Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Sci China Life Sci ; 67(3): 475-487, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-37219765

RESUMO

Cardiopulmonary bypass has been speculated to elicit systemic inflammation to initiate acute lung injury (ALI), including acute respiratory distress syndrome (ARDS), in patients after cardiac surgery. We previously found that post-operative patients showed an increase in endothelial cell-derived extracellular vesicles (eEVs) with components of coagulation and acute inflammatory responses. However, the mechanism underlying the onset of ALI owing to the release of eEVs after cardiopulmonary bypass, remains unclear. Plasma plasminogen-activated inhibitor-1 (PAI-1) and eEV levels were measured in patients with cardiopulmonary bypass. Endothelial cells and mice (C57BL/6, Toll-like receptor 4 knockout (TLR4-/-) and inducible nitric oxide synthase knockout (iNOS-/-)) were challenged with eEVs isolated from PAI-1-stimulated endothelial cells. Plasma PAI-1 and eEVs were remarkably enhanced after cardiopulmonary bypass. Plasma PAI-1 elevation was positively correlated with the increase in eEVs. The increase in plasma PAI-1 and eEV levels was associated with post-operative ARDS. The eEVs derived from PAI-1-stimulated endothelial cells could recognize TLR4 to stimulate a downstream signaling cascade identified as the Janus kinase 2/3 (JAK2/3)-signal transducer and activator of transcription 3 (STAT3)-interferon regulatory factor 1 (IRF-1) pathway, along with iNOS induction, and cytokine/chemokine production in vascular endothelial cells and C57BL/6 mice, ultimately contributing to ALI. ALI could be attenuated by JAK2/3 or STAT3 inhibitors (AG490 or S3I-201, respectively), and was relieved in TLR4-/- and iNOS-/- mice. eEVs activate the TLR4/JAK3/STAT3/IRF-1 signaling pathway to induce ALI/ARDS by delivering follistatin-like protein 1 (FSTL1), and FSTL1 knockdown in eEVs alleviates eEV-induced ALI/ARDS. Our data thus demonstrate that cardiopulmonary bypass may increase plasma PAI-1 levels to induce FSTL1-enriched eEVs, which target the TLR4-mediated JAK2/3/STAT3/IRF-1 signaling cascade and form a positive feedback loop, leading to ALI/ARDS after cardiac surgery. Our findings provide new insight into the molecular mechanisms and therapeutic targets for ALI/ARDS after cardiac surgery.


Assuntos
Lesão Pulmonar Aguda , Vesículas Extracelulares , Proteínas Relacionadas à Folistatina , Síndrome do Desconforto Respiratório , Animais , Humanos , Camundongos , Lesão Pulmonar Aguda/etiologia , Lesão Pulmonar Aguda/tratamento farmacológico , Lesão Pulmonar Aguda/metabolismo , Células Endoteliais/metabolismo , Vesículas Extracelulares/metabolismo , Proteínas Relacionadas à Folistatina/metabolismo , Proteínas Relacionadas à Folistatina/uso terapêutico , Inflamação/metabolismo , Lipopolissacarídeos/farmacologia , Pulmão/metabolismo , Camundongos Endogâmicos C57BL , Inibidor 1 de Ativador de Plasminogênio/metabolismo , Inibidor 1 de Ativador de Plasminogênio/uso terapêutico , Síndrome do Desconforto Respiratório/etiologia , Receptor 4 Toll-Like/metabolismo , Receptor 4 Toll-Like/uso terapêutico
2.
Cardiovasc Ther ; 2023: 5559368, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38024103

RESUMO

Objective: We demonstrated that circulating microparticles (MPs) are increased in patients with coronary heart disease (both chronic coronary syndrome (CCS) and acute coronary syndrome). Whether thrombolysis affects MPs in patients with ST-segment elevation myocardial infarction (STEMI) with or without percutaneous coronary intervention (PCI) is unknown. Methods: This study was divided into three groups: STEMI patients with thrombolysis (n = 18) were group T, patients with chronic coronary syndrome (n = 20) were group CCS, and healthy volunteers (n = 20) were the control group. Fasting venous blood was extracted from patients in the CCS and control groups, and venous blood was extracted from patients in the T group before (pre-T) and 2 hours after (post-T) thrombolysis. MPs from each group were obtained by centrifugation. After determining the concentration, the effects of MPs on endothelial nitric oxide synthase (eNOS) and inducible nitric oxide synthase (iNOS) in rat myocardial tissue in vitro were detected by immunohistochemistry and western blotting. Changes in nitric oxide (NO) and oxygen free radicals (O2•-) were also detected. The effect of MPs on vasodilation in isolated rat thoracic aortae was detected. Results: Compared with that in the control group (2.60 ± 0.38 mg/ml), the concentration of MPs was increased in patients with CCS (3.49 ± 0.72 mg/ml) and in STEMI patients before thrombolysis (4.17 ± 0.58 mg/ml). However, thrombolysis did not further increase MP levels (post-T, 4.23 ± 1.01 mg/ml) compared with those in STEMI patients before thrombolysis. Compared with those in the control group, MPs in both CCS and STEMI patients before thrombolysis inhibited the expression of eNOS (both immunohistochemistry and western blot analysis of phosphorylation at Ser1177), NO production in the isolated myocardium and vasodilation in vitro and stimulated the expression of iNOS (immunohistochemistry and western blot analysis of phosphorylation at Thr495), and the generation of O2•- in the isolated myocardium. The effects of MPs were further enhanced by MPs from STEMI patients 2 hours after thrombolysis. Conclusion: Changes in MP function after thrombolysis may be one of the mechanisms leading to ischemia-reperfusion after thrombolysis.


