Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Cardiovasc Pharmacol ; 82(3): 221-228, 2023 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-37381169

RESUMO

ABSTRACT: Excessive proliferation and migration of vascular smooth muscle cells (VSMCs) cause neointimal hyperplasia after percutaneous vascular interventions. Nuclear receptor subfamily 1 group D member 1 (NR1D1), a crucial member of circadian clock, is involved in the regulation of atherosclerosis and cellular proliferation. However, whether NR1D1 affects vascular neointimal hyperplasia remains unclear. In this study, we found that activating NR1D1 reduced injury-induced vascular neointimal hyperplasia. Overexpression of NR1D1 reduced the number of Ki-67-positive VSMCs and migrated VSMCs after platelet-derived growth factor (PDGF)-BB treatment. Mechanistically, NR1D1 suppressed the phosphorylation of AKT and 2 main effectors of the mammalian target of rapamycin complex 1 (mTORC1), S6, and 4EBP1 in PDGF-BB-challenged VSMCs. Re-activation of mTORC1 by Tuberous sclerosis 1 siRNA (si Tsc1 ) and re-activation of AKT by SC-79 abolished NR1D1-mediated inhibitory effects on proliferation and migration of VSMCs. Moreover, decreased mTORC1 activity induced by NR1D1 was also reversed by SC-79. Simultaneously, Tsc1 knockdown abolished the vascular protective effects of NR1D1 in vivo. In conclusion, NR1D1 reduces vascular neointimal hyperplasia by suppressing proliferation and migration of VSMCs in an AKT/mTORC1-dependent manner.


Assuntos
Músculo Liso Vascular , Lesões do Sistema Vascular , Humanos , Hiperplasia/metabolismo , Músculo Liso Vascular/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proliferação de Células , Becaplermina/farmacologia , Lesões do Sistema Vascular/patologia , Neointima/metabolismo , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo , Miócitos de Músculo Liso/metabolismo , Movimento Celular , Células Cultivadas
2.
Clin Exp Hypertens ; 45(1): 2178659, 2023 Dec 31.
Artigo em Inglês | MEDLINE | ID: mdl-36794491

RESUMO

BACKGROUND: In-stent restenosis hardly limits the therapeutic effect of the percutaneous vascular intervention. Although the restenosis is significantly ameliorated after the application of new drug-eluting stents, the incidence of restenosis remains at a high level. OBJECTIVE: Vascular adventitial fibroblasts (AFs) play an important role in intimal hyperplasia and subsequent restenosis. The current study was aimed to investigate the role of nuclear receptor subfamily 1, group D, member 1 (NR1D1) in the vascular intimal hyperplasia. METHODS AND RESULTS: We observed increased expression of NR1D1 after the transduction of adenovirus carrying Nr1d1 gene (Ad-Nr1d1) in AFs. Ad-Nr1d1 transduction significantly reduced the numbers of total AFs, Ki-67-positive AFs, and the migration rate of AFs. NR1D1 overexpression decreased the expression level of ß-catenin and attenuated the phosphorylation of the effectors of mammalian target of rapamycin complex 1 (mTORC1), including mammalian target of rapamycin (mTOR) and 4E binding protein 1 (4EBP1). Restoration of ß-catenin by SKL2001 abolished the inhibitory effects of NR1D1 overexpression on the proliferation and migration of AFs. Surprisingly, the restoration of mTORC1 activity by insulin could also reverse the decreased expression of ß-catenin, attenuated proliferation, and migration in AFs induced by NR1D1 overexpression. In vivo, we found that SR9009 (an agonist of NR1D1) ameliorated the intimal hyperplasia at days 28 after injury of carotid artery. We further observed that SR9009 attenuated the increased Ki-67-positive AFs, an essential part of vascular restenosis at days 7 after injury to the carotid artery. CONCLUSION: These data suggest that NR1D1 inhibits intimal hyperplasia by suppressing the proliferation and migration of AFs in a mTORC1/ß-catenin-dependent manner.


Assuntos
Músculo Liso Vascular , Membro 1 do Grupo D da Subfamília 1 de Receptores Nucleares , beta Catenina , beta Catenina/metabolismo , Movimento Celular , Proliferação de Células , Células Cultivadas , Fibroblastos , Hiperplasia/metabolismo , Hiperplasia/patologia , Antígeno Ki-67/metabolismo , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Miócitos de Músculo Liso , Neointima/genética , Neointima/metabolismo , Neointima/patologia , Membro 1 do Grupo D da Subfamília 1 de Receptores Nucleares/metabolismo , Serina-Treonina Quinases TOR/metabolismo
3.
Tohoku J Exp Med ; 256(2): 131-139, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-35197406

RESUMO

Type 2 diabetes mellitus (T2DM)-associated mitochondrial impairment may a key factor leading to liver injury. Transient receptor potential receptor vanilloid 1 (TRPV1) regulates the energy expenditure and cholesterol metabolism in hepatocytes and protects against oxidative toxicity. Optic atrophy 1 (OPA1) is involved in the protection of TRPV1 on cardiac microvascular and lung injury. The aim of this study is to identify the role of TRPV1 in redox signals and liver protection via OPA1. TRPV1 knockout (TRPV1-/-) mice were used. And T2DM associated liver injury was induced by high glucose and high fatty acid (HG/HF) treatment. Mechanisms were studied by TUNEL staining, transmission electron microscope (TEM) analysis, reverse transcription polymerase chain reaction (RT-PCR) and Western blotting in vivo and in vitro. We determined that HG/HF treatment increased TRPV1 expression in liver tissues and AML12 cells. The knockout of TRPV1 increased the apoptotic hepatocytes rate. The inhibition of TRPV1 by 5'-iRTX in HG/HF group elevated the reactive oxygen species (ROS) levels, whereas TRPV1 agonist capsaicin reduced ROS. Our studies also showed that the OPA1 expression was lower in livers from HG/HF treated mice than the control, and genetic ablation of TRPV1 decreased OPA1 expression to a greater extent than the HG/HF mice. The protective effects of TRPV1 on mitochondrial were blocked by OPA1 siRNA. In conclusion, our study showed that the identified regulation of TRPV1 to OPA1 has important implication to the pathogenesis of T2DM-associated liver injury. Targeting the action of TRPV1 and OPA1 presents a potential therapeutic intervention.


Assuntos
Doença Hepática Crônica Induzida por Substâncias e Drogas , Diabetes Mellitus Tipo 2 , GTP Fosfo-Hidrolases , Hiperglicemia , Hiperlipidemias , Canais de Cátion TRPV , Animais , Apoptose , Hiperglicemia/complicações , Camundongos , Canais de Cátion TRPV/genética , Canais de Cátion TRPV/metabolismo
4.
Aging (Albany NY) ; 13(1): 493-515, 2020 11 26.
Artigo em Inglês | MEDLINE | ID: mdl-33259334

RESUMO

Mitochondrial calcium uptake 1 (MICU1) is a pivotal molecule in maintaining mitochondrial homeostasis under stress conditions. However, it is unclear whether MICU1 attenuates mitochondrial stress in angiotensin II (Ang-II)-induced cardiac hypertrophy or if it has a role in the function of melatonin. Here, small-interfering RNAs against MICU1 or adenovirus-based plasmids encoding MICU1 were delivered into left ventricles of mice or incubated with neonatal murine ventricular myocytes (NMVMs) for 48 h. MICU1 expression was depressed in hypertrophic myocardia and MICU1 knockdown aggravated Ang-II-induced cardiac hypertrophy in vivo and in vitro. In contrast, MICU1 upregulation decreased cardiomyocyte susceptibility to hypertrophic stress. Ang-II administration, particularly in NMVMs with MICU1 knockdown, led to significantly increased reactive oxygen species (ROS) overload, altered mitochondrial morphology, and suppressed mitochondrial function, all of which were reversed by MICU1 supplementation. Moreover, peroxisome proliferator-activated receptor gamma coactivator 1-α (PGC-1α)/MICU1 expression in hypertrophic myocardia increased with melatonin. Melatonin ameliorated excessive ROS generation, promoted mitochondrial function, and attenuated cardiac hypertrophy in control but not MICU1 knockdown NMVMs or mice. Collectively, our results demonstrate that MICU1 attenuates Ang-II-induced cardiac hypertrophy by inhibiting mitochondria-derived oxidative stress. MICU1 activation may be the mechanism underlying melatonin-induced protection against myocardial hypertrophy.


Assuntos
Antioxidantes/farmacologia , Proteínas de Ligação ao Cálcio/genética , Cardiomegalia/genética , Melatonina/farmacologia , Mitocôndrias/metabolismo , Proteínas de Transporte da Membrana Mitocondrial/genética , Miocárdio/metabolismo , Miócitos Cardíacos/metabolismo , Estresse Oxidativo/genética , Angiotensina II/toxicidade , Animais , Proteínas de Ligação ao Cálcio/efeitos dos fármacos , Proteínas de Ligação ao Cálcio/metabolismo , Cardiomegalia/induzido quimicamente , Cardiomegalia/metabolismo , Modelos Animais de Doenças , Técnicas de Silenciamento de Genes , Coração/efeitos dos fármacos , Técnicas In Vitro , Camundongos , Mitocôndrias/efeitos dos fármacos , Proteínas de Transporte da Membrana Mitocondrial/efeitos dos fármacos , Proteínas de Transporte da Membrana Mitocondrial/metabolismo , Miocárdio/patologia , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/patologia , Estresse Oxidativo/efeitos dos fármacos , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/efeitos dos fármacos , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Vasoconstritores/toxicidade
5.
Aging (Albany NY) ; 12(10): 9726-9744, 2020 05 18.
Artigo em Inglês | MEDLINE | ID: mdl-32422606

RESUMO

Neurofibromin 2 (NF2), a potent tumor suppressor, is reported to inhibit proliferation in several cell types. The role of NF2 in neointima hyperplasia after vascular injury is unknown. We explored the role of NF2 in proliferation, migration of vascular smooth muscle cell (VSMC) and neointima hyperplasia after vascular injury. NF2 phosphorylation was elevated in VSMC subjected to platelet-derived growth factor (PDGF)-BB and in artery subjected to vascular injury. Mice deficient for Nf2 in VSMC showed enhanced neointima hyperplasia after injury, increased proliferation and migration of VSMC after PDGF-BB treatment. Mechanistically, we observed increased nuclear p-NF2, declined p-Yes-Associated Protein (YAP), nuclear translocation of YAP after PDGF-BB treatment or injury. NF2 knockdown or YAP overexpression showed similar phenotype in VSMC proliferation, migration and neointima hyperplasia. YAP inhibition abolished the above effects mediated by NF2 knockdown. Finally, NF2 knockdown further promoted YAP-TEA Domain Transcription Factor 1 (TEAD1) interaction after PDGF-BB treatment. Inhibition of TEAD1 blocked PDGF-BB-induced VSMC proliferation and migration, which were not reversed by either NF2 knockdown or YAP overexpression. In conclusion, NF2 knockdown promotes VSMC proliferation, migration and neointima hyperplasia after vascular injury via inducing YAP-TEAD1 interaction.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas de Ciclo Celular/metabolismo , Proteínas de Ligação a DNA/metabolismo , Neointima/genética , Neurofibromina 2/deficiência , Fatores de Transcrição/metabolismo , Remodelação Vascular/genética , Lesões do Sistema Vascular/genética , Animais , Proliferação de Células/genética , Hiperplasia/genética , Camundongos , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patologia , Miócitos de Músculo Liso/metabolismo , Miócitos de Músculo Liso/patologia , Fatores de Transcrição de Domínio TEA , Proteínas de Sinalização YAP
6.
Atherosclerosis ; 301: 44-53, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32325260

RESUMO

BACKGROUND AND AIMS: TRPA1 is a calcium permeable non-selective cation channel, its expression is up-regulated in atherosclerosis plaque, yet its function in macrophages activation is unknown. We sought to establish the role of TRPA1 in inflammatory macrophages activation. METHODS: TRPA1-/-ApoE-/- mice and C57BL/6 J control were treated with a high-fat diet (HFD) and the TRPA1 agonist cinnamaldehyde (CIN). Third-order branches of superior aorta of patients and mice were collected. Oil Red O staining and hematoxylin and eosin staining were performed to measure atherosclerotic lesions. The RNA-seq was performed to identify TRPA1 function in atherosclerosis. The expression of bone marrow-derived macrophages (BMDMs) markers was tested by Western blot. In addition, the levels of inflammatory factors were checked by ELISA. Chromatin immunoprecipitation (ChIP)-PCR and luciferase reporter gene assays were used to explore if TRPA1 could regulate histone modifications. RESULTS: TRPA1-/-ApoE-/- mice showed a significant increase in atherosclerosis plaques compared to ApoE-/- mice after HFD treatment. Conversely, activation of TRPA1 by CIN sharply reduced atherosclerosis progression. Atherosclerosis was associated with a significant change in macrophage polarization toward the M1 proinflammatory phenotype. We found that inhibition of TRPA1 remarkably stimulated M1 marker genes expression, while repressed M2 marker genes expression. The interaction between TRPA1 and Ezh2, a subunit of polycomb repressive complex 2, suppressed the proteasome-dependent degradation of Ezh2. Thus, TRPA1 epigenetically regulated H3K27 trimethylation level in macrophages. CONCLUSIONS: Our results demonstrate that TRPA1, up-regulated in atherosclerosis plaque, could regulate the macrophages toward an inflammatory phenotype, thereby modulating atherosclerosis progression. Activation of TRPA1 might serve as an atherosclerosis therapeutic target.


Assuntos
Aterosclerose , Placa Aterosclerótica , Animais , Apolipoproteínas E/genética , Aterosclerose/genética , Modelos Animais de Doenças , Humanos , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fenótipo , Canal de Cátion TRPA1/genética
7.
Oxid Med Cell Longev ; 2019: 8690746, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31772713

RESUMO

BACKGROUND: Hyper-free fatty acidemia (HFFA) impairs cardiac capillaries, as well as type 2 diabetes mellitus (T2DM). Perilipin 5 (Plin5) maintains metabolic balance of free fatty acids (FFAs) in high oxidative tissues via the states of nonphosphorylation and phosphorylation. However, when facing to T2DM-HFFA, Plin5's role in cardiac microvascular endothelial cells (CMECs) is not defined. METHODS: In mice of WT or Plin5-/-, T2DM models were rendered by high-fat diet combined with intraperitoneal injection of streptozocin. CMECs isolated from left ventricles were incubated with high glucose (HG) and high FFAs (HFFAs). Plin5 phosphorylation was stimulated by isoproterenol. Plin5 expression was knocked down by small interfering RNA (siRNA). We determined cardiac function by small animal ultrasound, apoptotic rate by flow cytometry, microvessel quantity by immunohistochemistry, microvascular integrity by scanning electron microscopy, intracellular FFAs by spectrophotometry, lipid droplets (LDs) by Nile red staining, mRNAs by quantitative real-time polymerase chain reaction, proteins by western blots, nitric oxide (NO) and reactive oxygen species (ROS) by fluorescent dye staining and enzyme-linked immunosorbent assay kits. RESULTS: In CMECs, HFFAs aggravated cell injury induced by HG and activated Plin5 expression. In mice with T2DM-HFFA, Plin5 deficiency reduced number of cardiac capillaries, worsened structural incompleteness, and enhanced diastolic dysfunction. Moreover, in CMECs treated with HG-HFFAs, both ablation and phosphorylation of Plin5 reduced LDs content, increased intracellular FFAs, stimulated mitochondrial ß-oxidation, added ROS generation, and reduced the expression and activity of endothelial nitric oxide synthase (eNOS), eventually leading to increased apoptotic rate and decreased NO content, all of which were reversed by N-acetyl-L-cysteine. CONCLUSION: Plin5 preserves lipid balance and cell survival in diabetic CMECs by regulating FFAs metabolism bidirectionally via the states of nonphosphorylation and phosphorylation.


Assuntos
CME-Carbodi-Imida/metabolismo , Diabetes Mellitus Tipo 2/tratamento farmacológico , Ácidos Graxos não Esterificados/metabolismo , Expressão Gênica/genética , Perilipina-5/uso terapêutico , Espécies Reativas de Oxigênio/metabolismo , Animais , Camundongos , Perilipina-5/farmacologia , Transfecção
8.
Toxicol Appl Pharmacol ; 383: 114747, 2019 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-31499192

RESUMO

Abnormal proliferation of vascular smooth muscle cells (VSMCs) is a hallmark of vascular restenosis. We investigated whether polypyrimidine tract-binding protein 1 (PTBP1), a novel regulator of cell proliferation and differentiation, is implicated in VSMC proliferation and neointima hyperplasia responding to injury. C57BL/6 J mice of 10-12 weeks old were randomly divided into sham and carotid artery injury group. Primary VSMCs obtained from thoracic aortas of 10- to 12-week-old mice were treated with physiological saline and platelet derived growth factor-BB (PDGF-BB). Adenovirus expressing shCon, shPTBP1 or shYY2 were transfected into the injured common carotid artery or VSMCs. qRT-PCR and immunoblotting were used to determine the mRNA and protein expression levels, respectively. Immunohistochemical staining of H&E and Ki-67 were used to evaluate restenosis of vessels. Cell counting kit-8 assay and Ki-67 immunofluorescent staining were utilized to evaluate the rate of VSMC proliferation. The expression of PTBP1 were upregulated both in injured arteries and in PDGF-BB-treated VSMCs. PTBP1 inhibition significantly attenuated neointima hyperplasia and Ki-67 positive area induced by injury. Knockdown of PTBP1 in vitro also suppressed VSMC proliferation after PDGF-BB treatment. The effects of PTBP1 inhibition mentioned above were all abolished by knockdown of YY2. Finally, we identified four cell cycle regulators (p53, p21, Cdkn1c, Cdkn2b) that were regulated by PTBP1/YY2 axis both in vitro and in vivo. These findings demonstrated that PTBP1 is a critical regulator of VSMC proliferation and neointima hyperplasia via modulating the expression of YY2.


Assuntos
Proliferação de Células/fisiologia , Ribonucleoproteínas Nucleares Heterogêneas/fisiologia , Hiperplasia/metabolismo , Músculo Liso Vascular/metabolismo , Neointima/metabolismo , Proteína de Ligação a Regiões Ricas em Polipirimidinas/fisiologia , Fatores de Transcrição/biossíntese , Animais , Becaplermina/farmacologia , Proliferação de Células/efeitos dos fármacos , Ribonucleoproteínas Nucleares Heterogêneas/antagonistas & inibidores , Hiperplasia/patologia , Camundongos , Camundongos Endogâmicos C57BL , Músculo Liso Vascular/efeitos dos fármacos , Músculo Liso Vascular/patologia , Neointima/patologia , Proteína de Ligação a Regiões Ricas em Polipirimidinas/antagonistas & inibidores , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia
9.
Hypertens Res ; 42(10): 1544-1551, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31133682

RESUMO

The cross-talk between skeletal muscle and adipose tissue has been identified to play a key role in the regulation of blood pressure and the development of hypertension. The role of different adipokines and myokines in hypertension and hypertension-related complications remains unclear. In the present study, 98 hypertensive patients and 24 normotensive controls were recruited, and additional subgroup analyses of hypertension-related complications were also performed. The levels of the circulating bone-derived factors leptin, apelin, fractalkine, brain-derived neurotrophic factor (BDNF), leukemia inhibitory factor (LIF), myostatin, fatty-acid-binding protein 3 (FABP3), irisin, follistatin-related protein 1 (FSTL1), oncostatin M, fibroblast growth factor 21 (FGF21) and musclin were measured by a protein liquid chip assay. The circulating levels of BDNF and musclin were decreased, whereas the leptin and irisin levels were increased, in hypertensive patients compared with those in the control individuals. Further logistic analysis indicated that the irisin level was positively correlated with SBP and an independent predictor for hypertension after adjustment. In nonobese subjects, the concentrations of DKK1, BDNF and FSTL1 were decreased, whereas the concentrations of leptin and irisin were increased. Irisin and DKK1 might be associated with hypertension. Additional subgroup analyses showed that irisin is significantly associated with hypertension-related stroke. In conclusion, we found that increased irisin levels are associated with hypertension and hypertension-related stroke. These findings indicate that irisin may be involved in the pathophysiology of hypertension.


Assuntos
Adipocinas/fisiologia , Fibronectinas/fisiologia , Hipertensão/etiologia , Adipocinas/sangue , Adulto , Idoso , Pressão Sanguínea , Fator Neurotrófico Derivado do Encéfalo/sangue , Estudos de Casos e Controles , Feminino , Fibronectinas/sangue , Humanos , Hipertensão/sangue , Hipertensão/complicações , Modelos Logísticos , Masculino , Pessoa de Meia-Idade , Proteínas Musculares/sangue , Estresse Oxidativo , Fatores de Transcrição/sangue
10.
J Hypertens ; 37(11): 2256-2268, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31136458

RESUMO

OBJECTIVES: Vascular smooth muscle cell (VSMC) proliferation is a crucial cause of vascular neointima hyperplasia and restenosis, thus limiting the long-term efficacy of percutaneous vascular intervention. We explored the role of wild-type p53-induced phosphatase 1 (Wip1), a potent regulator of tumorigenesis and atherosclerosis, in VSMC proliferation and neointima hyperplasia. METHODS AND RESULTS: Animal model of vascular restenosis was established in wild type C57BL/6J and VSMC-specific Tuberous Sclerosis 1 (TSC1)-knockdown mice by wire injury. We observed increased protein levels of Wip1, phospho (p)-S6 Ribosomal Protein (S6), p-4EBP1 but decreased p-adenosine 5'-monophosphate-activated protein kinase (AMPK)α both in carotid artery at day 28 after injury and in VSMCs after 48 h of platelet derived growth factor-BB (PDGF-BB) treatment. By using hematoxylin-eosin staining, Ki-67 immunohistochemical staining, cell counting kit-8 assay and Ki-67 immunofluorescence staining, we found Wip1 antagonist GSK2830371 (GSK) or mammalian target of rapamycin complex 1 (mTORC1) inhibitor rapamycin both obviously reversed the neointima formation and VSMC proliferation induced by wire injury and PDGF-BB, respectively. GSK also reversed the increase in mRNA level of Collagen I after wire injury. However, GSK had no obvious effects on VSMC migration induced by PDGF-BB. Simultaneously, TSC1 knockdown as well as AMPK inhibition by Compound C abolished the vascular protective and anti-proliferative effects of Wip1 inhibition. Additionally, suppression of AMPK also reversed the declined mTORC1 activity by GSK. CONCLUSION: Wip1 promotes VSMC proliferation and neointima hyperplasia after wire injury via affecting AMPK/mTORC1 pathway.


Assuntos
Aminopiridinas/uso terapêutico , Dipeptídeos/uso terapêutico , Miócitos de Músculo Liso/efeitos dos fármacos , Neointima/prevenção & controle , Proteína Fosfatase 2C/metabolismo , Lesões do Sistema Vascular/enzimologia , Proteínas Quinases Ativadas por AMP/metabolismo , Aminopiridinas/farmacologia , Animais , Becaplermina , Artéria Carótida Primitiva/patologia , Proliferação de Células/efeitos dos fármacos , Dipeptídeos/farmacologia , Modelos Animais de Doenças , Avaliação Pré-Clínica de Medicamentos , Hiperplasia , Masculino , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Músculo Liso Vascular , Neointima/etiologia , Proteína Fosfatase 2C/antagonistas & inibidores , Proteína Supressora de Tumor p53/metabolismo , Lesões do Sistema Vascular/complicações
11.
Cell Physiol Biochem ; 47(2): 817-826, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29807359

RESUMO

BACKGROUND/AIMS: Activation of stromal interaction molecule 1 (STIM1) and Orai1 participates in the development of cardiac hypertrophy. Store-operated Ca2+ entry-associated regulatory factor (SARAF) is an intrinsic inhibitor of STIM1-Orai1 interaction. Thus, we hypothesized that SARAF could prevent cardiac hypertrophy. METHODS: Male C57BL/6 mice, aged 8 weeks, were randomly divided into sham and abdominal aortic constriction surgery groups and were infected with lentiviruses expressing SARAF and GFP (Lenti-SARAF) or GFP alone (Lenti-GFP) via intramyocardial injection. At 4 weeks after aortic constriction, left ventricular structure and function were assessed by echocardiography and hemodynamic assays. The gene and protein expressions of SARAF, STIM1, and Orai1 were measured by quantitative PCR and Western blot, respectively. RESULTS: Gene and protein expressions of SARAF were significantly decreased, while STIM1 and Orai1 were increased in the heart tissue compared with sham group. Overexpression of SARAF in the heart prevented the upregulation of STIM1 and Orai1, and importantly, attenuated aortic constriction-induced decrease in maximal rate of left ventricular pressure decay and increases in thickness of interventricular septum and left ventricular posterior wall, heart weight/body weight ratio, and size of cardiomyocytes. Blood pressure detected through the carotid artery and left ventricular systolic function were not affected by SARAF overexpression. In addition, overexpression of SARAF also attenuated angiotensin II-induced upregulation of STIM1 and Orai1 and hypertrophy of cultured cardiomyocytes. CONCLUSION: Overexpression of SARAF in the heart prevents cardiac hypertrophy, probably through suppressing the upregulation of STIM1/Orai1.


Assuntos
Cardiomegalia/prevenção & controle , Proteínas de Membrana/metabolismo , Proteína ORAI1/metabolismo , Molécula 1 de Interação Estromal/metabolismo , Angiotensina II/farmacologia , Animais , Aorta Abdominal/cirurgia , Fator Natriurético Atrial/genética , Fator Natriurético Atrial/metabolismo , Cardiomegalia/metabolismo , Cardiomegalia/patologia , Linhagem Celular , Ecocardiografia , Masculino , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Miocárdio/metabolismo , Miocárdio/patologia , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Cadeias Pesadas de Miosina/genética , Cadeias Pesadas de Miosina/metabolismo , Peptídeo Natriurético Encefálico/genética , Peptídeo Natriurético Encefálico/metabolismo , Proteína ORAI1/genética , Pressão , Molécula 1 de Interação Estromal/genética , Regulação para Cima/efeitos dos fármacos , Função Ventricular Esquerda/fisiologia
12.
J Am Heart Assoc ; 6(10)2017 Oct 12.
Artigo em Inglês | MEDLINE | ID: mdl-29025747

RESUMO

BACKGROUND: Intracoronary stent restenosis, characterized by excessive smooth muscle cell (SMC) proliferation and myointimal hyperplasia, remains a clinical challenge. Mitochondrial membrane potential has been linked to the proliferative rate of SMCs. This study aimed to screen a critical gene regulating mitochondrial potential and to confirm its effects on myointimal formation in preclinical animal models. METHODS AND RESULTS: We performed transcriptome screening for genes differentially expressed in ligated versus unligated mouse carotid arteries. We observed that uncoupling protein 2 gene (Ucp2) mRNA, encoding UCP2, was transiently upregulated during the first 3 days after ligation and then significantly downregulated from day 7 through day 21, during which time neointima formed remarkably. The UCP2 protein level also declined after day 7 of ligation. In ligated carotid arteries, Ucp2-/- mice, compared with wild-type littermates, exhibited accelerated myointimal formation, which was associated with increased superoxide production and can be attenuated by treatment with antioxidant 4-hydroxy-2,2,6,6-tetramethyl-piperidinoxyl (TEMPOL). Knockdown of UCP2 enhanced human aortic SMC migration and proliferation that can also be attenuated by TEMPOL, whereas UCP2 overexpression inhibited SMC migration and proliferation, along with decreased activity of nuclear factor-κB. Moreover, nuclear factor-κB inhibitor attenuated UCP2 knockdown-enhanced SMC proliferation. Adenovirus-mediated overexpression of UCP2 inhibited myointimal formation in balloon-injured carotid arteries of rats and rabbits and in-stent stenosis of porcine coronary arteries. Moreover, UCP2 overexpression also suppressed neointimal hyperplasia in cultured human saphenous vein ex vivo. CONCLUSIONS: UCP2 inhibits myointimal hyperplasia after vascular injury, probably through suppressing nuclear factor-κB-dependent SMC proliferation and migration, rendering UCP2 a potential therapeutic target against restenosis.


Assuntos
Lesões das Artérias Carótidas/prevenção & controle , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Neointima , Proteína Desacopladora 2/metabolismo , Animais , Artérias Carótidas/metabolismo , Artérias Carótidas/patologia , Lesões das Artérias Carótidas/genética , Lesões das Artérias Carótidas/metabolismo , Lesões das Artérias Carótidas/patologia , Movimento Celular , Proliferação de Células , Células Cultivadas , Modelos Animais de Doenças , Predisposição Genética para Doença , Humanos , Hiperplasia , Masculino , Potencial da Membrana Mitocondrial , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mitocôndrias Musculares/metabolismo , Mitocôndrias Musculares/patologia , Músculo Liso Vascular/lesões , Músculo Liso Vascular/patologia , Miócitos de Músculo Liso/patologia , NF-kappa B/metabolismo , Fenótipo , Coelhos , Ratos Sprague-Dawley , Suínos , Porco Miniatura , Fatores de Tempo , Técnicas de Cultura de Tecidos , Proteína Desacopladora 2/deficiência , Proteína Desacopladora 2/genética
13.
J Vasc Surg ; 65(1): 197-205.e2, 2017 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26947234

RESUMO

OBJECTIVE: Transient receptor potential vanilloid 1 (TRPV1) is an important regulator of endothelial function, but the effects of TRPV1 on endothelial recovery and neointimal formation after vascular injury remain elusive. We tested the effects of activating TRPV1 using capsaicin on re-endothelialization and neointimal formation after wire-induced injury of the carotid artery in mice. METHODS: The human umbilical vein endothelial cells (HUVECs) were treated with the TRPV1 agonist capsaicin, its antagonist capsazepine, intracellular calcium chelator BAPTA, or mitofusin 2 (Mfn2)-specific short interfering RNA (siRNA). The migration, proliferation, mitochondrial morphology, membrane potential, and adenosine triphosphate production were measured. The carotid artery wire injury procedure was performed in male TRPV1 knockout mice and C57BL/6J wild-type (WT) mice that were then treated with or without Mfn2 siRNA. The re-endothelialization and neointimal formation were evaluated. RESULTS: Capsaicin significantly enhanced the migration and proliferation of HUVECs. Both capsazepine and BAPTA abolished capsaicin-induced migration and proliferation of HUVECs. In addition, capsaicin stimulated the formation of reticular mitochondria, augmented mitochondrial membrane potential, increased adenosine triphosphate production, and upregulated Mfn2. However, these effects were attenuated by knockdown of Mfn2 with specific siRNA. Dietary capsaicin markedly accelerated re-endothelialization and inhibited neointimal formation in WT mice but not in TRPV1 knockout mice. Moreover, Mfn2 siRNA also attenuated capsaicin-induced enhancement of endothelial recovery and suppression of neointimal hyperplasia in WT mice. CONCLUSIONS: Activation of TRPV1 with capsaicin attenuates neointimal formation by accelerating re-endothelialization through upregulation of Mfn2.


Assuntos
Capsaicina/farmacologia , Lesões das Artérias Carótidas/tratamento farmacológico , Células Endoteliais/efeitos dos fármacos , Neointima , Reepitelização/efeitos dos fármacos , Canais de Cátion TRPV/agonistas , Canais de Cátion TRPV/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Quelantes de Cálcio/farmacologia , Capsaicina/análogos & derivados , Lesões das Artérias Carótidas/metabolismo , Lesões das Artérias Carótidas/patologia , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Modelos Animais de Doenças , Ácido Egtázico/análogos & derivados , Ácido Egtázico/farmacologia , Células Endoteliais/metabolismo , Células Endoteliais/patologia , GTP Fosfo-Hidrolases/genética , GTP Fosfo-Hidrolases/metabolismo , Células Endoteliais da Veia Umbilical Humana , Humanos , Hiperplasia , Masculino , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Mitocondriais/genética , Proteínas Mitocondriais/metabolismo , Interferência de RNA , Transdução de Sinais/efeitos dos fármacos , Canais de Cátion TRPV/deficiência , Canais de Cátion TRPV/genética , Transfecção
14.
Sci Rep ; 6: 29975, 2016 07 25.
Artigo em Inglês | MEDLINE | ID: mdl-27453531

RESUMO

Restenosis limits the efficacy of vascular percutaneous intervention, in which vascular smooth muscle cell (VSMC) proliferation and activation of inflammation are two primary causal factors. Calpains influence VSMC proliferation and collagen synthesis. However, the roles of calpastatin and calpains in vascular restenosis remain unclear. Here, restenosis was induced by ligating the left carotid artery, and VSMCs were pretreated with platelet-derived growth factor (PDGF)-BB. Adenovirus vector carrying MMP2 sequence and specific small interfering RNA against calpain-1/2 were introduced. Finally, restenosis enhanced the expression of calpain-1/2, but reduced calpastatin content. In calpastatin transgenic mice, lumen narrowing was attenuated gradually and peaked on days 14-21. Cell proliferation and migration as well as collagen synthesis were inhibited in transgenic mice, and expression of calpain-1/2 and MMP2/transforming growth factor-ß1 (TGF-ß1). Consistently, in VSMCs pretreated with PDGF-BB, calpastatin induction and calpains inhibition suppressed the proliferation and migration of VSMCs and collagen synthesis, and reduced expression of calpain-1/2 and MMP2/TGF-ß1. Moreover, simvastatin improved restenosis indicators by suppressing the HIF-1α/calpains/MMP2/TGF-ß1 pathway. However, MMP2 supplementation eliminated the vascular protection of calpastatin induction and simvastatin. Collectively, calpains inhibition plays crucial roles in vascular restenosis by preventing neointimal hyperplasia at the early stage via suppression of the MMP2/TGF-ß1 pathway.


Assuntos
Calpaína/antagonistas & inibidores , Reestenose Coronária/metabolismo , Reestenose Coronária/patologia , Metaloproteinase 2 da Matriz/metabolismo , Transdução de Sinais , Fator de Crescimento Transformador beta1/metabolismo , Animais , Proteínas de Ligação ao Cálcio/metabolismo , Calpaína/metabolismo , Artérias Carótidas/patologia , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Colágeno/biossíntese , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Modelos Biológicos , Músculo Liso Vascular/patologia , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/metabolismo , Miócitos de Músculo Liso/patologia , Fator de Crescimento Derivado de Plaquetas/farmacologia , RNA Interferente Pequeno/metabolismo , Transdução de Sinais/efeitos dos fármacos , Sinvastatina/farmacologia
15.
Int J Mol Med ; 38(2): 601-9, 2016 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-27314441

RESUMO

Transient receptor potential vanilloid subtype 1 (TRPV1) is a non-selective cation channel with high permeability to Ca2+. Intracellular Ca2+ signaling is an essential regulator of endothelial nitric oxide (NO) synthase (eNOS) that plays a beneficial role in myocardial fibrosis. The aim of the present study was to determine the role of TRPV1 in isoproterenol-induced myocardial fibrosis. Transgenic mice overexpressing TRPV1 were generated on a C57BL/6J genetic background. An animal model of myocardial fibrosis was created by subcutaneously injecting the mice with isoproterenol. We found that the wild-type mice exhibited a significant increase in heart/body weight ratio, left ventricle/body weight ratio, left ventricular end-diastolic pressure (LVEDP), the cardiac fibrotic lesion area and collagen content, as well as a marked decrease in eNOS phosphorylation and NO/cyclic guanosine monophosphate (cGMP) levels at 2 weeks after the administration of isoproterenol (all p<0.01). However, these changes were significantly attenuated in the TRPV1 transgenic mice (p<0.05 or p<0.01). Moreover, the beneficial effects on myocardial fibrosis exerted by the overexpression of TRPV1 were attenuated by the administration of the eNOS inhibitor, Nω-nitro-L-arginine methyl ester (L-NAME) (all p<0.05). Similar anti-fibrotic effects were observed in in vitro experiments with primary cultured cardiac fibroblasts. The findings of our study suggest that TRPV1 overexpression attenuates isoproterenol­induced myocardial fibrosis.


Assuntos
Miocárdio/metabolismo , Miocárdio/patologia , Canais de Cátion TRPV/metabolismo , Animais , GMP Cíclico/metabolismo , Fibroblastos/metabolismo , Fibroblastos/patologia , Fibrose , Hemodinâmica , Isoproterenol , Masculino , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo III/metabolismo , Tamanho do Órgão , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais , Função Ventricular
16.
Medicine (Baltimore) ; 95(16): e3442, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-27100441

RESUMO

Arrhythmogenic right ventricular cardiomyopathy (ARVC) is a crucial health problem. With sudden death often being the first presentation, early diagnosis for ARVC is essential. Up to date, electrocardiogram (ECG) is a widely used diagnostic method without invasive harms. To diagnose and treat ARVC as well as possible, we should clearly elucidate its pathophysiological alterations. A 66-year-old farmer presented to the Emergency Department with continuous palpitation, chest tightness, profuse sweating, and nausea with no obvious predisposing causes. An ECG indicated ventricular tachycardia (VT). The patient experienced a sudden drop in blood pressure and acute confusion. After an immediate electrical conversion, his consciousness was gradually restored, and symptoms relieved. The patient was then transferred to the Department of Cardiology to receive ECG, echocardiography, coronary angiogram, biochemical assays, endocardiac tracing, and radiofrequency ablation. In the end, he was diagnosed with ARVC, evidenced by bilateral ventricle dilation and epsilon waves in leads V1-V3. Appropriate therapies were provided for this patient including pharmacological intervention and radiofrequency ablation. Although the diagnosis of ARVC is not difficult, this patient's ECG manifested several interesting features and should be further investigated: T wave inversions were found extensively in the anterior and inferior leads, revealing the involvement of bilateral ventricles; VTs with different morphologies and cycle lengths were found, and some VTs manifested the feature of irregularly irregular rhythm, reminding us to carefully differentiate some special VTs from atrial fibrillation (AF); and epsilon waves gradually appeared in leads V1-V3 and avR since the onset of ARVC. Most importantly, the epsilon waves behind QRS complex appeared in both sinus rhythm and ventricular premature beats/VT originating from cardiac apex, whereas the epsilon waves prior to QRS complex occurred in VT originating from right ventricular outflow tract (RVOT). The features of T wave inversion and epsilon wave in ECGs and the appearance of VTs with different morphologies can reflect the progression of ARVC. The position relationship between epsilon wave and QRS complex in VT depends on ventricular activation sequence, that is, the localization of epsilon wave depends on where VT is originating from.


Assuntos
Displasia Arritmogênica Ventricular Direita/fisiopatologia , Eletrocardiografia , Taquicardia Ventricular/fisiopatologia , Função Ventricular Direita/fisiologia , Idoso , Displasia Arritmogênica Ventricular Direita/complicações , Displasia Arritmogênica Ventricular Direita/diagnóstico , Diagnóstico Diferencial , Progressão da Doença , Ecocardiografia , Ventrículos do Coração/fisiopatologia , Humanos , Masculino , Taquicardia Ventricular/diagnóstico , Taquicardia Ventricular/etiologia
17.
Atherosclerosis ; 243(2): 589-92, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26545012

RESUMO

BACKGROUND: The goal of this study was to establish an apolipoprotein E-deficient (ApoE(-/-)) rat model. METHODS: The ApoE(-/-) rat was created by TALEN-mediated gene targeting in the genetic background of Sprague Dawley rat. Six-to eight-week-old male rats were used in the experiments (n = 10 in each group). RESULTS: After fed with high-cholesterol diet (HCD) for 12 weeks, the ApoE(-/-) rats displayed typical dyslipidemia. In contrast, HCD failed to induce hypercholesterolemia in wild-type rats. However, there was no obvious atherosclerotic lesion in oil red O-stained en face aortas and the aortic root sections in both genetic types of rats. Interestingly, partial ligation caused the formation of plaques consist of lipid and macrophages in carotid arteries from ApoE(-/-) rats, but induced the neointimal hyperplasia in wild-type rats. Additionally, we found that HCD slightly increased the expression of adhesion molecules, while partial ligation dramatically upregulated these molecules. CONCLUSIONS: The ApoE(-/-) rat is a novel model for dyslipidemia and could also be used in the research of atherosclerosis.


Assuntos
Apolipoproteínas E/deficiência , Artérias Carótidas/patologia , Doenças das Artérias Carótidas/patologia , Hipercolesterolemia/patologia , Placa Aterosclerótica , Animais , Apolipoproteínas E/genética , Artérias Carótidas/metabolismo , Artérias Carótidas/cirurgia , Doenças das Artérias Carótidas/sangue , Doenças das Artérias Carótidas/genética , Moléculas de Adesão Celular/sangue , Colesterol na Dieta , Dieta Hiperlipídica , Modelos Animais de Doenças , Predisposição Genética para Doença , Hipercolesterolemia/sangue , Hipercolesterolemia/genética , Hiperplasia , Mediadores da Inflamação/sangue , Ligadura , Lipídeos/sangue , Macrófagos/metabolismo , Macrófagos/patologia , Masculino , Neointima , Fenótipo , Ratos Sprague-Dawley , Ratos Transgênicos
18.
Pacing Clin Electrophysiol ; 38(10): 1201-9, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26172535

RESUMO

BACKGROUND: Left ventricular (LV) pacing in cardiac resynchronization therapy (CRT) can be achieved via a transvenous or epicardial route. A surgically implanted epicardial LV (eLV) lead is used after a standard transvenous LV (tLV) lead implantation has failed. However, studies of clinical outcomes in patients with eLV leads and comparisons of outcome between tLV and eLV-CRT are sparse. Therefore, the purpose of this study is to compare clinical response between tLV-CRT and eLV-CRT, as well as to understand the differences within the eLV-CRT population. METHODS: Forty-four patients received eLV-CRT following unsuccessful attempts of tLV-CRT implantation between 2002 and 2013 at the University of California, San Diego (UCSD) and Mayo Clinics. These patients were matched for age, gender, and etiology of cardiomyopathy in a 1:2 ratio with a cohort of patients who received tLV-CRT during the same time period. RESULTS: During a mean follow-up of 57 months, similar clinical outcomes and survival rate were noted between tLV and eLV-CRT patients (all P > 0.05). Within the eLV-CRT group, dilated cardiomyopathy patients had significant improvement in New York Heart Association class and ejection fraction (both P < 0.05), while ischemic cardiomyopathy patients did not (both P > 0.05). eLV-CRT patients with nonanterior lead location had significantly improved survival (P < 0.001). There was also a trend for improved survival in those with nonapical lead location (P = 0.09). CONCLUSION: In this case-matched two-centered study, comparable improvements were noted in patients with tLV-CRT and eLV-CRT. Operators should target nonanterior and nonapical locations during eLV-CRT implantation. Use of eLV-CRT should be considered a viable alternative for CRT candidates.


Assuntos
Terapia de Ressincronização Cardíaca/mortalidade , Terapia de Ressincronização Cardíaca/métodos , Cardiomiopatias/mortalidade , Cardiomiopatias/prevenção & controle , Insuficiência Cardíaca/mortalidade , Insuficiência Cardíaca/prevenção & controle , Distribuição por Idade , Idoso de 80 Anos ou mais , California/epidemiologia , Comorbidade , Eletrodos , Feminino , Seguimentos , Humanos , Estudos Longitudinais , Masculino , Minnesota/epidemiologia , Estudos Retrospectivos , Distribuição por Sexo , Taxa de Sobrevida , Resultado do Tratamento
19.
Free Radic Biol Med ; 82: 114-21, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25680284

RESUMO

TNF-α inhibitor reportedly protects against myocardial ischemia/reperfusion (MI/R) injury. It can also increase Notch1 expression in inflammatory bowel disease, revealing the regulation of Notch1 signaling by TNF-α inhibitor. However, the interaction between TNF-α inhibitor and Notch1 signaling in MI/R remains unclear. This study aimed to determine the involvement of TNF-α inhibitor with Notch1 in MI/R and delineate the related mechanism. Notch1-specific small interfering RNA (20 µg) or Jagged1 (a Notch ligand, 12 µg) was delivered through intramyocardial injection. Forty-eight hours after injection, mice received 30 min of myocardial ischemia followed by 3 h (for cell apoptosis and oxidative/nitrative stress) or 24h (for infarct size and cardiac function) of reperfusion. Ten minutes before reperfusion, mice randomly received an intraperitoneal injection of vehicle, etanercept, diphenyleneiodonium, 1400W, or EUK134. Finally, downregulation of Notch1 significantly reversed the alleviation of MI/R injury induced by etanercept, as evidenced by enlarged myocardial infarct size, suppressed cardiac function, and increased myocardial apoptosis. Moreover, Notch1 blockade increased the expression of inducible NO synthase (iNOS) and gp(91)(phox), enhanced NO and superoxide production, and accelerated their cytotoxic reaction product, peroxynitrite. Furthermore, NADPH inhibition with diphenyleneiodonium or iNOS suppression with 1400W mitigated the aggravation of MI/R injury induced by Notch1 downregulation in mice treated with etanercept. Additionally, either Notch1 activation with Jagged1 or peroxynitrite decomposition with EUK134 reduced nitrotyrosine content and attenuated MI/R injury. These data indicate that MI/R injury can be attenuated by TNF-α inhibitor, partly via Notch1 signaling-mediated suppression of oxidative/nitrative stress.


Assuntos
Infarto do Miocárdio/tratamento farmacológico , Traumatismo por Reperfusão Miocárdica/tratamento farmacológico , Estresse Oxidativo/fisiologia , Receptor Notch1/genética , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Animais , Proteínas de Ligação ao Cálcio/genética , Modelos Animais de Doenças , Regulação para Baixo , Ativação Enzimática , Etanercepte , Peptídeos e Proteínas de Sinalização Intercelular/genética , Proteína Jagged-1 , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Infarto do Miocárdio/induzido quimicamente , Infarto do Miocárdio/patologia , Traumatismo por Reperfusão Miocárdica/induzido quimicamente , Traumatismo por Reperfusão Miocárdica/patologia , Miocárdio/patologia , NADP/antagonistas & inibidores , Óxido Nítrico Sintase Tipo II/antagonistas & inibidores , Óxido Nítrico Sintase Tipo II/biossíntese , Oniocompostos/farmacologia , Compostos Organometálicos/farmacologia , Ácido Peroxinitroso/biossíntese , Interferência de RNA , RNA Interferente Pequeno , Espécies Reativas de Nitrogênio/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Receptor Notch1/biossíntese , Receptor Notch1/metabolismo , Salicilatos/farmacologia , Proteínas Serrate-Jagged
20.
Am J Hypertens ; 27(12): 1521-9, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24858305

RESUMO

BACKGROUND: Dietary capsaicin plays a protective role in hypertension, atherosclerosis, obesity, and hyperlipidemia through activating the transient receptor potential vanilloid type 1 (TRPV1), a nonselective cation channel. This study was designed to investigate the role of capsaicin in cardiac hypertrophy and fibrosis in a pressure overload model. METHODS: TRPV1 knockout (KO) mice and their wild-type (WT) littermates, aged 8 weeks, were randomly divided into sham and aortic banding surgery groups and were fed with chow or chow plus capsaicin for 10 weeks. RESULTS: Dietary capsaicin significantly attenuates pressure overload-induced increase in heart weight index, enlargement of ventricular volume, decrease in cardiac function, and increase in cardiac fibrosis in WT mice. However, these effects of capsaicin were absent in TRPV1 KO mice. Additionally, capsaicin blunted pressure overload-induced upregulation of transforming growth factor ß, connective tissue growth factor, and the phosphorylation of Smad2/3 in WT mice but not in TRPV1 KO mice. Moreover, capsaicin attenuated pressure overload-induced overexpression of metalloproteinase (MMP)-2, MMP-9 and MMP-13 in WT mice but not in TRPV1 KO mice. Capsaicin also attenuated angiotensin II-induced proliferation of cardiac fibroblasts from mice with the TRPV1 channel. CONCLUSIONS: Our results suggest that dietary capsaicin protects against cardiac hypertrophy and fibrosis in pressure overload mice through TRPV1.


Assuntos
Pressão Sanguínea/efeitos dos fármacos , Capsaicina/administração & dosagem , Cardiomegalia/dietoterapia , Cardiomiopatias/dietoterapia , Suplementos Nutricionais , Hipertensão/dietoterapia , Canais de Cátion TRPV/biossíntese , Animais , Western Blotting , Cardiomegalia/etiologia , Cardiomegalia/fisiopatologia , Cardiomiopatias/etiologia , Cardiomiopatias/fisiopatologia , Proliferação de Células , Modelos Animais de Doenças , Fibrose , Citometria de Fluxo , Hipertensão/complicações , Hipertensão/fisiopatologia , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Knockout , Miocárdio/patologia , Transdução de Sinais/efeitos dos fármacos , Canais de Cátion TRPV/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA