Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 64
Filtrar
1.
Acta Pharmacol Sin ; 45(4): 728-737, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38086898

RESUMO

Stimulation of adult cardiomyocyte proliferation is a promising strategy for treating myocardial infarction (MI). Earlier studies have shown increased CCL2 levels in plasma and cardiac tissue both in MI patients and mouse models. In present study we investigated the role of CCL2 in cardiac regeneration and the underlying mechanisms. MI was induced in adult mice by permanent ligation of the left anterior descending artery, we showed that the serum and cardiac CCL2 levels were significantly increased in MI mice. Intramyocardial injection of recombinant CCL2 (rCCL2, 1 µg) immediately after the surgery significantly promoted cardiomyocyte proliferation, improved survival rate and cardiac function, and diminished scar sizes in post-MI mice. Alongside these beneficial effects, we observed an increased angiogenesis and decreased cardiomyocyte apoptosis in post-MI mice. Conversely, treatment with a selective CCL2 synthesis inhibitor Bindarit (30 µM) suppressed both CCL2 expression and cardiomyocyte proliferation in P1 neonatal rat ventricle myocytes (NRVMs). We demonstrated in NRVMs that the CCL2 stimulated cardiomyocyte proliferation through STAT3 signaling: treatment with rCCL2 (100 ng/mL) significantly increased the phosphorylation levels of STAT3, whereas a STAT3 phosphorylation inhibitor Stattic (30 µM) suppressed rCCL2-induced cardiomyocyte proliferation. In conclusion, this study suggests that CCL2 promotes cardiac regeneration via activation of STAT3 signaling, underscoring its potential as a therapeutic agent for managing MI and associated heart failure.


Assuntos
Insuficiência Cardíaca , Infarto do Miocárdio , Humanos , Camundongos , Animais , Ratos , Quimiocina CCL2/metabolismo , Infarto do Miocárdio/metabolismo , Miócitos Cardíacos , Insuficiência Cardíaca/metabolismo , Regeneração , Camundongos Endogâmicos C57BL , Apoptose , Fator de Transcrição STAT3/metabolismo
2.
J Mol Cell Cardiol ; 185: 50-64, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-37918322

RESUMO

Better understanding of the mechanisms regulating the proliferation of pre-existing cardiomyocyte (CM) should lead to better options for regenerating injured myocardium. The absence of a perfect research model to definitively identify newly formed mammalian CMs is lacking. However, methodologies are being developed to identify and enrich proliferative CMs. These methods take advantages of the different proliferative states of CMs during postnatal development, before and after injury in the neonatal heart. New approaches use CMs labeled in lineage tracing animals or single cell technique-based CM clusters. This review aims to provide a timely update on the characteristics of the proliferative CMs, including their structural, functional, genetic, epigenetic and metabolic characteristics versus non-proliferative CMs. A better understanding of the characteristics of proliferative CMs should lead to the mechanisms for inducing endogenous CMs to self-renew, which is a promising therapeutic strategy to treat cardiac diseases that cause CM death in humans.


Assuntos
Cardiopatias , Miócitos Cardíacos , Animais , Recém-Nascido , Humanos , Miócitos Cardíacos/metabolismo , Proliferação de Células , Coração/fisiologia , Miocárdio , Mamíferos , Cardiopatias/metabolismo
3.
Acta Pharmacol Sin ; 44(6): 1149-1160, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-36473990

RESUMO

Heart failure with preserved ejection fraction (HFpEF) is highly prevalent, and lacks effective treatment. The aberration of WNT pathway underlies many pathological processes including cardiac fibrosis and hypertrophy, while porcupine is an acyltransferase essential for the secretion of WNT ligands. In this study we investigated the role of WNT signaling pathway in HFpEF as well as whether blocking WNT signaling by a novel porcupine inhibitor CGX1321 alleviated HFpEF. We established two experimental HFpEF mouse models, namely the UNX/DOCA model and high fat diet/L-NAME ("two-hit") model. The UNX/DOCA and "two-hit" mice were treated with CGX1321 (3 mg·kg-1·d-1) for 4 and 10 weeks, respectively. We showed that CGX1321 treatment significantly alleviated cardiac hypertrophy and fibrosis, thereby improving cardiac diastolic function and exercise performance in both models. Furthermore, both canonical and non-canonical WNT signaling pathways were activated, and most WNT proteins, especially WNT3a and WNT5a, were upregulated during the development of HEpEF in mice. CGX1321 treatment inhibited the secretion of WNT ligands and repressed both canonical and non-canonical WNT pathways, evidenced by the reduced phosphorylation of c-Jun and the nuclear translocation of ß-catenin and NFATc3. In an in vitro HFpEF model, MCM and ISO-treated cardiomyocytes, knockdown of porcupine by siRNA leads to a similar inhibitory effect on WNT pathways, cardiomyocyte hypertrophy and cardiac fibroblast activation as CGX1321 did, whereas supplementation of WNT3a and WNT5a reversed the anti-hypertrophy and anti-fibrosis effect of CGX1321. We conclude that WNT signaling activation plays an essential role in the pathogenesis of HFpEF, and porcupine inhibitor CGX1321 exerts a therapeutic effect on HFpEF in mice by attenuating cardiac hypertrophy, alleviating cardiac fibrosis and improving cardiac diastolic function.


Assuntos
Cardiomiopatias , Acetato de Desoxicorticosterona , Insuficiência Cardíaca , Animais , Camundongos , Cardiomegalia/patologia , Cardiomiopatias/patologia , Acetato de Desoxicorticosterona/farmacologia , Acetato de Desoxicorticosterona/uso terapêutico , Fibrose , Insuficiência Cardíaca/metabolismo , Miócitos Cardíacos , Volume Sistólico/fisiologia , Via de Sinalização Wnt
5.
BMC Cardiovasc Disord ; 22(1): 568, 2022 12 26.
Artigo em Inglês | MEDLINE | ID: mdl-36572851

RESUMO

BACKGROUND: GRACE risk score models are capable of predicting all-cause mortality of non-ST elevation myocardial infarction (NSTEMI) patients. However, its utility for evaluating major adverse cardiovascular events (MACE) in NSTEMI patients with multivessel disease (MVD) remains unclear. METHODS AND RESULTS: This study was designed as a retrospective cohort study that recruited patients with NSTEMI and multivessel disease between September 2013 and December 2018 in Daping Hospital, Chongqing, China. The primary outcome was a composite outcome that included all-cause mortality, recurrent angina, non-fatal myocardial infarction, coronary re-vascularization, and non-fatal strokes. Of the 827 patients with NSTEMI, 32 did not complete follow-up and 430 were excluded because of single-vessel disease. The remaining 365 NSTEMI patients with MVD had a median follow-up of 3.0 (IQR 2.6-3.3) years, 78 patients experienced outcomes. The GRACE risk score predicted the MACE (hazard ratio 1.014, 95% CI 1.006-1.021, P < 0.001). The GRACE risk score performed well in predicting all-cause mortality (c-statistic 0.72, 95% CI 0.59-0.85, P = 0.001) in MVD but was less powerful in predicting MACE (c-statistic 0.69, 95% CI 0.62-0.75, P < 0.001). When combining the GRACE risk score with the SYNTAX score, and blood urea nitrogen for predicting all-cause mortality and MACE events, the c-statistic value increased to 0.82 and 0.81 (P < 0.001). CONCLUSION: In NSTEMI patients with MVD, the GRACE score showed an acceptable predictive value for all-cause mortality, but it was less powerful in predicting MACE. Blood urea nitrogen may be valuable in assessing long-term cardiovascular events in patients with MVD.


Assuntos
Infarto do Miocárdio , Infarto do Miocárdio sem Supradesnível do Segmento ST , Humanos , Prognóstico , Infarto do Miocárdio sem Supradesnível do Segmento ST/diagnóstico , Infarto do Miocárdio sem Supradesnível do Segmento ST/terapia , Estudos Retrospectivos , Medição de Risco , Fatores de Risco
6.
Int Heart J ; 63(5): 814-820, 2022 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-36104232

RESUMO

Nonalcoholic fatty liver disease (NAFLD) is an emerging driver of cardiac arrhythmias. However, the relationship between NAFLD and malignant arrhythmia in non-ST-segment elevation myocardial infarction (NSTEMI) patients is still unclear.In this study, 358 NSTEMI inpatients were enrolled. They all received 24-hour Holter monitoring after percutaneous coronary intervention. All inpatients were divided into two groups: the non-NAFLD group (236 cases, 65.9%) and the NAFLD group (122 cases, 34.1%). Compared with the non-NAFLD group, the NAFLD group had a significantly higher incidence of PVCs/hour > 5 (premature ventricular complexes, 32.0% versus 9.3%, P < 0.001), ventricular tachycardia (VT, 22.1% versus 5.9%, P < 0.001), and sinus arrest (SA, 7.4% versus 1.3%, P = 0.002). We found that NAFLD was closely associated with the occurrence of VT [unadjusted odds ratio (OR) 4.507, 95% confidence interval (CI) 2.263-8.974, P < 0.001] and SA (OR 6.186, 95%CI 1.643-23.291, P = 0.007). After adjusting for age, sex, body mass index, and other confounding factors, the above differences were still statistically significant (VT: OR 4.808, 95%CI 2.254-10.253, P < 0.001; SA: OR 9.589, 95%CI 2.027-45.367, P = 0.004).NAFLD is associated with the occurrence of VT and SA in NSTEMI patients. It indicates that NAFLD might be a risk factor for malignant arrhythmias in post-NSTEMI patients.


Assuntos
Parada Cardíaca , Infarto do Miocárdio sem Supradesnível do Segmento ST , Hepatopatia Gordurosa não Alcoólica , Intervenção Coronária Percutânea , Infarto do Miocárdio com Supradesnível do Segmento ST , Taquicardia Ventricular , Complexos Ventriculares Prematuros , Parada Cardíaca/complicações , Humanos , Infarto do Miocárdio sem Supradesnível do Segmento ST/complicações , Hepatopatia Gordurosa não Alcoólica/complicações , Intervenção Coronária Percutânea/efeitos adversos , Fatores de Risco , Infarto do Miocárdio com Supradesnível do Segmento ST/complicações , Taquicardia Ventricular/complicações , Taquicardia Ventricular/etiologia , Complexos Ventriculares Prematuros/etiologia
7.
Basic Res Cardiol ; 117(1): 10, 2022 03 05.
Artigo em Inglês | MEDLINE | ID: mdl-35247074

RESUMO

The role of long non-coding RNA (lncRNA) in endogenous cardiac regeneration remains largely elusive. The mammalian cardiomyocyte is capable of regeneration for a brief period after birth. This fact allows the exploration of the roles of critical lncRNAs in the regulation of cardiac regeneration. Through a cardiac regeneration model by apical resection (AR) of the left ventricle in neonatal mice, we identified an lncRNA named natriuretic peptide A antisense RNA 1 (NPPA-AS1), which negatively regulated cardiomyocyte proliferation. In neonates, NPPA-AS1 deletion did not affect heart development, but was sufficient to prolong the postnatal window of regeneration after AR. In adult mice, NPPA-AS1 deletion improved cardiac function and reduced infarct size after myocardial infarction (MI), associated with a significant improvement in cardiomyocyte proliferation. Further analysis showed that NPPA-AS1 interacted with DNA repair-related molecule splicing factor, proline- and glutamine-rich (SFPQ). A heteromer of SFPQ and non-POU domain-containing octamer-binding protein (NONO) was required for double-strand DNA break repair, but NPPA-AS1 was competitively bound with SFPQ due to the overlapped binding sites of SFPQ and NONO. NPPA-AS1 deletion promoted the binding of SFPQ-NONO heteromer, decreased DNA damage, and activated cardiomyocyte cell cycle re-entry. Together, loss of NPPA-AS1 promoted cardiomyocyte proliferation by stabilizing SFPQ-NONO heteromer-induced DNA repair and exerted a therapeutic effect against MI in adult mice. Consequently, NPPA-AS1 may be a novel target for stimulating cardiac regeneration to treat MI.


Assuntos
Infarto do Miocárdio , RNA Longo não Codificante , Animais , Fator Natriurético Atrial , Proliferação de Células , Reparo do DNA , Proteínas de Ligação a DNA , Mamíferos , Camundongos , Infarto do Miocárdio/genética , Miócitos Cardíacos , Procainamida/análogos & derivados , RNA Longo não Codificante/genética , Proteínas de Ligação a RNA , Regeneração
8.
Biochem Biophys Res Commun ; 589: 267-274, 2022 01 22.
Artigo em Inglês | MEDLINE | ID: mdl-34933200

RESUMO

The deprivation of myocardial nutrition causes cardiomyocyte death and disturbance of energy metabolism. IKKε plays an important regulatory role in many biological events such as inflammation, redox reaction, cell death, etc. However, the more in-depth mechanism by which IKKε contributes to cardiomyocytes death in nutrition deprivation remains poorly understood. IKKε expression was knocked down by siRNA in H9c2 cells, and cells were cultured under starvation conditions to simulate ischemic conditions. Starvation triggered greater NLRP3 activation, accompanied by more IL-1ß, IL-18 and caspase-1 release in the siIKKε H9c2 cells compared with the control H9c2 cells. Western blot and immunofluorescence showed that the IKKε konckdown promoted NLRP3 expressions and ROS release under starvation conditions. Furthermore, electron micrography and JC-1 analysis revealed that IKKε konckdown resulted in aggravated mitochondrial damage and more mitochondrial ROS (mtROS) released in vitro. Notably, Western blot analysis showed that IKKε deficiency activated the TBK1 and IRF3 signaling pathways to promote pyroptosis in vitro. Collectively, our results indicate that IKKε protects against cardiomyocyte injury by reducing mitochondrial damage and NLRP3 expression following nutrition deprivation via regulation of the TBK1/IRF3 signaling pathway. This study further revealed the mechanism of IKKε in inflammation and myocardial nutrition deprivation.


Assuntos
Citoproteção , Quinase I-kappa B/metabolismo , Inflamassomos/metabolismo , Mitocôndrias/patologia , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Piroptose , Animais , Linhagem Celular , Técnicas de Silenciamento de Genes , Quinase I-kappa B/deficiência , Fator Regulador 3 de Interferon/metabolismo , Fosforilação , Proteínas Serina-Treonina Quinases/metabolismo , Ratos
9.
Front Cell Dev Biol ; 9: 673993, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34790655

RESUMO

Infection is thought to be involved in the pathogenesis of atherosclerosis. Studies have shown the association between helicobacter pylori (H. pylori) and coronary artery disease. It is interesting to find H. pylori DNA and cytotoxin-associated gene A (CagA) protein in atherosclerotic plaque. Outer membrane vesicles (OMVs), secreted by H. pylori, exert effects in the distant organ or tissue. However, whether or not OMVs from H. pylori are involved in the pathogenesis of atherosclerosis remains unknown. Our present study found that treatment with OMVs from CagA-positive H. pylori accelerated atherosclerosis plaque formation in ApoE-/- mice. H. pylori-derived OMVs inhibited proliferation and promoted apoptosis of human umbilical vein endothelial cells (HUVECs), which was also reflected in in vivo studies. These effects were normalized to some degree after treatment with lipopolysaccharide (LPS)-depleted CagA-positive OMVs or CagA-negative OMVs. Treatment with H. pylori-derived OMVs increased reactive oxygen species (ROS) levels and enhanced the activation of nuclear factor-κB (NF-κB) in HUVECs, which were reversed to some degree in the presence of a superoxide dismutase mimetic TEMPOL and a NF-κB inhibitor BAY11-7082. Expressions of interleukin-6 (IL-6) and tumor necrosis factor alpha (TNF-α), two inflammatory factors, were augmented after treatment with OMVs from H. pylori. These suggest that H. pylori-derived OMVs accelerate atherosclerosis plaque formation via endothelium injury. CagA and LPS from H. pylori-OMVs, at least in part, participate in these processes, which may be involved with the activation of ROS/NF-κB signaling pathway. These may provide a novel strategy to reduce the incidence and development of atherosclerosis.

10.
J Am Heart Assoc ; 10(21): e021895, 2021 11 02.
Artigo em Inglês | MEDLINE | ID: mdl-34713723

RESUMO

Background Exploring potential therapeutic target is of great significance for myocardial infarction (MI) and post-MI heart failure. Transcription factor Yin-Yang 1 (YY1) is an essential regulator of apoptosis and angiogenesis, but its role in MI is unclear. Methods and Results The expression of YY1 was assessed in the C57BL/6J mouse heart following MI. Overexpression or silencing of YY1 in the mouse heart was achieved by adeno-associated virus 9 injection. The survival, cardiac function, and scar size, as well as the apoptosis, angiogenesis, cardiac fibrosis, T helper 2 lymphocyte cytokine production, and macrophage polarization were assessed. The effects of YY1 on Akt phosphorylation and vascular endothelial growth factor production were also investigated. The expression of YY1 in heart was significantly stimulated by MI. The survival rate, cardiac function, scar size, and left ventricular volume of mice were improved by YY1 overexpression but worsened by YY1 silencing. YY1 alleviated cardiac apoptosis and fibrosis, promoted angiogenesis, T helper 2 cytokine production, and M2 macrophage polarization in the post-MI heart, it also enhanced the tube formation and migration ability of endothelial cells. Enhanced Akt phosphorylation, along with the increased vascular endothelial growth factor levels were observed in presence of YY1 overexpression. Conclusions YY1 ameliorates cardiac injury and remodeling after MI by repressing cardiomyocyte apoptosis and boosting angiogenesis, which might be ascribed to the enhancement of Akt phosphorylation and the subsequent vascular endothelial growth factor up-regulation. Increased T helper 2 cytokine production and M2 macrophage polarization may also be involved in YY1's cardioprotective effects. These findings supported YY1 as a potential target for therapeutic investigation of MI.


Assuntos
Traumatismos Cardíacos , Infarto do Miocárdio , Animais , Apoptose , Cicatriz , Citocinas , Modelos Animais de Doenças , Células Endoteliais , Camundongos , Camundongos Endogâmicos C57BL , Infarto do Miocárdio/prevenção & controle , Miócitos Cardíacos , Proteínas Proto-Oncogênicas c-akt , Fator A de Crescimento do Endotélio Vascular , Remodelação Ventricular , Yin-Yang
11.
Sci Rep ; 11(1): 15836, 2021 08 04.
Artigo em Inglês | MEDLINE | ID: mdl-34349170

RESUMO

Acute myocardial infarction (MI) is one of the leading causes of death in humans. Our previous studies showed that gastrin alleviated acute myocardial ischaemia-reperfusion injury. We hypothesize that gastrin might protect against heart injury after MI by promoting angiogenesis. An MI model was simulated by ligating the anterior descending coronary artery in adult male C57BL/6J mice. Gastrin was administered twice daily by intraperitoneal injection for 2 weeks after MI. We found that gastrin reduced mortality, improved myocardial function with reduced infarct size and promoted angiogenesis. Gastrin increased HIF-1α and VEGF expression. Downregulation of HIF-1α expression by siRNA reduced the proliferation, migration and tube formation of human umbilical vein endothelial cells. These results indicate that gastrin restores cardiac function after MI by promoting angiogenesis via the HIF-1α/VEGF pathway.


Assuntos
Gastrinas/farmacologia , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Infarto do Miocárdio/tratamento farmacológico , Neovascularização Patológica/prevenção & controle , Fator A de Crescimento do Endotélio Vascular/metabolismo , Animais , Proliferação de Células , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Infarto do Miocárdio/patologia , Transdução de Sinais , Fator A de Crescimento do Endotélio Vascular/genética
12.
Exp Ther Med ; 21(4): 358, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-33732331

RESUMO

Νon-small cell lung cancer (NSCLC) is the most frequently diagnosed type of cancer, and the most prevalent cause of cancer-associated mortality. The present study aimed to investigate whether microRNA (miR)-564 influences NSCLC progression by regulating NSCLC cell growth and migration, via targeting plexin A4. Therefore, the expression levels of miR-564 and plexin A4 were evaluated in NSCLC specimens or cells using reverse transcription-quantitative PCR. Furthermore, colony formation and Cell Counting Kit-8 assays were performed to determine the proliferative ability of NSCLC cells. The cell migration capacity was assessed using a Transwell assay. In addition, to examine the binding ability of miR-564 on the plexin A4 3'-untranslated region (3'UTR), a dual-luciferase reporter assay was performed. A mouse xenograft model was established to evaluate the effect of miR-564 knockdown on tumor growth in vivo, whereas the expression of plexin A4 and Ki67 in NSCLC tissues was detected using immunohistochemistry. Notably, miR-564 was downregulated in both NSCLC cell lines and tissues, while its overexpression, following transfection with miR-564 mimics, attenuated the proliferation and proliferation, migration and invasion of NSCLC cells. By contrast, silencing of miR-564 using a miR-564 inhibitor promoted NSCLC cell proliferation, migration and invasion. The luciferase assay revealed that miR-564 directly targeted the plexin A4 3'UTR in A549 and H460 cells. Additionally, the overexpression of plexin A4 rescued the effect of miR-564 on NSCLC cell proliferation, migration and invasion abilities. Further in vivo studies demonstrated that miR-564 knockdown promoted NSCLC growth, while miR-564 overexpression resulted in the opposite effect in nude mice. Overall, the results of the present study revealed that miR-564 promotes the proliferation and migration of NSCLC cells, both in vitro and in vivo, via targeting plexin A4. Therefore, miR-564 may be considered as a possible therapeutic target for NSCLC.

13.
Oxid Med Cell Longev ; 2021: 6429197, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33628362

RESUMO

I-κB kinase-ε (IKKε) is a member of the IKK complex and a proinflammatory regulator that is active in many diseases. Angiotensin II (Ang II) is a vasoconstricting peptide hormone, and Ang II-induced myocardial hypertrophy is a common cardiovascular disease that can result in heart failure. In this study, we sought to determine the role of IKKε in the development of Ang II-induced myocardial hypertrophy in mice. Wild-type (WT) and IKKε-knockout (IKKε-KO) mice were generated and infused with saline or Ang II for 8 weeks. We found that WT mouse hearts have increased IKKε expression after 8 weeks of Ang II infusion. Our results further indicated that IKKε-KO mice have attenuated myocardial hypertrophy and alleviated heart failure compared with WT mice. Additionally, Ang II-induced expression of proinflammatory and collagen factors was much lower in the IKKε-KO mice than in the WT mice. Apoptosis and pyroptosis were also ameliorated in IKKε-KO mice. Mechanistically, IKKε bound to extracellular signal-regulated kinase (ERK) and the mitogen-activated protein kinase p38, resulting in MAPK/ERK kinase (MEK) phosphorylation, and IKKε deficiency inhibited the phosphorylation of MEK-ERK1/2 and p38 in mouse heart tissues after 8 weeks of Ang II infusion. The findings of our study reveal that IKKε plays an important role in the development of Ang II-induced myocardial hypertrophy and may represent a potential therapeutic target for the management of myocardial hypertrophy.


Assuntos
Cardiomegalia/enzimologia , Cardiomegalia/patologia , Quinase I-kappa B/deficiência , Miocárdio/patologia , Angiotensina II , Animais , Colágeno/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Fibrose , Insuficiência Cardíaca/complicações , Quinase I-kappa B/metabolismo , Inflamação/complicações , Inflamação/patologia , Sistema de Sinalização das MAP Quinases , Camundongos Knockout , Piroptose , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
14.
Interact Cardiovasc Thorac Surg ; 32(6): 855-863, 2021 05 27.
Artigo em Inglês | MEDLINE | ID: mdl-33611582

RESUMO

OBJECTIVES: Left ventricular systolic dysfunction (LVSD) is common and associated with adverse events in patients receiving coronary artery bypass grafting (CABG). However, the prognosis of mild LVSD has not been clearly described. We aimed to evaluate the mid-term outcomes of patients with mild LVSD following CABG. METHODS: This multicentre cohort study using propensity score matching took place from December 2012 to October 2019 in Jiangsu Province, China, with a mean and maximum follow-up of 3.2 and 7.2 years, respectively. Patients were classified to normal left ventricular systolic function (left ventricular ejection fraction ≥53%) and mild LVSD (left ventricular ejection fraction >40%/<53%). The primary outcomes were death from all causes and death from cardiovascular causes. The secondary outcomes were heart failure, myocardial infarction, repeat revascularization and a composite of all mentioned outcomes, including death from all causes (major adverse events). RESULTS: A total of 581 pairs were formed after matching. In-hospital death (1.5% vs 2.1%, P = 0.51) did not differ between 2 cohorts. Throughout 7 years, mild LVSD was associated with higher rates of death from all causes [hazard ratio (HR) 0.59, 95% confidence interval (CI) 0.39-0.89; P = 0.012], death from cardiovascular causes (HR 0.55, 95% CI 0.36-0.90; P = 0.017), heart failure (HR 0.60, 95% CI 0.37-0.93; P = 0.023) and major adverse events (HR 0.66, 95% CI 0.49-0.91; P = 0.009). There was no difference in the rates of myocardial infarction and repeat revascularization. CONCLUSIONS: Mild LVSD was associated with a worse mid-term prognosis in patients following CABG.


Assuntos
Doença da Artéria Coronariana , Disfunção Ventricular Esquerda , Estudos de Coortes , Ponte de Artéria Coronária/efeitos adversos , Doença da Artéria Coronariana/complicações , Doença da Artéria Coronariana/diagnóstico por imagem , Doença da Artéria Coronariana/cirurgia , Mortalidade Hospitalar , Humanos , Estudos Retrospectivos , Volume Sistólico , Resultado do Tratamento , Disfunção Ventricular Esquerda/etiologia , Função Ventricular Esquerda
15.
Acta Histochem ; 123(1): 151653, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-33246224

RESUMO

OBJECTIVE: Cardiac hypertrophy is an adaptive response to physiological and pathological stimuli, the latter of which frequently progresses to valvulopathy, heart failure and sudden death. Recent reports revealed that pyroptosis is involved in regulating multiple cardiovascular diseases progression, including cardiac hypertrophy. However, the underlying mechanisms remain poorly understood. This study aims to extensively investigate the regulation of miR-133a-3p on pyroptosis in angiotensin II (Ang II)-induced cardiac hypertrophyin vitro. METHODS: The in vitro model of cardiac hypertrophy was induced by Ang II, which was validated by qPCR combined with measurement of cell surface area by immunofluorescence assay. CCK-8 assay and Hochest33342/PI staining was performed to assess pyroptosis. Dual luciferase reporter system was used to verify the direct interaction between miR-133a-3p and IKKε. The effects of miR-133a-3p/IKKε on pyroptosis activation and cardiac hypertrophy markers (Caspase-1, NLRP3, IL-1ß, IL-18, GSDMD, ASC, ANP, BNP and ß-MHC) were evaluated by western blot, ELISA and qPCR. RESULTS: Ang II treatment could induce cardiomyocyte hypertrophy and pyroptosis. The expression of miR-133a-3p was repressed in Ang II-treated HCM cells, and its overexpression could attenuate both pyroptosis and cardiac hypertrophyin vitro. Additionally, IKKε expression was significantly up-regulated in Ang II-induced HCM cells. Dual luciferase reporter system and qPCR validated that miR-133a-3p directly targeted the 3'-UTR of IKKε and suppressed its expression. Moreover, IKKε overexpression impaired the protective function of miR-133a-3p in cardiomyocyte hypertrophy. CONCLUSION: Collectively, miR-133a-3p attenuates Ang II induced cardiomyocyte hypertrophy via inhibition of pyroptosis by targeting IKKε. Therefore, miR-133a-3p up-regulation may be a promising strategy for cardiac hypertrophy treatment.


Assuntos
Cardiomegalia/metabolismo , Regulação Enzimológica da Expressão Gênica , Quinase I-kappa B/biossíntese , MicroRNAs/metabolismo , Miócitos Cardíacos/metabolismo , Piroptose , Linhagem Celular , Citocinas/metabolismo , Ativação Enzimática , Humanos , Miócitos Cardíacos/patologia
16.
Eur Heart J ; 42(14): 1415-1430, 2021 04 07.
Artigo em Inglês | MEDLINE | ID: mdl-33280021

RESUMO

AIMS: G protein-coupled receptor kinase 4 (GRK4) has been reported to play an important role in hypertension, but little is known about its role in cardiomyocytes and myocardial infarction (MI). The goal of present study is to explore the role of GRK4 in the pathogenesis and progression of MI. METHODS AND RESULTS: We studied the expression and distribution pattern of GRK4 in mouse heart after MI. GRK4 A486V transgenic mice, inducible cardiomyocyte-specific GRK4 knockout mice, were generated and subjected to MI with their control mice. Cardiac infarction, cardiac function, cardiomyocyte apoptosis, autophagic activity, and HDAC4 phosphorylation were assessed. The mRNA and protein levels of GRK4 in the heart were increased after MI. Transgenic mice with the overexpression of human GRK4 wild type (WT) or human GRK4 A486V variant had increased cardiac infarction, exaggerated cardiac dysfunction and remodelling. In contrast, the MI-induced cardiac dysfunction and remodelling were ameliorated in cardiomyocyte-specific GRK4 knockout mice. GRK4 overexpression in cardiomyocytes aggravated apoptosis, repressed autophagy, and decreased beclin-1 expression, which were partially rescued by the autophagy agonist rapamycin. MI also induced the nuclear translocation of GRK4, which inhibited autophagy by increasing HDAC4 phosphorylation and decreasing its binding to the beclin-1 promoter. HDAC4 S632A mutation partially restored the GRK4-induced inhibition of autophagy. MI caused greater impairment of cardiac function in patients carrying the GRK4 A486V variant than in WT carriers. CONCLUSION: GRK4 increases cardiomyocyte injury during MI by inhibiting autophagy and promoting cardiomyocyte apoptosis. These effects are mediated by the phosphorylation of HDAC4 and a decrease in beclin-1 expression.


Assuntos
Quinase 4 de Receptor Acoplado a Proteína G/fisiologia , Infarto do Miocárdio , Miócitos Cardíacos , Animais , Apoptose , Autofagia , Proteína Beclina-1 , Histona Desacetilases , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Remodelação Ventricular
17.
Front Cell Dev Biol ; 8: 570252, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33117800

RESUMO

To identify and analyze the live proliferating cardiomyocytes is crucial for deciphering the mechanisms controlling endogenous cardiac regeneration. Traditional methods confuse cell division with multinucleation in postnatal cardiomyocytes. Recent efforts have achieved significant progress on discerning cytokinesis from only nuclear division. However, those methods were either designed to label post-cytokinesis progeny or challenging to sort the live proliferating cardiomyocytes. In this study, we highlighted an Aurora kinase B reporter-based mouse system with a tdTomato fluorescence labeling. It could efficiently identify proliferating cardiomyocytes in neonates. The analysis of sorting tdTomato+ cardiomyocytes with different ploidy indicated that mononucleated cardiomyocytes might not possess significantly higher proliferating potential than other cardiomyocytes when most cardiomyocytes have become post-mitotic. Moreover, tdTomato+ cardiomyocytes were significantly increased and enriched at injury border zone after apex resection in neonates, while there were no increased tdTomato+ cardiomyocytes after myocardial infarction in adults.

18.
Cell Prolif ; 53(11): e12910, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-33047378

RESUMO

OBJECTIVES: The mechanisms responsible for the postnatal loss of mammalian cardiac regenerative capacity are not fully elucidated. The aim of the present study is to investigate the role of progesterone in cardiac regeneration and explore underlying mechanism. MATERIALS AND METHODS: Effect of progesterone on cardiomyocyte proliferation was analysed by immunofluorescent staining. RNA sequencing was performed to screen key target genes of progesterone, and yes-associated protein (YAP) was knocked down to demonstrate its role in pro-proliferative effect of progesterone. Effect of progesterone on activity of YAP promoter was measured by luciferase assay and interaction between progesterone receptor and YAP promoter by electrophoretic mobility shift assay (EMSA) and chromatin immunoprecipitation (ChIP). Adult mice were subjected to myocardial infarction, and then, effects of progesterone on adult cardiac regeneration were analysed. RESULTS: Progesterone supplementation enhanced cardiomyocyte proliferation in a progesterone receptor-dependent manner. Progesterone up-regulated YAP expression and knockdown of YAP by small interfering RNA reduced progesterone-mediated cardiomyocyte proliferative effect. Progesterone receptor interacted with the YAP promoter, determined by ChIP and EMSA; progesterone increased luciferase activity of YAP promoter and up-regulated YAP target genes. Progesterone administration also promoted adult cardiomyocyte proliferation and improved cardiac function in myocardial infarction. CONCLUSION: Our data uncover a role of circulating progesterone withdrawal as a novel mechanism for the postnatal loss of mammalian cardiac regenerative potential. Progesterone promotes both neonatal and adult cardiomyocyte proliferation by up-regulating YAP expression.


Assuntos
Proteínas Reguladoras de Apoptose/genética , Cardiotônicos/farmacologia , Proliferação de Células/efeitos dos fármacos , Miócitos Cardíacos/efeitos dos fármacos , Progesterona/farmacologia , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Cardiotônicos/uso terapêutico , Proteínas de Ciclo Celular/genética , Células Cultivadas , Masculino , Camundongos Endogâmicos C57BL , Infarto do Miocárdio/tratamento farmacológico , Infarto do Miocárdio/genética , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Progesterona/uso terapêutico , Ratos Sprague-Dawley , Regulação para Cima/efeitos dos fármacos , Proteínas de Sinalização YAP
19.
Oxid Med Cell Longev ; 2020: 7046923, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32724494

RESUMO

Autophagy of cardiomyocytes after myocardial infarction (MI) is an important factor affecting the prognosis of MI. Excessive autophagy can lead to massive death of cardiomyocytes, which will seriously affect cardiac function. IKKε plays a crucial role in the occurrence of autophagy, but the functional role in MI remains largely unknown. To evaluate the impact of IKKε on the autophagy of cardiomyocytes after MI, MI was induced by surgical left anterior descending coronary artery ligation in IKKε knockout (KO) mice and wild-type (WT) mice. Starvation of H9c2 cells with IKKε siRNA and rescued with IKKε overexpressed afterwards to test the mechanism of IKKε in autophagy in vitro. Our results demonstrated that the expression of IKKε was upregulated in mice myocardial tissues which were consistent with cardiomyocyte autophagy after MI. Significantly, the IKKε KO mice showed increased infarct size, decreased viable cardiomyocytes, and exacerbated cardiac dysfunction when compared with the wild-type mice. Western blot and electron micrography analysis also revealed that loss of IKKε induces excessive cardiomyocyte autophagy and reduced the expression of p-Akt and p-mTOR. Similar results were observed in IKKε siRNA H9c2 cells in vitro which were under starvation injury. Notably, the levels of p-Akt and p-mTOR can restore in IKKε rescued cells. In conclusion, our results indicated that IKKε protects cardiomyocyte survival by reduced autophagy following MI via regulation of the Akt/mTOR signaling pathway. Thus, our study suggests that IKKε might represent a potential therapeutic target for the treatment of MI.


Assuntos
Quinase I-kappa B/metabolismo , Miócitos Cardíacos/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Animais , Autofagia , Humanos , Camundongos , Infarto do Miocárdio
20.
J Cardiothorac Surg ; 15(1): 183, 2020 Jul 23.
Artigo em Inglês | MEDLINE | ID: mdl-32703274

RESUMO

BACKGROUND: This study compared limited aortic repair (ascending, and /or hemi-arch replacement) versus extended-arch repair (ascending, arch and proximal descending aortic replacement) used for patients aged 65 or older, who had type A acute aortic dissection (AAD), analyzing the influence of the extent of aortic repair on outcomes. METHODS: From January, 2001 to December, 2015, 103 patients aged 65 or older underwent operation due to type A AAD in Nanjing First Hospital. The cohort was divided into two subgroups according to the surgical approaches, including limited aortic replacement (LAR, n = 41) and total arch replacement + stent elephant trunk implantation (TAR+SET, n = 62). RESULTS: There was no significant difference in gender, age, hypertension, diabetes, smoking, PCI history, atrial fibrillation, pericardial effusion, aortic valve insufficiency (≥ moderate), shock situation before operation, and Euro-score II between the two groups except limb malperfusion and tear location. The cross-clamp time, CPB time, intubation time, ICU stay time and hospital time were all significantly less in the LAR group than in the TAR+SET group. A total of 89 patients were discharged home successfully after operation, with a difference of hospital mortality (P = 0.04). The overall survival rates at 5-year follow-up were 82.5 ± 6.0% in LAR group and 75.2 ± 5.6% in TAR+SET group, but with no difference (p = 0.151). The freedom from adverse aortic events at 5-year was 84.3 ± 6.5% in LAR group versus 97.9 ± 2.1% in TAR+SET group, with a statistical difference (p = 0.03). CONCLUSION: These findings support limited aortic resection is acceptable for elderly patients with type A AAD if surgical principles allow.


Assuntos
Aorta/cirurgia , Aneurisma Aórtico/cirurgia , Dissecção Aórtica/cirurgia , Procedimentos Cirúrgicos Vasculares/métodos , Fatores Etários , Idoso , Idoso de 80 Anos ou mais , Dissecção Aórtica/mortalidade , Aneurisma Aórtico/mortalidade , Implante de Prótese Vascular , Feminino , Seguimentos , Humanos , Masculino , Estudos Retrospectivos , Taxa de Sobrevida , Resultado do Tratamento
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...