Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Exp Med ; 214(10): 2999-3014, 2017 Oct 02.
Artigo em Inglês | MEDLINE | ID: mdl-28916644

RESUMO

Innate lymphoid cells (ILCs) patrol environmental interfaces to defend against infection and protect barrier integrity. Using a genetic tuning model, we demonstrate that the signal-dependent transcription factor (TF) STAT5 is critical for accumulation of all known ILC subsets in mice and reveal a hierarchy of STAT5 dependency for populating lymphoid and nonlymphoid tissues. We apply transcriptome and genomic distribution analyses to define a STAT5 gene signature in natural killer (NK) cells, the prototypical ILC subset, and provide a systems-based molecular rationale for its key functions downstream of IL-15. We also uncover surprising features of STAT5 behavior, most notably the wholesale redistribution that occurs when NK cells shift from tonic signaling to acute cytokine-driven signaling, and genome-wide coordination with T-bet, another key TF in ILC biology. Collectively, our data position STAT5 as a central node in the TF network that instructs ILC development, homeostasis, and function and provide mechanistic insights on how it works at cellular and molecular levels.


Assuntos
Homeostase/fisiologia , Linfócitos/fisiologia , Fator de Transcrição STAT5/fisiologia , Animais , Citometria de Fluxo , Perfilação da Expressão Gênica , Imunidade Celular/fisiologia , Imunidade Inata/fisiologia , Células Matadoras Naturais/metabolismo , Células Matadoras Naturais/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fator de Transcrição STAT5/genética
2.
Mol Cell Biol ; 36(16): 2108-20, 2016 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-27215382

RESUMO

Establishment of the mammary luminal cell lineage is controlled primarily by hormones and through specific transcription factors (TFs). Previous studies have linked histone methyltransferases to the differentiation of mammary epithelium, thus opening the possibility of biological significance of counteracting demethylases. We have now demonstrated an essential role for the H3K27me3 demethylase KDM6A in generating a balanced alveolar compartment. Deletion of Kdm6a in the mammary luminal cell lineage led to a paucity of luminal cells and an excessive expansion of basal cells, both in vivo and in vitro The inability to form structurally normal ducts and alveoli during pregnancy resulted in lactation failure. Mutant luminal cells did not exhibit their distinctive transcription factor pattern and displayed basal characteristics. The genomic H3K27me3 landscape was unaltered in mutant tissue, and support for a demethylase-independent mechanism came from mice expressing a catalytically inactive KDM6A. Mammary tissue developed normally in these mice. Chromatin immunoprecipitation sequencing (ChIP-seq) experiments demonstrated KDM6A binding to putative enhancers enriched for key mammary TFs and H3K27ac. This study demonstrated for the first time that the mammary luminal lineage relies on KDM6A to ensure a transcription program leading to differentiated alveoli. Failure to fully implement this program results in structurally and functionally impaired mammary tissue.


Assuntos
Células Epiteliais/citologia , Histona Desmetilases/genética , Histona Desmetilases/metabolismo , Histonas/genética , Glândulas Mamárias Animais/citologia , Fatores de Transcrição/genética , Animais , Diferenciação Celular , Linhagem da Célula , Células Cultivadas , Células Epiteliais/metabolismo , Feminino , Técnicas de Inativação de Genes , Glândulas Mamárias Animais/metabolismo , Camundongos , Gravidez , Análise de Sequência de RNA/métodos
3.
Elife ; 52016 03 21.
Artigo em Inglês | MEDLINE | ID: mdl-26999798

RESUMO

The transcription factor STAT5 is fundamental to the mammalian immune system. However, the relationship between its two paralogs, STAT5A and STAT5B, and the extent to which they are functionally distinct, remain uncertain. Using mouse models of paralog deficiency, we demonstrate that they are not equivalent for CD4(+) 'helper' T cells, the principal orchestrators of adaptive immunity. Instead, we find that STAT5B is dominant for both effector and regulatory (Treg) responses and, therefore, uniquely necessary for immunological tolerance. Comparative analysis of genomic distribution and transcriptomic output confirm that STAT5B has fargreater impact but, surprisingly, the data point towards asymmetric expression (i.e. paralog dose), rather than distinct functional properties, as the key distinguishing feature. Thus, we propose a quantitative model of STAT5 paralog activity whereby relative abundance imposes functional specificity (or dominance) in the face of widespread structural homology.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Fator de Transcrição STAT5/metabolismo , Animais , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fator de Transcrição STAT5/genética
4.
Nucleic Acids Res ; 43(18): 8774-89, 2015 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-26250110

RESUMO

Establishment and differentiation of mammary alveoli during pregnancy are controlled by prolactin through the transcription factors STAT5A and STAT5B (STAT5), which also regulate temporal activation of mammary signature genes. This study addressed the question whether the methyltransferase and transcriptional co-activator EZH2 controls the differentiation clock of mammary epithelium. Ablation of Ezh2 from mammary stem cells resulted in precocious differentiation of alveolar epithelium during pregnancy and the activation of mammary-specific STAT5 target genes. This coincided with enhanced occupancy of these loci by STAT5, EZH1 and RNA Pol II. Limited activation of differentiation-specific genes was observed in mammary epithelium lacking both EZH2 and STAT5, suggesting a modulating but not mandatory role for STAT5. Loss of EZH2 did not result in overt changes in genome-wide and gene-specific H3K27me3 profiles, suggesting compensation through enhanced EZH1 recruitment. Differentiated mammary epithelia did not form in the combined absence of EZH1 and EZH2. Transplantation experiments failed to demonstrate a role for EZH2 in the activity of mammary stem and progenitor cells. In summary, while EZH1 and EZH2 serve redundant functions in the establishment of H3K27me3 marks and the formation of mammary alveoli, the presence of EZH2 is required to control progressive differentiation of milk secreting epithelium during pregnancy.


Assuntos
Lactação/genética , Glândulas Mamárias Animais/metabolismo , Complexo Repressor Polycomb 2/fisiologia , Fator de Transcrição STAT5/metabolismo , Ativação Transcricional , Animais , Proteína Potenciadora do Homólogo 2 de Zeste , Epitélio/metabolismo , Feminino , Código das Histonas , Glândulas Mamárias Animais/crescimento & desenvolvimento , Camundongos Transgênicos , Complexo Repressor Polycomb 2/genética , Complexo Repressor Polycomb 2/metabolismo , Gravidez , RNA Polimerase II/metabolismo , Células-Tronco/metabolismo
5.
Dev Biol ; 395(2): 245-54, 2014 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-25236432

RESUMO

The transcription factor STAT5 mediates prolactin signaling and controls functional development of mammary tissue during pregnancy. This study has identified the miR-193b locus, also encoding miRNAs 365-1 and 6365, as a STAT5 target in mammary epithelium. While the locus was characterized by active histone marks in mammary tissue, STAT5 binding and expression during pregnancy, it was silent in most non-mammary cells. Inactivation of the miR-193b locus in mice resulted in elevated mammary stem/progenitor cell activity as judged by limiting dilution transplantation experiments of primary mammary epithelial cells. Colonies formed by mutant cells were larger and contained more Ki-67 positive cells. Differentiation of mammary epithelium lacking the miR-193b locus was accelerated during puberty and pregnancy, which coincided with the loss of Cav3 and elevated levels of Elf5. Normal colony development was partially obtained upon ectopically expressing Cav3 or upon siRNA-mediated reduction of Elf5 in miR-193b-null primary mammary epithelial cells. This study reveals a previously unknown link between the mammary-defining transcription factor STAT5 and a microRNA cluster in controlling mammary epithelial differentiation and the activity of mammary stem and progenitor cells.


Assuntos
Diferenciação Celular/fisiologia , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Loci Gênicos/genética , Glândulas Mamárias Animais/crescimento & desenvolvimento , MicroRNAs/metabolismo , Fator de Transcrição STAT5/metabolismo , Células-Tronco/fisiologia , Animais , Imunoprecipitação da Cromatina , Ensaio de Unidades Formadoras de Colônias , Feminino , Citometria de Fluxo , Regulação da Expressão Gênica no Desenvolvimento/genética , Camundongos , MicroRNAs/genética , Modelos Biológicos , Gravidez , Análise de Sequência de RNA
6.
Mol Endocrinol ; 28(5): 758-67, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24678731

RESUMO

Mammary-specific genetic programs are activated during pregnancy by the common transcription factor signal transducer and activator of transcription (STAT) 5. More than one third of these genes carry nuclear factor I/B (NFIB) binding motifs that coincide with STAT5 in vivo binding, suggesting functional synergy between these two transcription factors. The role of NFIB in this governance was investigated in mice from which Nfib had been inactivated in mammary stem cells or in differentiating alveolar epithelium. Although NFIB was not required for alveolar expansion, the combined absence of NFIB and STAT5 prevented the formation of functional alveoli. NFIB controlled the expression of mammary-specific and STAT5-regulated genes and chromatin immunoprecipitation-sequencing established STAT5 and NFIB binding at composite regulatory elements containing histone H3 lysine dimethylation enhancer marks and progesterone receptor binding. By integrating previously published chromatin immunoprecipitation-sequencing data sets, the presence of NFIB-STAT5 modules in other cell types was investigated. Notably, genomic sites bound by NFIB in hair follicle stem cells were also occupied by STAT5 in mammary epithelium and coincided with enhancer marks. Many of these genes were under NFIB control in both hair follicle stem cells and mammary alveolar epithelium. We propose that NFIB-STAT5 modules, possibly in conjunction with other transcription factors, control cell-specific genetic programs.


Assuntos
Fatores de Transcrição NFI/fisiologia , Fator de Transcrição STAT5/fisiologia , Animais , Sequência de Bases , Sítios de Ligação , Sequência Consenso , Feminino , Regulação da Expressão Gênica , Ontologia Genética , Lactação , Glândulas Mamárias Animais/citologia , Glândulas Mamárias Animais/fisiologia , Camundongos Nus , Camundongos Transgênicos , Gravidez , Transcriptoma
7.
PLoS One ; 9(1): e85123, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24497923

RESUMO

Development of mammary alveolar epithelium during pregnancy is controlled by prolactin, through the transcription factors STAT5A/B that activate specific sets of target genes. Here we asked whether some of STAT5's functions are mediated by microRNAs. The miR-21 promoter sequence contains a bona-fide STAT5 binding site and miR-21 levels increased in HC11 mammary cells upon prolactin treatment. In vivo miR-21 was abundantly expressed in mammary epithelium at day 6 of pregnancy. Analysis of mice lacking miR-21 revealed that their mammary tissue developed normally during pregnancy and dams were able to nurse their pups. Our study demonstrated that although expression of miR-21 is under prolactin control through the transcription factors STAT5A/B its presence is dispensable for mammary development and lactation.


Assuntos
Lactação/metabolismo , Glândulas Mamárias Animais/crescimento & desenvolvimento , MicroRNAs/genética , Fator de Transcrição STAT5/fisiologia , Animais , Diferenciação Celular , Linhagem Celular , Proliferação de Células , Células Epiteliais/fisiologia , Feminino , Glândulas Mamárias Animais/citologia , Glândulas Mamárias Animais/fisiologia , Camundongos Knockout , Camundongos Nus , MicroRNAs/metabolismo , Gravidez , Regiões Promotoras Genéticas , Ligação Proteica , Ativação Transcricional , Transcriptoma
8.
Mol Cell Biol ; 34(3): 464-73, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24277936

RESUMO

Differentiation of mammary secretory epithelium during pregnancy is characterized by sequential activation of genes over several orders of magnitude. Although the transcription factor STAT5 is key to alveolar development, it is not clear to what extent it controls temporal activation of genetic programs in secretory epithelium. To uncover molecular mechanisms effecting progressive differentiation, we explored genome-wide STAT5 binding and H3K4me3 (i.e., trimethylated histone H3 at K4) marks in mammary tissues at early and midpregnancy and at parturition. STAT5 binding to genes induced during pregnancy was low in immature mammary tissue but increased with epithelial differentiation. Increased STAT5 binding was associated with the establishment of H3K4me3 marks and transcriptional activation. STAT5 binding preceded the formation of H3K4me3 marks in some mammary-specific genes. De novo STAT5 binding was also found at distal sites, indicating enhancers. Furthermore, we established an exhaustive mammary transcriptome. Through integration of RNA-seq and STAT5 and H3K4me4 ChIP-seq data, we discovered novel mammary-specific alternative promoters and genes, including noncoding RNAs. Our findings suggest that STAT5 is an early step in establishing transcription complexes on genes specifically expressed in mammary epithelium. This is the first study in an organ that links progressive chromatin occupancy of STAT5 to the acquisition of H3K4me3 marks and transcription during hormone-induced differentiation.


Assuntos
Histonas/metabolismo , Glândulas Mamárias Animais/metabolismo , Fator de Transcrição STAT5/genética , Ativação Transcricional , Animais , Animais Recém-Nascidos , Cromatina/genética , Cromatina/metabolismo , Epitélio/crescimento & desenvolvimento , Epitélio/metabolismo , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Ontologia Genética , Lisina/metabolismo , Glândulas Mamárias Animais/crescimento & desenvolvimento , Metilação , Camundongos , Análise de Sequência com Séries de Oligonucleotídeos , Fosforilação , Gravidez , Regiões Promotoras Genéticas/genética , Ligação Proteica , Fator de Transcrição STAT5/metabolismo , Transcriptoma
9.
RNA Biol ; 10(12): 1807-14, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24356587

RESUMO

Generating heat and maintaining body temperature is the primary function of brown adipose tissue (BAT). Previous studies have implicated microRNAs, including miR-193b and miR-365-1, in BAT differentiation. We used mouse genetics to further understand the specific contributions of these two miRs. BAT function in mice with an inactivated miR-193b-365-1 locus, as determined by their response to the selective ß3 adrenergic receptor agonist CL316.243 and their tolerance to cold exposure, was normal and expression of genes associated with functional BAT, including Prdm16 and Ucp1, was unaffected. In addition, genome-wide expression profiles of miRNAs and mRNAs in BAT in the presence and absence of miR-193b-365-1 were determined. In summary, these data demonstrate, in contrast to earlier work, that the development, differentiation, and function of BAT do not require the presence of miR-193b and miR-365-1.


Assuntos
Tecido Adiposo Marrom/crescimento & desenvolvimento , Tecido Adiposo Marrom/fisiologia , MicroRNAs/fisiologia , Agonistas de Receptores Adrenérgicos beta 3/farmacologia , Animais , Diferenciação Celular , Células Cultivadas , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Dioxóis/farmacologia , Regulação da Expressão Gênica , Canais Iônicos/genética , Canais Iônicos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Mitocondriais/genética , Proteínas Mitocondriais/metabolismo , Mutação , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Proteína Desacopladora 1
10.
BMC Genomics ; 14: 4, 2013 Jan 16.
Artigo em Inglês | MEDLINE | ID: mdl-23324445

RESUMO

BACKGROUND: Cytokine-activated transcription factors from the STAT (Signal Transducers and Activators of Transcription) family control common and context-specific genetic programs. It is not clear to what extent cell-specific features determine the binding capacity of seven STAT members and to what degree they share genetic targets. Molecular insight into the biology of STATs was gained from a meta-analysis of 29 available ChIP-seq data sets covering genome-wide occupancy of STATs 1, 3, 4, 5A, 5B and 6 in several cell types. RESULTS: We determined that the genomic binding capacity of STATs is primarily defined by the cell type and to a lesser extent by individual family members. For example, the overlap of shared binding sites between STATs 3 and 5 in T cells is greater than that between STAT5 in T cells and non-T cells. Even for the top 1,000 highly enriched STAT binding sites, ~15% of STAT5 binding sites in mouse female liver are shared by other STATs in different cell types while in T cells ~90% of STAT5 binding sites are co-occupied by STAT3, STAT4 and STAT6. In addition, we identified 116 cis-regulatory modules (CRM), which are recognized by all STAT members across cell types defining a common JAK-STAT signature. Lastly, in liver STAT5 binding significantly coincides with binding of the cell-specific transcription factors HNF4A, FOXA1 and FOXA2 and is associated with cell-type specific gene transcription. CONCLUSIONS: Our results suggest that genomic binding of STATs is primarily determined by the cell type and further specificity is achieved in part by juxtaposed binding of cell-specific transcription factors.


Assuntos
Genômica , Sequências Reguladoras de Ácido Nucleico/genética , Fatores de Transcrição STAT/metabolismo , Animais , Bases de Dados Genéticas , Humanos , Motivos de Nucleotídeos , Ligação Proteica
11.
Nucleic Acids Res ; 41(3): 1622-36, 2013 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-23275557

RESUMO

The transcription factors Signal Transducer and Activator of Transcription (STAT) 5A/B mediate prolactin-induced mammary development during pregnancy. However, it is not clear how the different processes, expansion and maturation of alveolar precursor cells and the differential induction of milk protein genes are regulated on a molecular level. We have used mouse genetics and genome-wide analyses to determine how altering concentrations of STAT5A and STAT5B impacts mammary epithelial development during pregnancy and the regulation of target genes. The presence of only a single Stat5a or Stat5b allele was sufficient for the establishment of histologically undifferentiated alveolar units and two alleles permitted the execution of a differentiation program similar to that found with all four alleles. While one copy of Stat5 induced limited expression of target genes, two copies activated a lactation-like gene signature. Using ChIP-seq analyses on intact tissue under physiological conditions, we found that highly expressed and regulated genes were bound by STAT5 in their promoter proximal regions, whereas upstream binding had minor biological consequences. Remarkably, 80% of the genes bound by STAT5 in vivo were not under STAT5 control. RNA polymerase II intensity was directly proportional to STAT5 concentration only on STAT5 regulated genes providing mechanistic insight by which STAT5 activates mammary specific genes.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Lactação/genética , Glândulas Mamárias Animais/metabolismo , Fator de Transcrição STAT5/metabolismo , Animais , Sítios de Ligação , Epitélio/anatomia & histologia , Epitélio/crescimento & desenvolvimento , Epitélio/metabolismo , Feminino , Genótipo , Glândulas Mamárias Animais/anatomia & histologia , Glândulas Mamárias Animais/crescimento & desenvolvimento , Camundongos , Camundongos Knockout , Camundongos Nus , Gravidez , RNA Polimerase II/metabolismo , Fator de Transcrição STAT5/genética
12.
Cancer Res ; 73(1): 373-84, 2013 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-23149921

RESUMO

MOZ-TIF2 is a leukemogenic fusion oncoprotein that confers self-renewal capability to hematopoietic progenitor cells and induces acute myelogenous leukemia (AML) with long latency in bone marrow transplantation assays. Here, we report that FLT3-ITD transforms hematopoietic cells in cooperation with MOZ-TIF2 in vitro and in vivo. Coexpression of FLT3-ITD confers growth factor independent survival/proliferation, shortens disease latency, and results in an increase in the number of leukemic stem cells (LSC). We show that STAT5, a major effector of aberrant FLT3-ITD signal transduction, is both necessary and sufficient for this cooperative effect. In addition, STAT5 signaling is essential for MOZ-TIF2-induced leukemic transformation itself. Lack of STAT5 in fetal liver cells caused rapid differentiation and loss of replating capacity of MOZ-TIF2-transduced cells enriched for LSCs. Furthermore, mice serially transplanted with Stat5(-/-) MOZ-TIF2 leukemic cells develop AML with longer disease latency and finally incomplete penetrance when compared with mice transplanted with Stat5(+/+) MOZ-TIF2 leukemic cells. These data suggest that STAT5AB is required for the self-renewal of LSCs and represents a combined signaling node of FLT3-ITD and MOZ-TIF2 driven leukemogenesis. Therefore, targeting aberrantly activated STAT5 or rewired downstream signaling pathways may be a promising therapeutic option.


Assuntos
Transformação Celular Neoplásica/metabolismo , Leucemia Mieloide Aguda/metabolismo , Células-Tronco Neoplásicas/metabolismo , Proteínas de Fusão Oncogênica/metabolismo , Fator de Transcrição STAT5/metabolismo , Animais , Southern Blotting , Citometria de Fluxo , Camundongos , Camundongos Endogâmicos BALB C , Transdução de Sinais/fisiologia
14.
Genesis ; 50(9): 665-71, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22389215

RESUMO

Genome wide analysis revealed the miR-17/92 cluster as a STAT5 target. This cluster encodes six microRNAs, which predictably target genes that play a role in mammary gland development. In this study, we have deleted the miR-17/92 cluster in mammary stem cells and evaluated in the mouse its function during mammary gland development. Loss of the miR-17/92 cluster did not affect mammary development from prepuberty to lactation. Our studies demonstrated that, while expression of the miR-17/92 cluster is under control of the key mammary transcription factor STAT5, its presence is not required for normal mammary development or lactation.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento/genética , Glândulas Mamárias Animais/crescimento & desenvolvimento , MicroRNAs/genética , Família Multigênica/fisiologia , Fator de Transcrição STAT5/genética , Animais , Células Cultivadas , Imunoprecipitação da Cromatina , Células Epiteliais/metabolismo , Feminino , Deleção de Genes , Sequenciamento de Nucleotídeos em Larga Escala , Lactação/genética , Glândulas Mamárias Animais/citologia , Camundongos , Camundongos Nus , Gravidez , RNA Mensageiro/genética , Fator de Transcrição STAT5/metabolismo , Análise de Sequência de DNA , Células-Tronco
15.
Anal Biochem ; 412(1): 92-5, 2011 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-21255551

RESUMO

CRE-loxP-mediated inactivation and activation of genes in mouse mammary epithelium have been widely used to study genetic pathways in normal development and neoplastic transformation in vivo. In 1997, we generated three distinct mouse lines carrying an identical MMTV-Cre transgene (lines A, D, and F). Because the presence of CRE recombinase can adversely affect the physiology of nonmammary cells, we explored whether transgenic females display lactational defects. Whereas dams from line D nurse their pups and display overtly normal mammary development, line A shows some impairment during lactation and females from line F completely fail to nurse their litters. The ability to nurse a litter correlates with the extent of alveolar development and differentiation. This study demonstrates the importance of including appropriate "Cre-only" controls and provides guidelines to avoid problems in data interpretation.


Assuntos
Integrases/genética , Glândulas Mamárias Animais/metabolismo , Vírus do Tumor Mamário do Camundongo/genética , Transgenes , Animais , Animais Geneticamente Modificados , Feminino , Deleção de Genes , Integrases/metabolismo , Lactação , Glândulas Mamárias Animais/anormalidades , Vírus do Tumor Mamário do Camundongo/metabolismo , Camundongos , Camundongos Transgênicos/genética
16.
Neoplasia ; 12(11): 899-905, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21076615

RESUMO

Type I and type II classes of interferons (IFNs) signal through the JAK/STAT1 pathway and are known to be important in adaptive and innate immune responses and in protection against tumors. Although STAT1 is widely considered a tumor suppressor, it remains unclear, however, if this function occurs in tumor cells (cell autonomous) or if STAT1 acts primarily through immune cells. Here, the question of whether STAT1 has a cell autonomous role in mammary tumor formation was addressed in a mouse model of ERBB2/neu-induced breast cancer in the absence and presence of STAT1. For this purpose, mice that carry floxed Stat1 alleles, which permit cell-specific removal of STAT1, were generated. To induce tumors only in mammary cells lacking STAT1, Stat1 floxed mice were crossed with transgenic mice that express cre recombinase and the neu oncogene under the mouse mammary tumor virus LTR (Stat1fl/fl NIC). Stat1 was effectively deleted in mammary epithelium of virgin Stat1fl/fl NIC females. Time-to-tumor onset was significantly shorter in Stat1fl/fl NIC females than in WT NIC (Wilcoxon rank sum test, P = .02). The median time-to-tumor onset in the Stat1fl/fl NIC mice was 49.4 weeks, whereas it was 62.4 weeks in the WT NIC mice. These results suggest that STAT1 in mammary epithelial cells may play a role in suppressing tumorigenesis. The Stat1 floxed allele described in this study is also a unique resource to determine the cellular targets of IFNs and STAT1 action, which should aid our understanding and appreciation of these pathways.


Assuntos
Glândulas Mamárias Animais/metabolismo , Neoplasias Mamárias Animais/genética , Receptor ErbB-2/genética , Fator de Transcrição STAT1/genética , Animais , Western Blotting , Núcleo Celular/metabolismo , Células Cultivadas , Embrião de Mamíferos/citologia , Epitélio/metabolismo , Feminino , Fibroblastos/metabolismo , Estimativa de Kaplan-Meier , Masculino , Neoplasias Mamárias Animais/metabolismo , Neoplasias Mamárias Animais/patologia , Camundongos , Camundongos da Linhagem 129 , Camundongos Knockout , Camundongos Transgênicos , Receptor ErbB-2/metabolismo , Fator de Transcrição STAT1/metabolismo , Carga Tumoral , Células Tumorais Cultivadas
17.
Hepatology ; 52(5): 1808-18, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21038417

RESUMO

UNLABELLED: Although the cytokine-inducible transcription factor signal transducer and activator of transcription 5 (STAT5) promotes proliferation of a wide range of cell types, there are cell-specific and context-specific cases in which loss of STAT5 results in enhanced cell proliferation. Here, we report that loss of STAT5 from mouse embryonic fibroblasts (MEFs) leads to enhanced proliferation, which was linked to reduced levels of the cell cycle inhibitors p15(INK4B) and p21(CIP1). We further demonstrate that growth hormone, through the transcription factor STAT5, enhances expression of the Cdkn2b (cyclin-dependent kinase inhibitor 2B) gene and that STAT5A binds to interferon-gamma-activated sequence sites within the promoter. We recently demonstrated that ablation of STAT5 from liver results in hepatocellular carcinoma upon CCl4 treatment. We now establish that STAT5, like in MEFs, activates expression of the Cdkn2b gene in liver tissue. Loss of STAT5 led to diminished p15(INK4B) and increased hepatocyte proliferation. CONCLUSION: This study for the first time demonstrates that cytokines, through STAT5, induce the expression of a key cell cycle inhibitor. These experiments therefore shed mechanistic light on the context-specific role of STAT5 as tumor suppressor.


Assuntos
Inibidor de Quinase Dependente de Ciclina p19/genética , Inibidor de Quinase Dependente de Ciclina p21/genética , Fator de Transcrição STAT5/genética , Animais , Tetracloreto de Carbono/toxicidade , Carcinoma Hepatocelular/induzido quimicamente , Ciclo Celular , Divisão Celular , Embrião de Mamíferos , Fibroblastos/citologia , Fibroblastos/fisiologia , Homeostase , Cirrose Hepática/induzido quimicamente , Cirrose Hepática/genética , Neoplasias Hepáticas/induzido quimicamente , Neoplasias Hepáticas/patologia , Masculino , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Análise de Sequência com Séries de Oligonucleotídeos , Fator de Transcrição STAT5/deficiência , Proteínas Supressoras de Tumor/genética
18.
Proc Natl Acad Sci U S A ; 107(45): 19455-60, 2010 Nov 09.
Artigo em Inglês | MEDLINE | ID: mdl-20974963

RESUMO

Thymic stromal lymphopoietin (TSLP) is a type I cytokine that plays essential roles in allergic/inflammatory skin and airway disorders, in helminth infections, and in regulating intestinal immunity. TSLP signals via IL-7Rα and a specific TSLPR subunit that is highly related to the common cytokine receptor γ chain, γ(c). Although TSLP has effects on a broad range of hematopoetic cells and can induce STAT5 phosphorylation, TSLP was reported to not signal via JAK kinases, and the mechanism by which TSLP regulates STAT5 phosphorylation has been unclear. We now demonstrate the role of JAK1 and JAK2 in TSLP-mediated STAT5 phosphorylation in mouse and human primary CD4(+) T cells, in contrast to the known activation of JAK1 and JAK3 by the related cytokine, IL-7. We also show that just as JAK1 interacts with IL-7Rα, JAK2 is associated with TSLPR protein. Moreover, we demonstrate the importance of STAT5 activation for TSLP-mediated survival and proliferation of CD4(+) T cells. These findings clarify the basis for TSLP-mediated signaling and provide an example wherein a cytokine uses JAK1 and JAK2 to mediate the activation of STAT5.


Assuntos
Citocinas/metabolismo , Interleucina-7/metabolismo , Janus Quinase 1/metabolismo , Janus Quinase 2/metabolismo , Fator de Transcrição STAT5/metabolismo , Animais , Linfócitos T CD4-Positivos/citologia , Linfócitos T CD4-Positivos/imunologia , Proliferação de Células , Sobrevivência Celular/imunologia , Células Cultivadas , Humanos , Camundongos , Fosforilação , Transdução de Sinais , Linfopoietina do Estroma do Timo
19.
J Biol Chem ; 285(43): 32704-32709, 2010 Oct 22.
Artigo em Inglês | MEDLINE | ID: mdl-20720003

RESUMO

Cytokines control the biology of hematopoietic stem cells (HSCs) and progenitor cells in part through the transcription factors STAT5A/B. To investigate the target genes of STAT5A/B activated by cytokines in HSCs and progenitors, we performed microarray analyses using Lineage(-) Sca-1(+) c-Kit(+) (KSL) cells in the presence and absence of STAT5A/B. Stimulation with a mixture containing IL-3, IL-6, stem cell factor, thrombopoietin, and Flt3 ligand induced Ccn3/Nov mRNA over 100-fold in WT (control) but not Stat5a/b-null KSL cells. CCN3/NOV is a positive regulator of human HSC self-renewal and development of committed blood cells. Without stimulation, the Ccn3/Nov signal level was low in control KSL cells similar to Stat5a/b-null KSL cells. To determine which cytokine activates the Ccn3/Nov gene, we analyzed Lineage(-) c-Kit(+) (KL) and 32D cells using quantitative PCR and ChIP assays. Although stimulation with a mixture lacking IL-3 prevented the induction of Ccn3/Nov in control KL cells, IL-3 alone could induce Ccn3/Nov mRNA in control KL and 32D cells. ChIP assays using 32D cells revealed IL-3-induced binding of STAT5A/B to a γ-interferon-activated sequences site in the Ccn3/Nov gene promoter. This is the first report that Ccn3/Nov is directly induced by cytokines through STAT5A/B.


Assuntos
Citocinas/metabolismo , Regulação da Expressão Gênica/fisiologia , Células-Tronco Hematopoéticas/metabolismo , Proteína Sobre-Expressa em Nefroblastoma/biossíntese , Fator de Transcrição STAT5/metabolismo , Animais , Linhagem Celular , Citocinas/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Células-Tronco Hematopoéticas/citologia , Humanos , Camundongos , Camundongos Mutantes , Proteína Sobre-Expressa em Nefroblastoma/genética , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Fator de Transcrição STAT5/genética , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/metabolismo
20.
Genes Dev ; 23(20): 2382-7, 2009 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-19833766

RESUMO

Mammary alveologenesis is abrogated in the absence of the transcription factors STAT5A/5B, which mediate cytokine signaling. To reveal the underlying causes for this developmental block, we studied mammary stem and progenitor cells. While loss of STAT5A/5B did not affect the stem cell population and its ability to form mammary ducts, luminal progenitors were greatly reduced and unable to form alveoli during pregnancy. Temporally controlled expression of transgenic STAT5A in mammary epithelium lacking STAT5A/5B restored the luminal progenitor population and rescued alveologenesis in a reversible fashion in vivo. Thus, STAT5A is necessary and sufficient for the establishment of luminal progenitor cells.


Assuntos
Glândulas Mamárias Animais/citologia , Fator de Transcrição STAT5/metabolismo , Células-Tronco/citologia , Animais , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Gravidez , Fator de Transcrição STAT5/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...