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.
Cell Death Dis ; 14(12): 838, 2023 12 18.
Artigo em Inglês | MEDLINE | ID: mdl-38110339

RESUMO

Acute infection and chronic infection are the two most common fates of pathogenic virus infections. While several factors that contribute to these fates are described, the critical control points and the mechanisms that underlie infection fate regulation are incompletely understood. Using the acute and chronic lymphocytic choriomeningitis virus (LCMV) infection model of mice, we find that the early dynamic pattern of the IFN-I response is a differentiating trait between both infection fates. Acute-infected mice generate a 2-wave IFN-I response while chronic-infected mice generate only a 1-wave response. The underlying cause is a temporal difference in CD8 T cell-mediated killing of splenic marginal zone CD169+ macrophages. It occurs later in acute infection and thus enables CD169+ marginal zone macrophages to produce the 2nd IFN-I wave. This is required for subsequent immune events including induction of inflammatory macrophages, generation of effector CD8+ T cells and virus clearance. Importantly, these benefits come at a cost for the host in the form of spleen fibrosis. Due to an earlier marginal zone destruction, these ordered immune events are deregulated in chronic infection. Our findings demonstrate the critical importance of kinetically well-coordinated sequential immune events for acute infection control and highlights that it may come at a cost for the host organism.


Assuntos
Coriomeningite Linfocítica , Camundongos , Animais , Coriomeningite Linfocítica/patologia , Vírus da Coriomeningite Linfocítica/fisiologia , Infecção Persistente , Camundongos Endogâmicos C57BL , Linfócitos T CD8-Positivos , Macrófagos/patologia
2.
Nat Immunol ; 24(10): 1645-1653, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37709986

RESUMO

Persistent exposure to antigen during chronic infection or cancer renders T cells dysfunctional. The molecular mechanisms regulating this state of exhaustion are thought to be common in infection and cancer, despite obvious differences in their microenvironments. Here we found that NFAT5, an NFAT family transcription factor that lacks an AP-1 docking site, was highly expressed in exhausted CD8+ T cells in the context of chronic infections and tumors but was selectively required in tumor-induced CD8+ T cell exhaustion. Overexpression of NFAT5 in CD8+ T cells reduced tumor control, while deletion of NFAT5 improved tumor control by promoting the accumulation of tumor-specific CD8+ T cells that had reduced expression of the exhaustion-associated proteins TOX and PD-1 and produced more cytokines, such as IFNÉ£ and TNF, than cells with wild-type levels of NFAT5, specifically in the precursor exhausted PD-1+TCF1+TIM-3-CD8+ T cell population. NFAT5 did not promote T cell exhaustion during chronic infection with clone 13 of lymphocytic choriomeningitis virus. Expression of NFAT5 was induced by TCR triggering, but its transcriptional activity was specific to the tumor microenvironment and required hyperosmolarity. Thus, NFAT5 promoted the exhaustion of CD8+ T cells in a tumor-selective fashion.


Assuntos
Coriomeningite Linfocítica , Neoplasias , Humanos , Fatores de Transcrição/metabolismo , Linfócitos T CD8-Positivos , Exaustão das Células T , Infecção Persistente , Microambiente Tumoral , Receptor de Morte Celular Programada 1/genética , Receptor de Morte Celular Programada 1/metabolismo , Vírus da Coriomeningite Linfocítica , Neoplasias/metabolismo
4.
J Exp Med ; 217(3)2020 03 02.
Artigo em Inglês | MEDLINE | ID: mdl-31816635

RESUMO

Type I interferon (IFN-I) provides effective antiviral immunity but can exacerbate harmful inflammatory reactions and cause hematopoietic stem cell (HSC) exhaustion; therefore, IFN-I expression must be tightly controlled. While signaling mechanisms that limit IFN-I induction and function have been extensively studied, less is known about transcriptional repressors acting directly on IFN-I regulatory regions. We show that NFAT5, an activator of macrophage pro-inflammatory responses, represses Toll-like receptor 3 and virus-induced expression of IFN-I in macrophages and dendritic cells. Mice lacking NFAT5 exhibit increased IFN-I production and better control of viral burden upon LCMV infection but show exacerbated HSC activation under systemic poly(I:C)-induced inflammation. We identify IFNß as a primary target repressed by NFAT5, which opposes the master IFN-I inducer IRF3 by binding to an evolutionarily conserved sequence in the IFNB1 enhanceosome that overlaps a key IRF site. These findings illustrate how IFN-I responses are balanced by simultaneously opposing transcription factors.