Assuntos
Síndrome Coronariana Aguda , Intervenção Coronária Percutânea , Infarto do Miocárdio com Supradesnível do Segmento ST , Humanos , Ratos , Animais , Infarto do Miocárdio com Supradesnível do Segmento ST/diagnóstico , Infarto do Miocárdio com Supradesnível do Segmento ST/tratamento farmacológico , Intervenção Coronária Percutânea/efeitos adversos , Terapia Trombolítica , Resultado do Tratamento
3.
Cardiovasc Ther ; 2022: 1057772, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36072560

RESUMO

Objective: High-density lipoprotein (HDL) was found vasoprotective, but numbers of patients with acute myocardial infarction (AMI) have normal or even high levels of pathological HDL (pHDL). So, we investigate the mechanism of pHDL in AMI patients on angiogenesis. Methods: HDL with normal levels from healthy subjects (nHDL, control group, n = 20) and patients with AMI (pHDL, experimental groups, n = 30) were obtained by super high speed centrifugation. Then, effects of HDL on proliferation, migration, angiogenesis, and expression of ERK1/2 and its phosphorylation in human umbilical vein endothelial cells (HUVEC) with or without PD98059 (inhibitor of ERK1/2) preincubation were detected. Results: Compared with the control group (nHDL), HDL from the experimental group (pHDL) significantly inhibited the phosphorylation of ERK1/2, proliferation, migration, and angiogenesis of HUVEC (P < 0.05), while these effects of HDL could substantially be blocked by preincubation of PD98059 (P < 0.05). Conclusion: HDL in AMI patients affects angiogenesis by inhibiting ERK1/2 activation free from HDL levels.


Assuntos
Lipoproteínas HDL , Infarto do Miocárdio , Células Endoteliais da Veia Umbilical Humana , Humanos , Lipoproteínas HDL/metabolismo , Sistema de Sinalização das MAP Quinases , Infarto do Miocárdio/metabolismo , Fosforilação
4.
Mol Med Rep ; 23(6)2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-33899122

RESUMO

As a common factor of both type 2 diabetes mellitus (T2DM) and acute coronary syndrome (ACS), circulating microparticles (MPs) may provide a link between these two diseases. The present study compared the content and function of MPs from patients with ACS with or without T2DM. MPs from healthy subjects (n=20), patients with ACS (n=24), patients with T2DM (n=20) and patients with combined ACS and T2DM (n=24) were obtained. After incubating rat thoracic tissue with MPs, the effect of MPs on endothelial­dependent vasodilatation, expression of caveolin­1 and endothelial nitric oxide synthase (eNOS), phosphorylation of eNOS at the S1177 and T495 sites and its association with heat shock protein 90 (Hsp90), and the generation of NO and superoxide anion (O2˙­) were determined. MP concentrations were higher in patients with T2DM and patients with ACS with or without T2DM than in healthy subjects. Moreover, MPs from patients with T2DM or ACS led to impairment in endothelial­dependent vasodilatation, decreased expression of NO, as well as eNOS and its phosphorylation at Ser1177 and association with Hsp90, but increased eNOS phosphorylation at T495, caveolin­1 expression and O2˙­ generation. These effects were strengthened by MPs from patients with ACS combined with T2DM. T2DM not only increased MP content but also resulted in greater vascular impairment effects in ACS. These results may provide novel insight into the treatment of patients with ACS and T2DM.


Assuntos
Síndrome Coronariana Aguda/sangue , Micropartículas Derivadas de Células/metabolismo , Diabetes Mellitus Tipo 2/sangue , Angiopatias Diabéticas/sangue , Síndrome Coronariana Aguda/complicações , Síndrome Coronariana Aguda/patologia , Adulto , Animais , Caveolina 1/metabolismo , Células Cultivadas , Diabetes Mellitus Tipo 2/patologia , Angiopatias Diabéticas/patologia , Endotélio Vascular/metabolismo , Endotélio Vascular/patologia , Endotélio Vascular/fisiopatologia , Feminino , Proteínas de Choque Térmico HSP90/metabolismo , Humanos , Masculino , Pessoa de Meia-Idade , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo III/metabolismo , Ratos , Vasodilatação
5.
Redox Biol ; 36: 101642, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32863238

RESUMO

Normal high-density lipoprotein (nHDL) in normal, healthy subjects is able to promote angiogenesis, but the mechanism remains incompletely understood. HDL from patients with coronary artery disease may undergo a variety of oxidative modifications, rendering it dysfunctional; whether the angiogenic effect is mitigated by such dysfunctional HDL (dHDL) is unknown. We hypothesized that dHDL compromises angiogenesis. The angiogenic effects of nHDL and dHDL were assessed using endothelial cell culture, endothelial sprouts from cardiac tissue from C57BL/6 mice, zebrafish model for vascular growth and a model of impaired vascular growth in hypercholesterolemic low-density lipoprotein receptor null(LDLr-/-)mice. MiRNA microarray and proteomic analyses were used to determine the mechanisms. Lipid hydroperoxides were greater in dHDL than in nHDL. While nHDL stimulated angiogenesis, dHDL attenuated these responses. Protein and miRNA profiles in endothelial cells differed between nHDL and dHDL treatments. Moreover, nHDL suppressed miR-24-3p expression to increase vinculin expression resulting in nitric oxide (NO) production, whereas dHDL delivered miR-24-3p to inhibit vinculin expression leading to superoxide anion (O2•-) generation via scavenger receptor class B type 1. Vinculin was required for endothelial nitric oxide synthase (eNOS) expression and activation and modulated the PI3K/AKT/eNOS and ERK1/2 signaling pathways to regulate nHDL- and VEGF-induced angiogenesis. Vinculin overexpression or miR-24-3p inhibition reversed dHDL-impaired angiogenesis. The expressions of vinculin and eNOS and angiogenesis were decreased, but the expression of miR-24-3p and lipid hydroperoxides in HDL were increased in the ischemic lower limbs of hypercholesterolemic LDLr-/- mice. Overexpression of vinculin or miR-24-3p antagomir restored the impaired-angiogenesis in ischemic hypercholesterolemic LDLr-/- mice. Collectively, nHDL stimulated vinculin and eNOS expression to increase NO production by suppressing miR-24-3p to induce angiogenesis, whereas dHDL inhibited vinculin and eNOS expression to enhance O2•- generation by delivering miR-24-3p to impair angiogenesis, and that vinculin and miR-24-3p may be therapeutic targets for dHDL-impaired angiogenesis.


Assuntos
Doença da Artéria Coronariana , MicroRNAs , Animais , Doença da Artéria Coronariana/tratamento farmacológico , Doença da Artéria Coronariana/genética , Células Endoteliais , Voluntários Saudáveis , Humanos , Lipoproteínas HDL , Camundongos , Camundongos Endogâmicos C57BL , MicroRNAs/genética , Neovascularização Fisiológica , Óxido Nítrico Sintase Tipo III/genética , Fosfatidilinositol 3-Quinases , Proteômica , Peixe-Zebra
6.
World J Clin Cases ; 8(16): 3601-3607, 2020 Aug 26.
Artigo em Inglês | MEDLINE | ID: mdl-32913870

RESUMO

BACKGROUND: Coronary artery spasm is a major cause of myocardial ischemia. Although coronary artery spasm has been known for a long time, its mechanism has not yet been identified. Many clinicians, especially young clinicians pay less attention to coronary artery spasm, which may lead to some patients not being appropriately diagnosed and treated in time. We report a patient with spontaneous multivessel coronary artery spasm for more than 30 years diagnosed with intravascular ultrasound (IVUS) imaging. CASE SUMMARY: A 66-year-old Chinese male patient had chest squeezing at rest for more than 30 years. He had a history of cigarette smoking for more than 40 years and hypertension for 10 years. Before presenting at our institution, the patient had undergone coronary angiography 4 times and percutaneous transluminal coronary angioplasty procedures twice at other hospitals without a diagnosis of coronary artery spasm. However, his chest symptoms worsened. Spontaneous multivessel coronary artery spasm occurred during IVUS without provocation testing, and the IVUS image was recorded. Thus, the diagnosis of multifocal spontaneous coronary artery spasm was confirmed. The patient was placed on oral diltiazem, isosorbide mononitrate, and nicorandil to suppress coronary artery spasms. All medications were given at the maximum dosages tolerated by the patient. He was discharged after 5 d without complications. During the six-month follow-up period, the patient was symptom-free. CONCLUSION: Coronary artery spasm is still prevalent in Eastern countries. It is essential for clinicians to be aware of coronary artery spasm, which may be hard to detect and can be lethal, in order to diagnose and treat patients appropriately.

7.
Med Sci Monit ; 25: 240-247, 2019 Jan 08.
Artigo em Inglês | MEDLINE | ID: mdl-30617247

RESUMO

BACKGROUND This study observed the incidence of in-stent restenosis (ISR) after percutaneous coronary intervention (PCI) and discusses the risk factors of ISR based on clinical data, coronary angiography, and stent features, to provide a theoretical basis for the prevention and treatment of ISR. MATERIAL AND METHODS We selected 1132 cases who received stent implantation at the Shaanxi People's Hospital from June 2014 to June 2016 and were followed up by coronary angiography within 1 year. Based on coronary angiography, the cases were divided into ISR and non-ISR groups. ISR was defined as a reduction in lumen diameter by over 50% after PCI. The ISR group consisted of 93 cases and the non-ISR group consisted of 1039 cases. Medical history, biochemical indicators, features of coronary artery lesions, and stent status were analyzed retrospectively. Risk factors of ISR were identified by univariate and multivariate logistic regression analyses. RESULTS Among 1132 cases, 93 cases had ISR, with the overall incidence of 8.21%. Univariate and multivariate logistic regression analyses indicated that postoperative hypersensitive C-reactive protein (hs-CRP) levels (OR=2.309, 1.579-3.375 mg/L), postoperative homocysteine (HCY) levels (OR=2.202, 1.268-3.826 µmol/L), history of diabetes (OR=1.955,1.272-3.003), coronary bifurcation lesions (OR=3.785, 2.246-6.377), and stent length (OR=1.269, 1.179-1.365 mm) were independent risk factors of ISR after PCI (P<0.05). CONCLUSIONS Elevated hs-CRP and HCY levels after PCI, history of diabetes, coronary bifurcation lesions, and greater stent length were associated with a higher risk of ISR. Patients with a higher risk of ISR should receive routine follow-up and intense medication management after PCI to control the risk factors and to reduce ISR.


Assuntos
Reestenose Coronária/etiologia , Intervenção Coronária Percutânea/efeitos adversos , Stents/efeitos adversos , Idoso , Proteína C-Reativa/análise , China , Angiografia Coronária/efeitos adversos , Doença da Artéria Coronariana/complicações , Vasos Coronários/fisiopatologia , Feminino , Seguimentos , Homocisteína/análise , Humanos , Masculino , Pessoa de Meia-Idade , Estudos Retrospectivos , Fatores de Risco
8.
Int Immunopharmacol ; 62: 203-211, 2018 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-30015240

RESUMO

Monocytes recruited and adhering to the inflamed arteries are crucial for atherosclerosis development. Here, we report the role of zinc (Zn2+) homeostasis in monocyte adhesion and recruitment. By comparing the expression levels of Zn2+ transporters between non-adhering and adhering monocytes, we found that the Zn2+ importer ZIP8 was specifically upregulated in monocytes adhering to the aortas ex-vivo. Although the overexpression of ZIP8 increased the absorption of Zn2+, Fe2+ and Cd2+ in monocytes, only Zn2+ supplementation was demonstrated capable of promoting the adhesion of monocytes to endothelial monolayers in vitro. In addition, we confirmed the role of ZIP8-dependent Zn2+ influx in promoting monocyte adhesion to the aortas ex-vivo. More importantly, the enforced expression of ZIP8 increased monocyte adhesion and recruitment to the nascent atherosclerotic lesions in ApoE-/- mice. Overall, our results suggest that the Zn2+ influx in monocytes regulated by ZIP8 is a novel factor determining their adhesion and recruitment to atherosclerotic lesions, and that targeting ZIP8 or Zn2+ homeostasis may represent a novel strategy to interfere these activities.


Assuntos
Aorta/metabolismo , Proteínas de Transporte de Cátions/metabolismo , Adesão Celular , Doença da Artéria Coronariana/metabolismo , Monócitos/metabolismo , Zinco/metabolismo , Animais , Aorta/patologia , Apolipoproteínas E/genética , Proteínas de Transporte de Cátions/genética , Adesão Celular/genética , Linhagem Celular , Doença da Artéria Coronariana/genética , Doença da Artéria Coronariana/patologia , Modelos Animais de Doenças , Camundongos Endogâmicos C57BL , Camundongos Knockout , Monócitos/patologia , Regulação para Cima
9.
Cardiovasc Ther ; 35(4)2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28520220

RESUMO

INTRODUCTION: Platelet activation participates in the development of both coronary artery disease (CAD) and circulating microparticles (MPs). As a commonly used medicine for coronary heart disease, whether aspirin affects the function of MPs remains unclear. AIMS: This study was designed to test MPs from healthy subjects, and stable angina (SA) patients before and after aspirin administration were obtained. MPs origins were tested by flow cytometry. Rat thoracic aortas were incubated with MPs (with or without aspirin) to determine the effects of MPs on expression of ERK1/2, JNKs, and p38 MAPK. Affect on levels of NF-κB, VCAM-1, NO, and O2-. RESULTS: Compared with healthy subjects, MPs concentrations increased in SA patients, but decreased after aspirin administration. According to flow cytometry, aspirin mainly decreased platelet-derived MP. MPs from SA patients decreased the expression of ERK1/2, increased expression of p38 MAPKs, JNKs. Increased NF-κB, VCAM-1, and (O2-) levels decreased NO content. Aspirin therapy significantly inhibited function of MPs from SA patients, and pathway inhibitors (ERK1/2 inhibitor PD98059, p38 MAPKs inhibitor SB203580, NF-kB inhibitor PDTC) show similar effects with aspirin. CONCLUSION: These results indicate that the pro-inflammatory, oxidative stress, procoagulant, and adhesion properties of MPs can be partly blocked by aspirin via the ERK-NO/O2- and p38 -NF-κB-VCAM-1 signal pathway, which clarified other functions beyond anti-atherothrombotic of aspirin.


Assuntos
Angina Estável/patologia , Aspirina/uso terapêutico , Micropartículas Derivadas de Células , Endotélio Vascular/lesões , Endotélio Vascular/patologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Inibidores da Agregação Plaquetária/uso terapêutico , Animais , Aorta Torácica/patologia , Endotélio Vascular/efeitos dos fármacos , Voluntários Saudáveis , Humanos , Masculino , NF-kappa B/metabolismo , Óxido Nítrico/biossíntese , Ratos , Superóxidos/metabolismo , Molécula 1 de Adesão de Célula Vascular/biossíntese , Molécula 1 de Adesão de Célula Vascular/genética
10.
Cardiology ; 132(4): 252-60, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26329646

RESUMO

OBJECTIVES: Endothelial dysfunction is involved in the development of the acute coronary syndrome (ACS). Plasma microparticles(MPs) from other diseases have been demonstrated to initiate coagulation and endothelial dysfunction.However, whether MPs from ACS patients impair vasodilatation and endothelial function remains unclear. METHODS: Patients(n = 62) with ACS and healthy controls (n = 30) were recruited for MP isolation. Rat thoracic aortas were incubated with MPs from ACS patients or healthy controls to determine the effects of MPs on endothelial-dependent vasodilatation,the phosphorylation of Akt and endothelial nitric oxide synthase (eNOS), the interaction of eNOS with heat shock protein 90 (Hsp90), and nitric oxide (NO) and superoxide anion(O 2 ­ ) production. The origin of MPs was assessed by flow cytometry. RESULTS: MP concentrations were increased in patients with ACS compared with healthy controls. They were positively correlated with the degree of coronary artery stenosis. MPs from ACS patients impair endothelial-dependent vasodilatation, decrease both Akt and eNOS phosphorylation,decrease the interaction between eNOS and Hsp90,and decrease NO production but increase O 2 ­ generation in rat thoracic aortas. Endothelial-derived MPs and platelet-derived MPs made up nearly 75% of MPs. CONCLUSIONS: Our data indicate that MPs from ACS patients negatively affect endothelial-dependent vasodilatation via Akt/eNOS-Hsp90 pathways.


Assuntos
Síndrome Coronariana Aguda/metabolismo , Micropartículas Derivadas de Células/metabolismo , Endotélio Vascular/fisiopatologia , Proteínas de Choque Térmico HSP90/metabolismo , Óxido Nítrico Sintase Tipo III/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Vasodilatação/fisiologia , Síndrome Coronariana Aguda/cirurgia , Adulto , Idoso , Animais , Estudos de Casos e Controles , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Óxido Nítrico/metabolismo , Fosforilação , Ratos , Transdução de Sinais , Superóxidos/metabolismo
11.
J Thorac Cardiovasc Surg ; 150(3): 666-72, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26145768

RESUMO

OBJECTIVE: Vascular function is very important for maintaining circulation after cardiac surgery. Circulating microparticles (MPs) generated in various diseases play important roles in causing inflammation, coagulation, and vascular injury. However, the impact of MPs generated from patients who have valvular heart disease (VHD), before and after cardiac surgery, on vascular function remains unknown. This study is designed to investigate the impact of such MPs on vasodilation. METHODS: Microparticles were isolated from age-matched healthy subjects and patients who had VHD, before cardiac surgery, and at 12 hours and 72 hours afterward. The number of MPs was measured and compared. Effects evaluated were of the impact of MPs on: vasodilation of mice aorta; the phosphorylation and expression of Akt, endothelial nitric oxide synthase (eNOS), protein kinase C-ßII (PKC-ßII), and p70 ribosomal protein S6 kinase (p70S6K); expression of caveolin-1; the association of eNOS with heat shock protein 90 (HSP90); and generation of nitric oxide and superoxide anion of human umbilical vein endothelial cells. RESULTS: Compared with the healthy subjects, VHD patients had significantly higher levels of circulating MPs and those MPs before cardiac surgery can: impair endothelium-dependent vasodilation; inhibit phosphorylation of Akt and eNOS; increase activation of PKC-ßII and p70S6K; enhance expression of caveolin-1; reduce the association of HSP90 with eNOS; decrease nitric oxide production, and increase superoxide anion generation. These deleterious effects were even stronger in postoperative MPs. CONCLUSIONS: Our data demonstrate that MPs generated from VHD patients before and after cardiac surgery contributed to endothelial dysfunction, by uncoupling and inhibiting eNOS. Circulating MPs are potential therapeutic targets for the maintenance of vascular function postoperatively.


Assuntos
Aorta/metabolismo , Procedimentos Cirúrgicos Cardíacos , Micropartículas Derivadas de Células/metabolismo , Endotélio Vascular/metabolismo , Doenças das Valvas Cardíacas/sangue , Doenças das Valvas Cardíacas/cirurgia , Vasodilatação , Adulto , Animais , Aorta/fisiopatologia , Estudos de Casos e Controles , Células Cultivadas , Endotélio Vascular/fisiopatologia , Feminino , Doenças das Valvas Cardíacas/fisiopatologia , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Técnicas In Vitro , Masculino , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo III/metabolismo , Estresse Oxidativo , Transdução de Sinais
12.
J Mol Cell Cardiol ; 74: 209-19, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24887036

RESUMO

Normal high density lipoprotein (HDL) protects vascular function; however these protective effects of HDL may absent in valvular heart disease (VHD). Because vascular function plays an important role in maintaining the circulation post-cardiac surgery and some patients are difficult to stabilize, we hypothesized that a deleterious vascular effect of HDL may contribute to vascular dysfunction in VHD patients following surgery. HDL was isolated from age-match 28 healthy subjects and 84 patients with VHD and during cardiac surgery. HDL pro-inflammation index was measured and the effects of HDL on vasodilation, protein interaction, generation of nitric oxide (NO) and superoxide were determined. Patients with VHD received either simvastatin (20mg/d) or routine medications, and endothelial effects of HDL were characterized. HDL inflammation index significantly increased in VHD patients and post-cardiac surgery. HDL from VHD patients and post-cardiac surgery significantly impaired endothelium-dependent vasodilation, inhibited both Akt and endothelial nitric oxide synthase (eNOS) phosphorylation at S1177, eNOS associated with heat shock protein 90 (HSP90), NO production and increased eNOS phosphorylation at T495 and superoxide generation. Simvastatin therapy partially reduced HDL inflammation index, improved the capacity of HDL to stimulate eNOS and Akt phosphorylation at S1177, eNOS associated with HSP90, NO production, reduced eNOS phosphorylation at T495 and superoxide generation, and improved endothelium-dependent vasodilation. Our data demonstrated that HDL from VHD patients and cardiac surgery contributed to endothelial dysfunction through uncoupling of eNOS. This deleterious effect can be reversed by simvastatin, which improves the vasoprotective effects of HDL. Targeting HDL may be a therapeutic strategy for maintaining vascular function and improving the outcomes post-cardiac surgery.


Assuntos
Doenças das Valvas Cardíacas/metabolismo , Valvas Cardíacas/metabolismo , Lipoproteínas HDL/metabolismo , Adulto , Idoso , Animais , Vasos Sanguíneos/efeitos dos fármacos , Vasos Sanguíneos/patologia , Estudos de Casos e Controles , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/patologia , Feminino , Regulação da Expressão Gênica , Proteínas de Choque Térmico HSP90/genética , Proteínas de Choque Térmico HSP90/metabolismo , Doenças das Valvas Cardíacas/tratamento farmacológico , Doenças das Valvas Cardíacas/genética , Doenças das Valvas Cardíacas/patologia , Valvas Cardíacas/efeitos dos fármacos , Valvas Cardíacas/patologia , Humanos , Hipolipemiantes/uso terapêutico , Lipoproteínas HDL/farmacologia , Masculino , Camundongos , Pessoa de Meia-Idade , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo III/genética , Óxido Nítrico Sintase Tipo III/metabolismo , Fosforilação/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais , Sinvastatina/uso terapêutico , Superóxidos/antagonistas & inibidores , Superóxidos/metabolismo , Vasodilatação/efeitos dos fármacos
13.
J Cardiovasc Pharmacol ; 61(5): 430-6, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23429589

RESUMO

Mouse models of myocardial ischemic preconditioning (IPC) and ischemic postconditioning (IPD) have proven to be very useful models of cardiovascular diseases. In 2010, Gao described a novel procedure without the aid of mechanical ventilation. However, the technique of heart externalization could not be applied to mouse models of IPC or IPD due to the limited time frame of the technique. We proposed a modified simple and safe method using lung recruitment and short-term ventilation to perform the procedure in mice with IPC or IPD. The mice were randomly divided into 4 groups: the modified groups, M-IPC and M-IPD, and the conventional groups, C-IPC and C-IPD. In the 2 modified groups, the mice were removed from the ventilator and allowed to resume breathing spontaneously upon completion of the lung recruitment and the rapid closure of the thorax. Our study demonstrated that the postoperative recovery time was significantly reduced for the modified groups compared with the 2 conventional groups. Moreover, the inflammatory damages were attenuated by the modified method compared with the conventional method. In addition, the modified method significantly increased the survival rates of mice with IPC or IPD. The modified method improved the survival rates of mouse models of myocardial ischemia.


Assuntos
Modelos Animais de Doenças , Pós-Condicionamento Isquêmico/métodos , Precondicionamento Isquêmico Miocárdico/métodos , Isquemia Miocárdica/terapia , Animais , Arritmias Cardíacas/epidemiologia , Arritmias Cardíacas/etiologia , Vasos Coronários , Incidência , Interleucina-6/sangue , Ligadura , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Isquemia Miocárdica/complicações , Isquemia Miocárdica/mortalidade , Isquemia Miocárdica/patologia , Taxa de Sobrevida , Fatores de Tempo , Resultado do Tratamento
14.
Am J Physiol Endocrinol Metab ; 304(7): E695-702, 2013 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-23384770

RESUMO

Mitral valve endothelial cells are important for maintaining lifelong mitral valve integrity and function. Plasma endothelial microparticles (EMPs) increased in various pathological conditions related to activation of endothelial cells. However, whether EMPs will increase in mitral valve disease and their relationship remains unclear. Here, 81 patients with mitral valve disease and 45 healthy subjects were analyzed for the generation of EMPs by flow cytometry. Human mitral valve endothelial cells (HMVECs) were treated with EMPs. The phosphorylation of Akt and endothelial nitric oxide synthase (eNOS), the association of eNOS and heat shock protein 90 (HSP90), and the generation of nitric oxide (NO) and superoxide anion (O(2)(∙-)) were measured. EMPs were increased significantly in patients with mitral valve disease compared with those in healthy subjects. EMPs were negatively correlated with mitral valve area in patients with isolated mitral stenosis. EMPs were significantly higher in the group with severe mitral regurgitation than those in the group with mild and moderate mitral regurgitation. Furthermore, EMPs were decreased dramatically in both Akt and eNOS phosphorylation and the association of HSP90 with eNOS in HMVECs. EMPs decreased NO production but increased O(2)(∙-) generation in HMVECs. Our data demonstrated that EMPs were significantly increased in patients with mitral valve disease. The increase of EMPs can in turn impair HMVEC function by inhibiting the Akt/eNOS-HSP90 signaling pathway. These findings suggest that EMPs may be a therapeutic target for mitral valve disease.


Assuntos
Micropartículas Derivadas de Células/metabolismo , Células Endoteliais , Insuficiência da Valva Mitral/metabolismo , Estenose da Valva Mitral/metabolismo , Valva Mitral , Óxido Nítrico Sintase Tipo III/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Adulto , Estudos de Casos e Controles , Células Cultivadas , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Endotélio Vascular/citologia , Endotélio Vascular/metabolismo , Feminino , Proteínas de Choque Térmico HSP90/metabolismo , Humanos , Masculino , Pessoa de Meia-Idade , Valva Mitral/citologia , Valva Mitral/metabolismo , Óxido Nítrico/metabolismo , Fosforilação , Transdução de Sinais , Superóxidos/metabolismo
15.
Arterioscler Thromb Vasc Biol ; 32(9): 2304-13, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22796581

RESUMO

OBJECTIVE: Myocardial injury during cardiac surgery is a major cause of perioperative morbidity and mortality. We determined whether perioperative statin therapy is cardioprotective in patients undergoing noncoronary artery cardiac surgery and the potential mechanisms. METHODS AND RESULTS: One hundred fifty-one patients undergoing noncoronary artery cardiac surgery were randomly assigned to either a statin group (n=77) or a control group (n=74). Simvastatin (20 mg) was administered preoperatively and postoperatively. Plasma were analyzed for troponin T, isoenzyme of creatine kinase, C-reaction protein, interleukin-6, interleukin-8, creatinine, and blood urea nitrogen. Cardiac echocardiography was performed. Endothelial nitric oxide synthase (eNOS), Akt, p38, heat shock protein 90, caveolin-1, and nitric oxide (NO) in the heart were detected. Simvastatin significantly reduced plasma troponin T, isoenzyme of creatine kinase, C-reaction protein, blood urea nitrogen , creatinine, interleukin-6, interleukin-8, and the requirement of inotropic postoperatively. Simvastatin increased NO production, the expression of eNOS and phosphorylation at serine1177, phosphorylation of Akt, expression of heat shock protein 90, heat shock protein 90 association with eNOS and decreased eNOS phosphorylation at threonine 495, phosphorylation of p38, and expression of caveolin-1. Simvastatin also improved cardiac function postoperatively. CONCLUSIONS: Perioperative statin therapy can improve cardiac function and renal function by reducing myocardial injury and inflammatory response through activating Akt-eNOS and attenuating p38 signaling pathways in patients undergoing noncoronary artery cardiac surgery. Clinical Trial Registration- URL: http://www.clinicaltrials.gov. Unique identifier: NCT01178710.


Assuntos
Procedimentos Cirúrgicos Cardíacos/efeitos adversos , Traumatismos Cardíacos/prevenção & controle , Inibidores de Hidroximetilglutaril-CoA Redutases/administração & dosagem , Miocárdio/patologia , Sinvastatina/administração & dosagem , Adulto , Análise de Variância , Biomarcadores/sangue , Nitrogênio da Ureia Sanguínea , Proteína C-Reativa/metabolismo , Cardiotônicos/uso terapêutico , Caveolina 1/metabolismo , China , Creatina Quinase/sangue , Creatinina/sangue , Feminino , Proteínas de Choque Térmico HSP90/metabolismo , Traumatismos Cardíacos/sangue , Traumatismos Cardíacos/diagnóstico por imagem , Traumatismos Cardíacos/etiologia , Traumatismos Cardíacos/fisiopatologia , Humanos , Interleucina-6/sangue , Interleucina-8/sangue , Masculino , Pessoa de Meia-Idade , Miocárdio/metabolismo , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo III/metabolismo , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo , Método Simples-Cego , Volume Sistólico/efeitos dos fármacos , Fatores de Tempo , Resultado do Tratamento , Troponina T/sangue , Ultrassonografia , Função Ventricular Esquerda/efeitos dos fármacos , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
16.
J Thorac Cardiovasc Surg ; 144(3): 697-703, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22336756

RESUMO

OBJECTIVES: Endogenous nitric oxide synthase inhibitor asymmetric dimethylarginine is a cardiovascular risk factor that is elevated in patients with coronary artery disease. We hypothesized that novel endothelial nitric oxide synthase enhancer AVE3085 might improve the endothelial function altered by asymmetric dimethylarginine in the human internal thoracic artery. METHODS: Cumulative concentration-relaxation curves to acetylcholine (-11 to -5 log mol/L) were established in left internal thoracic artery rings (n = 65) from 27 patients undergoing coronary artery bypass grafting in precontraction induced by U46619 (-8 log mol/L) in the absence or presence of asymmetric dimethylarginine (100 µmol/L) or AVE3085 (30 µmol/L). Protein expressions of endothelial nitric oxide synthase and levels of superoxide anion production were detected. RESULTS: Maximal relaxation induced by acetylcholine was significantly attenuated by asymmetric dimethylarginine (12.7% ± 2.3% vs 35.3% ± 5.0% in control; P < .05) and significantly restored by AVE3085 (23.4% ± 2.8%; P < .05). AVE3085 also markedly restored endothelial nitric oxide synthase expression (0.29 ± 0.008; P = .012) reduced by asymmetric dimethylarginine (0.05 ± 0.04 vs 0.36 ± 0.03 in control; P = .014). Increased superoxide anion production by asymmetric dimethylarginine (2.97 ± 0.25 vs 0.51 ± 0.10 relative light units/[s/mg] in control; P < .05) was inhibited by AVE3805 (0.62 ± 0.104 relative light units/[s/mg]; P < .05). CONCLUSIONS: AVE3085 may restore endothelium-dependent relaxation reduced by asymmetric dimethylarginine through upregulation of endothelial nitric oxide synthase expression and inhibition of production of superoxide anion in human internal thoracic artery. These findings provide new insights into endothelial protection of coronary bypass grafting vessels to improve long-term patency of grafts.


Assuntos
Arginina/análogos & derivados , Benzodioxóis/farmacologia , Ativadores de Enzimas/farmacologia , Indanos/farmacologia , Artéria Torácica Interna/efeitos dos fármacos , Óxido Nítrico Sintase Tipo III/metabolismo , Vasodilatação/efeitos dos fármacos , Vasodilatadores/farmacologia , Acetilcolina/farmacologia , Idoso , Antioxidantes/farmacologia , Arginina/metabolismo , Western Blotting , Citoproteção , Relação Dose-Resposta a Droga , Ativação Enzimática , Feminino , Humanos , Técnicas In Vitro , Masculino , Artéria Torácica Interna/enzimologia , Artéria Torácica Interna/fisiopatologia , Pessoa de Meia-Idade , Miografia , Superóxidos/metabolismo , Grau de Desobstrução Vascular/efeitos dos fármacos
17.
J Cardiovasc Pharmacol ; 59(4): 352-62, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22286127

RESUMO

We have previously demonstrated that dl-3n-butylphthalide (NBP) has a potential angiogenic activity. In this study, we investigated the angiogenic effect of NBP and the molecular mechanisms underlying NBP-mediated angiogenesis. Zebrafish embryos and human umbilical vein endothelial cells were treated with various doses of NBP and several signaling pathway inhibitors. NBP induced ectopic subintestinal vessel production in zebrafish embryos and induced invasion, migration, and endothelial cell tube formation of human umbilical vein endothelial cells in a dose-dependent manner. These NBP-induced angiogenic effects were partially suppressed by SU5402, a fibroblast growth factor receptor 1 inhibitor; U0126, an extracellular signal-regulated kinase 1/2 (ERK1/2) inhibitor; LY294002, a phosphatidylinositol 3-kinase inhibitor; 1L6-hydroxymethyl-chiro-inositol-2-(R)-2-O-methyl-3-O-octadecyl-sn-glycerocarbonate, an Akt inhibitor; cavtratin, an endothelial nitric oxide synthase (eNOS) inhibitor and completely inhibited by a combination of U0126 and LY294002. NBP enhanced phosphorylation of ERK1/2 and fibroblast growth factor receptor 2 expression, which were inhibited by U0126. NBP increased the phosphorylation of Akt and eNOS at serine 1177, which was blocked by LY294002. NBP-stimulated nitric oxide production, which was reduced by LY294002. Our data demonstrated that (1) NBP promoted angiogenesis and (2) the angiogenic effects of NBP were mediated by the ERK1/2 and phosphatidylinositol 3-kinase/Akt-eNOS signaling pathways. Our findings suggest that NBP could be a novel agent for therapeutic angiogenesis in ischemic diseases.


Assuntos
Benzofuranos/farmacologia , Neovascularização Fisiológica/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Transdução de Sinais/efeitos dos fármacos , Animais , Benzofuranos/administração & dosagem , Relação Dose-Resposta a Droga , Células Endoteliais da Veia Umbilical Humana , Humanos , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Fármacos Neuroprotetores/administração & dosagem , Óxido Nítrico Sintase Tipo III/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo , Peixe-Zebra/embriologia
18.
Heart ; 98(5): 384-8, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22107759

RESUMO

OBJECTIVE: To determine whether remote ischaemic preconditioning (RIPC) is cardioprotective in patients undergoing heart valve replacement. DESIGN: Single-blinded, randomised controlled trial. SETTING: Tertiary referral hospital in China. PATIENTS: Adult patients (31-72 years) undergoing mitral valve, aortic valve or tricuspid valve surgery. INTERVENTIONS: Patients were randomised to either the RIPC (n=38) or control (n=35) group. After induction of anaesthesia, patients in the RIPC group underwent three 5 min cycles of right upper limb ischaemia, induced by an automated cuff-inflator placed on the upper arm and inflated to 200 mm Hg. Each cycle was interrupted by a 5 min period of reperfusion during which time the cuff was deflated. The control group had only a deflated cuff placed on the upper arm for 30 min. MAIN OUTCOME MEASURES: Serum troponin I concentration was measured before surgery and at 6, 12, 24, 48, and 72 h postoperatively. The cardiac function of all patients was followed postoperatively. RESULTS: Troponin I concentration was reduced in the RIPC group (398.7±179.3 µg/l) compared with the control group (708.4±242.5 µg/l). Mean difference was 309.7±50.8 (95% CI 210.1 to 409.3, p<0.0001). A greater improvement in postsurgical cardiac function was noted in the RIPC group than in the control group. CONCLUSIONS: These data indicate that RIPC reduces myocardial injury and improves cardiac function in patients undergoing heart valve surgery. TRIAL REGISTRATION NUMBER: NCT01175681.


Assuntos
Doenças das Valvas Cardíacas/cirurgia , Implante de Prótese de Valva Cardíaca , Precondicionamento Isquêmico Miocárdico/métodos , Isquemia Miocárdica/prevenção & controle , Telemedicina/métodos , Adulto , Idoso , Biomarcadores/sangue , China/epidemiologia , Feminino , Seguimentos , Doenças das Valvas Cardíacas/sangue , Humanos , Incidência , Masculino , Pessoa de Meia-Idade , Isquemia Miocárdica/epidemiologia , Isquemia Miocárdica/etiologia , Miocárdio/metabolismo , Prognóstico , Estudos Retrospectivos , Método Simples-Cego , Troponina I/sangue
19.
Am J Physiol Endocrinol Metab ; 300(4): E661-8, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21245463

RESUMO

Therapeutic angiogenesis remains unsuccessful in coronary artery disease. It is known that plasma endothelium-derived microparticles (EMPs) are increased in coronary artery disease and that hypercholesterolemia can inhibit angiogenesis. We evaluated the relationship between EMPs and hypercholesterolemia in the impairment of angiogenesis. EMPs isolated from human umbilical vein endothelial cells were injected into low-density lipoprotein receptor-null (LDLr(-/-)) mice fed a Western diet for 2 wk and C57BL6 mice for 6 h or were directly added to the tissue culture media. Hearts isolated from mice were sectioned and cultured, and endothelial tube formation was measured. The expression and phosphorylation of endothelial NO synthase (eNOS) and the generation of NO in the hearts were determined. Angiogenesis was inhibited by pathophysiological concentrations of EMPs but not physiological concentrations of EMPs in hearts from C57BL6 mice. However, angiogenesis was inhibited by EMPs at both physiological and pathophysiological concentrations of EMPs in hearts from hypercholesterolemic LDLr(-/-) mice. Pathophysiological concentrations of EMPs decreased eNOS phosphorylation at Ser(1177) and NO generation without altering eNOS expression in hearts from C57BL6 mice. Both physiological and pathophysiological concentrations of EMPs decreased not only eNOS phosphorylation at Ser(1177) and NO generation, but eNOS expression in hypercholesterolemic hearts from LDLr(-/-) mice. These data demonstrated that pathophysiological concentrations of EMPs could inhibit angiogenesis in hearts by decreasing eNOS activity. EMPs and hypercholesterolemia mutually enhanced their inhibitory effect of angiogenesis by inducing eNOS dysfunction. Our findings suggest a novel mechanism by which hypercholesterolemia impairs angiogenesis.


Assuntos
Micropartículas Derivadas de Células/fisiologia , Vasos Coronários/crescimento & desenvolvimento , Endotélio Vascular/citologia , Hipercolesterolemia/complicações , Neovascularização Fisiológica , Animais , Doenças Cardiovasculares/etiologia , Doenças Cardiovasculares/patologia , Doenças Cardiovasculares/fisiopatologia , Células Cultivadas , Regulação para Baixo , Coração/fisiopatologia , Humanos , Hipercolesterolemia/patologia , Hipercolesterolemia/fisiopatologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neovascularização Fisiológica/fisiologia , Receptores de LDL/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA