Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 52
Filtrar
1.
Autophagy ; 15(1): 98-112, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30153076

RESUMO

Macroautophagy (hereafter autophagy) is a lysosomal degradation pathway critical for maintaining cellular homeostasis and viability, and is predominantly regarded as a rapid and dynamic cytoplasmic process. To increase our understanding of the transcriptional and epigenetic events associated with autophagy, we performed extensive genome-wide transcriptomic and epigenomic profiling after nutrient deprivation in human autophagy-proficient and autophagy-deficient cells. We observed that nutrient deprivation leads to the transcriptional induction of numerous autophagy-associated genes. These transcriptional changes are reflected at the epigenetic level (H3K4me3, H3K27ac, and H3K56ac) and are independent of autophagic flux. As a proof of principle that this resource can be used to identify novel autophagy regulators, we followed up on one identified target: EGR1 (early growth response 1), which indeed appears to be a central transcriptional regulator of autophagy by affecting autophagy-associated gene expression and autophagic flux. Taken together, these data stress the relevance of transcriptional and epigenetic regulation of autophagy and can be used as a resource to identify (novel) factors involved in autophagy regulation.


Assuntos
Autofagia/fisiologia , Proteína 1 de Resposta de Crescimento Precoce/metabolismo , Epigênese Genética , Perfilação da Expressão Gênica , Lisossomos/metabolismo , Proteína 1 de Resposta de Crescimento Precoce/genética , Regulação da Expressão Gênica , Células HEK293 , Humanos , Nutrientes
2.
Oncogene ; 36(37): 5221-5230, 2017 09 14.
Artigo em Inglês | MEDLINE | ID: mdl-28504718

RESUMO

The transcription factor CCAAT/enhancer-binding protein alpha (C/EBPα) plays a critical role during embryogenesis and is thereafter required for homeostatic glucose metabolism, adipogenesis and myeloid development. Its ability to regulate the expression of lineage-specific genes and induce growth arrest contributes to the terminal differentiation of several cell types, including hepatocytes, adipocytes and granulocytes. CEBPA loss of-function mutations contribute to the development of ~10% of acute myeloid leukemia (AML), stablishing a tumor suppressor role for C/EBPα. Deregulation of C/EBPα expression has also been reported in a variety of additional human neoplasias, including liver, breast and lung cancer. However, functional CEBPA mutations have not been found in solid tumors, suggesting that abrogation of C/EBPα function in non-hematopoietic tissues is regulated by alternative mechanisms. Here we review the function of C/EBPα in solid tumors and focus on the molecular mechanisms underlying its tumor suppressive role.


Assuntos
Proteína alfa Estimuladora de Ligação a CCAAT/genética , Genes Supressores de Tumor , Neoplasias/genética , Animais , Diferenciação Celular/genética , Humanos , Neoplasias/sangue , Neoplasias/metabolismo , Neoplasias/patologia
3.
Autophagy ; 12(10): 1804-1816, 2016 10 02.
Artigo em Inglês | MEDLINE | ID: mdl-27532863

RESUMO

Bone remodeling is a continuous physiological process that requires constant generation of new osteoblasts from mesenchymal stem cells (MSCs). Differentiation of MSCs to osteoblast requires a metabolic switch from glycolysis to increased mitochondrial respiration to ensure the sufficient energy supply to complete this process. As a consequence of this increased mitochondrial metabolism, the levels of endogenous reactive oxygen species (ROS) rise. In the current study we analyzed the role of forkhead box O3 (FOXO3) in the control of ROS levels in human MSCs (hMSCs) during osteogenic differentiation. Treatment of hMSCs with H2O2 induced FOXO3 phosphorylation at Ser294 and nuclear translocation. This ROS-mediated activation of FOXO3 was dependent on mitogen-activated protein kinase 8 (MAPK8/JNK) activity. Upon FOXO3 downregulation, osteoblastic differentiation was impaired and hMSCs lost their ability to control elevated ROS levels. Our results also demonstrate that in response to elevated ROS levels, FOXO3 induces autophagy in hMSCs. In line with this, impairment of autophagy by autophagy-related 7 (ATG7) knockdown resulted in a reduced capacity of hMSCs to regulate elevated ROS levels, together with a reduced osteoblast differentiation. Taken together our findings are consistent with a model where in hMSCs, FOXO3 is required to induce autophagy and thereby reduce elevated ROS levels resulting from the increased mitochondrial respiration during osteoblast differentiation. These new molecular insights provide an important contribution to our better understanding of bone physiology.


Assuntos
Autofagia , Diferenciação Celular , Proteína Forkhead Box O3/metabolismo , Homeostase , Osteogênese , Autofagia/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Homeostase/efeitos dos fármacos , Humanos , Peróxido de Hidrogênio/farmacologia , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Osteogênese/efeitos dos fármacos , Oxirredução/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo
4.
Oncogene ; 35(31): 4141-8, 2016 08 04.
Artigo em Inglês | MEDLINE | ID: mdl-26686090

RESUMO

T-cell acute lymphoblastic leukemia (T-ALL) frequently involves aberrant expression of TAL1 (T-cell acute lymphocytic leukemia 1) and LMO2, oncogenic members of the TAL1 transcriptional complex. Transcriptional activity of the TAL1-complex is thought to have a pivotal role in the transformation of thymocytes and is associated with a differentiation block and self-renewal. The transcription factor Forkhead Box P3 (FOXP3) was recently described to be expressed in a variety of malignancies including T-ALL. Here we show that increased FOXP3 levels negatively correlate with expression of genes regulated by the oncogenic TAL1-complex in human T-ALL patient samples as well as a T-ALL cell line ectopically expressing FOXP3. In these cells, FOXP3 expression results in altered regulation of cell cycle progression and reduced cell viability. Finally, we demonstrate that FOXP3 binds LMO2 in vitro, resulting in decreased interaction between LMO2 and TAL1, providing a molecular mechanism for FOXP3-mediated transcriptional modulation in T-ALL. Collectively, our findings provide initial evidence for a novel role of FOXP3 as a tumor suppressor in T-ALL through modulation of TAL1 transcriptional activity.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Forkhead/fisiologia , Proteínas com Domínio LIM/fisiologia , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/fisiologia , Ciclo Celular , Fatores de Transcrição Forkhead/análise , Humanos , Leucemia-Linfoma Linfoblástico de Células T Precursoras/patologia , Proteína 1 de Leucemia Linfocítica Aguda de Células T , Proteínas Supressoras de Tumor/fisiologia
5.
Cell Death Differ ; 20(9): 1219-29, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23832113

RESUMO

Transcriptional activity of Forkhead box transcription factor class O (FOXO) proteins can result in a variety of cellular outcomes depending on cell type and activating stimulus. These transcription factors are negatively regulated by the phosphoinositol 3-kinase (PI3K)-protein kinase B (PKB) signaling pathway, which is thought to have a pivotal role in regulating survival of tumor cells in a variety of cancers. Recently, it has become clear that FOXO proteins can promote resistance to anti-cancer therapeutics, designed to inhibit PI3K-PKB activity, by inducing the expression of proteins that provide feedback at different levels of this pathway. We questioned whether such a feedback mechanism may also exist directly at the level of FOXO-induced transcription. To identify critical modulators of FOXO transcriptional output, we performed gene expression analyses after conditional activation of key components of the PI3K-PKB-FOXO signaling pathway and identified FOXP1 as a direct FOXO transcriptional target. Using chromatin immunoprecipitation followed by next-generation sequencing, we show that FOXP1 binds enhancers that are pre-occupied by FOXO3. By sequencing the transcriptomes of cells in which FOXO is specifically activated in the absence of FOXP1, we demonstrate that FOXP1 can modulate the expression of a specific subset of FOXO target genes, including inhibiting expression of the pro-apoptotic gene BIK. FOXO activation in FOXP1-knockdown cells resulted in increased cell death, demonstrating that FOXP1 prevents FOXO-induced apoptosis. We therefore propose that FOXP1 represents an important modulator of FOXO-induced transcription, promoting cellular survival.


Assuntos
Apoptose/genética , Resistencia a Medicamentos Antineoplásicos/genética , Fatores de Transcrição Forkhead/metabolismo , Neoplasias/metabolismo , Proteínas Repressoras/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/biossíntese , Animais , Proteínas Reguladoras de Apoptose , Sequência de Bases , Linhagem Celular , Sobrevivência Celular/genética , Proteína Forkhead Box O1 , Fatores de Transcrição Forkhead/genética , Regulação Neoplásica da Expressão Gênica , Células HEK293 , Humanos , Camundongos , Proteínas Mitocondriais/biossíntese , Neoplasias/tratamento farmacológico , Fosfatidilinositol 3-Quinases/metabolismo , Ligação Proteica , Proteínas Proto-Oncogênicas c-akt/metabolismo , Interferência de RNA , RNA Interferente Pequeno , Proteínas Repressoras/genética , Análise de Sequência de DNA , Transdução de Sinais , Transcrição Gênica
6.
Oncogene ; 32(29): 3397-409, 2013 Jul 18.
Artigo em Inglês | MEDLINE | ID: mdl-23246969

RESUMO

Development and progression of cancer are mediated by alterations in transcriptional networks, resulting in a disturbed balance between the activity of oncogenes and tumor suppressor genes. Transcription factors have the capacity to regulate global transcriptional profiles, and are consequently often found to be deregulated in their expression and function during tumorigenesis. Sex-determining region Y-related high-mobility-group box transcription factor 4 (SOX4) is a member of the group C subfamily of the SOX transcription factors and has a critical role during embryogenesis, where its expression is widespread and controls the development of numerous tissues. SOX4 expression is elevated in a wide variety of tumors, including leukemia, colorectal cancer, lung cancer and breast cancer, suggesting a fundamental role in the development of these malignancies. In many cancers, deregulated expression of this developmental factor has been correlated with increased cancer cell proliferation, cell survival, inhibition of apoptosis and tumor progression through the induction of an epithelial-to-mesenchymal transition and metastasis. However, in a limited subset of tumors, SOX4 has also been reported to act as a tumor suppressor. These opposing roles suggest that the outcome of SOX4 activation depends on the cellular context and the tumor origin. Indeed, SOX4 expression, transcriptional activity and target gene specificity can be controlled by signaling pathways, including the transforming growth factor-ß and the WNT pathway, as well as at the post-translational level through regulation of protein stability and interaction with specific cofactors, such as TCF, syntenin-1 and p53. Here, we provide an overview of our current knowledge concerning the role of SOX4 in tumor development and progression.


Assuntos
Transformação Celular Neoplásica/metabolismo , Invasividade Neoplásica , Fatores de Transcrição SOXC/metabolismo , Transdução de Sinais/fisiologia , Animais , Humanos
7.
Oncogene ; 31(21): 2668-79, 2012 May 24.
Artigo em Inglês | MEDLINE | ID: mdl-21986941

RESUMO

The transcription factor Sox4 is aberrantly expressed in many human tumors and can modulate tumorigenesis and metastases of murine tumors in vivo. However, mechanisms that control Sox4 function remain poorly defined. It has recently been observed that DNA damage increases Sox4 protein expression independently of Sox4 mRNA levels, suggesting an as yet undefined post-transcriptional mechanism regulating Sox4 expression and functionality. Here, we show that Sox4 protein is rapidly degraded by the proteasome as indicated by pharmacological inhibition with Mg132 and epoxymycin. Sox4 half-life was found to be less than 1 h as evident by inhibition of protein synthesis using cycloheximide. Ectopic expression of Sox4 deletion mutants revealed that the C-terminal 33 residues of Sox4 were critical in modulating its degradation in a polyubiquitin-independent manner. Syntenin, a Sox4 binding partner, associates with this domain and was found to stabilize Sox4 expression. Syntenin-induced stabilization of Sox4 correlated with Sox4-syntenin relocalization to the nucleus, where both proteins accumulate. Syntenin overexpression or knockdown in human tumor cell lines was found to reciprocally modulate Sox4 protein expression and transcriptional activity implicating its role as a regulator of Sox4. Taken together, our data demonstrate that the Sox4 C-terminal domain regulates polyubiquitin-independent proteasomal degradation of Sox4 that can be modulated by interaction with syntenin. As aberrant Sox4 expression has been found associated with many human cancers, modulation of Sox4 proteasomal degradation may impact oncogenesis and metastatic properties of tumors.


Assuntos
Complexo de Endopeptidases do Proteassoma/metabolismo , Fatores de Transcrição SOXC/metabolismo , Sinteninas/metabolismo , Motivos de Aminoácidos , Linhagem Celular Tumoral , Núcleo Celular/metabolismo , Meia-Vida , Humanos , Processamento Pós-Transcricional do RNA , Ativação Transcricional
8.
Leukemia ; 25(10): 1578-86, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21647153

RESUMO

The transcription factor T-cell acute lymphocytic leukemia (TAL)-1 is a major T-cell oncogene associated with poor prognosis in T-cell acute lymphoblastic leukemia (T-ALL). TAL1 binds histone deacetylase 1 and incubation with histone deacetylase inhibitors (HDACis) promotes apoptosis of leukemia cells obtained from TAL1 transgenic mice. Here, we show for the first time that TAL1 protein expression is strikingly downregulated upon histone deacetylase inhibition in T-ALL cells. This is due to decreased TAL1 gene transcription in cells with native TAL1 promoter, and due to impaired TAL1 mRNA translation in cells that harbor the TAL1(d) microdeletion and consequently express TAL1 under the control of the SCL/TAL1 interrupting locus (SIL) promoter. Notably, HDACi-triggered apoptosis of T-ALL cells is significantly reversed by TAL1 forced overexpression. Our results indicate that the HDACi-mediated apoptotic program in T-ALL cells is partially dependent on their capacity to downregulate TAL1 and provide support for the therapeutic use of HDACi in T-ALL.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Regulação para Baixo , Inibidores de Histona Desacetilases/farmacologia , Leucemia-Linfoma Linfoblástico de Células T Precursoras/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Regulação para Cima , Imunoprecipitação da Cromatina , Humanos , Leucemia-Linfoma Linfoblástico de Células T Precursoras/enzimologia , Leucemia-Linfoma Linfoblástico de Células T Precursoras/patologia , Proteína 1 de Leucemia Linfocítica Aguda de Células T , Células Tumorais Cultivadas
9.
Oncogene ; 28(1): 95-106, 2009 Jan 08.
Artigo em Inglês | MEDLINE | ID: mdl-18836482

RESUMO

Eukaryotic translation initiation factor 4B (eIF4B) plays a critical role during the initiation of protein synthesis and its activity can be regulated by multiple phosphorylation events. In a search for novel protein kinase B (PKB/c-akt) substrates, we identified eIF4B as a potential target. Using an in vitro kinase assay, we found that PKB can directly phosphorylate eIF4B on serine 422 (ser422). Activation of a conditional PKB mutant, interleukin-3 (IL-3) or insulin stimulation resulted in PKB-dependent phosphorylation of this residue in vivo. This was prevented by pretreatment of cells with the phosphatidylinositol 3-kinase (PI3K) inhibitor LY294002 or pharmacological inhibition of PKB. Pretreatment of cells with rapamycin, inhibiting mTOR or U0126 to inhibit MEK, had little effect on eIF4B ser422 phosphorylation. In contrast, following amino-acid refeeding, eIF4B ser422 phosphorylation was found to be mammalian target of rapamycin (mTOR)-dependent. We further identified eIF4B ser406 as a novel mitogen-regulated phosphorylation site. Insulin-induced phosphorylation of eIF4B ser406 was dependent on both MEK and mTOR activity. Utilizing a novel translational control luciferase assay, we could further demonstrate that phosphorylation of ser406 or ser422 is essential for optimal translational activity of eIF4B. These data provide novel insights into complex multikinase regulation of eIF4B phosphorylation and reveal an important mechanism by which PKB can regulate translation, potentially critical for the transforming capacity of this AGC kinase family member.


Assuntos
Transformação Celular Neoplásica/metabolismo , Fatores de Iniciação em Eucariotos/metabolismo , Iniciação Traducional da Cadeia Peptídica , Proteínas Proto-Oncogênicas c-akt/metabolismo , Sequência de Aminoácidos , Animais , Linhagem Celular , Transformação Celular Neoplásica/genética , Cromonas/farmacologia , Fatores de Iniciação em Eucariotos/genética , Insulina/metabolismo , Insulina/farmacologia , Camundongos , Dados de Sequência Molecular , Morfolinas/farmacologia , Inibidores de Fosfoinositídeo-3 Quinase , Fosforilação , Proteínas Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Serina/metabolismo , Especificidade por Substrato , Serina-Treonina Quinases TOR
10.
Oncogene ; 27(16): 2289-99, 2008 Apr 07.
Artigo em Inglês | MEDLINE | ID: mdl-18391971

RESUMO

Modulation FOXO transcription factor activities can lead to a variety of cellular outputs resulting in changes in proliferation, apoptosis, differentiation and metabolic responses. Although FOXO proteins all contain an identical DNA-binding domain their cellular functions appear to be distinct, as exemplified by differences in the phenotype of Foxo1, Foxo3 and Foxo4 null mutant mice. While some of these differences may be attributable to the differential expression patterns of these transcription factors, many cells and tissues express several FOXO isoforms. Recently it has become clear that FOXO proteins can regulate transcriptional responses independently of direct DNA-binding. It has been demonstrated that FOXOs can associate with a variety of unrelated transcription factors, regulating activation or repression of diverse target genes. The complement of transcription factors expressed in a particular cell type is thus critical in determining the functional end point of FOXO activity. These interactions greatly expand the possibilities for FOXO-mediated regulation of transcriptional programmes. This review details currently described FOXO-binding partners and examines the role of these interactions in regulating cell fate decisions.


Assuntos
Fatores de Transcrição Forkhead/metabolismo , Animais , Humanos , Ligação Proteica
11.
Thorax ; 60(7): 538-44, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15994259

RESUMO

BACKGROUND: The inflammatory process in chronic obstructive pulmonary disease (COPD) is characterised by the presence of neutrophils in the lung that are able to synthesise de novo several inflammatory mediators. The local chronic persistent inflammatory response is accompanied by systemic effects such as cytokine induced priming of peripheral leucocytes and muscle wasting. The preactivation or priming of peripheral blood neutrophils was used to gain more insight into the mechanisms of this systemic inflammatory response. METHODS: Gene arrays were performed on peripheral blood neutrophils obtained from healthy donors after stimulation in vitro with tumour necrosis factor (TNF)-alpha, granulocyte-macrophage colony stimulating factor (GM-CSF), or both. The expression of many inflammatory genes was regulated in these cells following stimulation. The expression of inflammatory genes in peripheral blood neutrophils in healthy subjects and those with COPD was measured by real time RT-PCR after stimulation with TNFalpha, GM-CSF, interleukin (IL)-8, fMLP, TNFalpha + GM-CSF, and lipopolysaccharide (LPS). RESULTS: The genes regulated in the gene array with TNFalpha/GM-CSF stimulated neutrophils included cytokines (such as IL-1beta), chemokines (such as IL-8), and adhesion molecules (such as ICAM-1). Disease severity as measured by forced expiratory volume in 1 second (FEV(1)) in COPD patients correlated with expression of several of these genes including IL-1beta (r = -0.540; p = 0.008), MIP-1beta (r = -0.583; p = 0.003), CD83 (r = -0.514; p = 0.012), IL-1 receptor 2 (r = -0.546; p = 0.007), and IL-1 receptor antagonist (r = -0.612; p = 0.002). CONCLUSIONS: These data are consistent with the hypothesis that progression of COPD is associated with the activation of neutrophils in the systemic compartment. De novo expression of inflammatory mediators by peripheral blood neutrophils suggests a pro-inflammatory role for these cells in the pathogenesis of COPD.


Assuntos
Fator Estimulador de Colônias de Granulócitos e Macrófagos/genética , Neutrófilos/metabolismo , Doença Pulmonar Obstrutiva Crônica/patologia , Fator de Necrose Tumoral alfa/genética , Citocinas/metabolismo , Feminino , Volume Expiratório Forçado/fisiologia , Expressão Gênica , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Humanos , Inflamação/genética , Inflamação/metabolismo , Masculino , Pessoa de Meia-Idade , Reação em Cadeia da Polimerase/métodos , Doença Pulmonar Obstrutiva Crônica/genética , Doença Pulmonar Obstrutiva Crônica/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Capacidade Vital/fisiologia
12.
Biochem Soc Trans ; 32(Pt3): 480-4, 2004 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15157166

RESUMO

Granulocytes are critical components of the innate immune system whose lifespan is limited by an intrinsic, constitutive, apoptotic pathway. However, the lifespan of these cells can be extended at an inflammatory locus through interaction with survival factors. Although a wide variety of factors can modulate granulocyte survival, they often utilize a common subset of intracellular signal transduction pathways. Over the last decade, evidence has accumulated that the PI3K (phosphatidylinositol 3-kinase) family of lipid kinases may be critical in regulating the ability of granulocytes to survive at inflammatory loci. Studies utilizing both pharmacological inhibitors of PI3K and isoform-specific knockout mice have demonstrated that this enzyme is needed for the anti-apoptotic effects of granulocyte survival factors. More recently, a serine/threonine protein kinase, termed protein kinase B (also known as c-akt), has been demonstrated to be important in modulating the prosurvival effects of PI3K activation. This can occur through modulation of the expression or phosphorylation of members of the Bcl-2 (B-cell lymphocytic-leukaemia proto-oncogene 2) family of apoptosis regulators. This review summarizes recent results that have implicated a role for PI3K in regulating granulocyte survival.


Assuntos
Apoptose , Granulócitos/patologia , Fosfatidilinositol 3-Quinases/metabolismo , Androstadienos/farmacologia , Animais , Linhagem da Célula , Ativação Enzimática , Granulócitos/enzimologia , Humanos , Inflamação , Camundongos , Camundongos Knockout , Modelos Biológicos , Fosfotransferases/metabolismo , Isoformas de Proteínas , Proteínas Serina-Treonina Quinases/metabolismo , Proto-Oncogene Mas , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-akt , Transdução de Sinais , Wortmanina
13.
Biochem Soc Trans ; 31(Pt 1): 292-7, 2003 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-12546704

RESUMO

Recently, the FOXO (Forkhead box, class O) subfamily of Forkhead transcription factors has been identified as direct targets of phosphoinositide 3-kinase-mediated signal transduction. The AFX (acute-lymphocytic-leukaemia-1 fused gene from chromosome X), FKHR (Forkhead in rhabdomyosarcoma) and FKHR-L1 (FKHR-like 1) transcription factors are directly phosphorylated by protein kinase B, resulting in nuclear export and inhibition of transcription. This signalling pathway was first identified in the nematode worm Caenorhabditis elegans, where it has a role in regulation of the life span of the organism. Studies have shown that this evolutionarily conserved signalling module has a role in regulation of both cell-cycle progression and cell survival in higher eukaryotes. These effects are co-ordinated by FOXO-mediated induction of a variety of specific target genes that are only now beginning to be identified. Interestingly, FOXO transcription factors appear to be able to regulate transcription through both DNA-binding-dependent and -independent mechanisms. Our understanding of the regulation of FOXO activity, and defining specific transcriptional targets, may provide clues to the molecular mechanisms controlling cell fate decisions to divide, differentiate or die.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Proteínas de Ligação a DNA/fisiologia , Proteínas Serina-Treonina Quinases , Fatores de Transcrição , Fatores de Transcrição/metabolismo , Fatores de Transcrição/fisiologia , Animais , Caenorhabditis elegans , Diferenciação Celular , Divisão Celular , Linhagem da Célula , Núcleo Celular/metabolismo , Sobrevivência Celular , Proteínas de Ligação a DNA/química , Modelos Biológicos , Fosforilação , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-akt , Transdução de Sinais , Fatores de Transcrição/química , Transcrição Gênica
14.
Blood ; 98(7): 2014-21, 2001 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-11567984

RESUMO

Inhibition of eosinophil apoptosis by exposure to interleukin-5 (IL-5) is associated with the development of tissue eosinophilia and may contribute to the inflammation characteristic of asthma. Analysis of the signaling events associated with this process has been hampered by the inability to efficiently manipulate eosinophils by the introduction of active or inhibitory effector molecules. Evidence is provided, using a dominant-negative N17 H-Ras protein (dn-H-Ras) and MEK inhibitor U0126, that activation of the Ras-Raf-MEK-ERK pathway plays a determining role in the prolongation of eosinophil survival by IL-5. For these studies, a small region of the human immunodeficiency virus Tat protein, a protein transduction domain known to enter mammalian cells efficiently, was fused to the N-terminus of dn-H-Ras. The Tat-dn-H-Ras protein generated from this construct transduced isolated human blood eosinophils at more than 95% efficiency. When Tat-dn-H-Ras-transduced eosinophils were treated with IL-5, they exhibited a time- and dosage-dependent reduction in extracellular regulated kinase 1 and 2 activation and an inhibition of p90 Rsk1 phosphorylation and IL-5-mediated eosinophil survival in vitro. In contrast, Tat-dn-H-Ras did not inhibit CD11b up-regulation or STAT5 tyrosine phosphorylation. These data demonstrate that Tat dominant-negative protein transduction can serve as an important and novel tool in studying primary myeloid cell signal transduction in primary leukocytes and can implicate the Ras-Raf-MEK-ERK pathway in IL-5-initiated eosinophil survival.


Assuntos
Sobrevivência Celular/efeitos dos fármacos , Eosinófilos/efeitos dos fármacos , Genes ras/genética , Interleucina-5/farmacologia , Fosfotransferases/efeitos dos fármacos , Transdução Genética , Ativação Enzimática/efeitos dos fármacos , Eosinófilos/citologia , Eosinófilos/metabolismo , Produtos do Gene tat/genética , Genes Dominantes , Genes ras/fisiologia , Humanos , Proteínas Quinases Ativadas por Mitógeno/efeitos dos fármacos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Fosfotransferases/metabolismo , Proteínas Recombinantes de Fusão/efeitos dos fármacos , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Recombinantes de Fusão/farmacologia , Proteínas Quinases S6 Ribossômicas/efeitos dos fármacos , Proteínas Quinases S6 Ribossômicas/metabolismo , Transdução de Sinais , Proteínas ras/efeitos dos fármacos , Proteínas ras/metabolismo , Proteínas ras/farmacologia
15.
Science ; 293(5532): 1136-8, 2001 Aug 10.
Artigo em Inglês | MEDLINE | ID: mdl-11498591

RESUMO

Cytokine receptors consist of multiple subunits, which are often shared between different receptors, resulting in the functional redundancy sometimes observed between cytokines. The interleukin 5 (IL-5) receptor consists of an IL-5-specific alpha-subunit (IL-5Ralpha) and a signal-transducing beta-subunit (betac) shared with the IL-3 and granulocyte-macrophage colony-stimulating factor (GM-CSF) receptors. In this study, we sought to find a role for the cytoplasmic domain of IL-5Ralpha. We show that syntenin, a protein containing PSD-95/Discs large/zO-1 (PDZ) domains, associates with the cytoplasmic tail of the IL-5Ralpha. Syntenin was found to directly associate with the transcription factor Sox4. Association of syntenin with IL-5Ralpha was required for IL-5-mediated activation of Sox4. These studies identify a mechanism of transcriptional activation by cytokine-specific receptor subunits.


Assuntos
Linfócitos B/metabolismo , Proteínas de Transporte/metabolismo , Proteínas de Grupo de Alta Mobilidade/metabolismo , Interleucina-5/farmacologia , Peptídeos e Proteínas de Sinalização Intracelular , Proteínas de Membrana , Receptores de Interleucina/metabolismo , Transativadores/metabolismo , Ativação Transcricional , Animais , Linfócitos B/imunologia , Células COS , Proteínas de Transporte/química , Linhagem Celular , Genes Reporter , Humanos , Camundongos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Fosforilação , Mutação Puntual , Estrutura Terciária de Proteína , Receptores de Interleucina/química , Receptores de Interleucina/genética , Receptores de Interleucina-5 , Proteínas Recombinantes de Fusão/metabolismo , Fatores de Transcrição SOXC , Deleção de Sequência , Transdução de Sinais , Sinteninas , Transfecção , Técnicas do Sistema de Duplo-Híbrido
16.
Blood ; 97(11): 3478-83, 2001 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-11369640

RESUMO

Fc receptors play an important role in leukocyte activation and the modulation of ligand binding ("activation") is a critical point of regulation. Previous studies demonstrated that the Fc receptor for IgA (FcalphaRI/CD89) is regulated by cytokine stimulation, switching it to a high-binding state. To investigate the mechanism by which cytokine-induced signal transduction pathways result in FcalphaRI activation, cell lines expressing various receptor mutants were generated. Binding studies indicated that truncation of the C-terminus of the FcalphaRI resulted in constitutive IgA binding, removing the need for cytokine stimulation. Furthermore, mutagenesis of a single C-terminal serine residue (S263) to alanine (S>A) (single-letter amino acid codes) also resulted in constitutive IgA binding, whereas a serine to aspartate (S>D) mutation was no longer functional. The role of S263 might be in regulating the interaction with the cytoskeleton, because disruption of the cytoskeleton results in reduced IgA binding to both FcalphaRwt and FcalphaR_S>A. In addition, overexpression of a membrane-targeted intracellular domain of FcalphaR, and the introduction of cell-permeable CD89 fusion proteins blocked IgA binding, implying a competition for endogenous proteins. The proposal is made that Fc receptors are activated by cytokines via an inside-out mechanism converging at the cytoplasmic tail of these receptors. (Blood. 2001;97:3478-3483)


Assuntos
Antígenos CD/química , Antígenos CD/fisiologia , Citocinas/farmacologia , Receptores Fc/química , Receptores Fc/fisiologia , Serina , Relação Estrutura-Atividade , Antígenos CD/genética , Citoplasma/química , Citoesqueleto/fisiologia , Expressão Gênica , Humanos , Imunoglobulina A/metabolismo , Mutagênese , Fosforilação , Receptores Fc/genética , Proteínas Recombinantes de Fusão/metabolismo , Transdução de Sinais , Transfecção
17.
Cytokine Growth Factor Rev ; 12(1): 19-25, 2001 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11312115

RESUMO

Cytokines mediate the transduction of proliferative, differentiation and survival signals in the hematopoietic system. Although the cytokine family is large and diverse, many different cytokines display broadly overlapping functions. This can be explained by the fact that cytokine receptors often share multiple subunits. Specificity in signal transduction can however be achieved through several mechanisms. This review focuses on how signal specificity can be achieved within the IL-3, IL-5 and GM-CSF receptor family. This is discussed in terms of receptor expression, recent advances in our understanding of intracellular signalling components, and analysis of null mutant knock-out mice.


Assuntos
Citocinas/metabolismo , Receptores de Fator Estimulador das Colônias de Granulócitos e Macrófagos/metabolismo , Receptores de Interleucina-3/metabolismo , Receptores de Interleucina/metabolismo , Transdução de Sinais , Animais , Humanos , Camundongos , Camundongos Knockout , Receptores de Fator Estimulador das Colônias de Granulócitos e Macrófagos/química , Receptores de Fator Estimulador das Colônias de Granulócitos e Macrófagos/genética , Receptores de Interleucina/química , Receptores de Interleucina/genética , Receptores de Interleucina-3/química , Receptores de Interleucina-3/genética , Receptores de Interleucina-5 , Transgenes
19.
J Leukoc Biol ; 68(5): 655-61, 2000 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11073104

RESUMO

Inflammation in allergic asthma is characterized by an influx of eosinophils and the presence of eosinophil products in the bronchial tissue. Orchestration of this inflammatory response is in part mediated by cytokines and chemoattractants, but final activation can require additional stimuli. IgA, the most abundant immunoglobulin at mucosal surfaces, is potentially a potent trigger for eosinophil activation. Previously, we have shown that binding IgA-coated targets is dependent on in vitro stimulation of cells with cytokines. Here, we demonstrate that eosinophils isolated from the blood of allergic asthmatic patients bind IgA beads independently of prior in vitro stimulation. Furthermore, we found that the proinflammatory cytokine, TNF-alpha, is a potent enhancer of IgA binding to eosinophils from allergic asthmatics, and it does not activate FcalphaR on eosinophils isolated from normal donors. The difference in IgA binding by FcalphaRs on normal and patient eosinophils might be explained by the activation of different signal transduction pathways. Studying intracellular signaling, we found an enhanced basal activity of phosphatidylinositol 3-kinase (PI3K) in eosinophils derived from allergic asthmatics. Moreover, inhibition of PI3K in these cells blocked the background and the TNF-alpha-induced IgA binding completely. In summary, these data demonstrate that the responsiveness of human eosinophils to TNF-alpha might be an important contribution for fine-tuning the allergic inflammatory reaction. Furthermore, the preactivation of PI3K results in a broader sensitivity to subsequent challenge with inflammatory cytokines.


Assuntos
Antígenos CD/fisiologia , Asma/sangue , Eosinófilos/fisiologia , Receptores Fc/fisiologia , Adolescente , Adulto , Antígenos CD/imunologia , Antígenos CD/metabolismo , Asma/imunologia , Cromonas/farmacologia , Ativação Enzimática , Inibidores Enzimáticos/farmacologia , Eosinófilos/imunologia , Eosinófilos/metabolismo , Feminino , Humanos , Imidazóis/farmacologia , Imunoglobulina A/imunologia , Imunoglobulina A/metabolismo , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/fisiologia , Masculino , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Morfolinas/farmacologia , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-akt , Piridinas/farmacologia , Receptores Fc/imunologia , Receptores Fc/metabolismo , Receptores do Fator de Necrose Tumoral/metabolismo , Receptores Tipo I de Fatores de Necrose Tumoral , Receptores Tipo II do Fator de Necrose Tumoral , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Fator de Necrose Tumoral alfa/farmacologia , Proteínas Quinases p38 Ativadas por Mitógeno
20.
Mol Cell Biol ; 20(24): 9138-48, 2000 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11094066

RESUMO

Interleukin-3 (IL-3), IL-5, and granulocyte-macrophage colony-stimulating factor regulate the survival, proliferation, and differentiation of hematopoietic lineages. Phosphatidylinositol 3-kinase (PI3K) has been implicated in the regulation of these processes. Here we investigate the molecular mechanism by which PI3K regulates cytokine-mediated proliferation and survival in the murine pre-B-cell line Ba/F3. IL-3 was found to repress the expression of the cyclin-dependent kinase inhibitor p27(KIP1) through activation of PI3K, and this occurs at the level of transcription. This transcriptional regulation occurs through modulation of the forkhead transcription factor FKHR-L1, and IL-3 inhibited FKHR-L1 activity in a PI3K-dependent manner. We have generated Ba/F3 cell lines expressing a tamoxifen-inducible active FKHR-L1 mutant [FKHR-L1(A3):ER*]. Tamoxifen-mediated activation of FKHR-L1(A3):ER* resulted in a striking increase in p27(KIP1) promoter activity and mRNA and protein levels as well as induction of the apoptotic program. The level of p27(KIP1) appears to be critical in the regulation of cell survival since mere ectopic expression of p27(KIP1) was sufficient to induce Ba/F3 apoptosis. Moreover, cell survival was increased in cytokine-starved bone marrow-derived stem cells from p27(KIP1) null-mutant mice compared to that in cells from wild-type mice. Taken together, these observations indicate that inhibition of p27(KIP1) transcription through PI3K-induced FKHR-L1 phosphorylation provides a novel mechanism of regulating cytokine-mediated survival and proliferation.


Assuntos
Proteínas de Ciclo Celular , Citocinas/metabolismo , Proteínas de Ligação a DNA/metabolismo , Inibidores Enzimáticos/metabolismo , Interleucina-3/metabolismo , Proteínas Associadas aos Microtúbulos/genética , Fosfatidilinositol 3-Quinases/metabolismo , Fatores de Transcrição/metabolismo , Transcrição Gênica/genética , Proteínas Supressoras de Tumor , Animais , Apoptose/efeitos dos fármacos , Apoptose/genética , Linfócitos B/citologia , Linfócitos B/metabolismo , Linhagem Celular , Células Cultivadas , Inibidor de Quinase Dependente de Ciclina p27 , Quinases Ciclina-Dependentes/antagonistas & inibidores , Proteínas de Ligação a DNA/antagonistas & inibidores , Proteínas de Ligação a DNA/genética , Inibidores Enzimáticos/farmacologia , Eosinófilos/efeitos dos fármacos , Eosinófilos/metabolismo , Proteína Forkhead Box O1 , Fatores de Transcrição Forkhead , Regulação da Expressão Gênica , Genes Reporter/genética , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Humanos , Hidroxitestosteronas/farmacologia , Interleucina-3/genética , Camundongos , Proteínas Associadas aos Microtúbulos/metabolismo , Mutação , PTEN Fosfo-Hidrolase , Monoéster Fosfórico Hidrolases/genética , Monoéster Fosfórico Hidrolases/metabolismo , Regiões Promotoras Genéticas/efeitos dos fármacos , RNA/metabolismo , Transdução de Sinais , Sirolimo/farmacologia , Tamoxifeno/farmacologia , Fatores de Transcrição/antagonistas & inibidores , Fatores de Transcrição/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...