Assuntos
Interferon Tipo I/imunologia , Fatores de Transcrição/imunologia , Animais , Células Dendríticas/imunologia , Feminino , Inflamação/imunologia , Fator Regulador 3 de Interferon/imunologia , Interferon gama/imunologia , Macrófagos/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Poli I-C/imunologia , Transdução de Sinais/imunologia , Receptores Toll-Like/imunologia , Transcrição Gênica/imunologia
5.
Front Immunol ; 10: 535, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30949179

RESUMO

The transcription factor NFAT5, also known as TonEBP, belongs to the family of Rel homology domain-containing factors, which comprises the NF-κB proteins and the calcineurin-dependent NFAT1 to NFAT4. NFAT5 shares several structural and functional features with other Rel-family factors, for instance it recognizes DNA elements with the same core sequence as those bound by NFAT1 to 4, and like NF-κB it responds to Toll-like receptors (TLR) and activates macrophage responses to microbial products. On the other hand, NFAT5 is quite unique among Rel-family factors as it can be activated by hyperosmotic stress caused by elevated concentrations of extracellular sodium ions. NFAT5 regulates specific genes but also others that are inducible by NF-κB and NFAT1 to 4. The ability of NFAT5 to do so in response to hypertonicity, microbial products, and inflammatory stimuli may extend the capabilities of immune cells to mount effective anti-pathogen responses in diverse microenvironment and signaling conditions. Recent studies identifying osmostress-dependent and -independent functions of NFAT5 have broadened our understanding of how NFAT5 may modulate immune function. In this review we focus on the role of NFAT5 in macrophages and T cells in different contexts, discussing findings from in vivo mouse models of NFAT5 deficiency and reviewing current knowledge on its mechanisms of regulation. Finally, we propose several questions for future research.


Assuntos
Macrófagos/imunologia , Linfócitos T/imunologia , Fatores de Transcrição/imunologia , Animais , Humanos , Hipernatremia/imunologia , Pressão Osmótica
6.
J Exp Med ; 215(11): 2901-2918, 2018 11 05.
Artigo em Inglês | MEDLINE | ID: mdl-30327417

RESUMO

MHCII in antigen-presenting cells (APCs) is a key regulator of adaptive immune responses. Expression of MHCII genes is controlled by the transcription coactivator CIITA, itself regulated through cell type-specific promoters. Here we show that the transcription factor NFAT5 is needed for expression of Ciita and MHCII in macrophages, but not in dendritic cells and other APCs. NFAT5-deficient macrophages showed defective activation of MHCII-dependent responses in CD4+ T lymphocytes and attenuated capacity to elicit graft rejection in vivo. Ultrasequencing analysis of NFAT5-immunoprecipitated chromatin uncovered an NFAT5-regulated region distally upstream of Ciita This region was required for CIITA and hence MHCII expression, exhibited NFAT5-dependent characteristics of active enhancers such as H3K27 acetylation marks, and required NFAT5 to interact with Ciita myeloid promoter I. Our results uncover an NFAT5-regulated mechanism that maintains CIITA and MHCII expression in macrophages and thus modulates their T lymphocyte priming capacity.


Assuntos
Elementos Facilitadores Genéticos/imunologia , Regulação da Expressão Gênica/imunologia , Antígenos de Histocompatibilidade Classe II/imunologia , Macrófagos/imunologia , Proteínas Nucleares/imunologia , Transativadores/imunologia , Fatores de Transcrição/imunologia , Animais , Linfócitos T CD4-Positivos/citologia , Linfócitos T CD4-Positivos/metabolismo , Rearranjo Gênico/imunologia , Antígenos de Histocompatibilidade Classe II/genética , Macrófagos/citologia , Camundongos , Camundongos Knockout , Proteínas Nucleares/genética , Transativadores/genética , Fatores de Transcrição/genética
7.
J Immunol ; 200(1): 305-315, 2018 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-29150563

RESUMO

Macrophages are exquisite sensors of tissue homeostasis that can rapidly switch between pro- and anti-inflammatory or regulatory modes to respond to perturbations in their microenvironment. This functional plasticity involves a precise orchestration of gene expression patterns whose transcriptional regulators have not been fully characterized. We had previously identified the transcription factor NFAT5 as an activator of TLR-induced responses, and in this study we explore its contribution to macrophage functions in different polarization settings. We found that both in classically and alternatively polarized macrophages, NFAT5 enhanced functions associated with a proinflammatory profile such as bactericidal capacity and the ability to promote Th1 polarization over Th2 responses. In this regard, NFAT5 upregulated the Th1-stimulatory cytokine IL-12 in classically activated macrophages, whereas in alternatively polarized ones it enhanced the expression of the pro-Th1 mediators Fizz-1 and arginase 1, indicating that it could promote proinflammatory readiness by regulating independent genes in differently polarized macrophages. Finally, adoptive transfer assays in vivo revealed a reduced antitumor capacity in NFAT5-deficient macrophages against syngeneic Lewis lung carcinoma and ID8 ovarian carcinoma cells, a defect that in the ID8 model was associated with a reduced accumulation of effector CD8 T cells at the tumor site. Altogether, detailed analysis of the effect of NFAT5 in pro- and anti-inflammatory macrophages uncovered its ability to regulate distinct genes under both polarization modes and revealed its predominant role in promoting proinflammatory macrophage functions.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Inflamação/imunologia , Macrófagos/imunologia , Neoplasias Experimentais/imunologia , Fatores de Transcrição/metabolismo , Animais , Arginase/metabolismo , Carcinoma Pulmonar de Lewis , Diferenciação Celular , Homeostase , Mediadores da Inflamação/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Interleucina-12/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células Th1/imunologia , Células Th2/imunologia , Fatores de Transcrição/genética , Regulação para Cima
8.
Sci Signal ; 7(332): re2, 2014 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-24985347

RESUMO

The kinase mammalian target of rapamycin (mTOR) is a central regulator of cell growth and proliferation that integrates inputs from growth factor receptors, nutrient availability, intracellular ATP (adenosine 5'-triphosphate), and a variety of stressors. Since early works in the mid-1990s uncovered the role of mTOR in stimulating protein translation, this kinase has emerged as a rather multifaceted regulator of numerous processes. Whereas mTOR is generally activated by growth- and proliferation-stimulating signals, its activity can be reduced and even suppressed when cells are exposed to a variety of stress conditions. However, cells can also adapt to stress while maintaining their growth capacity and mTOR function. Despite knowledge accumulated on how stress represses mTOR, less is known about mTOR influencing stress responses. In this review, we discuss the capability of mTOR, in particular mTOR complex 1 (mTORC1), to activate stress-responsive transcription factors, and we outline open questions for future investigation.


Assuntos
Proliferação de Células/fisiologia , Biossíntese de Proteínas/fisiologia , Transdução de Sinais/fisiologia , Estresse Fisiológico/fisiologia , Serina-Treonina Quinases TOR/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Humanos , Alvo Mecanístico do Complexo 1 de Rapamicina , Complexos Multiproteicos/metabolismo , Fatores de Transcrição/metabolismo
9.
Clin Orthop Surg ; 6(1): 20-5, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24605185

RESUMO

BACKGROUND: To analyze the incidence and clinical-microbiological characteristics of osteomyelitis (OM) in a tertiary Spanish hospital. METHODS: All cases diagnosed with OM between January 2007 and December 2010 were retrospectively reviewed. The variables examined include epidemiological characteristics, risk factors, affected bone, radiographic changes, histology, microbiological culture results, antibiotic treatment, and the need for surgery. RESULTS: Sixty-three cases of OM were diagnosed. Twenty-six patients (41.3%) had acute OM whereas 37 patients (58.7%) were classified as chronic OM. OM may result from haematogenous or contiguous microbial seeding. In this group, 49 patients (77.8%) presented with OM secondary to a contiguous source of infection and 14 patients had hematogenous OM (22.2%). Staphylococcus aureus was the most commonly found microorganism. CONCLUSIONS: OM mainly affected patients with risk factors related to the presence of vascular diseases. Antibiotic treatment must be guided by susceptibility patterns of individual microorganisms, although it must be performed together with surgery in most of the cases.


Assuntos
Osteomielite , Doença Aguda , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Antibacterianos/uso terapêutico , Criança , Pré-Escolar , Doença Crônica , Feminino , Humanos , Lactente , Masculino , Pessoa de Meia-Idade , Osteomielite/tratamento farmacológico , Osteomielite/epidemiologia , Osteomielite/microbiologia , Estudos Retrospectivos , Fatores de Risco , Espanha/epidemiologia , Infecções Estafilocócicas , Staphylococcus aureus/isolamento & purificação , Centros de Atenção Terciária , Adulto Jovem
10.
Proc Natl Acad Sci U S A ; 110(40): 16091-6, 2013 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-24043824

RESUMO

The Rel-like transcription factors nuclear factor kappa B (NF-κB) and the calcineurin-dependent nuclear factor of activated T cells (NFATc) control specific points of thymocyte maturation. Thymocytes also express a distinct member of the Rel family, the calcineurin-independent, osmostress response regulator NFAT5. Here we show that IKKß regulates the expression of NFAT5 in thymocytes, which in turn contributes to the survival of T-cell receptor αß thymocytes and the transition from the ß-selection checkpoint to the double-positive stage in an osmostress-independent manner. NFAT5-deficient thymocytes had normal expression and proximal signaling of the pre-T-cell receptor but exhibited a partial defect in ß-chain allelic exclusion and increased apoptosis. Further analysis showed that NFAT5 regulated the expression of the prosurvival factors A1 and Bcl2 and attenuated the proapoptotic p53/Noxa axis. These findings position NFAT5 as a target of the IKKß/NF-κB pathway in thymocytes and as a downstream effector of the prosurvival role of the pre-T-cell receptor.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento/imunologia , Glicoproteínas de Membrana/metabolismo , Fatores de Transcrição NFATC/metabolismo , Receptores de Antígenos de Linfócitos T alfa-beta/metabolismo , Transdução de Sinais/imunologia , Timócitos/imunologia , Animais , Apoptose/imunologia , Glicoproteínas de Membrana/imunologia , Camundongos , Camundongos Transgênicos , Fatores de Transcrição NFATC/genética , Fatores de Transcrição NFATC/imunologia , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Receptores de Antígenos de Linfócitos T alfa-beta/imunologia , Timócitos/citologia , Proteína Supressora de Tumor p53/metabolismo
11.
J Exp Med ; 209(2): 379-93, 2012 Feb 13.
Artigo em Inglês | MEDLINE | ID: mdl-22312110

RESUMO

Toll-like receptors (TLRs) engage networks of transcriptional regulators to induce genes essential for antimicrobial immunity. We report that NFAT5, previously characterized as an osmostress responsive factor, regulates the expression of multiple TLR-induced genes in macrophages independently of osmotic stress. NFAT5 was essential for the induction of the key antimicrobial gene Nos2 (inducible nitric oxide synthase [iNOS]) in response to low and high doses of TLR agonists but is required for Tnf and Il6 mainly under mild stimulatory conditions, indicating that NFAT5 could regulate specific gene patterns depending on pathogen burden intensity. NFAT5 exhibited two modes of association with target genes, as it was constitutively bound to Tnf and other genes regardless of TLR stimulation, whereas its recruitment to Nos2 or Il6 required TLR activation. Further analysis revealed that TLR-induced recruitment of NFAT5 to Nos2 was dependent on inhibitor of κB kinase (IKK) ß activity and de novo protein synthesis, and was sensitive to histone deacetylases. In vivo, NFAT5 was necessary for effective immunity against Leishmania major, a parasite whose clearance requires TLRs and iNOS expression in macrophages. These findings identify NFAT5 as a novel regulator of mammalian anti-pathogen responses.


Assuntos
Regulação da Expressão Gênica/imunologia , Redes Reguladoras de Genes/imunologia , Macrófagos/metabolismo , Receptores Toll-Like/metabolismo , Fatores de Transcrição/metabolismo , Animais , Imunoprecipitação da Cromatina , Primers do DNA/genética , Citometria de Fluxo , Regulação da Expressão Gênica/genética , Redes Reguladoras de Genes/genética , Quinase I-kappa B/metabolismo , Immunoblotting , Interleucina-6/metabolismo , Leishmania/imunologia , Luciferases , Camundongos , Camundongos Knockout , Análise em Microsséries , Óxido Nítrico Sintase Tipo II/metabolismo , Nitritos/metabolismo , Plasmídeos/genética , Fatores de Transcrição/genética , Fator de Necrose Tumoral alfa/metabolismo
12.
J Immunol ; 185(11): 6624-35, 2010 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-21037089

RESUMO

Immune cells rely on the transcription factor NFAT5 to adapt to hypertonic stress. The hypertonicity-dependent role of NFAT5 in T cells in vivo remains unclear because mouse models of NFAT5 deficiency have produced substantially different T cell phenotypes. In this study, we analyzed the T cell compartment in NFAT5-null and T cell-specific NFAT5 knockout mice. We found that NFAT5-null mice had constitutive, pronounced hypernatremia and suffered a severe immunodeficiency, with T cell lymphopenia, altered CD8 naive/memory homeostasis, and inability to reject allogeneic tumors. By contrast, T cell-specific NFAT5 knockout mice had normal plasma tonicity, rejected allogeneic tumors, and exhibited only a mild, low-penetrance memory bias in CD8 cells. Notably, when T cells from these mice were cultured ex vivo in hypernatremic media, they exhibited features found in NFAT5-null mice, with pronounced naive/memory imbalance and impaired homeostatic survival in response to IL-7, as well as a severe inhibition of their mitogen-induced proliferation. By analyzing surface receptors whose expression might be affected in NFAT5-deficient cells, we identified CD24 as a novel NFAT5 target induced by hypertonicity both in vitro and in vivo, and required to sustain T cell expansion under osmostress. NFAT5 bound to the Cd24 promoter in response to hypertonicity facilitated the local derepression of chromatin and enhanced the expression of CD24 mRNA and protein. Altogether, our results indicate that the systemic hypernatremia of NFAT5-null mice is a major contributor to their immunodeficiency, and highlight the role of NFAT5 and CD24 in the homeostasis of T cells under osmostress in vivo.


Assuntos
Antígeno CD24/fisiologia , Diferenciação Celular/imunologia , Homeostase/imunologia , Hipernatremia/imunologia , Hipernatremia/patologia , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/patologia , Fatores de Transcrição/fisiologia , Animais , Antígeno CD24/biossíntese , Diferenciação Celular/genética , Linhagem Celular Tumoral , Modelos Animais de Doenças , Rejeição de Enxerto/genética , Rejeição de Enxerto/imunologia , Homeostase/genética , Hipernatremia/genética , Memória Imunológica/genética , Memória Imunológica/imunologia , Linfopenia/genética , Linfopenia/imunologia , Linfopenia/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Pressão Osmótica , Plasmocitoma/genética , Plasmocitoma/imunologia , Plasmocitoma/patologia , Imunodeficiência Combinada Severa/genética , Imunodeficiência Combinada Severa/imunologia , Imunodeficiência Combinada Severa/patologia , Subpopulações de Linfócitos T/metabolismo , Fatores de Transcrição/deficiência , Fatores de Transcrição/genética
13.
Sci Signal ; 2(65): pe20, 2009 Apr 07.
Artigo em Inglês | MEDLINE | ID: mdl-19351952

RESUMO

Nuclear factor of activated T cells 5 (NFAT5) is a member of the Rel family of transcription factors and is an essential inducer of osmoprotective gene products in mammalian cells. Its activation by hypertonicity requires p38 mitogen-activated protein kinase (MAPK) signaling and other pathways. A study now elucidates a signaling cascade regulated by the guanine nucleotide exchange factor Brx that leads to the activation of p38alpha MAPK and the induction of nfat5 messenger RNA in response to osmotic stress in lymphocytes and renal medullary cells. Brx-deficient lymphocytes showed impaired responses to hypertonicity, and brx(+/-) mice exhibited immune defects similar to those of nfat5-deficient mice. These findings support a major role for Brx in regulating the osmoprotective function of NFAT5 in different cell types.


Assuntos
Proteínas de Ancoragem à Quinase A/metabolismo , Regulação da Expressão Gênica/fisiologia , Modelos Biológicos , Fatores de Transcrição NFATC/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Transdução de Sinais/fisiologia , Animais , Humanos , Linfócitos/metabolismo , Camundongos , Antígenos de Histocompatibilidade Menor , Concentração Osmolar
14.
Biochem Pharmacol ; 72(11): 1597-604, 2006 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-16904650

RESUMO

Stress, be it from environmental factors or intrinsic to the cell as result of growth and metabolism, can be harmful to cells. Mammalian cells have developed numerous mechanisms to respond to diverse forms of stress. These mechanisms combine signaling cascades and activation of gene expression programs to orchestrate an adaptive response that will allow the cell to survive and resume its normal functioning. In this review we will focus on the transcription factor NFAT5, a fundamental regulator of the response to osmotic stress in mammalian cells. Identified in 1999, NFAT5 is the latest addition to the Rel family, which comprises the NF-kappaB and NFATc proteins. Though in some of its structural and functional features NFAT5 is a hybrid between these two major groups of Rel proteins, it has unique characteristics that make it stand on its own as a third type of Rel transcription factor. Since its discovery, NFAT5 has been studied mostly in the context of the hypertonicity stress response. The advent of mouse models deficient in NFAT5 and other recent advances have confirmed a fundamental osmoprotective role for this factor in mammals, but also revealed features that suggest it may have a wider range of functions.


Assuntos
Adaptação Fisiológica , Regulação da Expressão Gênica , Fatores de Transcrição NFATC/genética , Fator de Transcrição RelB/genética , Fatores de Transcrição/genética , Sequência de Aminoácidos , Animais , Humanos , Camundongos , Camundongos Knockout , Dados de Sequência Molecular , Fatores de Transcrição NFATC/metabolismo , Pressão Osmótica , Alinhamento de Sequência , Fator de Transcrição RelB/metabolismo , Fatores de Transcrição/metabolismo
15.
Nucleic Acids Res ; 33(12): 3845-54, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-16027109

RESUMO

Tumor necrosis factor (TNF) is a pro-inflammatory cytokine that plays an important role in a variety of infectious and autoimmune disorders. Its transcription is regulated in a stimulus- and cell-type-specific manner via the recruitment of distinct DNA/activator complexes forming secondary structures or enhanceosomes. NFATp, a member of the nuclear factor of activated T cells (NFAT) family of transcription factors, plays a critical role in TNF gene regulation under a variety of conditions. In this study, we show that NFAT5, the most recently described NFAT family member, binds to the TNF promoter in a manner distinct from other NFAT proteins and is a key mediator in the activation of TNF gene transcription during hypertonic stress alone.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Proteínas Nucleares/metabolismo , Regiões Promotoras Genéticas , Fatores de Transcrição/metabolismo , Ativação Transcricional , Fator de Necrose Tumoral alfa/genética , Animais , Sítios de Ligação , Linhagem Celular , Proteínas de Ligação a DNA/antagonistas & inibidores , Proteínas de Ligação a DNA/genética , Genes Reporter , Camundongos , Mutação , Fatores de Transcrição NFATC , Proteínas Nucleares/antagonistas & inibidores , Proteínas Nucleares/genética , Pressão Osmótica , Interferência de RNA , RNA Mensageiro/biossíntese , Fatores de Transcrição/antagonistas & inibidores , Fatores de Transcrição/genética , Fator de Necrose Tumoral alfa/biossíntese
16.
Nat Cell Biol ; 4(7): 540-4, 2002 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12080349

RESUMO

Integrins, receptors for extracellular matrix ligands, are critical regulators of the invasive phenotype. Specifically, the alpha(6)beta(4) integrin has been linked with epithelial cell motility, cellular survival and carcinoma invasion, hallmarks of metastatic tumours. Previous studies have also shown that antagonists of the NFAT (nuclear factor of activated T-cells) family of transcription factors exhibit strong anti-tumour-promoting activity. This suggests that NFAT may function in tumour metastasis. Here, we investigate the involvement of NFAT in promoting carcinoma invasion downstream of the alpha(6)beta(4) integrin. We provide evidence that both NFAT1, and the recently identified NFAT5 isoform, are expressed in invasive human ductal breast carcinomas and participate in promoting carcinoma invasion using cell lines derived from human breast and colon carcinomas. NFAT1 and NFAT5 activity correlates with the expression of the alpha(6)beta(4) integrin. In addition, the transcriptional activity of NFAT5 is induced by alpha(6)beta(4) clustering in the presence of chemo-attractants, resulting in enhanced cell migration. These observations show that NFATs are targets of alpha(6)beta(4) integrin signalling and are involved in promoting carcinoma invasion, highlighting a novel function for this family of transcription factors in human cancer.


Assuntos
Antígenos de Superfície/fisiologia , Proteínas de Ligação a DNA/fisiologia , Integrinas/fisiologia , Invasividade Neoplásica/fisiopatologia , Proteínas Nucleares , Fatores de Transcrição/fisiologia , Neoplasias da Mama/patologia , Neoplasias do Colo/patologia , Feminino , Humanos , Integrina alfa6beta4 , Fatores de Transcrição NFATC , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA