Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Free Radic Biol Med ; 210: 25-41, 2024 01.
Article in English | MEDLINE | ID: mdl-37956909

ABSTRACT

Metabolic reprogramming of vascular smooth muscle cell (VSMC) plays a critical role in the pathogenesis of thoracic aortic dissection (TAD). Previous researches have mainly focused on dysregulation of fatty acid or glucose metabolism, while the impact of amino acids catabolic disorder in VSMCs during the development of TAD remains elusive. Here, we identified branched-chain amino acid (BCAA) catabolic defect as a metabolic hallmark of TAD. The bioinformatics analysis and data from human aorta revealed impaired BCAA catabolism in TAD individuals. This was accompanied by upregulated branched-chain α-ketoacid dehydrogenase kinase (BCKDK) expression and BCKD E1 subunit alpha (BCKDHA) phosphorylation, enhanced vascular inflammation, and hyperactivation of mTOR signaling. Further in vivo experiments demonstrated that inhibition of BCKDK with BT2 (a BCKDK allosteric inhibitor) treatment dephosphorylated BCKDHA and re-activated BCAA catabolism, attenuated VSMCs phenotypic switching, alleviated aortic remodeling, mitochondrial reactive oxygen species (ROS) damage and vascular inflammation. Additionally, the beneficial actions of BT2 were validated in a TNF-α challenged murine VSMC cell line. Meanwhile, rapamycin conferred similar beneficial effects against VSMC phenotypic switching, cellular ROS damage as well as inflammatory response. However, co-treatment with MHY1485 (a classic mTOR activator) reversed the beneficial effects of BT2 by reactivating mTOR signaling. Taken together, the in vivo and in vitro evidence showed that impairment of BCAA catabolism resulted in aortic accumulation of BCAA and further caused VSMC phenotypic switching, mitochondrial ROS damage and inflammatory response via mTOR hyperactivation. BCKDK and mTOR signaling may serve as the potential drug targets for the prevention and treatment of TAD.


Subject(s)
Dissection, Thoracic Aorta , Muscle, Smooth, Vascular , Animals , Humans , Mice , Amino Acids, Branched-Chain/metabolism , Inflammation/pathology , Muscle, Smooth, Vascular/metabolism , Reactive Oxygen Species , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism
2.
Biotechnol Genet Eng Rev ; : 1-13, 2023 Mar 23.
Article in English | MEDLINE | ID: mdl-36951429

ABSTRACT

To explore the efficiency of nutritional support therapy . Pharmacists led the construction of an individualized nutritional computing system and were involved in the process of treatment. After obtaining relevant professional knowledge and instruction on how to operate the system, MDT members intervened in the incorrect treatment process during nutritional support therapy. The Department of Radiation Oncology and the Intensive Care Unit (ICU) were selected as pilot departments to compare and analyze the rationality of nutrition risk screening and the use of enteral nutrition (EN) and parenteral nutrition (PN) in treatment before and after intervention. The individualized nutritional computing system significantly improved work efficiency, promoted nutrition risk screening, and saved 10-15 minutes in the treatment of each patient. After intervention in the Department of Radiation Oncology, the use rate of Total Nutrient Admixture (TNA) increased by 7.17%, and the single-bottle infusion rate of PN preparation decreased by 17.94% in patients at risk of malnutrition. The use rate of EN and single-bottle infusion rate of PN preparation in patients without risk of malnutrition decreased by 15.17% and 20.81%, respectively. Overall, 98.75% of ICU patients were at risk of malnutrition. The use rates of EN and TNA increased by 12.79% and 12.14%, respectively, and the single-bottle infusion rate of PN preparation decreased by 10.06%. Streamlined and mobile MDT, the use of an individualized nutritional computing system, and the effective work of pharmacists in the process significantly improved the efficiency and rationality of nutritional support therapy .

3.
Redox Biol ; 59: 102594, 2023 02.
Article in English | MEDLINE | ID: mdl-36603528

ABSTRACT

The potential coexistence of Alzheimer's disease (AD) and atrial fibrillation (AF) is increasingly common as aging-related diseases. However, little is known about mechanisms responsible for atrial remodeling in AD pathogenesis. α7 nicotinic acetylcholine receptors (α7nAChR) has been shown to have profound effects on mitochondrial oxidative stress in both organ diseases. Here, we investigate the role of α7nAChR in mediating the effects of amyloid-ß (Aß) in cultured mouse atrial cardiomyocytes (HL-1 cells) and AD model mice (APP/PS1). In vitro, apoptosis, oxidative stress and mitochondrial dysfunction induced by Aß long-term (72h) in HL-1 cells were prevented by α-Bungarotoxin(α-BTX), an antagonist of α7nAChR. This cardioprotective effect was due to reinstating Ca2+ mishandling by decreasing the activation of CaMKII and MAPK signaling pathway, especially the oxidation of CaMKII (oxi-CaMKII). In vivo studies demonstrated that targeting knockdown of α7nAChR in cardiomyocytes could ameliorate AF progression in late-stage (12 months) APP/PS1 mice. Moreover, α7nAChR deficiency in cardiomyocytes attenuated APP/PS1-mutant induced atrial remodeling characterized by reducing fibrosis, atrial dilation, conduction dysfunction, and inflammatory mediator activities via suppressing oxi-CaMKII/MAPK/AP-1. Taken together, our findings suggest that diminished α7nAChR could rescue Aß-induced atrial remodeling through oxi-CaMKII/MAPK/AP-1-mediated mitochondrial oxidative stress in atrial cells and AD mice.


