Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 65
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Opt Lett ; 47(5): 1037-1040, 2022 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-35230284

RESUMO

A tip nanofocusing light field, with high electric-field intensity and nanoscale mode volume, can significantly improve nonlinear light scattering efficiency, thereby greatly promoting the development of strong-field nano-optics. Here, tip-enhanced four-wave mixing (FWM) is theoretically analyzed through two ultrafast radial vector beams internally illuminating an Ag-coated silica tip (ACST). Two femtosecond pulses, with radial electric vectors and pulse width of 100 fs, are adopted as excitation sources to illuminate the ACST. Degenerate tip-enhanced FWM (ωFWM = 2ω1-ω2) with a nonlinear conversion efficiency of ∼10-5 is achieved. The peak electric-field amplitude of the two pump pulses is 5 × 107 V/m, which is two orders of magnitude lower than that of the external excitation method. Further theoretical analysis shows that the conversion efficiency of the tip-enhanced FWM has strict frequency detuning dependence characteristics, and is closely related to the frequency response of the tip nanofocusing light field. This plasmonic tip provides an approach for enhancing nonlinear nano-optics, and may be used in the field of tip-based FWM nanoscopy.

2.
Opt Lett ; 46(22): 5554-5557, 2021 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-34780404

RESUMO

Plasmonic tip nanofocusing has gained much attention owing to its wide application in the field of nanospectroscopy. Here, we present the Au nanosphere (AuNS)-assisted coupling ultrafast surface plasmon polaritons (SPP) background-free tip nanofocusing. The plasmonic tip was prepared by attaching an AuNS on the shaft of an Au conical tip fabricated by electrochemical etching. The AuNS was adopted as an antenna to couple the far-field excitation light to the propagating SPP along the shaft to the tip apex for achieving power compression. Importantly, we experimentally and theoretically demonstrate that such a plasmonic tip can realize background-free ultrafast SPP tip nanofocusing with radially polarized features in a wide spectral range based on the localized SPP resonance effect supported by AuNS. Furthermore, the intensity of the tip nanofocusing light field has strong polarization dependence under linearly polarized light excitation, providing a powerful platform for spatiotemporal light control on the nanoscale. Our technique realizes remote excitation of background-free tip nanofocusing with a structured light feature, and it holds promising potential for tip-enhanced nanospectroscopies, nonlinear nanophotonics, etc.

3.
J Mol Cell Cardiol ; 149: 1-14, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32941882

RESUMO

BRD4 is a member of the BET family of epigenetic regulators. Inhibition of BRD4 by the selective bromodomain inhibitor JQ1, alleviates thoracic aortic constriction-induced cardiac hypertrophy and heart failure. However, whether BRD4 inhibition by JQ1 has therapeutic effect on diabetic cardiomyopathy, a major cause of heart failure in patients with Type 2 diabetes, remains unknown. Here, we discover a novel link between BRD4 and PINK1/Parkin-mediated mitophagy during diabetic cardiomyopathy. Upregulation of BRD4 in diabetic mouse hearts inhibits PINK1/Parkin-mediated mitophagy, resulting in accumulation of damaged mitochondria and subsequent impairment of cardiac structure and function. BRD4 inhibition by JQ1 improves mitochondrial function, and repairs the cardiac structure and function of the diabetic heart. These effects depended on rewiring of the BRD4-driven transcription and repression of PINK1. Deletion of Pink1 suppresses mitophagy, exacerbates cardiomyopathy, and abrogates the therapeutic effect of JQ1 on diabetic cardiomyopathy. Our results illustrate a valid therapeutic strategy for treating diabetic cardiomyopathy by inhibition of BRD4.


Assuntos
Azepinas/farmacologia , Cardiomiopatias Diabéticas/patologia , Dieta Hiperlipídica , Mitofagia , Proteínas Nucleares/antagonistas & inibidores , Proteínas Quinases/metabolismo , Fatores de Transcrição/antagonistas & inibidores , Triazóis/farmacologia , Ubiquitina-Proteína Ligases/metabolismo , Animais , Animais Recém-Nascidos , Diabetes Mellitus Tipo 2/complicações , Deleção de Genes , Camundongos Endogâmicos C57BL , Mitocôndrias Cardíacas/efeitos dos fármacos , Mitocôndrias Cardíacas/metabolismo , Mitocôndrias Cardíacas/patologia , Mitofagia/efeitos dos fármacos , Proteínas Nucleares/metabolismo , Regiões Promotoras Genéticas/genética , Proteínas Quinases/genética , Fatores de Transcrição/metabolismo , Regulação para Cima/efeitos dos fármacos
4.
Circulation ; 139(16): 1913-1936, 2019 04 16.
Artigo em Inglês | MEDLINE | ID: mdl-30646747

RESUMO

BACKGROUND: Fundc1 (FUN14 domain containing 1), an outer mitochondrial membrane protein, is important for mitophagy and mitochondria-associated endoplasmic reticulum membranes (MAMs). The roles of Fundc1 and MAMs in diabetic hearts remain unknown. The aims of this study, therefore, were to determine whether the diabetes mellitus-induced Fundc1 expression could increase MAM formation, and whether disruption of MAM formation improves diabetic cardiac function. METHODS: Levels of FUNDC1 were examined in the hearts from diabetic patients and nondiabetic donors. Levels of Fundc1-induced MAMs and mitochondrial and heart function were examined in mouse neonatal cardiomyocytes exposed to high glucose (HG, 30 mmol/L d-glucose for 48 hours), and in streptozotocin-treated cardiac-specific Fundc1 knockout mice and cardiac-specific Fundc1 knockout diabetic Akita mice, as well. RESULTS: FUNDC1 levels were significantly elevated in cardiac tissues from diabetic patients in comparison with those from nondiabetic donors. In cultured mouse neonatal cardiomyocytes, HG conditions increased levels of Fundc1, the inositol 1,4,5-trisphosphate type 2 receptor (Ip3r2), and MAMs. Genetic downregulation of either Fundc1 or Ip3r2 inhibited MAM formation, reduced endoplasmic reticulum-mitochondrial Ca2+ flux, and improved mitochondrial function in HG-treated cardiomyocytes. Consistently, adenoviral overexpression of Fundc1 promoted MAM formation, mitochondrial Ca2+ increase, and mitochondrial dysfunction in cardiomyocytes exposed to normal glucose (5.5 mmol/L d-glucose). In comparison with nondiabetic controls, levels of Fundc1, Ip3r2, and MAMs were significantly increased in hearts from streptozotocin-treated mice and Akita mice. Furthermore, in comparison with control hearts, diabetes mellitus markedly increased coimmunoprecipitation of Fundc1 and Ip3r2. The binding of Fundc1 to Ip3r2 inhibits Ip3r2 ubiquitination and proteasome-mediated degradation. Cardiomyocyte-specific Fundc1 deletion ablated diabetes mellitus-induced MAM formation, prevented mitochondrial Ca2+ increase, mitochondrial fragmentation, and apoptosis with improved mitochondrial functional capacity and cardiac function. In mouse neonatal cardiomyocytes, HG suppressed AMP-activated protein kinase activity. Furthermore, in cardiomyocytes of Prkaa2 knockout mice, expression of Fundc1, MAM formation, and mitochondrial Ca2+ levels were significantly increased. Finally, adenoviral overexpression of a constitutively active mutant AMP-activated protein kinase ablated HG-induced MAM formation and mitochondrial dysfunction. CONCLUSIONS: We conclude that diabetes mellitus suppresses AMP-activated protein kinase, initiating Fundc1-mediated MAM formation, mitochondrial dysfunction, and cardiomyopathy, suggesting that AMP-activated protein kinase-induced Fundc1 suppression is a valid target to treat diabetic cardiomyopathy.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Cardiomiopatias Diabéticas/metabolismo , Retículo Endoplasmático/metabolismo , Proteínas de Membrana/metabolismo , Mitocôndrias/fisiologia , Membranas Mitocondriais/metabolismo , Proteínas Mitocondriais/metabolismo , Miócitos Cardíacos/fisiologia , Proteínas Quinases Ativadas por AMP/genética , Adulto , Idoso , Animais , Sinalização do Cálcio , Linhagem Celular , Cardiomiopatias Diabéticas/patologia , Retículo Endoplasmático/ultraestrutura , Feminino , Regulação da Expressão Gênica , Humanos , Masculino , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pessoa de Meia-Idade , Membranas Mitocondriais/ultraestrutura , Proteínas Mitocondriais/genética , Contração Miocárdica/genética , Ratos
5.
Arterioscler Thromb Vasc Biol ; 39(7): 1419-1431, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31092012

RESUMO

Objective- Inhibition of SIRT (sirtuin)-1, a nicotinamide adenine dinucleotide-dependent protein deacetylase, is linked to cigarette smoking-induced arterial stiffness, but the underlying mechanisms remain largely unknown. The aim of the present study was to determine the effects and mechanisms of nicotine, a major component of cigarette smoke, on SIRT1 activity and arterial stiffness. Approach and Results- Arterial stiffness, peroxynitrite (ONOO-) formation, SIRT1 expression and activity were monitored in mouse aortas of 8-week-old C57BL/6 mice (wild-type) or Sirt1-overexpressing ( Sirt1 Super) mice with or without nicotine for 4 weeks. In aortas of wild-type mice, nicotine reduced SIRT1 protein and activity by ≈50% without affecting its mRNA levels. In those from Sirt1 Super mice, nicotine also markedly reduced SIRT1 protein and activity to the levels that were comparable to those in wild-type mice. Nicotine infusion significantly induced collagen I, fibronectin, and arterial stiffness in wild-type but not Sirt1 Super mice. Nicotine increased the levels of iNOS (inducible nitric oxide synthase) and the co-staining of SIRT1 and 3-nitrotyrosine, a footprint of ONOO- in aortas. Tempol, which ablated ONOO- by scavenging superoxide anion, reduced the effects of nicotine on SIRT1 and collagen. Mutation of zinc-binding cysteine 395 or 398 in SIRT1 into serine (C395S) or (C398S) abolished SIRT1 activity. Furthermore, ONOO- dose-dependently inhibited the enzyme and increased zinc release in recombinant SIRT1. Finally, we found SIRT1 inactivation by ONOO- activated the YAP (Yes-associated protein) resulting in abnormal ECM (extracellular matrix) remodeling. Conclusions- Nicotine induces ONOO-, which selectively inhibits SIRT1 resulting in a YAP-mediated ECM remodeling. Visual Overview- An online visual overview is available for this article.


Assuntos
Nicotina/farmacologia , Ácido Peroxinitroso/fisiologia , Sirtuína 1/antagonistas & inibidores , Rigidez Vascular/efeitos dos fármacos , Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Animais , Proteínas de Ciclo Celular/fisiologia , Células Cultivadas , Matriz Extracelular/metabolismo , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Espécies Reativas de Nitrogênio/metabolismo , Sirtuína 1/fisiologia , Proteínas de Sinalização YAP
6.
Arterioscler Thromb Vasc Biol ; 38(2): 373-385, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29242271

RESUMO

OBJECTIVE: SNRK (sucrose nonfermenting 1-related kinase) is a novel member of the AMPK (adenosine monophosphate-activated protein kinase)-related superfamily that is activated in the process of angiogenesis. Currently, little is known about the function of SNRK in angiogenesis in the physiological and pathological conditions. APPROACH AND RESULTS: In this study, in Snrk global heterozygous knockout mice, retina angiogenesis and neovessel formation after hindlimb ischemia were suppressed. Consistently, mice with endothelial cell (EC)-specific Snrk deletion exhibited impaired retina angiogenesis, and delayed perfusion recovery and exacerbated muscle apoptosis in ischemic hindlimbs, compared with those of littermate wide-type mice. Endothelial SNRK expression was increased in the extremity vessel samples from nonischemic human. In ECs cultured in hypoxic conditions, HIF1α (hypoxia inducible factor 1α) bound to the SNRK promoter to upregulate SNRK expression. In the nuclei of hypoxic ECs, SNRK complexed with SP1 (specificity protein 1), and together, they bound to an SP1-binding motif in the ITGB1 (ß1 integrin) promoter, resulting in enhanced ITGB1 expression and promoted EC migration. Furthermore, SNRK or SP1 deficiency in ECs ameliorated hypoxia-induced ITGB1 expression and, consequently, inhibited EC migration and angiogenesis. CONCLUSIONS: Taken together, our data have revealed that SNRK/SP1-ITGB1 signaling axis promotes angiogenesis in vivo.


Assuntos
Células Endoteliais/enzimologia , Isquemia/enzimologia , Pulmão/irrigação sanguínea , Músculo Esquelético/irrigação sanguínea , Neovascularização Fisiológica , Proteínas Serina-Treonina Quinases/metabolismo , Vasos Retinianos/enzimologia , Animais , Antígenos CD/genética , Antígenos CD/metabolismo , Apoptose , Velocidade do Fluxo Sanguíneo , Caderinas/genética , Caderinas/metabolismo , Movimento Celular , Proliferação de Células , Células Cultivadas , Modelos Animais de Doenças , Células Endoteliais/patologia , Regulação Enzimológica da Expressão Gênica , Membro Posterior , Células Endoteliais da Veia Umbilical Humana/enzimologia , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Integrina beta1/genética , Integrina beta1/metabolismo , Isquemia/genética , Isquemia/fisiopatologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Regiões Promotoras Genéticas , Proteínas Serina-Treonina Quinases/deficiência , Proteínas Serina-Treonina Quinases/genética , Fluxo Sanguíneo Regional , Fator de Transcrição Sp1/genética , Fator de Transcrição Sp1/metabolismo
7.
Circulation ; 136(23): 2248-2266, 2017 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-28942427

RESUMO

BACKGROUND: FUN14 domain containing 1 (FUNDC1) is a highly conserved outer mitochondrial membrane protein. The aim of this study is to examine whether FUNDC1 modulates the mitochondria-associated endoplasmic reticulum (ER) membranes (MAMs), mitochondrial morphology, and function in cardiomyocytes and intact hearts. METHODS: The impacts of FUNDC1 on MAMs formation and cardiac functions were studied in mouse neonatal cardiomyocytes, in mice with cardiomyocyte-specific Fundc1 gene knockout (Fundc1f/Y/CreαMyHC+/- ), and in the cardiac tissues of the patients with heart failure. RESULTS: In mouse neonatal cardiomyocytes and intact hearts, FUNDC1 was localized in MAMs by binding to ER-resided inositol 1,4,5-trisphosphate type 2 receptor (IP3R2). Fundc1 ablation disrupted MAMs and reduced the levels of IP3R2 and Ca2+ in both mitochondria and cytosol, whereas overexpression of Fundc1 increased the levels of IP3R2 and Ca2+ in both mitochondria and cytosol. Consistently, Fundc1 ablation increased Ca2+ levels in ER, whereas Fundc1 overexpression lowered ER Ca2+ levels. Further, Fundc1 ablation in cardiomyocytes elongated mitochondria and compromised mitochondrial functions. Mechanistically, we found that Fundc1 ablation-induced reduction of intracellular Ca2+ levels suppressed mitochondrial fission 1 protein (Fis1) expression and mitochondrial fission by reducing the binding of the cAMP response element binding protein (CREB) in the Fis1 promoter. Fundc1f/Y/CreαMyHC+/- mice but not their littermate control mice (Fundc1wt/Y/CreαMyHC+/- ) exhibited cardiac dysfunction. The ligation of the left ventricle artery of Fundc1f/Y/CreαMyHC+/- mice caused more severe cardiac dysfunction than those in sham-treated Fundc1f/Y/CreαMyHC+/- mice. Finally, we found that the FUNDC1/MAMs/CREB/Fis1 signaling axis was significantly suppressed in patients with heart failure. CONCLUSIONS: We conclude that FUNDC1 binds to IP3R2 to modulate ER Ca2+ release into mitochondria and cytosol. Further, a disruption of the FUNDC1 and IP3R2 interaction lowers the levels of Ca2+ in mitochondria and cytosol, both of which instigate aberrant mitochondrial fission, mitochondrial dysfunction, cardiac dysfunction, and heart failure.


Assuntos
Retículo Endoplasmático/metabolismo , Insuficiência Cardíaca/metabolismo , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Membranas Intracelulares/metabolismo , Proteínas de Membrana/metabolismo , Mitocôndrias Cardíacas/metabolismo , Dinâmica Mitocondrial , Membranas Mitocondriais/metabolismo , Proteínas Mitocondriais/metabolismo , Miócitos Cardíacos/metabolismo , Animais , Sítios de Ligação , Cálcio/metabolismo , Estudos de Casos e Controles , Células Cultivadas , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Retículo Endoplasmático/patologia , Predisposição Genética para Doença , Insuficiência Cardíaca/genética , Insuficiência Cardíaca/patologia , Insuficiência Cardíaca/fisiopatologia , Humanos , Membranas Intracelulares/patologia , Proteínas de Membrana/deficiência , Proteínas de Membrana/genética , Camundongos Knockout , Mitocôndrias Cardíacas/patologia , Membranas Mitocondriais/patologia , Proteínas Mitocondriais/deficiência , Proteínas Mitocondriais/genética , Mitofagia , Miócitos Cardíacos/patologia , Fenótipo , Regiões Promotoras Genéticas , Ligação Proteica , Transdução de Sinais , Fatores de Tempo
11.
Biochim Biophys Acta ; 1842(9): 1844-54, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25016145

RESUMO

Lipid accumulation is a central event in the development of chronic metabolic diseases, including obesity and type 2 diabetes, but the mechanisms responsible for lipid accumulation are incompletely understood. This study was designed to investigate the mechanisms for excess nutrient-induced lipid accumulation and whether activation of AMP-activated protein kinase (AMPK) prevents the hepatic lipid accumulation in excess nutrient-treated HepG2 cells and high fat diet (HFD)-fed mice. Exposure of HepG2 cells to high levels of glucose or palmitate induced the endoplasmic reticulum (ER) stress response, activated sterol regulatory element-binding protein-1 (SREBP-1), and enhanced lipid accumulation, all of which were sensitive to ER stress inhibitor and gene silencing of eukaryotic initiation factor 2α. The increases in ER stress response and lipid accumulation were associated with activation of mammalian target of rapamycin complex 1 (mTORC1) signaling. Inhibition of mTORC1 signaling attenuated the ER stress response and lipid accumulation induced by high glucose or by deletion of tuberous sclerosis 2. In addition, AMPK activation prevented the mTORC1 activation, ER stress response, and lipid accumulation. This effect was mimicked or abrogated, respectively, by overexpression of constitutively active and dominant-negative AMPK mutants. Finally, treatment of HFD-fed mice with 5-aminoimidazole-4-carboxamide-1-ß-4-ribofuranoside inhibited the mTORC1 pathway, suppressed the ER stress response, and prevented insulin resistance and hepatic lipid accumulation. We conclude that activation of AMPK prevents excess nutrient-induced hepatic lipid accumulation by inhibiting mTORC1 and ER stress response.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Dieta Hiperlipídica , Estresse do Retículo Endoplasmático , Retículo Endoplasmático/metabolismo , Lipídeos/análise , Fígado/metabolismo , Complexos Multiproteicos/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Proteínas Quinases Ativadas por AMP/genética , Animais , Western Blotting , Colesterol/metabolismo , Retículo Endoplasmático/patologia , Glucose/farmacologia , Técnica Clamp de Glucose , Células Hep G2 , Humanos , Hiperinsulinismo/metabolismo , Hiperinsulinismo/patologia , Fígado/patologia , Masculino , Alvo Mecanístico do Complexo 1 de Rapamicina , Camundongos , Camundongos Endogâmicos C57BL , Complexos Multiproteicos/antagonistas & inibidores , Complexos Multiproteicos/genética , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteína de Ligação a Elemento Regulador de Esterol 1/genética , Proteína de Ligação a Elemento Regulador de Esterol 1/metabolismo , Edulcorantes/farmacologia , Serina-Treonina Quinases TOR/antagonistas & inibidores , Serina-Treonina Quinases TOR/genética , Triglicerídeos/metabolismo
12.
J Mol Cell Cardiol ; 71: 71-80, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24513079

RESUMO

Diabetic cardiomyopathy is characterized by ventricular dysfunction that occurs in diabetic patients independent of coronary artery disease, hypertension, and any other cardiovascular diseases. Diabetic cardiomyopathy has become a major cause of diabetes-related mortality. Thus, an urgent need exists to clarify the mechanism of pathogenesis. Emerging evidence demonstrates that diabetes induces cardiomyocyte apoptosis and suppresses cardiac autophagy, indicating that the interplay between the autophagy and apoptotic cell death pathways is important in the pathogenesis of diabetic cardiomyopathy. This review highlights recent advances in the crosstalk between autophagy and apoptosis and its importance in the development of diabetic cardiomyopathy. This article is part of a Special Issue entitled "Protein Quality Control, the Ubiquitin Proteasome System, and Autophagy".


Assuntos
Apoptose/fisiologia , Autofagia/fisiologia , Diabetes Mellitus/patologia , Cardiomiopatias Diabéticas/patologia , Animais , Humanos , Miócitos Cardíacos/patologia
13.
Am J Physiol Endocrinol Metab ; 306(2): E197-209, 2014 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-24302004

RESUMO

Nutrient overload is associated with the development of obesity, insulin resistance, and type 2 diabetes. However, the underlying mechanisms for developing insulin resistance in the presence of excess nutrients are incompletely understood. We investigated whether activation of AMP-activated protein kinase (AMPK) prevents the hepatic insulin resistance that is induced by the consumption of a high-protein diet (HPD) and the presence of excess amino acids. Exposure of HepG2 cells to excess amino acids reduced AMPK phosphorylation, upregulated Notch1 expression, and impaired the insulin-stimulated phosphorylation of Akt Ser(473) and insulin receptor substrate-1 (IRS-1) Tyr(612). Inhibition of Notch1 prevented amino acid-induced insulin resistance, which was accompanied by reduced expression of Rbp-Jk, hairy and enhancer of split-1, and forkhead box O1. Mechanistically, mTORC1 signaling was activated by excess amino acids, which then positively regulated Notch1 expression through the activation of the signal transducer and activator of transcription 3 (STAT3). Activation of AMPK by metformin inhibited mTORC1-STAT3 signaling, thereby preventing excess amino acid-impaired insulin signaling. Finally, HPD feeding suppressed AMPK activity, activated mTORC1/STAT3/Notch1 signaling, and induced insulin resistance. Chronic administration of either metformin or rapamycin inhibited the HPD-activated mTORC1/STAT3/Notch1 signaling pathway and prevented hepatic insulin resistance. We conclude that the upregulation of Notch1 expression by hyperactive mTORC1 signaling is an essential event in the development of hepatic insulin resistance in the presence of excess amino acids. Activation of AMPK prevents amino acid-induced insulin resistance through the suppression of the mTORC1/STAT3/Notch1 signaling pathway.


Assuntos
Proteínas Quinases Ativadas por AMP/fisiologia , Aminoácidos/metabolismo , Resistência à Insulina , Fígado/metabolismo , Complexos Multiproteicos/antagonistas & inibidores , Receptor Notch1/antagonistas & inibidores , Fator de Transcrição STAT3/antagonistas & inibidores , Serina-Treonina Quinases TOR/antagonistas & inibidores , Aminoácidos/farmacologia , Animais , Regulação para Baixo/efeitos dos fármacos , Ativação Enzimática , Células Hep G2 , Humanos , Fígado/efeitos dos fármacos , Masculino , Alvo Mecanístico do Complexo 1 de Rapamicina , Camundongos , Camundongos Endogâmicos C57BL , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia
14.
Am J Pathol ; 183(2): 626-37, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23770348

RESUMO

Oxidized lipoproteins stimulate autophagy in advanced atherosclerotic plaques. However, the mechanisms underlying autophagy induction and the role of autophagy in atherogenesis remain to be determined. This study was designed to investigate the mechanisms by which 7-ketocholesterol (7-KC), a major component of oxidized lipoproteins, induces autophagy. This study was also designed to determine the effect of autophagy induction on apoptosis, a central event in the development of atherosclerosis. Exposure of human aortic smooth muscle cells to 7-KC increased autophagic flux. Autophagy induction was suppressed by treating the cells with either a reactive oxygen species scavenger or an antioxidant. Administration of 7-KC concomitantly up-regulated Nox4 expression, increased intracellular hydrogen peroxide levels, and inhibited autophagy-related gene 4B activity. Catalase overexpression to remove hydrogen peroxide or Nox4 knockdown with siRNA reduced intracellular hydrogen peroxide levels, restored autophagy-related gene 4B activity, and consequently attenuated 7-KC-induced autophagy. Moreover, inhibition of autophagy aggravated both endoplasmic reticulum (ER) stress and cell death in response to 7-KC. In contrast, up-regulation of autophagic activity by rapamycin had opposite effects. Finally, activation of autophagy by chronic rapamycin treatment attenuated ER stress, apoptosis, and atherosclerosis in apolipoprotein E knockout (ApoE(-/-)) mouse aortas. In conclusion, we demonstrate that up-regulation of autophagy is a cellular protective response that attenuates 7-KC-induced cell death in human aortic smooth muscle cells.


Assuntos
Autofagia/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Cetocolesteróis/farmacologia , Animais , Aorta , Apoptose , Aterosclerose/prevenção & controle , Proteínas Relacionadas à Autofagia , Fármacos Cardiovasculares/farmacologia , Morte Celular/efeitos dos fármacos , Células Cultivadas , Cisteína Endopeptidases/efeitos dos fármacos , Cisteína Endopeptidases/metabolismo , Sequestradores de Radicais Livres/farmacologia , Humanos , Peróxido de Hidrogênio/metabolismo , Masculino , Camundongos , Proteínas Associadas aos Microtúbulos/metabolismo , Músculo Liso Vascular/efeitos dos fármacos , Músculo Liso Vascular/fisiologia , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/fisiologia , NADPH Oxidase 4 , NADPH Oxidases/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Sirolimo/farmacologia , Regulação para Cima
15.
Autophagy ; 20(3): 629-644, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-37963060

RESUMO

PYCARD (PYD and CARD domain containing), a pivotal adaptor protein in inflammasome assembly and activation, contributes to innate immunity, and plays an essential role in the pathogenesis of atherosclerosis and restenosis. However, its roles in microRNA biogenesis remain unknown. Therefore, this study aimed to investigate the roles of PYCARD in miRNA biogenesis and neointima formation using pycard knockout (pycard-/-) mice. Deficiency of Pycard reduced circulating miRNA profile and inhibited Mir17 seed family maturation. The systemic pycard knockout also selectively reduced the expression of AGO2 (argonaute RISC catalytic subunit 2), an important enzyme in regulating miRNA biogenesis, by promoting chaperone-mediated autophagy (CMA)-mediated degradation of AGO2, specifically in adipose tissue. Mechanistically, pycard knockout increased PRMT8 (protein arginine N-methyltransferase 8) expression in adipose tissue, which enhanced AGO2 methylation, and subsequently promoted its binding to HSPA8 (heat shock protein family A (Hsp70) member 8) that targeted AGO2 for lysosome degradation through chaperone-mediated autophagy. Finally, the reduction of AGO2 and Mir17 family expression prevented vascular injury-induced neointima formation in Pycard-deficient conditions. Overexpression of AGO2 or administration of mimic of Mir106b (a major member of the Mir17 family) prevented Pycard deficiency-mediated inhibition of neointima formation in response to vascular injury. These data demonstrate that PYCARD inhibits CMA-mediated degradation of AGO2, which promotes microRNA maturation, thereby playing a critical role in regulating neointima formation in response to vascular injury independently of inflammasome activity and suggest that modulating PYCARD expression and function may represent a powerful therapeutic strategy for neointima formation.Abbreviations: 6-AN: 6-aminonicotinamide; ACTB: actin, beta; aDMA: asymmetric dimethylarginine; AGO2: argonaute RISC catalytic subunit 2; CAL: carotid artery ligation; CALCOCO2: calcium binding and coiled-coil domain 2; CMA: chaperone-mediated autophagy; CTSB: cathepsin B; CTSD: cathepsin D; DGCR8: DGCR8 microprocessor complex subunit; DOCK2: dedicator of cyto-kinesis 2; EpiAdi: epididymal adipose tissue; HSPA8: heat shock protein family A (Hsp70) member 8; IHC: immunohistochemical; ISR: in-stent restenosis; KO: knockout; LAMP2: lysosomal-associated membrane protein 2; MAP1LC3/LC3: microtubule associated protein 1 light chain 3; miRNA: microRNA; NLRP3: NLR family pyrin domain containing 3; N/L: ammonium chloride combined with leupeptin; PRMT: protein arginine methyltransferase; PVAT: peri-vascular adipose tissues; PYCARD: PYD and CARD domain containing; sDMA: symmetric dimethylarginine; ULK1: unc-51 like kinase 1; VSMCs: vascular smooth muscle cells; WT: wild-type.


Assuntos
Autofagia Mediada por Chaperonas , MicroRNAs , Lesões do Sistema Vascular , Animais , Camundongos , MicroRNAs/genética , Inflamassomos/metabolismo , Autofagia/fisiologia , Neointima , Proteínas de Ligação a RNA , Proteínas de Choque Térmico/metabolismo , Proteínas Adaptadoras de Sinalização CARD/metabolismo , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Proteínas Ativadoras de GTPase/metabolismo , Proteína-Arginina N-Metiltransferases/metabolismo
16.
Autophagy ; 19(5): 1491-1511, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36300763

RESUMO

Ischemia-induced angiogenesis is critical for blood flow restoration and tissue regeneration, but the underlying molecular mechanism is not fully understood. ATG7 (autophagy related 7) is essential for classical degradative macroautophagy/autophagy and cell cycle regulation. However, whether and how ATG7 influences endothelial cell (EC) function and regulates post-ischemic angiogenesis remain unknown. Here, we showed that in mice subjected to femoral artery ligation, EC-specific deletion of Atg7 significantly impaired angiogenesis, delayed the recovery of blood flow reperfusion, and displayed reduction in HIF1A (hypoxia inducible factor 1 subunit alpha) expression. In addition, in cultured human umbilical vein endothelial cells (HUVECs), overexpression of HIF1A prevented ATG7 deficiency-reduced tube formation. Mechanistically, we identified STAT1 (signal transducer and activator of transcription 1) as a transcription suppressor of HIF1A and demonstrated that ablation of Atg7 upregulated STAT1 in an autophagy independent pathway, increased STAT1 binding to HIF1A promoter, and suppressed HIF1A expression. Moreover, lack of ATG7 in the cytoplasm disrupted the association between ATG7 and the transcription factor ZNF148/ZFP148/ZBP-89 (zinc finger protein 148) that is required for STAT1 constitutive expression, increased the binding between ZNF148/ZFP148/ZBP-89 and KPNB1 (karyopherin subunit beta 1), which promoted ZNF148/ZFP148/ZBP-89 nuclear translocation, and increased STAT1 expression. Finally, inhibition of STAT1 by fludarabine prevented the inhibition of HIF1A expression, angiogenesis, and blood flow recovery in atg7 KO mice. Our work reveals that lack of ATG7 inhibits angiogenesis by suppression of HIF1A expression through upregulation of STAT1 independently of autophagy under ischemic conditions, and suggest new therapeutic strategies for cancer and cardiovascular diseases.Abbreviations: ATG5: autophagy related 5; ATG7: autophagy related 7; atg7 KO: endothelial cell-specific atg7 knockout; BECN1: beclin 1; ChIP: chromatin immunoprecipitation; CQ: chloroquine; ECs: endothelial cells; EP300: E1A binding protein p300; HEK293: human embryonic kidney 293 cells; HIF1A: hypoxia inducible factor 1 subunit alpha; HUVECs: human umbilical vein endothelial cells; IFNG/IFN-γ: Interferon gamma; IRF9: interferon regulatory factor 9; KPNB1: karyopherin subunit beta 1; MAP1LC3A: microtubule associated protein 1 light chain 3 alpha; MEFs: mouse embryonic fibroblasts; MLECs: mouse lung endothelial cells; NAC: N-acetyl-l-cysteine; NFKB1/NFκB: nuclear factor kappa B subunit 1; PECAM1/CD31: platelet and endothelial cell adhesion molecule 1; RELA/p65: RELA proto-oncogene, NF-kB subunit; ROS: reactive oxygen species; SP1: Sp1 transcription factor; SQSTM1/p62: sequestosome 1; STAT1: signal transducer and activator of transcription 1; ULK1: unc-51 like autophagy activating kinase 1; ulk1 KO: endothelial cell-specific ulk1 knockout; VSMCs: mouse aortic smooth muscle cells; WT: wild type; ZNF148/ZFP148/ZBP-89: zinc finger protein 148.


Assuntos
Autofagia , Fibroblastos , Camundongos , Humanos , Animais , Autofagia/genética , Células HEK293 , Fator de Transcrição STAT1 , Células Endoteliais da Veia Umbilical Humana , Isquemia , Fator 1 Induzível por Hipóxia , Carioferinas , Proteínas de Ligação a DNA , Fatores de Transcrição , Subunidade alfa do Fator 1 Induzível por Hipóxia
17.
Am J Physiol Endocrinol Metab ; 303(10): E1234-44, 2012 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-22967499

RESUMO

Leucine supplementation has been shown to prevent high-fat diet (HFD)-induced obesity, hyperglycemia, and dyslipidemia in animal models, but the underlying mechanisms are not fully understood. Recent studies suggest that activation of Sirtuin 1 (SIRT1) is an important mechanism to maintain energy and metabolic homeostasis. We therefore examined the involvement of SIRT1 in leucine supplementation-prevented obesity and insulin resistance. To accomplish this goal, male C57BL/6J mice were fed normal diet or HFD, supplemented with or without leucine. After 2 mo of treatment, alterations in SIRT1 expression, insulin signaling, and energy metabolism were analyzed. Eight weeks of HFD induced obesity, fatty liver, mitochondrial dysfunction, hyperglycemia, and insulin resistance in mice. Addition of leucine to HFD correlated with increased expression of SIRT1 and NAMPT (nicotinamide phosphoribosyltransferase) as well as higher intracellular NAD(+) levels, which decreased acetylation of peroxisome proliferator-activated receptor-γ coactivator 1α (PGC1α) and forkhead box O1 (FoxO1). The deacetylation of PGC1α may contribute to upregulation of genes controlling mitochondrial biogenesis and fatty acid oxidation, thereby improving mitochondrial function and preventing HFD-induced obesity in mice. Moreover, decreased acetylation of FoxO1 was accompanied by decreased expression of pseudokinase tribble 3 (TRB3) and reduced the association between TRB3 and Akt, which enhanced insulin sensitivity and improved glucose metabolism. Finally, transfection of dominant negative AMPK prevented activation of SIRT1 signaling in HFD-Leu mice. These data suggest that increased expression of SIRT1 after leucine supplementation may lead to reduced acetylation of PGC1α and FoxO1, which is associated with attenuation of HFD-induced mitochondrial dysfunction, insulin resistance, and obesity.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Citocinas/metabolismo , Resistência à Insulina/fisiologia , Leucina/administração & dosagem , Nicotinamida Fosforribosiltransferase/metabolismo , Obesidade/metabolismo , Sirtuína 1/biossíntese , Proteínas Quinases Ativadas por AMP/genética , Animais , Western Blotting , Proteínas de Ciclo Celular , Citocinas/genética , DNA Mitocondrial/química , DNA Mitocondrial/genética , Suplementos Nutricionais , Proteína Forkhead Box O1 , Fatores de Transcrição Forkhead/metabolismo , Regulação da Expressão Gênica , Técnica Clamp de Glucose , Leucina/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Renovação Mitocondrial , Nicotinamida Fosforribosiltransferase/genética , Obesidade/genética , Obesidade/prevenção & controle , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo , RNA/química , RNA/genética , Distribuição Aleatória , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais , Sirtuína 1/genética , Sirtuína 1/metabolismo , Transativadores/metabolismo , Fatores de Transcrição
18.
Am J Pathol ; 179(6): 2835-44, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22015457

RESUMO

The risk of diabetic retinopathy is associated with the presence of both oxidative stress and toxic eicosanoids. Whether oxidative stress actually causes diabetic retinopathy via the generation of toxic eicosanoids, however, remains unknown. The aim of the present study was to determine whether tyrosine nitration of prostacyclin synthase (PGIS) contributes to retinal cell death in vitro and in vivo. Exposure of human retinal pericytes to heavily oxidized and glycated LDL (HOG-LDL), but not native forms of LDL (N-LDL), for 24 hours significantly increased pericyte apoptosis, accompanied by increased tyrosine nitration of PGIS and decreased PGIS activity. Inhibition of the thromboxane receptor or cyclooxygenase-2 dramatically attenuated HOG-LDL-induced apoptosis without restoring PGIS activity. Administration of superoxide dismutase (to scavenge superoxide anions) or L-N(G)-nitroarginine methyl ester (L-NAME, a nonselective nitric oxide synthase inhibitor) restored PGIS activity and attenuated pericyte apoptosis. In Akita mouse retinas, diabetes increased intraretinal levels of oxidized LDL and glycated LDL, induced PGIS nitration, enhanced apoptotic cell death, and impaired blood-retinal barrier function. Chronic administration of tempol, a superoxide scavenger, reduced intraretinal oxidized LDL and glycated LDL levels, PGIS nitration, and retina cell apoptosis, thereby preserving the integrity of blood-retinal barriers. In conclusion, oxidized LDL-mediated PGIS nitration and associated thromboxane receptor stimulation might be important in the initiation and progression of diabetic retinopathy.


Assuntos
Apoptose/efeitos dos fármacos , Sistema Enzimático do Citocromo P-450/metabolismo , Retinopatia Diabética/patologia , Oxirredutases Intramoleculares/metabolismo , Lipoproteínas LDL/farmacologia , Tirosina/farmacologia , Animais , Antioxidantes/farmacologia , Barreira Hematorretiniana , Células Cultivadas , Óxidos N-Cíclicos/farmacologia , Ciclo-Oxigenase 2/metabolismo , Produtos Finais de Glicação Avançada , Humanos , Lipoproteínas LDL/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , NG-Nitroarginina Metil Éster/farmacologia , Óxido Nítrico/biossíntese , Óxido Nítrico Sintase Tipo II/metabolismo , Pericitos/patologia , Receptores de Tromboxanos/metabolismo , Retina/metabolismo , Retina/patologia , Marcadores de Spin , Superóxido Dismutase/farmacologia , Junções Íntimas/efeitos dos fármacos , Tirosina/análogos & derivados
19.
Circ Res ; 106(6): 1117-28, 2010 Apr 02.
Artigo em Inglês | MEDLINE | ID: mdl-20167927

RESUMO

RATIONALE: AMP-activated protein kinase (AMPK) is an energy sensor and ubiquitously expressed in vascular cells. Recent studies suggest that AMPK activation improves endothelial function by counteracting oxidative stress in endothelial cells. How AMPK suppresses oxidative stress remains to be established. OBJECTIVE: The aim of this study is to examine the effects of AMPK in regulating NAD(P)H oxidase, oxidative stress, and endothelial function. METHODS AND RESULTS: The markers of oxidative stress, NAD(P)H oxidase subunit expression (gp91(phox), p47(phox), p67(phox), NOX1 to -4), NAD(P)H oxidase-mediated superoxide production, 26S proteasome activity, IkappaBalpha degradation, and nuclear translocation of nuclear factor (NF)-kappaB (p50 and p65) were examined in cultured human umbilical vein endothelial cells and mouse aortas isolated from AMPKalpha2 deficient mice. Compared to the wild type, acetylcholine-induced endothelium-dependent relaxation was significantly impaired in parallel with increased production of oxidants in AMPKalpha2(-/-) mice. Further, pretreatment of aorta with either superoxide dismutase (SOD) or tempol or apocynin significantly improved acetylcholine-induced endothelium-dependent relaxation in AMPKalpha2(-/-) mice. Analysis of aortic endothelial cells from AMPKalpha2(-/-) mice and human umbilical vein endothelial cells expressing dominant negative AMPK or AMPKalpha2-specific siRNA revealed that loss of AMPK activity increased NAD(P)H oxidase subunit expression (gp91(phox), p47(phox), p67(phox), NOX1 and -4), NAD(P)H oxidase-mediated superoxide production, 26S proteasome activity, IkappaBalpha degradation, and nuclear translocation of NF-kappaB (p50 and p65), whereas AMPK activation by AICAR or overexpression of constitutively active AMPK had the opposite effect. Consistently, we found that genetic deletion of AMPKalpha2 in low-density lipoprotein receptor knockout (LDLr(-/-)) strain markedly increased 26S proteasome activity, IkappaB degradation, NF-kappaB transactivation, NAD(P)H oxidase subunit overexpression, oxidative stress, and endothelial dysfunction, all of which were largely suppressed by chronic administration of MG132, a potent cell permeable proteasome inhibitor. CONCLUSIONS: We conclude that AMPKalpha2 functions as a physiological suppressor of NAD(P)H oxidase and ROS production in endothelial cells. In this way, AMPK maintains the nonatherogenic and noninflammatory phenotype of endothelial cells.


Assuntos
Proteínas Quinases Ativadas por AMP/deficiência , Células Endoteliais/enzimologia , Endotélio Vascular/enzimologia , NADPH Oxidases/metabolismo , Estresse Oxidativo , Complexo de Endopeptidases do Proteassoma/metabolismo , Vasodilatação , Proteínas Quinases Ativadas por AMP/antagonistas & inibidores , Proteínas Quinases Ativadas por AMP/genética , Acetofenonas/farmacologia , Acetilcolina/farmacologia , Aminoimidazol Carboxamida/análogos & derivados , Aminoimidazol Carboxamida/farmacologia , Animais , Antioxidantes/farmacologia , Células Cultivadas , Óxidos N-Cíclicos/farmacologia , Relação Dose-Resposta a Droga , Células Endoteliais/efeitos dos fármacos , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/fisiopatologia , Ativação Enzimática , Inibidores Enzimáticos/farmacologia , Deleção de Genes , Regulação Enzimológica da Expressão Gênica , Humanos , Proteínas I-kappa B/metabolismo , Mediadores da Inflamação/sangue , Interferon gama/sangue , Interleucina-2/sangue , Isoenzimas , Leupeptinas/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , NADPH Oxidases/antagonistas & inibidores , NADPH Oxidases/genética , Inibidor de NF-kappaB alfa , NF-kappa B/metabolismo , Nitroprussiato/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Inibidores de Proteassoma , Interferência de RNA , Receptores de LDL/deficiência , Receptores de LDL/genética , Ribonucleotídeos/farmacologia , Marcadores de Spin , Superóxido Dismutase/metabolismo , Superóxidos/metabolismo , Transfecção , Vasodilatação/efeitos dos fármacos , Vasodilatadores/farmacologia
20.
Nat Commun ; 13(1): 6371, 2022 10 26.
Artigo em Inglês | MEDLINE | ID: mdl-36289221

RESUMO

Indoleamine 2,3 dioxygenase-1 (IDO1) catalyzes tryptophan-kynurenine metabolism in many inflammatory and cancer diseases. Of note, acute inflammation that occurs immediately after heart injury is essential for neonatal cardiomyocyte proliferation and heart regeneration. However, the IDO1-catalyzed tryptophan metabolism during heart regeneration is largely unexplored. Here, we find that apical neonatal mouse heart resection surgery led to rapid and consistent increases in cardiac IDO1 expression and kynurenine accumulation. Cardiac deletion of Ido1 gene or chemical inhibition of IDO1 impairs heart regeneration. Mechanistically, elevated kynurenine triggers cardiomyocyte proliferation by activating the cytoplasmic aryl hydrocarbon receptor-SRC-YAP/ERK pathway. In addition, cardiomyocyte-derived kynurenine transports to endothelial cells and stimulates cardiac angiogenesis by promoting aryl hydrocarbon receptor nuclear translocation and enhancing vascular endothelial growth factor A expression. Notably, Ahr deletion prevents indoleamine 2,3 dioxygenase -kynurenine-associated heart regeneration. In summary, increasing indoleamine 2,3 dioxygenase-derived kynurenine level promotes cardiac regeneration by functioning as an endogenous regulator of cardiomyocyte proliferation and cardiac angiogenesis.


Assuntos
Cinurenina , Receptores de Hidrocarboneto Arílico , Camundongos , Animais , Cinurenina/metabolismo , Receptores de Hidrocarboneto Arílico/genética , Receptores de Hidrocarboneto Arílico/metabolismo , Fator A de Crescimento do Endotélio Vascular/genética , Triptofano/metabolismo , Indolamina-Pirrol 2,3,-Dioxigenase/metabolismo , Células Endoteliais/metabolismo , Miócitos Cardíacos/metabolismo , Transdução de Sinais/fisiologia , Proliferação de Células
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA