Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
ESC Heart Fail ; 10(2): 1077-1089, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36578105

RESUMO

AIM: The progression of atherosclerosis can lead to the occurrence of multiple cardiovascular diseases (coronary heart disease, etc.). E prostanoid receptor-3 (EP3) is known to participate in the progression of atherosclerosis. This study aimed to investigate the mechanism by which EP3 modulates the development of atherosclerosis. METHODS AND RESULTS: ApoE-/- mice were used to construct in vivo model of atherosclerosis. Human aortic smooth muscle cells (HASMCs) were stimulated with oxidized low-density lipoprotein (ox-LDL) to construct in vitro model of atherosclerosis. mRNA expressions were assessed by qRT-PCR, and western blot was applied to assess the protein levels. CCK-8 assay was applied to assess the cell viability. The inflammatory cytokines levels were assessed by enzyme-linked immunosorbent assay, and flow cytometry was applied to assess cell apoptosis. In vivo experiment was constructed to investigate the impact of EP3 in atherosclerosis development. L-798106 (EP3 inhibitor) significantly inhibited the levels of pro-inflammatory cytokines in atherosclerosis in vivo. EP3 inhibitor (L-798106) significantly reversed ox-LDL-caused HASMCs injury via inhibiting the apoptosis and inflammatory responses (P < 0.05). The levels of interleukin-17 (IL-17) and intercellular adhesion molecule-1 (ICAM-1) in HASMCs were elevated by ox-LDL, whereas L-798106 or knockdown of cyclic AMP (cAMP) response element-binding protein (CREB) notably restored this phenomenon (P < 0.05). EP3 overexpression further aggravated ox-LDL-induced inflammation in HASMCs, and EP3 up-regulated the levels of IL-17 and ICAM-1 in ox-LDL-treated HASMCs (P < 0.05). EP3 up-regulation promoted the inflammatory responses in ox-LDL-treated HASMCs through mediation of cAMP/protein kinase A (PKA)/CREB/IL-17/ICAM-1 axis (P < 0.05). CONCLUSIONS: EP3 inhibitor alleviates ox-LDL-induced HASMC inflammation via mediation of cAMP/PKA/CREB/IL-17/ICAM-1 axis. Our study might shed new lights on discovering novel strategies against atherosclerosis.


Assuntos
Aterosclerose , Molécula 1 de Adesão Intercelular , Animais , Humanos , Camundongos , Aterosclerose/genética , Moléculas de Adesão Celular/metabolismo , Citocinas/metabolismo , Inflamação/metabolismo , Molécula 1 de Adesão Intercelular/metabolismo , Interleucina-17/metabolismo , Lipoproteínas LDL/metabolismo , Miócitos de Músculo Liso/metabolismo , Prostaglandinas/metabolismo
2.
Arch Biochem Biophys ; 711: 109007, 2021 10 30.
Artigo em Inglês | MEDLINE | ID: mdl-34400144

RESUMO

Pituitary tumor-transforming gene 1 (PTTG1) has been found to be associated with the process of cell proliferation and invasion, and is highly expressed in aortic dissection (AD). However, its potential role and underlying mechanism in AD remain uncertain. This study aims at elucidating the roles of specificity protein 1 (SP1) and PTTG1 in the migration and phenotypic switching of aortic vascular smooth muscle cells (VSMCs) in AD. Aortic samples were collected from 35 patients with AD for examination of PTTG1 expression in the tissues by qPCR, western blot and immunofluorescence. Human aortic vascular smooth muscle cells (HAVSMCs) were stimulated with platelet-derived growth factor-BB (PDGF-BB) to establish the cellular model of AD. PTTG1 expression in VSMCs was also examined by qPCR and western blot. Cell viability was detected by CCK-8, cell proliferation by EdU staining and cell migration by wound healing and transwell. Western blot was then performed to assay migration-related proteins. After interference with PTTG1, the levels of smooth muscle pthenotypic switch markers smooth muscle protein 22 alpha (SM22-α) and osteopontin (OPN) were detected by qPCR, western blot and immunofluorescence. The binding of SP1 and PTTG1 was verified with dual-luciferase reporter assay and chromatin immunoprecipitation assay (ChIP). PTTG1 overexpression was found in AD patients. Interference with PTTG1 attenuated the proliferation and migration of PDGF-BB-stimulated HAVSMCs, in addition to their switching from contractile phenotype to synthetic phenotype. Transcription factor SP1 was up-regulated in PDGF-BB-stimulated HAVSMCs, combined with PTTG1 promoter sequence and regulated PTTG1 expression, whose overexpression reversed the effects of PTTG1 interference on cell proliferation, migration and phenotypic switching. SP1 transcriptional activation of PTTG1 activated MAPK/ERK signaling pathway. In conclusion, SP1 transcriptional activation of PTTG1 regulates the migration and phenotypic transformation of HAVSMCs in AD by MAPK Signaling.


Assuntos
Dissecção Aórtica/metabolismo , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Securina/metabolismo , Fator de Transcrição Sp1/metabolismo , Aorta/metabolismo , Becaplermina/farmacologia , Movimento Celular/fisiologia , Proliferação de Células/fisiologia , Células Cultivadas , Técnicas de Silenciamento de Genes , Humanos , Sistema de Sinalização das MAP Quinases/fisiologia , Pessoa de Meia-Idade , Músculo Liso Vascular/citologia , Músculo Liso Vascular/efeitos dos fármacos , Miócitos de Músculo Liso/efeitos dos fármacos , Securina/genética , Ativação Transcricional/fisiologia , Regulação para Cima/efeitos dos fármacos
3.
Medicine (Baltimore) ; 99(27): e20994, 2020 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-32629716

RESUMO

INTRODUCTION: Acute myocardial infarction with simultaneous coronary thrombosis has been rarely reported. This combination induces various arrhythmias and is a high-risk factor for cardiogenic shock. PATIENT CONCERNS: A 65-year-old man presented with sweating and a 3-h abrupt persistent back pain that radiated to the anterior. DIAGNOSIS: Multisite myocardial infarction, coronary thrombosis with and complex malignant arrhythmia INTERVENTIONS:: Prompt intervention includes cardiac pacing, percutaneous coronary intervention (PCI), thrombus aspiration and intra-aortic balloon pump (IABP). OUTCOMES: The patient was successfully rescued after PCI and thrombus aspiration. CONCLUSIONS: Recognition of dynamic electrocardiographic changes enhances our understanding of the pathogenesis of myocardial infarction.


Assuntos
Arritmias Cardíacas/complicações , Trombose Coronária/complicações , Infarto do Miocárdio/complicações , Idoso , Arritmias Cardíacas/cirurgia , Trombose Coronária/cirurgia , Eletrocardiografia , Humanos , Masculino , Intervenção Coronária Percutânea/métodos , Choque Cardiogênico/etiologia
4.
Front Physiol ; 10: 1247, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31611817

RESUMO

Endothelial dysfunction, which leads to ischemic events under atherosclerotic conditions, can be attenuated by antagonizing the thromboxane-prostanoid receptor (TP) that mediates the vasoconstrictor effect of prostanoids including prostacyclin (PGI2). This study aimed to determine whether antagonizing the E prostanoid receptor-3 (EP3; which can also be activated by PGI2) adds to the above effect of TP deficiency (TP-/-) under atherosclerotic conditions and if so, the underlying mechanism(s). Atherosclerosis was induced in ApoE-/- mice and those with ApoE-/- and TP-/-. Here, we show that in phenylephrine pre-contracted abdominal aortic rings with atherosclerotic lesions of ApoE-/-/TP-/- mice, although an increase of force (which was larger than that of non-atherosclerotic controls) evoked by the endothelial muscarinic agonist acetylcholine to blunt the concurrently activated relaxation in ApoE-/- counterparts was largely removed, the relaxation evoked by the agonist was still smaller than that of non-atherosclerotic TP-/- mice. EP3 antagonism not only increased the above relaxation, but also reversed the contractile response evoked by acetylcholine in NO synthase-inhibited atherosclerotic ApoE-/-/TP-/- rings into a relaxation sensitive to I prostanoid receptor antagonism. In ApoE-/- atherosclerotic vessels the expression of endothelial NO synthase was decreased, yet the production of PGI2 (which evokes contraction via both TP and EP3) evoked by acetylcholine was unaltered compared to non-atherosclerotic conditions. These results demonstrate that EP3 blockade adds to the effect of TP-/- in uncovering the dilator action of natively produced PGI2 to alleviate endothelial dysfunction in atherosclerotic conditions.

5.
Eur J Pharmacol ; 836: 11-17, 2018 Oct 05.
Artigo em Inglês | MEDLINE | ID: mdl-30107163

RESUMO

Prostaglandin (PG) D2, a prostanoid known to have hypotensive effect, can evoke increased in vitro prepartum myometrial contraction resulting from up-regulation of the F prostanoid (FP) receptor. The present study further determined postpartum rat uterine responses to PGD2 to evaluate the possibility of the prostanoid becoming a therapeutic for postpartum uterine atony, a major cause of postpartum hemorrhage that can lead to maternal morbidity. In vitro and in vivo postpartum uterine responses to PGD2 were determined and compared to those of prepartum rats. Here we show that in postpartum myometrial strips PGD2 did evoke a contraction sensitive to FP receptor antagonism. Interestingly, this response was not only to a greater extent than that of prepartum rats, but also comparable with the contraction obtained with PGF2α, a therapeutic for postpartum uterine atony but contradicted in conditions including hypertension. Indeed, PGD2 was also found to cause increases of basal uterine contraction under in vivo conditions. Western blots revealed that the expression of FP receptors in postpartum myometrium was higher than that of prepartum rats. Moreover, we noted that the amount of PGD2 produced in postpartum uteri, although lower than that of prepartum rats, was increased compared to non-pregnant conditions. These results thus demonstrate that due to a further up-regulation or high expression of myometrial FP receptors, PGD2 can evoke potent uterine contraction postpartum, and hence the prostanoid, which is naturally synthesized in uterine tissues, could be a potential therapeutic for postpartum uterine atony, especially in settings, such as hypertension.


Assuntos
Período Pós-Parto/efeitos dos fármacos , Período Pós-Parto/fisiologia , Prostaglandina D2/farmacologia , Contração Uterina/efeitos dos fármacos , Animais , Ciclo-Oxigenase 1/metabolismo , Feminino , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Masculino , Período Pós-Parto/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores de Prostaglandina/metabolismo
6.
Am J Med Sci ; 341(2): 119-25, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21042170

RESUMO

INTRODUCTION: Cardiac hypertrophy is a risk factor for QT prolongation and cardiac sudden death. In this study, the authors examined the expressional regulation on the rat human ether-a-go-go-related gene (HERG), which encodes a structural subunit of the rapid component of the delayed rectifier potassium current (I(Kr)), during myocardial hypertrophy using rat as a model system. METHODS: Cardiac hypertrophy was established in Sprague-Dawley rats by coarctation of the abdominal aorta [left ventricular hypertrophy (LVH) group]. Sham-operated rats were defined as control group (Ctrl group). Hemodynamic, morphologic and histologic parameters were recorded 6 weeks after operation. In addition, the expression of HERG was also determined using a combination of real-time polymerase chain reaction, Western blot and immunohistochemical analyses. RESULTS: Compared with the sham-operated Ctrl group, abdominal aortic coarctation induced LVH in the LVH group, as evidenced by significantly increased ratios of heart weight/left ventricular weight to body weight and enlarged left ventricular myocytes in the histologic sections. The hemodynamic profile revealed significant increases in heart rate and left ventricular end-diastolic pressure, as well as a decrease in the maximal rate of left ventricular pressure fall in the LVH rats, when compared with the Ctrl rats. Electrocardiograms showed prolonged QT and corrected QT intervals. On the molecular level, a significant reduction of HERG, messengerRNA and protein was observed in LVH group, which was inversely correlated with prolonged corrected QT (r = -0.842, P = 0.000). CONCLUSION: The expressional down-regulation of HERG gene may constitute a novel mechanism for QT prolongation during cardiac hypertrophy.


Assuntos
Canais de Potássio Éter-A-Go-Go/genética , Hipertrofia Ventricular Esquerda/genética , Animais , Aorta Abdominal , Coartação Aórtica/complicações , Sequência de Bases , Primers do DNA/genética , Modelos Animais de Doenças , Regulação para Baixo , Canal de Potássio ERG1 , Canais de Potássio Éter-A-Go-Go/metabolismo , Feminino , Humanos , Hipertrofia Ventricular Esquerda/complicações , Hipertrofia Ventricular Esquerda/metabolismo , Hipertrofia Ventricular Esquerda/patologia , Síndrome do QT Longo/etiologia , Síndrome do QT Longo/genética , Síndrome do QT Longo/metabolismo , Masculino , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...