Subject(s)
Alzheimer Disease , Atrial Fibrillation , Atrial Remodeling , Animals , Mice , alpha7 Nicotinic Acetylcholine Receptor/genetics , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Atrial Fibrillation/genetics , Atrial Fibrillation/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Mitochondria/metabolism , Oxidative Stress , Transcription Factor AP-1/metabolism , Mitogen-Activated Protein Kinases/metabolism
4.
Apoptosis ; 28(3-4): 607-626, 2023 04.
Article in English | MEDLINE | ID: mdl-36708428

ABSTRACT

Ferroptosis contributes to the pathogenesis of atrial fibrillation (AF), although the mechanisms are still largely uncovered. The current study was designed to explore the pharmacological effects of icariin against ethanol-induced atrial remodeling, if any, and the mechanisms involved with a focus on SIRT1 signaling. Excessive ethanol-treated animals were administered with Ferrostatin-1, Erastin or icariin to evaluate the potential effects of icariin or ferroptosis. Then, the underling mechanisms was further explored in the in vitro experiments using HL-1 atrial myocytes. Excessive ethanol administration caused significant atrial damage as evidenced by increased susceptibility to AF, altered atrial conduction pattern, atrial enlargement, and enhanced fibrotic markers. These detrimental effects were reversed by Ferrostatin-1 or icariin treatment, while Erastin co-administration markedly abolished the beneficial actions conferred by icariin. Mechanistically, ethanol-treated atria exhibited markedly up-regulated pro-ferroptotic protein (PTGS2, ACSL4, P53) and suppressed anti-ferroptotic molecules (GPX4, FTH1). Icariin treatment inhibited ethanol-induced atrial ferroptosis by reducing atrial mitochondrial damage, ROS accumulation and iron overload. Interestingly, the in vivo and in vitro data showed that icariin activated atrial SIRT1-Nrf-2-HO-1 signaling pathway, while EX527 not only reversed these effects, but also abolished the therapeutic effects of icariin. Moreover, the stimulatory effects on GPX4, SLC7A11 and the suppressive effects on ACSL4, P53 conferred by icariin were blunted by EX527 treatment. These data demonstrate that ferroptosis plays a causative role in the pathogenesis of ethanol-induced atrial remodeling and susceptibility to AF. Icariin protects against atrial damage by inhibiting ferroptosis via SIRT1 signaling. Its role as a prophylactic/therapeutic drug deserves further clinical study.


Subject(s)
Atrial Fibrillation , Atrial Remodeling , Ferroptosis , Animals , Atrial Fibrillation/chemically induced , Atrial Fibrillation/drug therapy , Apoptosis , Sirtuin 1/genetics , Tumor Suppressor Protein p53 , Ethanol/toxicity
5.
Front Cardiovasc Med ; 9: 968014, 2022.
Article in English | MEDLINE | ID: mdl-36312282

ABSTRACT

Background: Atrial fibrillation (AF) is the most frequent arrythmia managed in clinical practice. Several mechanisms have been proposed to contribute to the occurrence and persistence of AF, in which oxidative stress plays a non-negligible role. The endocannabinoid system (ECS) is involved in a variety physiological and pathological processes. Cannabinoid receptor 1 (CB1R) and cannabinoid receptor 2 (CB2R) are expressed in the heart, and studies have shown that activating CB2R has a protective effect on the myocardium. However, the role of CB2R in AF is unknown. Materials and methods: Angiotensin II (Ang II)-infused mice were treated with the CB2R agonist AM1241 intraperitoneally for 21 days. Atrial structural remodeling, AF inducibility, electrical transmission, oxidative stress and fibrosis were measured in mice. Results: The susceptibility to AF and the level of oxidative stress were increased significantly in Ang II-infused mice. In addition, nicotinamide adenine dinucleotide phosphate oxidase 2 (NOX2), NOX4, and oxidized Ca2+/calmodulin-dependent protein kinase II (ox-CaMKII) were highly expressed. More importantly, treatment with AM1241 activated CB2R, resulting in a protective effect. Conclusion: The present study demonstrates that pharmacological activation of CB2R exerts a protective effect against AF via a potential NOX/CaMKII mechanism. CB2R is a potential therapeutic target for AF.

6.
Biochim Biophys Acta Mol Basis Dis ; 1868(10): 166483, 2022 10 01.
Article in English | MEDLINE | ID: mdl-35798229

ABSTRACT

Excessive alcohol consumption has long been identified as a risk factor for adverse atrial remodeling and atrial fibrillation (AF). Icariin is a principal active component from traditional Chinese medicine Herba Epimedii and has been demonstrated to exert potential antiarrhythmic effect. The present study was designed to evaluate the effect of icariin against alcohol-induced atrial remodeling and disruption of mitochondrial dynamics and furthermore, to elucidate the underlying mechanisms. Excessive alcohol-treated C57BL/6 J mice were infected with serotype 9 adeno-associated virus (AAV9) carrying mouse SIRT3 gene or negative control virus. Meanwhile, icariin (50 mg/kg/d) was administered to the animals in the presence or absence of AAV9 carrying SIRT3 shRNA. We noted that 8 weeks of icariin treatment effectively attenuated alcohol consumption-induced atrial structural and electrical remodeling as evidenced by reduced AF inducibility and reversed atrial electrical conduction pattern as well as atrial enlargement. Furthermore, icariin-treated group exhibited significantly enhanced atrial SIRT3-AMPK signaling, decreased atrial mitoSOX fluorescence and mitochondrial fission markers, elevated mitochondrial fusion markers (MFN1, MFN2) as well as NRF-1-Tfam-mediated mitochondrial biogenesis. Importantly, these beneficial effects were mimicked by SIRT3 overexpression while abolished by SIRT3 knockdown. These data revealed that targeting atrial SIRT3-AMPK signaling and preserving mitochondrial dynamics might serve as the novel therapeutic strategy against alcohol-induced AF genesis. Additionally, icariin ameliorated atrial remodeling and mitochondrial dysfunction by activating SIRT3-AMPK signaling, highlighting the use of icariin as a promising antiarrhythmic agent in this circumstance.


Subject(s)
Atrial Fibrillation , Atrial Remodeling , Flavonoids , Sirtuin 3 , AMP-Activated Protein Kinases/genetics , Alcohol Drinking/adverse effects , Animals , Atrial Fibrillation/chemically induced , Atrial Fibrillation/drug therapy , Flavonoids/pharmacology , Mice , Mice, Inbred C57BL , Sirtuin 3/genetics
7.
Food Funct ; 13(13): 7302-7319, 2022 Jul 04.
Article in English | MEDLINE | ID: mdl-35726783

ABSTRACT

Polydatin has attracted much attention as a potential cardioprotective agent against ischemic heart disease and diabetic cardiomyopathy. However, the effect and mechanism of polydatin supplementation on alcoholic cardiomyopathy (ACM) are still unknown. This study aimed to determine the therapeutic effect of polydatin against ACM and to explore the molecular mechanisms with a focus on SIRT6-AMP-activated protein kinase (AMPK) signaling and mitochondrial function. The ACM model was established by feeding C57/BL6 mice with an ethanol Lieber-DeCarli diet for 12 weeks. The mice received polydatin (20 mg kg-1) or vehicle treatment. We showed that polydatin treatment not only improved cardiac function but also reduced myocardial fibrosis and dynamin-related protein 1 (Drp-1)-mediated mitochondrial fission, and enhanced PTEN-induced putative kinase 1 (PINK1)-Parkin-dependent mitophagy in alcohol-treated myocardium. Importantly, these beneficial effects were mimicked by SIRT6 overexpression but abolished by the infection of recombinant serotype 9 adeno-associated virus (AAV9) carrying SIRT6-specific small hairpin RNA. Mechanistically, alcohol consumption induced a gradual decrease in the myocardial SIRT6 level, while polydatin effectively activated SIRT6-AMPK signaling and modulated mitochondrial dynamics and mitophagy, thus reducing oxidative stress damage and preserving mitochondrial function. In summary, these data present new information regarding the therapeutic actions of polydatin, suggesting that the activation of SIRT6 signaling may represent a new approach for tackling ACM-related cardiac dysfunction.


Subject(s)
Alcoholism , Cardiomyopathy, Alcoholic , Sirtuins , AMP-Activated Protein Kinases/metabolism , Alcohol Drinking , Animals , Cardiomyopathy, Alcoholic/metabolism , Ethanol , Glucosides , Mice , Sirtuins/genetics , Sirtuins/metabolism , Stilbenes
8.
Curr Stem Cell Res Ther ; 17(8): 815-824, 2022.
Article in English | MEDLINE | ID: mdl-34844547

ABSTRACT

BACKGROUND: Human adipose-derived stem cells (hASCs) play an important role in regenerative medicine. OBJECTIVE: Exploring the mechanism of Rg1 in the promotion of the proliferation and adipogenic differentiation of hASCs is important in regenerative medicine research. METHODS: To observe ginsenoside Rg1 in promoting the proliferation and adipogenic differentiation of hASCs, Rg1 medium at different concentrations was established and tested using the cell counting kit-8 (CCK-8) assay, oil red O staining, alizarin red, and alcian blue. Compared to the control, differentially expressed genes (DEGs) were screened via DEG analysis, which was carried out in the Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses. To explore the relationship among mRNA, long non-coding RNA (lncRNA) and microRNA (miRNA), we constructed a competing endogenous RNA (ceRNA) network. RESULTS: In this study, Rg1 was observed to promote the proliferation and adipogenic differentiation of hASCs. Additionally, enriched BPs and KEGG pathways may be involved in the promotion process, where FXR1 and Lnc-GAS5-AS1 were found to be regulatory factors. The regulatory network suggested that Rg1 could regulate the adipocytokine signaling pathway and IL-17 signaling pathway via FXR1 and Lnc-GAS5-AS1, which served as the mechanism encompassing the promotion of Rg1 on the proliferation and adipogenic differentiation of hASCs. CONCLUSION: A comprehensive transcriptional regulatory network related to the promotion ability of Rg1 was constructed, revealing mechanisms regarding Rg1's promotion of the proliferation and adipogenic differentiation of hASCs. The present study provides a theoretical basis for optimizing the function of hASCs.


Subject(s)
Ginsenosides , MicroRNAs , RNA, Long Noncoding , Adipokines/metabolism , Alcian Blue/metabolism , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Ginsenosides/pharmacology , Humans , Interleukin-17/metabolism , MicroRNAs/genetics , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , RNA, Messenger/genetics , RNA-Binding Proteins/metabolism , Stem Cells/drug effects
9.
Free Radic Biol Med ; 178: 202-214, 2022 01.
Article in English | MEDLINE | ID: mdl-34864165

ABSTRACT

Mitochondrial reactive oxygen species (ROS) damage and atrial remodeling serve as the crucial substrates for the genesis of atrial fibrillation (AF). Branched-chain amino acids (BCAAs) catabolic defect plays critical roles in multiple cardiovascular diseases. However, the alteration of atrial BCAA catabolism and its role in AF remain largely unknown. This study aimed to explore the role of BCAA catabolism in the pathogenesis of AF and to further evaluate the therapeutic effect of melatonin with a focus on protein kinase G (PKG)-cAMP response element binding protein (CREB)-Krüppel-like factor 15 (KLF15) signaling. We found that angiotensin II-treated atria exhibited significantly elevated BCAA level, reduced BCAA catabolic enzyme activity, increased AF vulnerability, aggravated atrial electrical and structural remodeling, and enhanced mitochondrial ROS damage. These deleterious effects were attenuated by melatonin co-administration while exacerbated by BCAA oral supplementation. Melatonin treatment ameliorated BCAA-induced atrial damage and reversed BCAA-induced down-regulation of atrial PKGIα expression, CREB phosphorylation as well as KLF15 expression. However, inhibition of PKG partly abolished melatonin-induced beneficial actions. In summary, these data demonstrated that atrial BCAA catabolic defect contributed to the pathogenesis of AF by aggravating tissue fibrosis and mitochondrial ROS damage. Melatonin treatment ameliorated Ang II-induced atrial structural as well as electrical remodeling by activating PKG-CREB-KLF15. The present study reveals additional mechanisms contributing to AF genesis and highlights the opportunity of a novel therapy for AF by targeting BCAA catabolism. Melatonin may serve as a potential therapeutic agent for AF intervention.


Subject(s)
Atrial Fibrillation , Melatonin , Amino Acids, Branched-Chain , Angiotensin II , Atrial Fibrillation/chemically induced , Atrial Fibrillation/drug therapy , Atrial Fibrillation/genetics , Cyclic AMP Response Element-Binding Protein/genetics , Cyclic GMP-Dependent Protein Kinases/genetics , Humans , Kruppel-Like Transcription Factors , Melatonin/pharmacology
10.
J Pineal Res ; 70(1): e12698, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33016468

ABSTRACT

Targeting mitochondrial quality control with melatonin has been found promising for attenuating diabetic cardiomyopathy (DCM), although the underlying mechanisms remain largely undefined. Activation of SIRT6 and melatonin membrane receptors exerts cardioprotective effects while little is known about their roles during DCM. Using high-fat diet-streptozotocin-induced diabetic rat model, we found that prolonged diabetes significantly decreased nocturnal circulatory melatonin and heart melatonin levels, reduced the expressions of cardiac melatonin membrane receptors, and decreased myocardial SIRT6 and AMPK-PGC-1α-AKT signaling. 16 weeks of melatonin treatment inhibited the progression of DCM and the following myocardial ischemia-reperfusion (MI/R) injury by reducing mitochondrial fission, enhancing mitochondrial biogenesis and mitophagy via re-activating SIRT6 and AMPK-PGC-1α-AKT signaling. After the induction of diabetes, adeno-associated virus carrying SIRT6-specific small hairpin RNA or luzindole was delivered to the animals. We showed that SIRT6 knockdown or antagonizing melatonin receptors abolished the protective effects of melatonin against mitochondrial dysfunction as evidenced by aggravated mitochondrial fission and reduced mitochondrial biogenesis and mitophagy. Additionally, SIRT6 shRNA or luzindole inhibited melatonin-induced AMPK-PGC-1α-AKT activation as well as its cardioprotective actions. Collectively, we demonstrated that long-term melatonin treatment attenuated the progression of DCM and reduced myocardial vulnerability to MI/R injury through preserving mitochondrial quality control. Melatonin membrane receptor-mediated SIRT6-AMPK-PGC-1α-AKT axis played a key role in this process. Targeting SIRT6 with melatonin treatment may be a promising strategy for attenuating DCM and reducing myocardial vulnerability to ischemia-reperfusion injury in diabetic patients.


Subject(s)
Diabetic Cardiomyopathies/prevention & control , Melatonin/pharmacology , Mitochondria, Heart/drug effects , Myocardial Reperfusion Injury/prevention & control , Myocytes, Cardiac/drug effects , Organelle Biogenesis , Sirtuins/metabolism , AMP-Activated Protein Kinases/metabolism , Animals , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Type 2/complications , Diabetic Cardiomyopathies/enzymology , Diabetic Cardiomyopathies/etiology , Diabetic Cardiomyopathies/pathology , Forkhead Box Protein O3/metabolism , Male , Mitochondria, Heart/enzymology , Mitochondria, Heart/ultrastructure , Myocardial Reperfusion Injury/enzymology , Myocardial Reperfusion Injury/pathology , Myocytes, Cardiac/enzymology , Myocytes, Cardiac/ultrastructure , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rats, Sprague-Dawley , Signal Transduction , Sirtuins/genetics , Time Factors
11.
Cardiovasc Res ; 115(11): 1617-1628, 2019 Sep 01.
Article in English | MEDLINE | ID: mdl-30590389

ABSTRACT

AIMS: Myocardial dysfunction is an important cause of heart failure (HF). RNA polymerase II subunit 5 (RPB5)-mediating protein (RMP) is a transcriptional mediating protein which co-ordinates cellular processes including gene expression, metabolism, proliferation, and genome stability. However, its role in cardiac disease remains unknown. We aimed to determine the role and regulatory mechanisms of RMP in cardiomyocyte function and the development of HF. METHODS AND RESULTS: Myocardial RMP expression was examined in human heart tissues from healthy controls and patients with advanced HF. Compared to normal cardiac tissues, RMP levels were significantly decreased in the myocardium of patients with advanced HF. To investigate the role of RMP in cardiac function, Cre-loxP recombinase technology was used to generate tamoxifen-inducible cardiomyocyte-specific Rmp knockout mice. Unexpectedly, cardiomyocyte-specific deletion of Rmp in mice resulted in contractile dysfunction, cardiac dilatation, and fibrosis. Furthermore, the lifespan of cardiac-specific Rmp-deficient mice was significantly shortened when compared with littermates. Mechanistically, we found that chronic HF in Rmp-deficient mice was associated with impaired mitochondrial structure and function, which may be mediated via a transforming growth factor-ß/Smad3-proliferator-activated receptor coactivator1α (PGC1α)-dependent mechanism. PGC1α overexpression partially rescued chronic HF in cardiomyocyte-specific Rmp-deficient mice, and Smad3 blockade protected against the loss of PGC1α and adenosine triphosphate content that was induced by silencing RMP in vitro. CONCLUSIONS: RMP plays a protective role in chronic HF. RMP may protect cardiomyocytes from injury by maintaining PGC1α-dependent mitochondrial biogenesis and function. The results from this study suggest that RMP may be a potential therapeutic agent for treating HF.


Subject(s)
Heart Failure/metabolism , Mitochondria, Heart/metabolism , Myocardial Contraction , Myocytes, Cardiac/metabolism , Repressor Proteins/deficiency , Ventricular Function, Left , Ventricular Remodeling , Animals , Apoptosis , Case-Control Studies , Cell Line , Disease Models, Animal , Fibrosis , Heart Failure/genetics , Heart Failure/pathology , Heart Failure/physiopathology , Humans , Mice, Inbred C57BL , Mice, Knockout , Mitochondria, Heart/genetics , Mitochondria, Heart/ultrastructure , Myocytes, Cardiac/ultrastructure , Organelle Biogenesis , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Repressor Proteins/genetics , Signal Transduction , Smad3 Protein/metabolism , Transforming Growth Factor beta1/metabolism
12.
Int J Cardiol ; 266: 213-217, 2018 Sep 01.
Article in English | MEDLINE | ID: mdl-29887450

ABSTRACT

BACKGROUND: Myocardial infarction (MI) is one of the most common causes of cardiac morbidity and mortality. Many evidences suggest that hypothermia have a more pronounced impact as an adjunctive therapy for MI to reduce infarct size. However, the function of long non-coding RNAs (lncRNA) in therapeutic hypothermia for MI remains poorly understood. METHODS: In this study, we investigated the expression of lncRNA-UIHTC (upregulated in hypothermia treated cardiomyocytes, NONHSAT094064) in ischemic heart tissues. To investigate its function, overexpression of UIHTC was performed by adeno-associated virus vectors after MI model in rat. RESULTS: lncRNA-UIHTC was upregulated in ischemic or injury cardiomyocytes. Overexpression of lncRNA-UIHTC in peri-infarction attenuated cardiac dysfunction in vivo. Mechanistically, lncRNA-UIHTC enhanced the mitochondrial function via upregulation of PGC1α. Moreover, when we knocked down PGC1α, the mitochondrial maximal oxygen consumption and ATP levels enhanced by overexpression of UIHTC were nearly completely restored. CONCLUSIONS: Altogether we have provided a new mechanism whereby hypothermia protected heart against ischemic via lncRNA-UIHTC. The UIHTC provided a new potential therapeutic target for MI but prevented the complications of hypothermia.


Subject(s)
Hypothermia/metabolism , Mitochondria/physiology , Myocardial Infarction/metabolism , Myocardial Infarction/prevention & control , Myocytes, Cardiac/metabolism , RNA, Long Noncoding/biosynthesis , Animals , Cell Line , Humans , Mitochondria/drug effects , Myocytes, Cardiac/drug effects , RNA, Long Noncoding/pharmacology , RNA, Long Noncoding/therapeutic use , Rats , Up-Regulation/drug effects , Up-Regulation/physiology
13.
Data Brief ; 17: 610-625, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29552610

ABSTRACT

This article elaborates on cardioprotective action of hypothermia related long noncoding RNA against myocardial infarction through improving mitochondrial function, which preset by J Zhang. Herein, we provide the materials and methods used in that study. And provided the detail of dysregulation of lncRNAs under the treatment of hypothermia. Furthermore, we found that lnc-UIHTC (lncRNA upregulated in hypothermia treated cardiomyocyte, NONHSAT094064) attenuated cardiomyocytes apoptosis in vitro.

14.
Biochim Biophys Acta Mol Basis Dis ; 1864(2): 563-578, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29196237

ABSTRACT

It has been demonstrated that the anti-oxidative and cardioprotective effects of melatonin are, at least in part, mediated by its membrane receptors. However, the direct downstream signaling remains unknown. We previously found that melatonin ameliorated myocardial ischemia-reperfusion (MI/R) injury in diabetic animals, although the underlying mechanisms are also incompletely understood. This study was designed to determine the role of melatonin membrane receptors in melatonin's cardioprotective actions against diabetic MI/R injury with a focus on cGMP and its downstream effector PKG. Streptozotocin-induced diabetic Sprague-Dawley rats and high-glucose medium-incubated H9c2 cardiomyoblasts were utilized to determine the effects of melatonin against MI/R injury. Melatonin treatment preserved cardiac function and reduced oxidative damage and apoptosis. Additionally, melatonin increased intracellular cGMP level, PKGIα expression, p-VASP/VASP ratio and further modulated myocardial Nrf-2-HO-1 and MAPK signaling. However, these effects were blunted by KT5823 (a selective inhibitor of PKG) or PKGIα siRNA except that intracellular cGMP level did not changed significantly. Additionally, our in vitro study showed that luzindole (a nonselective melatonin membrane receptor antagonist) or 4P-PDOT (a selective MT2 receptor antagonist) not only blocked the cytoprotective effect of melatonin, but also attenuated the stimulatory effect of melatonin on cGMP-PKGIα signaling and its modulatory effect on Nrf-2-HO-1 and MAPK signaling. This study showed that melatonin ameliorated diabetic MI/R injury by modulating Nrf-2-HO-1 and MAPK signaling, thus reducing myocardial apoptosis and oxidative stress and preserving cardiac function. Importantly, melatonin membrane receptors (especially MT2 receptor)-dependent cGMP-PKGIα signaling played a critical role in this process.


Subject(s)
Cyclic GMP-Dependent Protein Kinases/metabolism , Cyclic GMP/metabolism , Heart/drug effects , Melatonin/pharmacology , Reperfusion Injury/metabolism , Acetylcysteine/metabolism , Animals , Apoptosis , Cell Membrane/metabolism , Cell Survival , Diabetes Mellitus, Experimental , Enzyme Activation , Gene Expression Regulation , Male , Myocardium/metabolism , Myocardium/pathology , NF-E2-Related Factor 2/metabolism , Oxidative Stress , RNA, Small Interfering/metabolism , Rats , Rats, Sprague-Dawley , Reperfusion Injury/drug therapy , Signal Transduction , Tryptamines/pharmacology
15.
Sci Rep ; 7(1): 7116, 2017 08 02.
Article in English | MEDLINE | ID: mdl-28769046

ABSTRACT

Endothelial cell injury can promote the development of various cardiovascular diseases, thus, fully understanding the mechanisms underlying the maintenance of vascular endothelial cell homoeostasis may help prevent and treat cardiovascular disease. Kaiso, a zinc finger and BTB domain containing transcription factor, is key to embryonic development and cancer, but how Kaiso interacts with vascular endothelium is not fully understood. We report that Kaiso has an anti-apoptotic function in human umbilical vein endothelial cells (HUVECs) and human microvascular endothelial cells (HMEC-1s). Overexpression of Kaiso significantly increased cell viability and inhibited hydrogen peroxide-induced apoptosis. Furthermore, Kaiso increased expression of B-cell CLL/lymphoma 2 (BCL2) and reduced expression of BCL2-associated X protein (BAX) and BCL2-interacting killer (BIK) by differentially regulating gene promoter activity. Methylated DNA and specific Kaiso binding site (KBS) contributed to gene regulatory activity of Kaiso. In addition, p120ctn functioned cooperatively in Kaiso-mediated transcriptional regulation.


Subject(s)
Apoptosis/genetics , Gene Expression Regulation , Human Umbilical Vein Endothelial Cells/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Transcription Factors/metabolism , Apoptosis/drug effects , Binding Sites , Biomarkers , DNA Methylation , Gene Expression Regulation/drug effects , Gene Silencing , Human Umbilical Vein Endothelial Cells/drug effects , Humans , Hydrogen Peroxide/pharmacology , Promoter Regions, Genetic , Protein Binding , RNA, Small Interfering/genetics , Transcription Factors/genetics , Transcription, Genetic
16.
Apoptosis ; 22(7): 942-954, 2017 07.
Article in English | MEDLINE | ID: mdl-28455824

ABSTRACT

Diallyl trisulfide (DATS) protects against apoptosis during myocardial ischemia-reperfusion (MI/R) injury in diabetic state, although the underlying mechanisms remain poorly defined. Previously, we and others demonstrated that silent information regulator 1 (SIRT1) activation inhibited oxidative stress and endoplasmic reticulum (ER) stress during MI/R injury. We hypothesize that DATS reduces diabetic MI/R injury by activating SIRT1 signaling. Streptozotocin (STZ)-induced type 1 diabetic rats were subjected to MI/R surgery with or without perioperative administration of DATS (40 mg/kg). We found that DATS treatment markedly improved left ventricular systolic pressure and the first derivative of left ventricular pressure, reduced myocardial infarct size as well as serum creatine kinase and lactate dehydrogenase activities. Furthermore, the myocardial apoptosis was also suppressed by DATS as evidenced by reduced apoptotic index and cleaved caspase-3 expression. However, these effects were abolished by EX527 (the inhibitor of SIRT1 signaling, 5 mg/kg). We further found that DATS effectively upregulated SIRT1 expression and its nuclear distribution. Additionally, PERK/eIF2α/ATF4/CHOP-mediated ER stress-induced apoptosis was suppressed by DATS treatment. Moreover, DATS significantly activated Nrf-2/HO-1 antioxidant signaling pathway, thus reducing Nox-2/4 expressions. However, the ameliorative effects of DATS on oxidative stress and ER stress-mediated myocardial apoptosis were inhibited by EX527 administration. Taken together, these data suggest that perioperative DATS treatment effectively ameliorates MI/R injury in type 1 diabetic setting by enhancing cardiac SIRT1 signaling. SIRT1 activation not only upregulated Nrf-2/HO-1-mediated antioxidant signaling pathway but also suppressed PERK/eIF2α/ATF4/CHOP-mediated ER stress level, thus reducing myocardial apoptosis and eventually preserving cardiac function.


Subject(s)
Allyl Compounds/administration & dosage , Myocardial Infarction/drug therapy , Reperfusion Injury/drug therapy , Sirtuin 1/genetics , Sulfides/administration & dosage , Animals , Antioxidants/administration & dosage , Apoptosis/drug effects , Caspase 3/genetics , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Type 1/drug therapy , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/pathology , Disease Models, Animal , Endoplasmic Reticulum Stress/drug effects , Gene Expression Regulation/drug effects , Heme Oxygenase-1/genetics , Humans , Myocardial Infarction/genetics , Myocardial Infarction/pathology , NF-E2-Related Factor 2/genetics , Oxidative Stress/drug effects , Rats , Reperfusion Injury/genetics , Reperfusion Injury/pathology , Sirtuin 1/antagonists & inhibitors
17.
Biomed Res Int ; 2017: 7951793, 2017.
Article in English | MEDLINE | ID: mdl-28299332

ABSTRACT

It has been shown that inflammation and oxidative stress are important factors in postoperative atrial fibrillation (POAF). Angiotensin converting enzyme (ACE) and apelin have a close relationship with inflammation and oxidative stress. The effect of ACE and apelin on POAF after off-pump coronary artery bypass grafting (OPCABG) remains a question. The concentrations of serum ACE, angiotensin II (Ang II), apelin, bradykinin (BK), malondialdehyde (MDA), and C reactive protein (CRP) were measured in the perioperative period of OPCABG. The levels of serum ACE in the POAF group were higher than in the no POAF group both preoperatively and postoperatively. Apelin in the POAF group was lower than in the no POAF group. There was a correlation between serum ACE and apelin. Postoperatively, CRP and MDA in the POAF group were higher than in the no POAF group; however, there was no difference before the operation. Preoperative ACE and apelin were both significant and independent risk factors for POAF. In conclusion, the high ACE and low apelin preoperatively led to CRP and MDA being increased postoperatively, which was probably associated with POAF after OPCABG. Apelin may be a new predictor for POAF.


Subject(s)
Atrial Fibrillation/etiology , Coronary Artery Bypass, Off-Pump/adverse effects , Intercellular Signaling Peptides and Proteins/blood , Peptidyl-Dipeptidase A/blood , Aged , Angiotensin II/blood , Apelin , Atrial Fibrillation/physiopathology , Bradykinin/blood , C-Reactive Protein/metabolism , Case-Control Studies , Female , Humans , Inflammation , Male , Malondialdehyde/blood , Middle Aged , Multivariate Analysis , Oxidative Stress , Postoperative Complications , Postoperative Period , Preoperative Period , Retrospective Studies , Risk Factors
18.
Sci Rep ; 7: 41337, 2017 01 25.
Article in English | MEDLINE | ID: mdl-28120943

ABSTRACT

Enhancing mitochondrial biogenesis and reducing mitochondrial oxidative stress have emerged as crucial therapeutic strategies to ameliorate diabetic myocardial ischemia/reperfusion (MI/R) injury. Melatonin has been reported to be a safe and potent cardioprotective agent. However, its role on mitochondrial biogenesis or reactive oxygen species (ROS) production in type 1 diabetic myocardium and the underlying mechanisms remain unknown. We hypothesize that melatonin ameliorates MI/R injury in type 1 diabetic rats by preserving mitochondrial function via AMPK-PGC-1α-SIRT3 signaling pathway. Both our in vivo and in vitro data showed that melatonin reduced MI/R injury by improving cardiac function, enhancing mitochondrial SOD activity, ATP production and oxidative phosphorylation complex (II, III and IV), reducing myocardial apoptosis and mitochondrial MDA, H2O2 generation. Importantly, melatonin also activated AMPK-PGC-1α-SIRT3 signaling and increased SOD2, NRF1 and TFAM expressions. However, these effects were abolished by Compound C (a specific AMPK signaling blocker) administration. Additionally, our cellular experiment showed that SIRT3 siRNA inhibited the cytoprotective effect of melatonin without affecting p-AMPK/AMPK ratio and PGC-1α expression. Taken together, we concluded that melatonin preserves mitochondrial function by reducing mitochondrial oxidative stress and enhancing its biogenesis, thus ameliorating MI/R injury in type 1 diabetic state. AMPK-PGC1α-SIRT3 axis plays an essential role in this process.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Type 1/drug therapy , Melatonin/therapeutic use , Mitochondria/metabolism , Myocardial Reperfusion Injury/drug therapy , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Sirtuin 3/metabolism , Animals , Apoptosis/drug effects , Cardiotonic Agents/pharmacology , Cardiotonic Agents/therapeutic use , Cytochromes c/metabolism , Cytosol/metabolism , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Type 1/complications , Diabetes Mellitus, Type 1/pathology , Glucose/toxicity , Glucose Tolerance Test , Male , Melatonin/pharmacology , Mice , Mitochondria/drug effects , Myocardial Reperfusion Injury/complications , Myocardial Reperfusion Injury/pathology , Myocardium/pathology , Oxidative Stress/drug effects , RNA, Small Interfering/metabolism , Rats, Sprague-Dawley , Signal Transduction , Streptozocin , Superoxide Dismutase/metabolism , Transcription Factors/metabolism
19.
Am J Cardiol ; 119(3): 473-477, 2017 Feb 01.
Article in English | MEDLINE | ID: mdl-27887690

ABSTRACT

Oxidative stress has been considered to be an important factor contributing to postoperative atrial fibrillation (PoAF). Mitochondrial DNA (mtDNA) copy number in peripheral blood has been found to be associated with a patient's oxidative stress. Therefore, we sought to determine whether there was association between mtDNA copy number and the onset of atrial fibrillation. mtDNA copy numbers were measured using the quantitative real-time polymerase chain reaction in peripheral blood from 485 consecutive patients with sinus rhythm undergoing coronary artery bypass grafting. The blood was collected before surgery. In the cohort, the incidence of PoAF was 20.8% (101/485). The mean mtDNA copy number was significantly higher in patents with PoAF than in those with sinus rhythm (36.43 vs 16.63, p <0.001). The receiver operating characteristic analysis proved that the mtDNA copy number could predict PoAF with good sensitivity and specificity (area under the curve = 0.814, cutoff = 20.91, sensitivity = 70.3%, specificity = 80.2%, p <0.001). On multivariate logistic and Cox regression analysis, mtDNA copy number was shown to be a significant independent risk factor for PoAF (odds ratio = 10.01, p <0.001 and hazard ratio = 7.011, p = 0.004). There was a strong positive correlation between mtDNA copy number and malondialdehyde in patients with PoAF (r = 0.449, p = 0.01). In conclusion, we showed that elevated mtDNA copy number in peripheral blood is associated with PoAF. Further investigation is needed to validate mtDNA copy number as a predictive biomarker for PoAF and to explore its potential role in arrhythmogenesis.


Subject(s)
Atrial Fibrillation/epidemiology , Coronary Artery Bypass, Off-Pump , Coronary Artery Disease/surgery , DNA, Mitochondrial/blood , Postoperative Complications/epidemiology , Aged , Atrial Fibrillation/blood , Female , Humans , Incidence , Logistic Models , Male , Middle Aged , Multivariate Analysis , Oxidative Stress , Postoperative Complications/blood , Proportional Hazards Models , Real-Time Polymerase Chain Reaction , Retrospective Studies , Risk Factors
20.
J Pineal Res ; 62(1)2017 Jan.
Article in English | MEDLINE | ID: mdl-27753144

ABSTRACT

Stress hyperglycemia is commonly observed in patients suffering from ischemic heart disease. It not only worsens cardiovascular prognosis but also attenuates the efficacies of various cardioprotective agents. This study aimed to investigate the protective effect of melatonin against myocardial ischemia-reperfusion (MI/R) injury in acute hyperglycemic state with a focus on Notch1/Hes1/Akt signaling and intracellular thioredoxin (Trx) system. Sprague Dawley rats were subjected to MI/R surgery and high-glucose (HG, 500 g/L) infusion (4 mL/kg/h) to induce temporary hyperglycemia. Rats were treated with or without melatonin (10 mg/kg/d) during the operation. Furthermore, HG (33 mmol/L)-incubated H9c2 cardiomyoblasts were treated in the presence or absence of luzindole (a competitive melatonin receptor antagonist), DAPT (a γ-secretase inhibitor), LY294002 (a PI3-kinase/Akt inhibitor), or thioredoxin-interacting protein (Txnip) adenoviral vectors. We found that acute hyperglycemia aggravated MI/R injury by suppressing Notch1/Hes1/Akt signaling and intracellular Trx activity. Melatonin treatment effectively ameliorated MI/R injury by reducing infarct size, myocardial apoptosis, and oxidative stress. Moreover, melatonin also markedly enhanced Notch1/Hes1/Akt signaling and rescued intracellular Trx system by upregulating Notch1, N1ICD, Hes1, and p-Akt expressions, increasing Trx activity, and downregulating Txnip expression. However, these effects were blunted by luzindole, DAPT, or LY294002. Additionally, Txnip overexpression not only decreased Trx activity, but also attenuated the cytoprotective effect of melatonin. We conclude that impaired Notch1 signaling aggravates MI/R injury in acute hyperglycemic state. Melatonin rescues Trx system by reducing Txnip expression via Notch1/Hes1/Akt signaling in a membrane receptor-dependent manner. Its role as a prophylactic/therapeutic drug deserves further clinical study.


Subject(s)
Hyperglycemia/complications , Melatonin/pharmacology , Myocardial Reperfusion Injury/prevention & control , Signal Transduction/drug effects , Thioredoxins/metabolism , Animals , Blotting, Western , Disease Models, Animal , Fluorescent Antibody Technique , Male , Myocardial Reperfusion Injury/complications , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Sprague-Dawley , Receptor, Notch1/metabolism , Transcription Factor HES-1/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...