Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 63
Filtrar
1.
Circ Res ; 131(11): 926-943, 2022 11 11.
Artigo em Inglês | MEDLINE | ID: mdl-36278398

RESUMO

BACKGROUND: Heart failure with preserved ejection fraction (HFpEF) is a growing health problem without effective therapies. Epidemiological studies indicate that diabetes is a strong risk factor for HFpEF, and about 45% of patients with HFpEF are suffering from diabetes, yet the underlying mechanisms remain elusive. METHODS: Using a combination of echocardiography, hemodynamics, RNA-sequencing, molecular biology, in vitro and in vivo approaches, we investigated the roles of SIRT6 (sirtuin 6) in regulation of endothelial fatty acid (FA) transport and HFpEF in diabetes. RESULTS: We first observed that endothelial SIRT6 expression was markedly diminished in cardiac tissues from heart failure patients with diabetes. We then established an experimental mouse model of HFpEF in diabetes induced by a combination of the long-term high-fat diet feeding and a low-dose streptozocin challenge. We also generated a unique humanized SIRT6 transgenic mouse model, in which a single copy of human SIRT6 transgene was engineered at mouse Rosa26 locus and conditionally induced with the Cre-loxP technology. We found that genetically restoring endothelial SIRT6 expression in the diabetic mice ameliorated diastolic dysfunction concurrently with decreased cardiac lipid accumulation. SIRT6 gain- or loss-of-function studies showed that SIRT6 downregulated endothelial FA uptake. Mechanistically, SIRT6 suppressed endothelial expression of PPARγ through SIRT6-dependent deacetylation of histone H3 lysine 9 around PPARγ promoter region; and PPARγ reduction mediated SIRT6-dependent inhibition of endothelial FA uptake. Importantly, oral administration of small molecule SIRT6 activator MDL-800 to diabetic mice mitigated cardiac lipid accumulation and diastolic dysfunction. CONCLUSIONS: The impairment of endothelial SIRT6 expression links diabetes to HFpEF through the alteration of FA transport across the endothelial barrier. Genetic and pharmacological strategies that restored endothelial SIRT6 function in mice with diabetes alleviated experimental HFpEF by limiting FA uptake and improving cardiac metabolism, thus warranting further clinical evaluation.


Assuntos
Diabetes Mellitus Experimental , Insuficiência Cardíaca , Sirtuínas , Humanos , Camundongos , Animais , Volume Sistólico/fisiologia , Insuficiência Cardíaca/metabolismo , PPAR gama , Modelos Animais de Doenças , Sirtuínas/genética , Lipídeos
2.
Eur Heart J ; 44(20): 1818-1833, 2023 05 21.
Artigo em Inglês | MEDLINE | ID: mdl-36469488

RESUMO

AIMS: Variants of the junctional cadherin 5 associated (JCAD) locus associate with acute coronary syndromes. JCAD promotes experimental atherosclerosis through the large tumor suppressor kinase 2 (LATS2)/Hippo pathway. This study investigates the role of JCAD in arterial thrombosis. METHODS AND RESULTS: JCAD knockout (Jcad-/-) mice underwent photochemically induced endothelial injury to trigger arterial thrombosis. Primary human aortic endothelial cells (HAECs) treated with JCAD small interfering RNA (siJCAD), LATS2 small interfering RNA (siLATS2) or control siRNA (siSCR) were employed for in vitro assays. Plasma JCAD was measured in patients with chronic coronary syndrome or ST-elevation myocardial infarction (STEMI). Jcad-/- mice displayed reduced thrombogenicity as reflected by delayed time to carotid occlusion. Mechanisms include reduced activation of the coagulation cascade [reduced tissue factor (TF) expression and activity] and increased fibrinolysis [higher thrombus embolization episodes and D-dimer levels, reduced vascular plasminogen activator inhibitor (PAI)-1 expression]. In vitro, JCAD silencing inhibited TF and PAI-1 expression in HAECs. JCAD-silenced HAECs (siJCAD) displayed increased levels of LATS2 kinase. Yet, double JCAD and LATS2 silencing did not restore the control phenotype. si-JCAD HAECs showed increased levels of phosphoinositide 3-kinases (PI3K)/ proteinkinase B (Akt) activation, known to downregulate procoagulant expression. The PI3K/Akt pathway inhibitor-wortmannin-prevented the effect of JCAD silencing on TF and PAI-1, indicating a causative role. Also, co-immunoprecipitation unveiled a direct interaction between JCAD and Akt. Confirming in vitro findings, PI3K/Akt and P-yes-associated protein levels were higher in Jcad-/- animals. Lastly, as compared with chronic coronary syndrome, STEMI patients showed higher plasma JCAD, which notably correlated positively with both TF and PAI-1 levels. CONCLUSIONS: JCAD promotes arterial thrombosis by modulating coagulation and fibrinolysis. Herein, reported translational data suggest JCAD as a potential therapeutic target for atherothrombosis.


Assuntos
Infarto do Miocárdio com Supradesnível do Segmento ST , Trombose , Animais , Humanos , Camundongos , Células Endoteliais/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Inibidor 1 de Ativador de Plasminogênio/metabolismo , Proteínas Serina-Treonina Quinases/genética , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , RNA Interferente Pequeno , Transdução de Sinais , Infarto do Miocárdio com Supradesnível do Segmento ST/metabolismo , Trombose/metabolismo , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/metabolismo
3.
Trends Biochem Sci ; 44(7): 561-564, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31036409

RESUMO

Hutchinson-Gilford progeria syndrome (HGPS) is an ultrarare and fatal disease with features of premature aging and cardiovascular diseases (atherosclerosis, myocardial infarction, and stroke). Several landmark studies in 2018-2019 have revealed novel mechanisms underlying cardiovascular pathologies in HGPS, and implicate future potential therapies for HGPS, and possibly physiological aging.


Assuntos
Doenças Cardiovasculares/complicações , Doenças Cardiovasculares/tratamento farmacológico , Piperidinas/uso terapêutico , Progéria/complicações , Progéria/tratamento farmacológico , Piridinas/uso terapêutico , Doenças Cardiovasculares/patologia , Humanos , Progéria/patologia
4.
Proc Natl Acad Sci U S A ; 117(9): 4792-4801, 2020 03 03.
Artigo em Inglês | MEDLINE | ID: mdl-32075915

RESUMO

Angiogenesis, the formation of new blood vessels, is tightly regulated by gene transcriptional programs. Yin Ying 1 (YY1) is a ubiquitously distributed transcription factor with diverse and complex biological functions; however, little is known about the cell-type-specific role of YY1 in vascular development and angiogenesis. Here we report that endothelial cell (EC)-specific YY1 deletion in mice led to embryonic lethality as a result of abnormal angiogenesis and vascular defects. Tamoxifen-inducible EC-specific YY1 knockout (YY1iΔEC ) mice exhibited a scarcity of retinal sprouting angiogenesis with fewer endothelial tip cells. YY1iΔEC mice also displayed severe impairment of retinal vessel maturation. In an ex vivo mouse aortic ring assay and a human EC culture system, YY1 depletion impaired endothelial sprouting and migration. Mechanistically, YY1 functions as a repressor protein of Notch signaling that controls EC tip-stalk fate determination. YY1 deficiency enhanced Notch-dependent gene expression and reduced tip cell formation. Specifically, YY1 bound to the N-terminal domain of RBPJ (recombination signal binding protein for Ig Kappa J region) and competed with the Notch coactivator MAML1 (mastermind-like protein 1) for binding to RBPJ, thereby impairing the NICD (intracellular domain of the Notch protein)/MAML1/RBPJ complex formation. Our study reveals an essential role of endothelial YY1 in controlling sprouting angiogenesis through directly interacting with RBPJ and forming a YY1-RBPJ nuclear repression complex.


Assuntos
Proteína de Ligação a Sequências Sinal de Recombinação J de Imunoglobina/metabolismo , Morfogênese/fisiologia , Neovascularização Patológica/metabolismo , Fator de Transcrição YY1/metabolismo , Animais , Proteínas de Transporte/metabolismo , Diferenciação Celular , Células Endoteliais/metabolismo , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Masculino , Camundongos/embriologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neovascularização Patológica/genética , Neovascularização Patológica/patologia , Neovascularização Fisiológica/genética , Neovascularização Fisiológica/fisiologia , Proteínas Nucleares , Ligação Proteica , Receptores Notch/metabolismo , Vasos Retinianos/metabolismo , Transdução de Sinais , Fatores de Transcrição , Fator de Transcrição YY1/genética
5.
Proc Natl Acad Sci U S A ; 116(2): 546-555, 2019 01 08.
Artigo em Inglês | MEDLINE | ID: mdl-30584103

RESUMO

SENCR is a human-specific, vascular cell-enriched long-noncoding RNA (lncRNA) that regulates vascular smooth muscle cell and endothelial cell (EC) phenotypes. The underlying mechanisms of action of SENCR in these and other cell types is unknown. Here, levels of SENCR RNA are shown to be elevated in several differentiated human EC lineages subjected to laminar shear stress. Increases in SENCR RNA are also observed in the laminar shear stress region of the adult aorta of humanized SENCR-expressing mice, but not in disturbed shear stress regions. SENCR loss-of-function studies disclose perturbations in EC membrane integrity resulting in increased EC permeability. Biotinylated RNA pull-down and mass spectrometry establish an abundant SENCR-binding protein, cytoskeletal-associated protein 4 (CKAP4); this ribonucleoprotein complex was further confirmed in an RNA immunoprecipitation experiment using an antibody to CKAP4. Structure-function studies demonstrate a noncanonical RNA-binding domain in CKAP4 that binds SENCR Upon SENCR knockdown, increasing levels of CKAP4 protein are detected in the EC surface fraction. Furthermore, an interaction between CKAP4 and CDH5 is enhanced in SENCR-depleted EC. This heightened association appears to destabilize the CDH5/CTNND1 complex and augment CDH5 internalization, resulting in impaired adherens junctions. These findings support SENCR as a flow-responsive lncRNA that promotes EC adherens junction integrity through physical association with CKAP4, thereby stabilizing cell membrane-bound CDH5.


Assuntos
Junções Aderentes/metabolismo , Células Endoteliais da Veia Umbilical Humana/metabolismo , Proteínas de Membrana/metabolismo , RNA Longo não Codificante/metabolismo , Junções Aderentes/genética , Antígenos CD/genética , Antígenos CD/metabolismo , Caderinas/genética , Caderinas/metabolismo , Cateninas/genética , Cateninas/metabolismo , Células Endoteliais da Veia Umbilical Humana/citologia , Humanos , Proteínas de Membrana/genética , Domínios Proteicos , RNA Longo não Codificante/genética , Resistência ao Cisalhamento/fisiologia , delta Catenina
6.
Hepatology ; 72(5): 1717-1734, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32048304

RESUMO

BACKGROUND AND AIMS: Liver fibrosis (LF) is a central pathological process that occurs in most types of chronic liver diseases. Advanced LF causes cirrhosis, hepatocellular carcinoma, and liver failure. However, the exact molecular mechanisms underlying the initiation and progression of LF remain largely unknown. APPROACH AND RESULTS: This study was designed to investigate the role of protein kinase D3 (PKD3; gene name Prkd3) in the regulation of liver homeostasis. We generated global Prkd3 knockout (Prkd3-/- ) mice and myeloid-cell-specific Prkd3 knockout (Prkd3∆LysM ) mice, and we found that both Prkd3-/- mice and Prkd3∆LysM mice displayed spontaneous LF. PKD3 deficiency also aggravated CCl4 -induced LF. PKD3 is highly expressed in hepatic macrophages (HMs), and PKD3 deficiency skewed macrophage polarization toward a profibrotic phenotype. Activated profibrotic macrophages produced transforming growth factor beta that, in turn, activates hepatic stellate cells to become matrix-producing myofibroblasts. Moreover, PKD3 deficiency decreased the phosphatase activity of SH2-containing protein tyrosine phosphatase-1 (a bona-fide PKD3 substrate), resulting in sustained signal transducer and activator of transcription 6 activation in macrophages. In addition, we observed that PKD3 expression in HMs was down-regulated in cirrhotic human liver tissues. CONCLUSIONS: PKD3 deletion in mice drives LF through the profibrotic macrophage activation.


Assuntos
Cirrose Hepática Experimental/patologia , Cirrose Hepática/patologia , Proteína Quinase C/deficiência , Animais , Tetracloreto de Carbono/toxicidade , Células Cultivadas , Progressão da Doença , Células Estreladas do Fígado/metabolismo , Humanos , Fígado/citologia , Fígado/patologia , Cirrose Hepática Experimental/induzido quimicamente , Cirrose Hepática Experimental/diagnóstico , Cirrose Hepática Experimental/genética , Macrófagos/metabolismo , Camundongos , Camundongos Knockout , Miofibroblastos/metabolismo , Cultura Primária de Células , Proteína Quinase C/genética , Proteína Tirosina Fosfatase não Receptora Tipo 6/metabolismo , Índice de Gravidade de Doença , Análise Serial de Tecidos , Fator de Crescimento Transformador beta/metabolismo
7.
Biochem Biophys Res Commun ; 521(2): 279-284, 2020 01 08.
Artigo em Inglês | MEDLINE | ID: mdl-31668369

RESUMO

Serum response factor (SRF), a key transcription factor, plays an important role in regulating cell functions such as proliferation and differentiation. Most proteins are unstable, and protein stability is regulated through the ubiquitin-proteasome system (UPS) or the autophagy lysosome pathway (ALP). Whether SRF is degraded and what mechanisms control SRF protein stability remain unexplored. Western blot analyses of cells treated with cycloheximide (CHX), a protein synthesis inhibitor, showed that SRF was degraded in a time-dependent manner. Moreover, we observed that SRF undergoes autophagy-dependent destruction, which is accelerated by serum deprivation. Through bioinformatics screening, we found that SRF contains the GSK3ß phosphorylation motif (T/SPPXS): SPDSPPRSDPT, which is conserved from zebrafish to humans. Serum deprivation stimulated GSK3ß activation that then potentiates SRF degradation through the autophagy lysosome pathway. Since SRF is important for numerous cellular activities, our results suggest that the autophagy-dependent SRF degradation pathway may provide a new avenue to modulate SRF-mediated cell functions.


Assuntos
Autofagia , Fator de Resposta Sérica/química , Animais , Células COS , Células Cultivadas , Chlorocebus aethiops , Meios de Cultura Livres de Soro/farmacologia , Cicloeximida/farmacologia , Glicogênio Sintase Quinase 3 beta/metabolismo , Humanos , Lisossomos/metabolismo , Estabilidade Proteica , Ratos , Fator de Resposta Sérica/metabolismo
8.
Proc Natl Acad Sci U S A ; 114(13): E2739-E2747, 2017 03 28.
Artigo em Inglês | MEDLINE | ID: mdl-28292896

RESUMO

Megacystis microcolon intestinal hypoperistalsis syndrome (MMIHS) is a congenital visceral myopathy characterized by severe dilation of the urinary bladder and defective intestinal motility. The genetic basis of MMIHS has been ascribed to spontaneous and autosomal dominant mutations in actin gamma 2 (ACTG2), a smooth muscle contractile gene. However, evidence suggesting a recessive origin of the disease also exists. Using combined homozygosity mapping and whole exome sequencing, a genetically isolated family was found to carry a premature termination codon in Leiomodin1 (LMOD1), a gene preferentially expressed in vascular and visceral smooth muscle cells. Parents heterozygous for the mutation exhibited no abnormalities, but a child homozygous for the premature termination codon displayed symptoms consistent with MMIHS. We used CRISPR-Cas9 (CRISPR-associated protein) genome editing of Lmod1 to generate a similar premature termination codon. Mice homozygous for the mutation showed loss of LMOD1 protein and pathology consistent with MMIHS, including late gestation expansion of the bladder, hydronephrosis, and rapid demise after parturition. Loss of LMOD1 resulted in a reduction of filamentous actin, elongated cytoskeletal dense bodies, and impaired intestinal smooth muscle contractility. These results define LMOD1 as a disease gene for MMIHS and suggest its role in establishing normal smooth muscle cytoskeletal-contractile coupling.


Assuntos
Anormalidades Múltiplas/genética , Autoantígenos/fisiologia , Colo/anormalidades , Proteínas do Citoesqueleto/fisiologia , Pseudo-Obstrução Intestinal/genética , Proteínas Musculares/fisiologia , Bexiga Urinária/anormalidades , Animais , Autoantígenos/genética , Autoantígenos/metabolismo , Códon sem Sentido , Proteínas do Citoesqueleto/genética , Proteínas do Citoesqueleto/metabolismo , Feminino , Humanos , Recém-Nascido , Camundongos , Contração Muscular/genética , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Músculo Liso/fisiologia
9.
Eur Heart J ; 40(29): 2398-2408, 2019 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-31539914

RESUMO

AIMS: Recent genome-wide association studies (GWAS) have identified that the JCAD locus is associated with risk of coronary artery disease (CAD) and myocardial infarction (MI). However, the mechanisms whereby candidate gene JCAD confers disease risk remain unclear. We addressed whether and how JCAD affects the development of atherosclerosis, the common cause of CAD. METHODS AND RESULTS: By mining data in the Genotype-Tissue Expression (GTEx) database, we found that CAD-associated risk variants at the JCAD locus are linked to increased JCAD gene expression in human arteries, implicating JCAD as a candidate causal CAD gene. We therefore generated global and endothelial cell (EC) specific-JCAD knockout mice, and observed that JCAD deficiency attenuated high fat diet-induced atherosclerosis in ApoE-deficient mice. JCAD-deficiency in mice also improved endothelium-dependent relaxation. Genome-wide transcriptional profiling of JCAD-depleted human coronary artery ECs showed that JCAD depletion inhibited the activation of YAP/TAZ pathway, and the expression of downstream pro-atherogenic genes, including CTGF and Cyr61. As a result, JCAD-deficient ECs attracted fewer monocytes in response to lipopolysaccharide (LPS) stimulation. Moreover, JCAD expression in ECs was decreased under unidirectional laminar flow in vitro and in vivo. Proteomics studies suggest that JCAD regulates YAP/TAZ activation by interacting with actin-binding protein TRIOBP, thereby stabilizing stress fiber formation. Finally, we observed that endothelial JCAD expression was increased in mouse and human atherosclerotic plaques. CONCLUSION: The present study demonstrates that the GWAS-identified CAD risk gene JCAD promotes endothelial dysfunction and atherosclerosis, thus highlighting the possibility of new therapeutic strategies for CAD by targeting JCAD.


Assuntos
Aterosclerose/genética , Moléculas de Adesão Celular/genética , Doença da Artéria Coronariana/genética , Endotélio Vascular/fisiopatologia , Predisposição Genética para Doença/genética , Animais , Apolipoproteínas E/genética , Dieta Ocidental/efeitos adversos , Endotélio Vascular/metabolismo , Feminino , Genes/genética , Estudo de Associação Genômica Ampla , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Transdução de Sinais
10.
Biochem Biophys Res Commun ; 514(3): 913-918, 2019 06 30.
Artigo em Inglês | MEDLINE | ID: mdl-31084931

RESUMO

Liver fibrosis is a common consequence of various chronic hepatitis and liver injuries. The myofibroblasts, through the accumulation of extracellular matrix (ECM) proteins, are closely associated with the progression of liver fibrosis. However, the molecular mechanisms underlying transcriptional regulation of fibrogenic genes and ECM proteins in myofibroblasts remain largely unknown. Using tamoxifen inducible myofibroblast-specific Cre-expressing mouse lines with selective deletion of the transcription factor Yin Yang 1 (YY1), here we show that YY1 deletion in myofibroblasts mitigates carbon tetrachloride-induced liver fibrosis. This protective effect of YY1 ablation on liver fibrosis was accompanied with reduced expression of profibrogenic genes and ECM proteins, including TNF-α, TGF-ß, PDGF, IL-6, α-SMA and Col1α1 in liver tissues from YY1 mutant mice. Moreover, using the human hepatic stellate cell (HSC) line LX-2, we found that knockdown of YY1 in myofibroblasts by siRNA treatment diminished myofibroblast proliferation, α-SMA expression, and collagen deposition. Collectively, our findings reveal a specific role of YY1 in hepatic myofibroblasts and suggest a new therapeutic strategy for hepatic fibrosis-associated liver diseases.


Assuntos
Cirrose Hepática/patologia , Miofibroblastos/patologia , Fator de Transcrição YY1/genética , Animais , Linhagem Celular , Deleção de Genes , Humanos , Cirrose Hepática/genética , Camundongos , Camundongos Endogâmicos C57BL , Miofibroblastos/metabolismo , Interferência de RNA
11.
Biochem Biophys Res Commun ; 498(3): 633-639, 2018 04 06.
Artigo em Inglês | MEDLINE | ID: mdl-29524414

RESUMO

Sirtuin 1 (SIRT1) is an NAD+-dependent protein deacetylase that plays a critical role in controlling energy metabolism, stress response and aging. Hence, enhancing SIRT1 activity could be a potential therapeutic strategy to treat metabolic diseases such as diabetes. However, pharmacological activators for SIRT1 are scarce to date. In this study, using the optimized high throughput screening, we identified E6155, a piperazine 1, 4- diamide compound, as a new small molecular activator of SIRT1. We further found that E6155 significantly upregulated glucose uptake in cultured normal liver cells and skeletal muscle cells through increasing SIRT1 deacetylase activity. In type 2 diabetic KKAy mice, E6155 treatment markedly decreased the level of fasting glucose. Moreover, E6155 improved oral glucose tolerance and insulin tolerance. Euglycemic clamp and the homeostasis model assessment of insulin resistance index showed that E6155 ameliorated the insulin resistance and increased insulin sensitivity in diabetic mice. Mechanistically, we observed that the antidiabetic effects of E6155 were involved in SIRT1 dependent activation of LKB1/AMPK and IRS1/AKT pathways. In conclusion, our findings identified E6155 as a novel SIRT1 activator and suggested that E6155 could be a promising drug candidate for treating insulin resistance and diabetes.


Assuntos
Diabetes Mellitus Tipo 2/tratamento farmacológico , Hipoglicemiantes/uso terapêutico , Resistência à Insulina , Piperazinas/uso terapêutico , Sirtuína 1/metabolismo , Animais , Glicemia/análise , Glicemia/metabolismo , Linhagem Celular , Diabetes Mellitus Tipo 2/sangue , Diabetes Mellitus Tipo 2/metabolismo , Feminino , Glucose/metabolismo , Teste de Tolerância a Glucose , Células Hep G2 , Humanos , Hipoglicemiantes/química , Insulina/metabolismo , Camundongos , Piperazinas/química , Bibliotecas de Moléculas Pequenas/química , Bibliotecas de Moléculas Pequenas/uso terapêutico
12.
Exp Eye Res ; 170: 169-176, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29501382

RESUMO

MicroRNA miR-126 has been shown to be required for proper angiogenesis in several models. However, its expression, regulation and function in the mouse choroid remain unclear. Our previous data has shown that miR-126 expression is enriched in the endothelial cells (ECs) of the mouse choroid. Here we report that a 5.5 kb Egfl7/miR-126 promoter drives the expression of miR-126 in the choroid ECs during choroidal vascular development. The expression of miR-126 in the ECs is regulated by flow stress likely through Krüppel-like transcriptional factors. miR-126-/- mice show mildly delayed choroidal vascular development, but adult knockout mice develop periphery choroidal vascular lesions. This study suggests that miR-126 is largely dispensable for mouse choroidal development but required for maintaining choroidal vasculature integrity.


Assuntos
Corioide/irrigação sanguínea , Corioide/embriologia , Células Endoteliais/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , MicroRNAs/genética , Neovascularização Fisiológica/genética , Animais , Proteínas de Ligação ao Cálcio , Células Cultivadas , Família de Proteínas EGF , Angiofluoresceinografia , Células Endoteliais da Veia Umbilical Humana , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Plasmídeos , Proteínas/genética , Reação em Cadeia da Polimerase em Tempo Real
13.
FASEB J ; 29(11): 4523-31, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26183772

RESUMO

Coxsackievirus B3 (CVB3), an important human causative pathogen for viral myocarditis, pancreatitis, and meningitis, has evolved different strategies to manipulate the host signaling machinery to ensure successful viral infection. We previously revealed a crucial role for the ERK1/2 signaling pathway in regulating viral infectivity. However, the detail mechanism remains largely unknown. Grb2-associated binder 1 (GAB1) is an important docking protein responsible for intracellular signaling assembly and transduction. In this study, we demonstrated that GAB1 was proteolytically cleaved after CVB3 infection at G175 and G436 by virus-encoded protease 2A(pro), independent of caspase activation. Knockdown of GAB1 resulted in a significant reduction of viral protein expression and virus titers. Moreover, we showed that virus-induced cleavage of GAB1 is beneficial to viral growth as the N-terminal proteolytic product of GAB1 (GAB1-N1-174) further enhances ERK1/2 activation and promotes viral replication. Our results collectively suggest that CVB3 targets host GAB1 to generate a GAB1-N1-174 fragment that enhances viral infectivity, at least in part, via activation of the ERK pathway. The findings in this study suggest a novel mechanism that CVB3 employs to subvert the host signaling and facilitate consequent viral replication.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Enterovirus Humano B/fisiologia , Sistema de Sinalização das MAP Quinases , Peptídeo Hidrolases/metabolismo , Proteínas Virais/metabolismo , Replicação Viral/fisiologia , Proteínas Adaptadoras de Transdução de Sinal/genética , Caspases/genética , Caspases/metabolismo , Infecções por Coxsackievirus/genética , Infecções por Coxsackievirus/metabolismo , Ativação Enzimática/genética , Células HeLa , Humanos , Proteína Quinase 1 Ativada por Mitógeno/genética , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/genética , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Peptídeo Hidrolases/genética , Proteínas Virais/genética
14.
Circ Res ; 110(1): 59-70, 2012 Jan 06.
Artigo em Inglês | MEDLINE | ID: mdl-22076634

RESUMO

RATIONALE: The Rad-Gem/Kir-related family (RGKs) consists of small GTP-binding proteins that strongly inhibit the activity of voltage-gated calcium channels. Among RGKs, Rem1 is strongly and specifically expressed in cardiac tissue. However, the physiological role and regulation of RGKs, and Rem1 in particular, are largely unknown. OBJECTIVE: To determine if Rem1 function is physiologically regulated by adrenergic signaling and thus impacts voltage-gated L-type calcium channel (VLCC) activity in the heart. METHODS AND RESULTS: We found that activation of protein kinase D1, a protein kinase downstream of α(1)-adrenergic signaling, leads to direct phosphorylation of Rem1 at Ser18. This results in an increase of the channel activity and plasma membrane expression observed by using a combination of electrophysiology, live cell confocal microscopy, and immunohistochemistry in heterologous expression system and neonatal cardiomyocytes. In addition, we show that stimulation of α(1)-adrenergic receptor-protein kinase D1-Rem1 signaling increases transverse-tubule VLCC expression that results in increased L-type Ca(2+) current density in adult ventricular myocytes. CONCLUSION: The α(1)-adrenergic stimulation releases Rem1 inhibition of VLCCs through direct phosphorylation of Rem1 at Ser18 by protein kinase D1, resulting in an increase of the channel activity and transverse-tubule expression. Our results uncover a novel molecular regulatory mechanism of VLCC trafficking and function in the heart and provide the first demonstration of physiological regulation of RGK function.


Assuntos
Canais de Cálcio Tipo L/fisiologia , Miócitos Cardíacos/fisiologia , Proteínas Quinases/fisiologia , Transporte Proteico/fisiologia , Receptores Adrenérgicos alfa 1/fisiologia , Transdução de Sinais/fisiologia , Animais , Membrana Celular/fisiologia , Células Cultivadas , Masculino , Microtúbulos/fisiologia , Modelos Animais , Proteínas Monoméricas de Ligação ao GTP/fisiologia , Miócitos Cardíacos/citologia , Técnicas de Patch-Clamp , Fosforilação , Proteína Quinase C , Ratos , Ratos Sprague-Dawley
15.
Inflammation ; 47(1): 323-332, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-37819455

RESUMO

Inflammatory response in the pulmonary endothelium drives the pathogenesis of acute lung injury and sepsis. Sirtuin 6 (SIRT6), a member of class III NAD+-dependent deacetylases belonging to the sirtuin family, regulates senescence, metabolism, and inflammation and extends lifespan in mice and model organisms. However, the role of SIRT6 in pulmonary endothelial inflammation is unknown. Thus, we hypothesized that SIRT6 suppresses inflammatory response in human lung microvascular cells (HLMEC) and ensues monocyte adhesion to endothelial cells. Primary HLMECs were treated with control or SIRT6 adenovirus or SIRT6 agonist, with or without lipopolysaccharide (LPS) treatment. We observed that treatment with LPS did not affect the protein expression of SIRT6 in HLMECs. However, adenovirus-mediated SIRT6 overexpression attenuated LPS-induced VCAM1 gene and protein expression, followed by decreased monocyte adhesion to endothelial cells. Similarly, activation of SIRT6 by a recently reported SIRT6 activator UBCS039, but not the regioisomer negative control compound UBCS060, ameliorated LPS-induced VCAM1 mRNA and protein expression as well as monocyte adhesion. Moreover, luciferase assay revealed that SIRT6 adenovirus decreased the activity of NF-κB, the master regulator of vascular inflammation. Taken together, these results indicate that molecular and pharmacological activation of SIRT6 protects against lung microvascular inflammation via suppressing NF-κB activation, implicating the therapeutic potential of the SIRT6 activators for lung disorders associated with microvascular inflammation.


Assuntos
Pneumonia , Sirtuínas , Humanos , Camundongos , Animais , NF-kappa B/metabolismo , Lipopolissacarídeos/farmacologia , Células Endoteliais/metabolismo , Inflamação/induzido quimicamente , Inflamação/prevenção & controle , Inflamação/metabolismo , Pneumonia/induzido quimicamente , Pneumonia/prevenção & controle , Pneumonia/metabolismo , Pulmão/metabolismo , Endotélio Vascular/metabolismo
16.
Cell Death Dis ; 15(4): 283, 2024 Apr 22.
Artigo em Inglês | MEDLINE | ID: mdl-38649362

RESUMO

Acute liver failure (ALF) is a deadly illness due to insufficient detoxification in liver induced by drugs, toxins, and other etiologies, and the effective treatment for ALF is very limited. Among the drug-induced ALF, acetaminophen (APAP) overdose is the most common cause. However, the molecular mechanisms underlying APAP hepatoxicity remain incompletely understood. Sirtuin 6 (Sirt6) is a stress responsive protein deacetylase and plays an important role in regulation of DNA repair, genomic stability, oxidative stress, and inflammation. Here, we report that genetic and pharmacological activation of Sirt6 protects against ALF in mice. We first observed that Sirt6 expression was significantly reduced in the liver tissues of human patients with ALF and mice treated with an overdose of APAP. Then we developed an inducible Sirt6 transgenic mice for Cre-mediated overexpression of the human Sirt6 gene in systemic (Sirt6-Tg) and hepatic-specific (Sirt6-HepTg) manners. Both Sirt6-Tg mice and Sirt6-HepTg mice exhibited the significant protection against APAP hepatoxicity. In contrast, hepatic-specific Sirt6 knockout mice exaggerated APAP-induced liver damages. Mechanistically, Sirt6 attenuated APAP-induced hepatocyte necrosis and apoptosis through downregulation of oxidative stress, inflammation, the stress-activated kinase JNK activation, and apoptotic caspase activation. Moreover, Sirt6 negatively modulated the level and activity of poly (ADP-ribose) polymerase 1 (PARP1) in APAP-treated mouse liver tissues. Importantly, the specific Sirt6 activator MDL-800 exhibited better therapeutic potential for APAP hepatoxicity than the current drug acetylcysteine. Furthermore, in the model of bile duct ligation induced ALF, hepatic Sirt6-KO exacerbated, but Sirt6-HepTg mitigated liver damage. Collectively, our results demonstrate that Sirt6 protects against ALF and suggest that targeting Sirt6 activation could be a new therapeutic strategy to alleviate ALF.


Assuntos
Acetaminofen , Hepatócitos , Falência Hepática Aguda , Sirtuínas , Animais , Humanos , Masculino , Camundongos , Acetaminofen/efeitos adversos , Apoptose/efeitos dos fármacos , Hepatócitos/metabolismo , Hepatócitos/efeitos dos fármacos , Fígado/metabolismo , Fígado/patologia , Fígado/efeitos dos fármacos , Falência Hepática Aguda/metabolismo , Falência Hepática Aguda/induzido quimicamente , Falência Hepática Aguda/patologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Estresse Oxidativo/efeitos dos fármacos , Sirtuínas/metabolismo , Sirtuínas/genética
17.
Arterioscler Thromb Vasc Biol ; 32(7): 1707-16, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22628435

RESUMO

OBJECTIVE: Vascular endothelial growth factor (VEGF) promotes leukocyte adhesion to endothelial cells (ECs). Angiopoietin-1 (Ang-1) inhibits this response. Nuclear receptor-77 (Nur77) is a proangiogenic nuclear receptor. In the present study, we assessed the influence of Ang-1 and VEGF on Nur77 expression in ECs, and evaluated its role in Ang-1/VEGF-mediated leukocyte adhesion. METHODS AND RESULTS: Expression of Nur77 was evaluated with real-time polymerase chain reaction and immunoblotting. Adhesion of leukocytes to ECs was monitored with inverted microscopy. Nur77 expression or activity was inhibited using adenoviruses expressing dominant-negative form of Nur77, retroviruses expressing Nur77 in the antisense direction, and small interfering RNA oligos. Both Ang-1 and VEGF induce Nur77 expression, by >5- and 30-fold, respectively. When combined, Ang-1 potentiates VEGF-induced Nur77 expression. Ang-1 induces Nur77 through the phosphoinositide 3-kinase and extracellular signal-regulated protein kinase 1/2 pathways. VEGF induces Nur77 expression through the protein kinase D/histone deacetylase 7/myocyte enhancer factor 2 and extracellular signal-regulated protein kinase 1/2 pathways. VEGF induces nuclear factor-kappaB transcription factor, vascular cell adhesion molecule-1, and E-selectin expressions, and promotes leukocyte adhesion to ECs. Ang-1 inhibits these responses. This inhibitory effect of Ang-1 disappears when Nur77 expression is disrupted, restoring the inductive effects of VEGF on adhesion molecule expression, and increased leukocyte adhesion to ECs. CONCLUSIONS: Nur77 promotes anti-inflammatory effects of Ang-1, and functions as a negative feedback inhibitor of VEGF-induced EC activation.


Assuntos
Angiopoietina-1/farmacologia , Células Endoteliais/efeitos dos fármacos , Leucócitos/efeitos dos fármacos , Membro 1 do Grupo A da Subfamília 4 de Receptores Nucleares/fisiologia , Fator A de Crescimento do Endotélio Vascular/farmacologia , Adesão Celular/efeitos dos fármacos , Células Cultivadas , Células Endoteliais/fisiologia , Histona Desacetilases/metabolismo , Humanos , Quinase I-kappa B/genética , Leucócitos/fisiologia , NF-kappa B/metabolismo , Membro 1 do Grupo A da Subfamília 4 de Receptores Nucleares/genética , Fosfatidilinositol 3-Quinases/fisiologia , Fosforilação , Proteínas Proto-Oncogênicas c-akt/fisiologia , Canais de Cátion TRPP/metabolismo , Células U937 , Molécula 1 de Adesão de Célula Vascular/genética
18.
Proc Natl Acad Sci U S A ; 107(35): 15467-72, 2010 Aug 31.
Artigo em Inglês | MEDLINE | ID: mdl-20716686

RESUMO

Dynamic nucleocytoplasmic shuttling of class IIa histone deacetylases (HDACs) is a fundamental mechanism regulating gene transcription. Recent studies have identified several protein kinases that phosphorylate HDAC5, leading to its exportation from the nucleus. However, the negative regulatory mechanisms for HDAC5 nuclear exclusion remain largely unknown. Here we show that cAMP-activated protein kinase A (PKA) specifically phosphorylates HDAC5 and prevents its export from the nucleus, leading to suppression of gene transcription. PKA interacts directly with HDAC5 and phosphorylates HDAC5 at serine 280, an evolutionarily conserved site. Phosphorylation of HDAC5 by PKA interrupts the association of HDAC5 with protein chaperone 14-3-3 and hence inhibits stress signal-induced nuclear export of HDAC5. An HDAC5 mutant that mimics PKA-dependent phosphorylation localizes in the nucleus and acts as a dominant inhibitor for myocyte enhancer factor 2 transcriptional activity. Molecular manipulations of HDAC5 show that PKA-phosphorylated HDAC5 inhibits cardiac fetal gene expression and cardiomyocyte hypertrophy. Our findings identify HDAC5 as a substrate of PKA and reveal a cAMP/PKA-dependent pathway that controls HDAC5 nucleocytoplasmic shuttling and represses gene transcription. This pathway may represent a mechanism by which cAMP/PKA signaling modulates a wide range of biological functions and human diseases such as cardiomyopathy.


Assuntos
Núcleo Celular/metabolismo , Subunidades Catalíticas da Proteína Quinase Dependente de AMP Cíclico/metabolismo , Histona Desacetilases/metabolismo , Miócitos Cardíacos/metabolismo , Transporte Ativo do Núcleo Celular/efeitos dos fármacos , Sequência de Aminoácidos , Animais , Animais Recém-Nascidos , Células COS , Forma Celular , Células Cultivadas , Chlorocebus aethiops , Colforsina/farmacologia , AMP Cíclico/farmacologia , Subunidades Catalíticas da Proteína Quinase Dependente de AMP Cíclico/genética , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Histona Desacetilases/genética , Humanos , Immunoblotting , Microscopia de Fluorescência , Dados de Sequência Molecular , Miócitos Cardíacos/citologia , Fosforilação , Ratos , Homologia de Sequência de Aminoácidos , Especificidade por Substrato , Transcrição Gênica
19.
Blood ; 115(14): 2971-9, 2010 Apr 08.
Artigo em Inglês | MEDLINE | ID: mdl-20042720

RESUMO

Fluid shear stress generated by steady laminar blood flow protects vessels from atherosclerosis. Krüppel-like factor 2 (KLF2) and endothelial nitric oxide synthase (eNOS) are fluid shear stress-responsive genes and key mediators in flow anti-inflammatory and antiatherosclerotic actions. However, the molecular mechanisms underlying flow induction of KLF2 and eNOS remain largely unknown. Here, we show a novel role of histone deacetylase 5 (HDAC5) in flow-mediated KLF2 and eNOS expression. We found for the first time that fluid shear stress stimulated HDAC5 phosphorylation and nuclear export in endothelial cells through a calcium/calmodulin-dependent pathway. Consequently, flow induced the dissociation of HDAC5 and myocyte enhancer factor-2 (MEF2) and enhanced MEF2 transcriptional activity, which leads to expression of KLF2 and eNOS. Adenoviral overexpression of a HDAC5 phosphorylation-defective mutant (Ser259/Ser498 were replaced by Ala259/Ala498, HDAC5-S/A), which shows resistance to flow-induced nuclear export, suppressed flow-mediated MEF2 transcriptional activity and expression of KLF2 and eNOS. Importantly, HDAC5-S/A attenuated the flow-inhibitory effect on monocyte adhesion to endothelial cells. Taken together, our results reveal that phosphorylation-dependent derepression of HDAC5 mediates flow-induced KLF2 and eNOS expression as well as flow anti-inflammation, and suggest that HDAC5 could be a potential therapeutic target for the prevention of atherosclerosis.


Assuntos
Núcleo Celular/metabolismo , Células Endoteliais/metabolismo , Regulação da Expressão Gênica , Fatores de Transcrição Kruppel-Like/biossíntese , Monócitos/metabolismo , Óxido Nítrico Sintase Tipo III/biossíntese , Estresse Fisiológico , Transporte Ativo do Núcleo Celular , Adenoviridae , Substituição de Aminoácidos , Aterosclerose/genética , Aterosclerose/metabolismo , Aterosclerose/patologia , Aterosclerose/prevenção & controle , Velocidade do Fluxo Sanguíneo , Cálcio/metabolismo , Calmodulina/genética , Calmodulina/metabolismo , Adesão Celular , Núcleo Celular/genética , Núcleo Celular/patologia , Células Endoteliais/patologia , Histona Desacetilases , Humanos , Fatores de Transcrição Kruppel-Like/genética , Monócitos/patologia , Mutação de Sentido Incorreto , Fosforilação/genética
20.
Arterioscler Thromb Vasc Biol ; 31(5): 1016-23, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21372298

RESUMO

OBJECTIVE: Grb2-associated binder 1 (Gab1), a scaffolding adaptor protein, plays an important role in transmitting key signals that control cell growth, differentiation, and function from multiple tyrosine kinase receptors. The study was designed to investigate the role of endothelial Gab1 in angiogenesis and its underlying molecular mechanisms. METHODS AND RESULTS: Using Cre-Lox recombination technology, we generated endothelial-specific Gab1 knockout (Gab1-ecKO) mice. Gab1-ecKO mice are viable and showed no obvious developmental defects in the vascular system. To analyze the role of Gab1 in postnatal angiogenesis, we used hindlimb ischemia and Matrigel plug models. We found that loss of endothelial Gab1 in mice dramatically impaired postnatal angiogenesis. Gab1-ecKO mice had impaired ischemia-initiated blood flow recovery, exhibited reduced angiogenesis, and were associated with marked limb necrosis. We further observed significant endothelial cell (EC) death in the ischemic hindlimb of Gab1-ecKO mice. Matrigel plug assay showed that hepatocyte growth factor (HGF)-mediated angiogenesis was inhibited in Gab1-ecKO mice. In vitro studies showed that Gab1 was required for HGF-induced EC migration, tube formation, and microvessel sprouting. Mechanistically, HGF stimulated Gab1 tyrosine phosphorylation in ECs, leading to activation of extracellular regulated MAP kinase 1/2 and Akt, which are angiogenic and survival signaling. CONCLUSIONS: Gab1 is essential for postnatal angiogenesis through mediating angiogenic and survival signaling.


Assuntos
Endotélio Vascular/metabolismo , Isquemia/metabolismo , Músculo Esquelético/irrigação sanguínea , Neovascularização Fisiológica , Fosfoproteínas/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Animais , Movimento Celular , Proliferação de Células , Sobrevivência Celular , Modelos Animais de Doenças , Endotélio Vascular/patologia , Endotélio Vascular/fisiopatologia , Fator de Crescimento de Hepatócito/metabolismo , Membro Posterior , Isquemia/genética , Isquemia/patologia , Isquemia/fisiopatologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Fosfoproteínas/deficiência , Fosfoproteínas/genética , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo , Recuperação de Função Fisiológica , Fluxo Sanguíneo Regional , Transdução de Sinais , Fatores de Tempo , Tirosina
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA