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











Base de dados
Intervalo de ano de publicação
1.
Cell Rep ; 43(4): 114114, 2024 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-38625791

RESUMO

Patients afflicted with Stimulator of interferon gene (STING) gain-of-function mutations frequently present with debilitating interstitial lung disease (ILD) that is recapitulated in mice expressing the STINGV154M mutation (VM). Prior radiation chimera studies revealed an unexpected and critical role for non-hematopoietic cells in initiating ILD. To identify STING-expressing non-hematopoietic cell types required for the development of ILD, we use a conditional knockin (CKI) model and direct expression of the VM allele to hematopoietic cells, fibroblasts, epithelial cells, or endothelial cells. Only endothelial cell-targeted VM expression results in enhanced recruitment of immune cells to the lung associated with elevated chemokine expression and the formation of bronchus-associated lymphoid tissue, as seen in the parental VM strain. These findings reveal the importance of endothelial cells as instigators of STING-driven lung disease and suggest that therapeutic targeting of STING inhibitors to endothelial cells could potentially mitigate inflammation in the lungs of STING-associated vasculopathy with onset in infancy (SAVI) patients or patients afflicted with other ILD-related disorders.


Assuntos
Células Endoteliais , Mutação com Ganho de Função , Pulmão , Proteínas de Membrana , Animais , Proteínas de Membrana/metabolismo , Proteínas de Membrana/genética , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Camundongos , Pulmão/patologia , Pulmão/metabolismo , Linfócitos/metabolismo , Doenças Pulmonares Intersticiais/patologia , Doenças Pulmonares Intersticiais/genética , Doenças Pulmonares Intersticiais/metabolismo , Camundongos Endogâmicos C57BL , Humanos
2.
bioRxiv ; 2023 Jul 27.
Artigo em Inglês | MEDLINE | ID: mdl-37547024

RESUMO

Patients afflicted with STING gain-of-function mutations frequently present with debilitating interstitial lung disease ( ILD ) that is recapitulated in mice expressing the STING V154M mutation ( VM ). Prior radiation chimera studies revealed an unexpected and critical role for non-hematopoietic cells in the initiation of ILD. To identify STING-expressing non-hematopoietic cell types relevant to ILD, we generated a conditional knock-in ( CKI ) model in which expression of the VM allele was directed to hematopoietic cells, fibroblasts, epithelial cells, or endothelial cells. Only endothelial cell-targeted expression of the mutant allele resulted in the recruitment of immune cells to the lung and the formation of bronchus-associated lymphoid tissue, as seen in the parental VM strain. These findings reveal the importance of endothelial cells as instigators of STING-driven lung disease and suggest that therapeutic targeting of STING inhibitors to endothelial cells could potentially mitigate inflammation in the lungs of SAVI patients or patients afflicted with other ILD-related disorders. Summary: Patients with STING gain-of-function (GOF) mutations develop life-threatening lung autoinflammation. In this study, Gao et al. utilize a mouse model of conditional STING GOF to demonstrate a role for endothelial STING GOF in initiating immune cell recruitment into lung tissues of SAVI mice.

5.
Circ Res ; 129(1): 131-135, 2021 06 25.
Artigo em Inglês | MEDLINE | ID: mdl-34166069

RESUMO

Vascular malformations, affecting ≈1% to 1.5% of the population, comprise a spectrum of developmental patterning defects of capillaries, arteries, veins, and/or lymphatics. The majority of vascular malformations occur sporadically; however, inherited malformations exist as a part of complex congenital diseases. The malformations, ranging from birthmarks to life-threatening conditions, are present at birth, but may reveal signs and symptoms-including pain, bleeding, disfigurement, and functional defects of vital organs-in infancy, childhood, or adulthood. Vascular malformations often exhibit recurrent patterns at affected sites due to the lack of curative treatments. This review series provides a state-of-the-art assessment of vascular malformation research at basic, clinical, genetic, and translational levels.


Assuntos
Vasos Sanguíneos/anormalidades , Anormalidades Linfáticas , Vasos Linfáticos/anormalidades , Malformações Vasculares , Animais , Vasos Sanguíneos/metabolismo , Predisposição Genética para Doença , Variação Genética , Humanos , Anormalidades Linfáticas/genética , Anormalidades Linfáticas/metabolismo , Anormalidades Linfáticas/patologia , Anormalidades Linfáticas/terapia , Vasos Linfáticos/metabolismo , Fenótipo , Fatores de Risco , Malformações Vasculares/genética , Malformações Vasculares/metabolismo , Malformações Vasculares/patologia , Malformações Vasculares/terapia
7.
J Exp Med ; 217(7)2020 07 06.
Artigo em Inglês | MEDLINE | ID: mdl-32405640

RESUMO

Human hepatic vascular cavernomas, the most common benign tumor of the liver, were described in the mid-1800s, yet the mechanisms for their formation and effective treatments remain unknown. Here, we demonstrate gain-of-function mutations in KRAS or BRAF genes within liver endothelial cells as a causal mechanism for hepatic vascular cavernomas. We identified gain-of-function mutations in KRAS or BRAF genes in pathological liver tissue samples from patients with hepatic vascular cavernomas. Mice expressing these human KRASG12D or BRAFV600E mutations in hepatic endothelial cells recapitulated the human hepatic vascular cavernoma phenotype of dilated sinusoidal capillaries with defective branching patterns. KRASG12D or BRAFV600E induced "zipper-like" contiguous expression of junctional proteins at sinusoidal endothelial cell-cell contacts, switching capillaries from branching to cavernous expansion. Pharmacological or genetic inhibition of the endothelial RAS-MAPK1 signaling pathway rescued hepatic vascular cavernoma formation in endothelial KRASG12D- or BRAFV600E-expressing mice. These results uncover a major cause of hepatic vascular cavernomas and provide a road map for their personalized treatment.


Assuntos
Fígado/irrigação sanguínea , Fígado/patologia , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Mutação/genética , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas B-raf/genética , Proteínas Proto-Oncogênicas p21(ras)/genética , Junções Aderentes/metabolismo , Adulto , Idoso de 80 Anos ou mais , Animais , Comunicação Celular/efeitos dos fármacos , Perda do Embrião/patologia , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Feminino , Mutação com Ganho de Função/genética , Humanos , Fígado/efeitos dos fármacos , Fígado/enzimologia , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Macrófagos/patologia , Masculino , Camundongos , Pessoa de Meia-Idade , Proteínas Quinases Ativadas por Mitógeno/metabolismo
9.
J Clin Invest ; 128(5): 2064-2075, 2018 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-29664014

RESUMO

Receptor interacting protein kinase 1 (RIPK1) has important kinase-dependent and kinase-independent scaffolding functions that activate or prevent apoptosis or necroptosis in a cell context-dependent manner. The kinase activity of RIPK1 mediates hypothermia and lethality in a mouse model of TNF-induced shock, reflecting the hyperinflammatory state of systemic inflammatory response syndrome (SIRS), where the proinflammatory "cytokine storm" has long been viewed as detrimental. Here, we demonstrate that cytokine and chemokine levels did not predict survival and, importantly, that kinase-inactive Ripk1D138N/D138N hematopoietic cells afforded little protection from TNF- or TNF/zVAD-induced shock in reconstituted mice. Unexpectedly, RIPK1 kinase-inactive mice transplanted with WT hematopoietic cells remained resistant to TNF-induced shock, revealing that a nonhematopoietic lineage mediated protection. TNF-treated Ripk1D138N/D138N mice exhibited no significant increases in intestinal or vascular permeability, nor did they activate the clotting cascade. We show that TNF administration damaged the liver vascular endothelium and induced phosphorylated mixed lineage kinase domain-like (phospho-MLKL) reactivity in endothelial cells isolated from TNF/zVAD-treated WT, but not Ripk1D138N/D138N, mice. These data reveal that the tissue damage present in this SIRS model is reflected, in part, by breaks in the vasculature due to endothelial cell necroptosis and thereby predict that RIPK1 kinase inhibitors may provide clinical benefit to shock and/or sepsis patients.


Assuntos
Endotélio Vascular/enzimologia , Fígado/enzimologia , Proteína Serina-Treonina Quinases de Interação com Receptores/metabolismo , Síndrome de Resposta Inflamatória Sistêmica/enzimologia , Clorometilcetonas de Aminoácidos/toxicidade , Animais , Endotélio Vascular/lesões , Endotélio Vascular/patologia , Células-Tronco Hematopoéticas , Fígado/patologia , Camundongos , Camundongos da Linhagem 129 , Camundongos Mutantes , Necrose , Proteína Serina-Treonina Quinases de Interação com Receptores/genética , Síndrome de Resposta Inflamatória Sistêmica/induzido quimicamente , Síndrome de Resposta Inflamatória Sistêmica/genética , Síndrome de Resposta Inflamatória Sistêmica/patologia , Fator de Necrose Tumoral alfa/toxicidade
10.
Nat Commun ; 6: 6727, 2015 Apr 13.
Artigo em Inglês | MEDLINE | ID: mdl-25865356

RESUMO

The plasticity of differentiated cells in adult tissues undergoing repair is an area of intense research. Pulmonary alveolar type II cells produce surfactant and function as progenitors in the adult, demonstrating both self-renewal and differentiation into gas exchanging type I cells. In vivo, type I cells are thought to be terminally differentiated and their ability to give rise to alternate lineages has not been reported. Here we show that Hopx becomes restricted to type I cells during development. However, unexpectedly, lineage-labelled Hopx(+) cells both proliferate and generate type II cells during adult alveolar regrowth following partial pneumonectomy. In clonal 3D culture, single Hopx(+) type I cells generate organoids composed of type I and type II cells, a process modulated by TGFß signalling. These findings demonstrate unanticipated plasticity of type I cells and a bidirectional lineage relationship between distinct differentiated alveolar epithelial cell types in vivo and in single-cell culture.


Assuntos
Linhagem da Célula/fisiologia , Células Epiteliais/citologia , Proteínas de Homeodomínio/genética , Alvéolos Pulmonares/citologia , Regeneração/fisiologia , Animais , Técnicas de Cultura de Células , Diferenciação Celular , Linhagem da Célula/efeitos dos fármacos , Proliferação de Células , Rastreamento de Células , Células Clonais , Cruzamentos Genéticos , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Expressão Gênica , Genes Reporter , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Proteínas de Homeodomínio/metabolismo , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Pneumonectomia , Alvéolos Pulmonares/efeitos dos fármacos , Alvéolos Pulmonares/metabolismo , Transdução de Sinais , Tamoxifeno/farmacologia , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta/metabolismo
11.
Cardiovasc Pathol ; 24(2): 80-93, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25434723

RESUMO

Despite high expression levels, the role of Tsc1 in cardiovascular tissue is ill defined. We launched this study to examine the role of Tsc1 in cardiac physiology and pathology. Mice in which Tsc1 was deleted in cardiac tissue and vascular smooth muscle (Tsc1c/cSM22cre(+/-)), developed progressive cardiomegaly and hypertension and died early. Hearts of Tsc1c/cSM22cre(+/-) mice displayed a progressive increase in cardiomyocyte number, and to a lesser extent, size between the ages of 1 and 6 weeks. In addition, compared to control hearts, proliferation markers (phospho-histone 3 and PCNA) were elevated in Tsc1c/cSM22cre(+/-) cardiomyocytes at 0-4 weeks, suggesting that cardiomyocyte proliferation was the predominant mechanism underlying cardiomegaly in Tsc1c/cSM22cre(+/-) mice. To examine the contribution of Tsc1 deletion in peripheral vascular smooth muscle to the cardiac phenotype, Tsc1c/cSM22cre(+/-) mice were treated with the antihypertensive, hydralazine. Prevention of hypertension had no effect on survival, cardiac size, or cardiomyocyte number in these mice. We furthermore generated mice in which Tsc1 was deleted only in vascular smooth muscle but not in cardiac tissue (Tsc1c/cSMAcre-ER(T2+/-)). The Tsc1c/cSMAcre-ER(T2+/-) mice also developed hypertension. However, their survival was normal and no cardiac abnormalities were observed. Our results suggest that loss of Tsc1 in the heart causes cardiomegaly, which is driven by increased cardiomyocyte proliferation that also appears to confer relative resistance to afterload reduction. These findings support a critical role for the Tsc1 gene as gatekeeper in the protection against uncontrolled cardiac growth.


Assuntos
Cardiomegalia/metabolismo , Proliferação de Células/genética , Miócitos Cardíacos/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Animais , Cardiomegalia/genética , Cardiomegalia/patologia , Modelos Animais de Doenças , Hemodinâmica/fisiologia , Hiperplasia/genética , Hiperplasia/metabolismo , Immunoblotting , Imuno-Histoquímica , Camundongos , Camundongos Knockout , Reação em Cadeia da Polimerase Multiplex , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patologia , Miócitos Cardíacos/patologia , Reação em Cadeia da Polimerase , Proteína 1 do Complexo Esclerose Tuberosa , Proteínas Supressoras de Tumor/genética
12.
Am J Respir Cell Mol Biol ; 46(2): 132-8, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22298527

RESUMO

Histone deacetylase (HDAC) inhibitors may offer novel approaches in the treatment of asthma. We postulate that trichostatin A (TSA), a Class 1 and 2 inhibitor of HDAC, inhibits airway hyperresponsiveness in antigen-challenged mice. Mice were sensitized and challenged with Aspergillus fumigatus antigen (AF) and treated with TSA, dexamethasone, or vehicle. Lung resistance (R(L)) and dynamic compliance were measured, and bronchial alveolar lavage fluid (BALF) was analyzed for numbers of leukocytes and concentrations of cytokines. Human precision-cut lung slices (PCLS) were treated with TSA and their agonist-induced bronchoconstriction was measured, and TSA-treated human airway smooth muscle (ASM) cells were evaluated for the agonist-induced activation of Rho and intracellular release of Ca(2+). The activity of HDAC in murine lungs was enhanced by antigen and abrogated by TSA. TSA also inhibited methacholine (Mch)-induced increases in R(L) and decreases in dynamic compliance in naive control mice and in AF-sensitized and -challenged mice. Total cell counts, concentrations of IL-4, and numbers of eosinophils in BALF were unchanged in mice treated with TSA or vehicle, whereas dexamethasone inhibited the numbers of eosinophils in BALF and concentrations of IL-4. TSA inhibited the carbachol-induced contraction of PCLS. Treatment with TSA inhibited the intracellular release of Ca(2+) in ASM cells in response to histamine, without affecting the activation of Rho. The inhibition of HDAC abrogates airway hyperresponsiveness to Mch in both naive and antigen-challenged mice. TSA inhibits the agonist-induced contraction of PCLS and mobilization of Ca(2+) in ASM cells. Thus, HDAC inhibitors demonstrate a mechanism of action distinct from that of anti-inflammatory agents such as steroids, and represent a promising therapeutic agent for airway disease.


Assuntos
Asma/fisiopatologia , Broncoconstrição/efeitos dos fármacos , Inibidores de Histona Desacetilases/farmacologia , Ácidos Hidroxâmicos/farmacologia , Inflamação/prevenção & controle , Modelos Biológicos , Animais , Western Blotting , Líquido da Lavagem Broncoalveolar , Cálcio/metabolismo , Carbacol/farmacologia , Dexametasona/farmacologia , Eletroforese em Gel de Poliacrilamida , Feminino , Humanos , Interleucina-4/metabolismo , Interleucina-6/metabolismo , Camundongos , Camundongos Endogâmicos C57BL
13.
Circ Res ; 109(11): 1240-9, 2011 Nov 11.
Artigo em Inglês | MEDLINE | ID: mdl-21959220

RESUMO

RATIONALE: The development of the cardiac outflow tract (OFT) and great vessels is a complex process that involves coordinated regulation of multiple progenitor cell populations. Among these populations, neural crest cells make important contributions to OFT formation and aortic arch remodeling. Although numerous signaling pathways, including Notch, have been implicated in this process, the role of epigenetics in OFT development remains largely unexplored. OBJECTIVE: Because histone deacetylases (Hdacs) play important roles in the epigenetic regulation of mammalian development, we have investigated the function of Hdac3, a class I Hdac, during cardiac neural crest development in mouse. METHODS AND RESULTS: Using 2 neural crest drivers, Wnt1-Cre and Pax3(Cre), we show that loss of Hdac3 in neural crest results in perinatal lethality and cardiovascular abnormalities, including interrupted aortic arch type B, aortic arch hypoplasia, double-outlet right ventricle, and ventricular septal defect. Affected embryos are deficient in aortic arch artery smooth muscle during midgestation, despite intact neural crest cell migration and preserved development of other cardiac and truncal neural crest derivatives. The Hdac3-dependent block in smooth muscle differentiation is cell autonomous and is associated with downregulation of the Notch ligand Jagged1, a key driver of smooth muscle differentiation in the aortic arch arteries. CONCLUSIONS: These results indicate that Hdac3 plays a critical and specific regulatory role in the neural crest-derived smooth muscle lineage and in formation of the OFT.


Assuntos
Coração Fetal/enzimologia , Cardiopatias Congênitas/enzimologia , Histona Desacetilases/fisiologia , Músculo Liso/patologia , Crista Neural/patologia , Timo/anormalidades , Medula Suprarrenal/embriologia , Animais , Aorta Torácica/anormalidades , Diferenciação Celular/fisiologia , Linhagem da Célula , Movimento Celular , Dupla Via de Saída do Ventrículo Direito/embriologia , Dupla Via de Saída do Ventrículo Direito/enzimologia , Dupla Via de Saída do Ventrículo Direito/genética , Feminino , Coração Fetal/crescimento & desenvolvimento , Regulação da Expressão Gênica no Desenvolvimento , Cardiopatias Congênitas/embriologia , Cardiopatias Congênitas/genética , Comunicação Interventricular/embriologia , Comunicação Interventricular/enzimologia , Comunicação Interventricular/genética , Ventrículos do Coração/embriologia , Ventrículos do Coração/enzimologia , Histona Desacetilases/deficiência , Histona Desacetilases/genética , Masculino , Camundongos , Camundongos Transgênicos , Fator de Transcrição PAX3 , Fatores de Transcrição Box Pareados/fisiologia , Receptores Notch/fisiologia , Proteína Wnt1/fisiologia
14.
Cell Stem Cell ; 8(4): 376-88, 2011 Apr 08.
Artigo em Inglês | MEDLINE | ID: mdl-21474102

RESUMO

Transcription factor-based cellular reprogramming has opened the way to converting somatic cells to a pluripotent state, but has faced limitations resulting from the requirement for transcription factors and the relative inefficiency of the process. We show here that expression of the miR302/367 cluster rapidly and efficiently reprograms mouse and human somatic cells to an iPSC state without a requirement for exogenous transcription factors. This miRNA-based reprogramming approach is two orders of magnitude more efficient than standard Oct4/Sox2/Klf4/Myc-mediated methods. Mouse and human miR302/367 iPSCs display similar characteristics to Oct4/Sox2/Klf4/Myc-iPSCs, including pluripotency marker expression, teratoma formation, and, for mouse cells, chimera contribution and germline contribution. We found that miR367 expression is required for miR302/367-mediated reprogramming and activates Oct4 gene expression, and that suppression of Hdac2 is also required. Thus, our data show that miRNA and Hdac-mediated pathways can cooperate in a powerful way to reprogram somatic cells to pluripotency.


Assuntos
Reprogramação Celular/genética , MicroRNAs/fisiologia , Células-Tronco Pluripotentes/citologia , Animais , Humanos , Fator 4 Semelhante a Kruppel , Camundongos , Fatores de Transcrição/fisiologia
15.
Mol Cell Biol ; 31(11): 2349-63, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21444723

RESUMO

Like phosphorylation, acetylation of lysine residues within a protein is considered a biologically relevant modification that controls the activity of target proteins. During stress of cells, massive protein acetylation takes place. Here, we show that p38 mitogen-activated protein kinase (MAPK), which controls many biological functions during stress, is reversibly acetylated by PCAF/p300 and HDAC3. We identified two acetylated lysine residues, K152 and K53, located in the substrate binding domain and in the ATP-binding pocket of p38, respectively. Acetylation of lysine 53 enhanced the activity of p38 by increasing its affinity for ATP binding. The enhanced acetylation and activation of p38 were found to be in parallel with reduced intracellular ATP levels in cardiomyocytes under stress, as well as in vivo models of cardiac hypertrophy. Thus, our data show, for the first time, that p38 activity is critically regulated by, in addition to phosphorylation, reversible acetylation of a lysine residue, which is conserved in other kinases, implying the possibility of a similar mechanism regulating their activity.


Assuntos
Miócitos Cardíacos/metabolismo , Processamento de Proteína Pós-Traducional , Proteínas Quinases p38 Ativadas por Mitógeno/química , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Acetilação , Acetiltransferases , Trifosfato de Adenosina/metabolismo , Animais , Células Cultivadas , Células HEK293 , Células HeLa , Histona Desacetilases/metabolismo , Humanos , Hipertrofia , Espectrometria de Massas , Camundongos , Miócitos Cardíacos/patologia , Miócitos Cardíacos/fisiologia , Ratos , Estresse Fisiológico , Fatores de Transcrição de p300-CBP/metabolismo
16.
Dev Cell ; 19(3): 450-9, 2010 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-20833366

RESUMO

Regulation of chromatin structure via histone modification has recently received intense attention. Here, we demonstrate that the chromatin-modifying enzyme histone deacetylase 2 (Hdac2) functions with a small homeodomain factor, Hopx, to mediate deacetylation of Gata4, which is expressed by cardiac progenitor cells and plays critical roles in the regulation of cardiogenesis. In the absence of Hopx and Hdac2 in mouse embryos, Gata4 hyperacetylation is associated with a marked increase in cardiac myocyte proliferation, upregulation of Gata4 target genes, and perinatal lethality. Hdac2 physically interacts with Gata4, and this interaction is stabilized by Hopx. The ability of Gata4 to transactivate cell cycle genes is impaired by Hopx/Hdac2-mediated deacetylation, and this effect is abrogated by loss of Hdac2-Gata4 interaction. These results suggest that Gata4 is a nonhistone target of Hdac2-mediated deacetylation and that Hdac2, Hopx, and Gata4 coordinately regulate cardiac myocyte proliferation during embryonic development.


Assuntos
Embrião de Mamíferos/metabolismo , Fator de Transcrição GATA4/metabolismo , Histona Desacetilase 2/fisiologia , Proteínas de Homeodomínio/fisiologia , Miócitos Cardíacos/metabolismo , Acetilação , Animais , Biomarcadores/metabolismo , Western Blotting , Proliferação de Células , Imunofluorescência , Perfilação da Expressão Gênica , Técnicas Imunoenzimáticas , Imunoprecipitação , Hibridização In Situ , Luciferases/metabolismo , Camundongos , Camundongos Knockout , Mutagênese Sítio-Dirigida , Miócitos Cardíacos/citologia , Análise de Sequência com Séries de Oligonucleotídeos , RNA Mensageiro/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa
17.
J Biol Chem ; 285(17): 13304-13, 2010 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-20185820

RESUMO

To determine the underlying mechanism of Foxp1/2/4-mediated transcriptional repression, a yeast two-hybrid screen was performed that identified p66beta, a transcriptional repressor and component of the NuRD chromatin-remodeling complex. We show that direct interactions between Foxp1/4 and p66beta are mediated by the CR2 domain within p66beta and the zinc finger/leucine zipper repression domain found in Foxp1/2/4. These direct interactions are functionally relevant as overexpression of p66beta in combination with Foxp factors cooperatively represses Foxp target gene expression, whereas loss of p66 and Foxp factors results in de-repression of endogenous Foxp target genes in lung epithelial cells. Moreover, the NuRD components HDAC1/2 associate in a macromolecular complex with Foxp proteins, and loss of expression or inhibition of HDAC1/2 activity leads to de-repression of Foxp target gene expression. Importantly, we show in vivo that Foxp1 and HDAC2 act cooperatively to regulate expression of the cytoprotective cytokine interleukin-6, which results in increased resistance to hyperoxic lung injury in Foxp1/HDAC2 compound mutant animals. These data reveal an important interaction between the Foxp transcription factors and the NuRD chromatin-remodeling complex that modulates transcriptional repression critical for the lung epithelial injury response.


Assuntos
Fatores de Transcrição Forkhead/metabolismo , Hiperóxia/metabolismo , Interleucina-6/biossíntese , Pulmão/metabolismo , Complexo Mi-2 de Remodelação de Nucleossomo e Desacetilase/metabolismo , Proteínas Repressoras/metabolismo , Mucosa Respiratória/lesões , Mucosa Respiratória/metabolismo , Animais , Fatores de Transcrição Forkhead/genética , Regulação da Expressão Gênica , Histona Desacetilase 1/genética , Histona Desacetilase 1/metabolismo , Histona Desacetilase 2/genética , Histona Desacetilase 2/metabolismo , Humanos , Hiperóxia/genética , Interleucina-6/genética , Complexo Mi-2 de Remodelação de Nucleossomo e Desacetilase/genética , Camundongos , Camundongos Mutantes , Células NIH 3T3 , Proteínas Repressoras/genética , Técnicas do Sistema de Duplo-Híbrido , Dedos de Zinco
18.
Circ Res ; 105(12): 1240-7, 2009 Dec 04.
Artigo em Inglês | MEDLINE | ID: mdl-19875726

RESUMO

RATIONALE: Cardiac hypertrophy occurs in response to a variety of extrinsic and intrinsic stimuli that impose increased biomechanical stress. The phosphatidylinositol 3-kinase (PI3K)/Akt pathway has previously been strongly associated with hypertrophic signaling in the heart, and with the control of cell size in multiple contexts. This pathway is tightly regulated by many factors, including a host of kinases and phosphatases that function at multiple steps in the signaling cascade. For example, the PTEN (phosphatase and tensin homolog) tumor suppressor protein is a phosphoinositide 3-phosphatase that, by metabolizing phosphatidylinositol 3,4,5-trisphosphate (PtdIns[3,4,5]P(3), PIP3), acts in direct antagonism to growth factor-stimulated PI3K. Inhibition of PTEN leads to cardiomyocyte hypertrophy. Another polyphoinositide phosphatase, inositol polyphosphate-5-phosphatase F (Inpp5f) has recently been implicated in regulation of cardiac hypertrophy. Like PTEN, this phosphatase can degrade PtdIns(3,4,5)P(3) and thus modulates the PI3K/Akt pathway. OBJECTIVE: To characterize the role of Inpp5f in regulating cardiac hypertrophy. METHODS AND RESULTS: We generated homozygous Inpp5f knockout mice and cardiac specific Inpp5f overexpression transgenic mice. We evaluated their hearts for biochemical, structural and functional changes. Inpp5f knockout mice have augmented hypertrophy and reactivation of the fetal gene program in response to stress when compared to wild-type littermates. Furthermore, cardiac overexpression of Inpp5f in transgenic mice reduces hypertrophic responsiveness. CONCLUSIONS: Our results suggest that Inpp5f is a functionally important endogenous modulator of cardiac myocyte size and of the cardiac response to stress.


Assuntos
Cardiomegalia/enzimologia , Miocárdio/enzimologia , Monoéster Fosfórico Hidrolases/metabolismo , Animais , Cardiomegalia/induzido quimicamente , Cardiomegalia/genética , Cardiomegalia/patologia , Tamanho Celular , Modelos Animais de Doenças , Regulação da Expressão Gênica , Humanos , Fosfatos de Inositol/metabolismo , Inositol Polifosfato 5-Fosfatases , Isoproterenol , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Miocárdio/patologia , Monoéster Fosfórico Hidrolases/deficiência , Monoéster Fosfórico Hidrolases/genética , RNA Mensageiro/metabolismo
19.
Gene ; 408(1-2): 187-95, 2008 Jan 31.
Artigo em Inglês | MEDLINE | ID: mdl-18068911

RESUMO

HOXA9 is a homeobox transcription factor expressed in endothelial cells (EC) and its expression is rapidly downregulated during EC activation by inflammatory signals like tumor necrosis factor-alpha (TNF-alpha) and lipopolysaccharide (LPS). Recently, we have shown that HOXA9 overexpression prevents EC activation by inhibiting NF-kappaB activity, which suggests that HOXA9 downregulation is an essential event for EC activation. The present study is directed towards understanding the mechanism of HOXA9 regulation during EC activation. Here we show that nuclear factor-kappaB (NF-kappaB) activation is an essential step for HOXA9 downregulation. Deletion analyses of HOXA9 promoter in EC and NF-kappaB knockout cells have shown that NF-kappaB is a major transcription factor that is absolutely required for HOXA9 downregulation. Our 5' deletion analysis of HOXA9 promoter shows that NF-kappaB response element is localized within first 400 nucleotides, while minimal basal promoter is within 100 nucleotides upstream of its transcriptional start site. We demonstrate that HOXA9 regulates its own expression by positive feedback mechanism. To define mechanism by which HOXA9 autoregulates its expression, we show that HOXA9 DNA binding and transactivation domains are essential.


Assuntos
Proteínas de Homeodomínio/genética , NF-kappa B/metabolismo , Regulação para Baixo , Células Endoteliais/metabolismo , Proteínas de Homeodomínio/metabolismo , Humanos , Regiões Promotoras Genéticas , Fator de Transcrição RelA/metabolismo , Ativação Transcricional , Transfecção , Fator de Necrose Tumoral alfa/metabolismo , Regulação para Cima
20.
Nat Med ; 13(3): 324-31, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17322895

RESUMO

In the adult heart, a variety of stresses induce re-expression of a fetal gene program in association with myocyte hypertrophy and heart failure. Here we show that histone deacetylase-2 (Hdac2) regulates expression of many fetal cardiac isoforms. Hdac2 deficiency or chemical histone deacetylase (HDAC) inhibition prevented the re-expression of fetal genes and attenuated cardiac hypertrophy in hearts exposed to hypertrophic stimuli. Resistance to hypertrophy was associated with increased expression of the gene encoding inositol polyphosphate-5-phosphatase f (Inpp5f) resulting in constitutive activation of glycogen synthase kinase 3beta (Gsk3beta) via inactivation of thymoma viral proto-oncogene (Akt) and 3-phosphoinositide-dependent protein kinase-1 (Pdk1). In contrast, Hdac2 transgenic mice had augmented hypertrophy associated with inactivated Gsk3beta. Chemical inhibition of activated Gsk3beta allowed Hdac2-deficient adults to become sensitive to hypertrophic stimulation. These results suggest that Hdac2 is an important molecular target of HDAC inhibitors in the heart and that Hdac2 and Gsk3beta are components of a regulatory pathway providing an attractive therapeutic target for the treatment of cardiac hypertrophy and heart failure.


Assuntos
Cardiomegalia/enzimologia , Quinase 3 da Glicogênio Sintase/metabolismo , Histona Desacetilases/fisiologia , Proteínas Repressoras/fisiologia , Animais , Cardiomegalia/embriologia , Cardiomegalia/genética , Ativação Enzimática/fisiologia , Feto , Glicogênio Sintase Quinase 3 beta , Histona Desacetilase 2 , Histona Desacetilases/biossíntese , Histona Desacetilases/deficiência , Histona Desacetilases/genética , Isoenzimas/metabolismo , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Dados de Sequência Molecular , Proteínas Repressoras/biossíntese , Proteínas Repressoras/genética , Transdução de Sinais/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA