lethality and is cell autonomous. Further, without Gfi1, multipotent progenitors do not maintain Notch1-activated global expression profiles typical for T-lineage precursors. In agreement with this, we find that both lymphoid-primed multipotent progenitors (LMPP) and early T lineage progenitors (ETP) do not properly form or function in Gfi1(-/-) mice. These defects correlate with an inability of Gfi1(-/-) progenitors to activate lymphoid genes, including IL7R, Rag1, Flt3 and Notch1. Our data indicate that Gfi1 is required for hematopoietic precursors to withstand Notch1 activation and to maintain Notch1 dependent transcriptional programming to determine early T-lymphoid lineage identity.


Assuntos
Proteínas de Ligação a DNA/genética , Receptor Notch1/genética , Transdução de Sinais/genética , Linfócitos T/citologia , Fatores de Transcrição/genética , Animais , Linhagem da Célula , Regulação Leucêmica da Expressão Gênica/imunologia , Redes Reguladoras de Genes , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Humanos , Linfócitos/citologia , Linfócitos/metabolismo , Camundongos , Linfócitos T/imunologia
13.
Semin Immunol ; 23(5): 368-78, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21920773

RESUMO

T- and B-lymphocytes are important elements in the immune defense repertoire of higher organisms. The development and function of lymphoid cells is regulated at many levels one being the control of gene expression by transcription factors. The zinc finger transcriptional repressor Gfi1 has emerged as a factor that is critically implicated in the commitment of precursor cells for the lymphoid lineage. In addition, Gfi1 controls distinct stages of early T- or B-lymphoid development and is also critical for their maturation, activation and effector function. From many years of work, a picture emerges in which Gfi1 is part of a complicated, but well orchestrated network of interdependent regulators, most of which impinge on lymphoid development and activation by transcriptional regulation. Biochemical studies show that Gfi1 is part of a large DNA binding multi-protein complex that enables histone modifications, but may also control alternative pre mRNA splicing. Many insights into the biological role of Gfi1 have been gained through the study of gene deficient mice that have defects in B- and T-cell differentiation, in T-cell selection and polarization processes and in the response of mature B- and T-cells towards antigen. Most importantly, the defects seen in Gfi1 deficient mice also point to roles of Gfi1 in diseases of the immune system that involve auto-immune responses and acute lymphoid leukemia and lymphoma.


Assuntos
Linfócitos B/imunologia , Proteínas de Ligação a DNA/metabolismo , Linfopoese/fisiologia , Linfócitos T/imunologia , Fatores de Transcrição/metabolismo , Animais , Humanos , Ativação Linfocitária , Camundongos , Modelos Biológicos
14.
Semin Immunol ; 23(5): 379-87, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22000024

RESUMO

The Myc interacting zinc finger protein 1 (Miz-1) is a BTB/POZ domain containing transcription factor that can function as an activator or repressor depending on its binding partners. In a complex with co-factors such as nuclophosmin or p300, Miz-1 stimulates transcription of genes that encode regulators of cell cycle progression such as p21(Cip1) or p15(Ink4b) or inhibitors of apoptosis such as Bcl-2. In contrast, Miz-1 becomes a transcriptional repressor when it binds to c-Myc or Bcl-6, which replace nucleophosmin or p300. During lymphocyte development, Miz-1 functions as a regulator of the IL-7 signaling pathway at very early steps in the bone marrow and thymus. When the IL-7 receptor (IL-7R) recognizes its cognate cytokine, a cascade of events is initiated that involves the recruitment of janus kinases (JAK) to the cytoplasmic part of the IL-7R, the phosphorylation of Stat5, its dimerization and relocation to the nucleus, enabling a transcriptional programming that governs commitment, survival and proliferation of lymphoid lineage cells. Miz-1 is critical in this signal transduction pathway, since it controls the expression of Socs1, an inhibitor of JAKs and thus of Stat5 activation and Bcl-2 expression. A lack of Miz-1 blocks IL-7 mediated signaling, which is detrimental for early B- and T-lymphoid development. These functions of Miz-1 during early lymphocyte development are c-Myc-independent. In contrast, when c-Myc is constitutively over-expressed, for instance during c-Myc induced lymphomagenesis, the interaction between Miz-1 and c-Myc becomes important and critical for the initiation and maintenance of c-Myc-dependent lymphoid malignancies.


Assuntos
Linfócitos B/imunologia , Fatores de Transcrição Kruppel-Like/metabolismo , Linfoma/patologia , Linfócitos T/imunologia , Linfócitos B/citologia , Proteínas de Ligação a DNA/metabolismo , Humanos , Modelos Biológicos , Proteínas Proto-Oncogênicas c-bcl-6 , Transdução de Sinais , Linfócitos T/citologia , Fatores de Transcrição/metabolismo
15.
Blood ; 120(2): 314-22, 2012 Jul 12.
Artigo em Inglês | MEDLINE | ID: mdl-22668850

RESUMO

Recent studies have established that during embryonic development, hematopoietic progenitors and stem cells are generated from hemogenic endothelium precursors through a process termed endothelial to hematopoietic transition (EHT). The transcription factor RUNX1 is essential for this process, but its main downstream effectors remain largely unknown. Here, we report the identification of Gfi1 and Gfi1b as direct targets of RUNX1 and critical regulators of EHT. GFI1 and GFI1B are able to trigger, in the absence of RUNX1, the down-regulation of endothelial markers and the formation of round cells, a morphologic change characteristic of EHT. Conversely, blood progenitors in Gfi1- and Gfi1b-deficient embryos maintain the expression of endothelial genes. Moreover, those cells are not released from the yolk sac and disseminated into embryonic tissues. Taken together, our findings demonstrate a critical and specific role of the GFI1 transcription factors in the first steps of the process leading to the generation of hematopoietic progenitors from hemogenic endothelium.


Assuntos
Proteínas de Ligação a DNA/fisiologia , Hemangioblastos/citologia , Hemangioblastos/fisiologia , Hematopoese/fisiologia , Proteínas Proto-Oncogênicas/fisiologia , Proteínas Repressoras/fisiologia , Fatores de Transcrição/fisiologia , Animais , Diferenciação Celular , Linhagem Celular , Subunidade alfa 2 de Fator de Ligação ao Core/deficiência , Subunidade alfa 2 de Fator de Ligação ao Core/genética , Subunidade alfa 2 de Fator de Ligação ao Core/fisiologia , Proteínas de Ligação a DNA/deficiência , Proteínas de Ligação a DNA/genética , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/fisiologia , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Masculino , Camundongos , Camundongos Knockout , Modelos Biológicos , Neovascularização Fisiológica , Gravidez , Proteínas Proto-Oncogênicas/deficiência , Proteínas Proto-Oncogênicas/genética , Proteínas Repressoras/deficiência , Proteínas Repressoras/genética , Fatores de Transcrição/deficiência , Fatores de Transcrição/genética , Saco Vitelino/citologia , Saco Vitelino/embriologia , Saco Vitelino/fisiologia
16.
Blood ; 120(19): 4006-17, 2012 Nov 08.
Artigo em Inglês | MEDLINE | ID: mdl-22932805

RESUMO

The coding single nucleotide polymorphism GFI136N in the human gene growth factor independence 1 (GFI1) is present in 3%-7% of whites and increases the risk for acute myeloid leukemia (AML) by 60%. We show here that GFI136N, in contrast to GFI136S, lacks the ability to bind to the Gfi1 target gene that encodes the leukemia-associated transcription factor Hoxa9 and fails to initiate histone modifications that regulate HoxA9 expression. Consistent with this, AML patients heterozygous for the GFI136N variant show increased HOXA9 expression compared with normal controls. Using ChipSeq, we demonstrate that GFI136N specific epigenetic changes are also present in other genes involved in the development of AML. Moreover, granulomonocytic progenitors, a bone marrow subset from which AML can arise in humans and mice, show a proliferative expansion in the presence of the GFI136N variant. In addition, granulomonocytic progenitors carrying the GFI136N variant allele have altered gene expression patterns and differ in their ability to grow after transplantation. Finally, GFI136N can accelerate a K-RAS driven fatal myeloproliferative disease in mice. Our data suggest that the presence of a GFI136N variant allele induces a preleukemic state in myeloid precursors by deregulating the expression of Hoxa9 and other AML-related genes.


Assuntos
Proteínas de Ligação a DNA/genética , Epigênese Genética , Proteínas de Homeodomínio/genética , Transtornos Mieloproliferativos/genética , Proteínas Proto-Oncogênicas p21(ras)/genética , Fatores de Transcrição/genética , Animais , Análise por Conglomerados , Proteínas de Ligação a DNA/metabolismo , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Predisposição Genética para Doença , Hematopoese/genética , Histonas/metabolismo , Humanos , Camundongos , Camundongos Transgênicos , Células Progenitoras Mieloides/metabolismo , Células Progenitoras Mieloides/patologia , Transtornos Mieloproliferativos/metabolismo , Transtornos Mieloproliferativos/mortalidade , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Fatores de Transcrição/metabolismo
17.
J Immunol ; 188(11): 5377-88, 2012 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-22523384

RESUMO

The regulation of posttranscriptional modifications of pre-mRNA by alternative splicing is important for cellular function, development, and immunity. The receptor tyrosine phosphatase CD45, which is expressed on all hematopoietic cells, is known for its role in the development and activation of T cells. CD45 is known to be alternatively spliced, a process that is partially regulated by heterogeneous nuclear ribonucleoprotein (hnRNP) L. To investigate the role of hnRNP L further, we have generated conditional hnRNP L knockout mice and found that LckCre-mediated deletion of hnRNP L results in a decreased thymic cellularity caused by a partial block at the transition stage between double-negative 4 and double-positive cells. In addition, hnRNP L(-/-) thymocytes express aberrant levels of the CD45RA splice isoforms and show high levels of phosphorylated Lck at the activator tyrosine Y394, but lack phosphorylation of the inhibitory tyrosine Y505. This indicated an increased basal Lck activity and correlated with higher proliferation rates of double-negative 4 cells in hnRNP L(-/-) mice. Deletion of hnRNP L also blocked the migration and egress of single-positive thymocytes to peripheral lymphoid organs in response to sphingosine-1-phosphate and the chemokines CCL21 and CXCL12 very likely as a result of aberrant splicing of genes encoding GTPase regulators and proteins affecting cytoskeletal organization. Our results indicate that hnRNP L regulates T cell differentiation and migration by regulating pre-TCR and chemokine receptor signaling.


Assuntos
Processamento Alternativo/imunologia , Diferenciação Celular/imunologia , Movimento Celular/imunologia , Proliferação de Células , Ribonucleoproteínas Nucleares Heterogêneas Grupo L/genética , Células-Tronco/imunologia , Subpopulações de Linfócitos T/imunologia , Timo/imunologia , Processamento Alternativo/genética , Animais , Diferenciação Celular/genética , Movimento Celular/genética , Ribonucleoproteínas Nucleares Heterogêneas Grupo L/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Células-Tronco/citologia , Células-Tronco/metabolismo , Subpopulações de Linfócitos T/citologia , Subpopulações de Linfócitos T/metabolismo , Timo/citologia , Timo/metabolismo
18.
Sci Immunol ; 9(95): eadj2654, 2024 May 31.
Artigo em Inglês | MEDLINE | ID: mdl-38820141

RESUMO

Tissue-resident innate lymphoid cells (ILCs) play a vital role in the frontline defense of various tissues, including the lung. The development of type 2 ILCs (ILC2s) depends on transcription factors such as GATA3, RORα, GFI1, and Bcl11b; however, the factors regulating lung-resident ILC2s remain unclear. Through fate mapping analysis of the paralog transcription factors GFI1 and GFI1B, we show that GFI1 is consistently expressed during the transition from progenitor to mature ILC2s. In contrast, GFI1B expression is limited to specific subsets of bone marrow progenitors and lung-resident ILC progenitors. We found that GFI1B+ lung ILC progenitors represent a multi-lineage subset with tissue-resident characteristics and the potential to form lung-derived ILC subsets and liver-resident ILC1s. Loss of GFI1B in bone marrow progenitors led to the selective loss of lung-resident IL-18R+ ILCs and mature ILC2, subsequently preventing the emergence of effector ILCs that could protect the lung against inflammatory or tumor challenge.


Assuntos
Imunidade Inata , Pulmão , Camundongos Endogâmicos C57BL , Proteínas Proto-Oncogênicas , Animais , Pulmão/imunologia , Pulmão/citologia , Camundongos , Imunidade Inata/imunologia , Proteínas Proto-Oncogênicas/imunologia , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/deficiência , Proteínas Proto-Oncogênicas/metabolismo , Células Progenitoras Linfoides/imunologia , Células Progenitoras Linfoides/citologia , Proteínas Repressoras/genética , Proteínas Repressoras/imunologia , Camundongos Knockout , Linfócitos/imunologia , Diferenciação Celular/imunologia , Proteínas de Ligação a DNA , Fatores de Transcrição
19.
Dev Biol ; 362(2): 194-218, 2012 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-22185794

RESUMO

Mix progenitors are short-lived multipotential cells formed as intestinal epithelial stem cells initiate a differentiation program. Clone dynamics indicates that various epithelial cell lineages arise from Mix via a sequence of progressively restricted progenitor states. Lateral inhibitory Notch signaling between the daughters of Mix (DOM) is thought to break their initial symmetry, thereby determining whether a DOM invokes a columnar (absorptive) or granulocytic (secretory) cell lineage program. This is supported by the absence of granulocytes following enforced Notch signaling or Atoh1 deletion. Conversely, granulocytes increase in frequency following inhibition of Notch signaling or Hes1 deletion. Thus reciprocal repression between Hes1 and Atoh1 is thought to implement a Notch signaling-driven cell-fate-determining binary switch in DOM. The brush (tuft) cells, a poorly understood chemosensory cell type, are not incorporated into this model. We report that brush cell numbers increase dramatically following conditional Atoh1-deletion, demonstrating that brush cell production, determination, differentiation and survival are Atoh1-independent. We also report that brush cells are derived from Gfi1b-expressing progenitors. These and related results suggest a model in which initially equivalent DOM progenitors have three metastable states defined by the transcription factors Hes1, Atoh1, and Gfi1b. Lateral inhibitory Notch signaling normally ensures that Hes1 dominates in one of the two DOMs, invoking a columnar lineage program, while either Atoh1 or Gfi1b dominates in the other DOM, invoking a granulocytic or brush cell lineage program, respectively, and thus implementing a cell fate-determining ternary switch.


Assuntos
Diferenciação Celular/fisiologia , Linhagem da Célula/fisiologia , Mucosa Intestinal/citologia , Microvilosidades/fisiologia , Células-Tronco Multipotentes/citologia , Transdução de Sinais/fisiologia , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/deficiência , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Primers do DNA , Citometria de Fluxo , Proteínas de Homeodomínio/metabolismo , Camundongos , Camundongos Knockout , Microscopia de Fluorescência , Reação em Cadeia da Polimerase , Proteínas Proto-Oncogênicas/metabolismo , Receptores Notch/metabolismo , Proteínas Repressoras/metabolismo , Fatores de Transcrição HES-1
20.
J Biol Chem ; 287(41): 34386-99, 2012 Oct 05.
Artigo em Inglês | MEDLINE | ID: mdl-22865857

RESUMO

Interleukin-7 receptor α (IL-7Rα) is essential for T cell survival and differentiation. Glucocorticoids are potent enhancers of IL-7Rα expression with diverse roles in T cell biology. Here we identify the transcriptional repressor, growth factor independent-1 (Gfi1), as a novel intermediary in glucocorticoid-induced IL-7Rα up-regulation. We found Gfi1 to be a major inhibitory target of dexamethasone by microarray expression profiling of 3B4.15 T-hybridoma cells. Concordantly, retroviral transduction of Gfi1 significantly blunted IL-7Rα up-regulation by dexamethasone. To further assess the role of Gfi1 in vivo, we generated bacterial artificial chromosome (BAC) transgenic mice, in which a modified Il7r locus expresses GFP to report Il7r gene transcription. By introducing this BAC reporter transgene into either Gfi1-deficient or Gfi1-transgenic mice, we document in vivo that IL-7Rα transcription is up-regulated in the absence of Gfi1 and down-regulated when Gfi1 is overexpressed. Strikingly, the in vivo regulatory role of Gfi1 was specific for CD8(+), and not CD4(+) T cells or immature thymocytes. These results identify Gfi1 as a specific transcriptional repressor of the Il7r gene in CD8 T lymphocytes in vivo.


Assuntos
Linfócitos T CD8-Positivos/metabolismo , Proteínas de Ligação a DNA/metabolismo , Regulação da Expressão Gênica/fisiologia , Receptores de Interleucina-7/biossíntese , Proteínas Repressoras/metabolismo , Fatores de Transcrição/metabolismo , Animais , Linfócitos T CD4-Positivos/citologia , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD8-Positivos/citologia , Proteínas de Ligação a DNA/genética , Dexametasona/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Glucocorticoides/farmacologia , Humanos , Camundongos , Camundongos Knockout , Especificidade de Órgãos/efeitos dos fármacos , Especificidade de Órgãos/fisiologia , Receptores de Interleucina-7/genética , Proteínas Repressoras/genética , Fatores de Transcrição/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA