Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 181
Filter
Add more filters

Publication year range
1.
FASEB J ; 38(10): e23655, 2024 May 31.
Article in English | MEDLINE | ID: mdl-38767449

ABSTRACT

The disruption of mitochondria homeostasis can impair the contractile function of cardiomyocytes, leading to cardiac dysfunction and an increased risk of heart failure. This study introduces a pioneering therapeutic strategy employing mitochondria derived from human umbilical cord mesenchymal stem cells (hu-MSC) (MSC-Mito) for heart failure treatment. Initially, we isolated MSC-Mito, confirming their functionality. Subsequently, we monitored the process of single mitochondria transplantation into recipient cells and observed a time-dependent uptake of mitochondria in vivo. Evidence of human-specific mitochondrial DNA (mtDNA) in murine cardiomyocytes was observed after MSC-Mito transplantation. Employing a doxorubicin (DOX)-induced heart failure model, we demonstrated that MSC-Mito transplantation could safeguard cardiac function and avert cardiomyocyte apoptosis, indicating metabolic compatibility between hu-MSC-derived mitochondria and recipient mitochondria. Finally, through RNA sequencing and validation experiments, we discovered that MSC-Mito transplantation potentially exerted cardioprotection by reinstating ATP production and curtailing AMPKα-mTOR-mediated excessive autophagy.


Subject(s)
AMP-Activated Protein Kinases , Apoptosis , Autophagy , Mesenchymal Stem Cells , Mitochondria , Myocytes, Cardiac , TOR Serine-Threonine Kinases , Animals , Humans , Male , Mice , AMP-Activated Protein Kinases/metabolism , Doxorubicin/pharmacology , Heart Failure/metabolism , Mesenchymal Stem Cells/metabolism , Mice, Inbred C57BL , Mitochondria/metabolism , Mitochondria/transplantation , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , TOR Serine-Threonine Kinases/metabolism
2.
FASEB J ; 38(18): e70070, 2024 Sep 30.
Article in English | MEDLINE | ID: mdl-39301939

ABSTRACT

The objective of the study was to assess the therapeutic efficacy of targeting remote zone cardiomyocytes with cardiosphere-derived cell (CDC) extracellular vesicles (EVs) delivered via intramyocardial and intravenous routes following acute myocardial infarction (MI). Cardiomyocyte (CM) cell death plays a significant role in left ventricular (LV) remodeling and cardiac dysfunction following MI. While EVs secreted by CDCs have shown efficacy in promoting cardiac repair in preclinical models of MI, their translational potential is limited by their biodistribution and requirement for intramyocardial delivery. We hypothesized that engineering the surface of EVs to target cardiomyocytes would enhance their therapeutic efficacy following systemic delivery in a model of acute MI. CDC-derived EVs were engineered to express a CM-specific binding peptide (CMP) on their surface and characterized for size, morphology, and protein expression. Mice with acute MI underwent both intramyocardial and intravenous delivery of EVs, CMP-EVs and placebo in a double-blind study. LVEF was assessed by echo at 2- and 28-days post-MI and tissue samples processed for assessment of EV biodistribution and histological endpoints. CMP-EVs demonstrated superior cardiac targeting and retention when compared with unmodified EVs 24 h post-MI. Mice treated with IV delivered CMP-EVs demonstrated a significant improvement in LVEF and a significant reduction in remote zone cardiomyocyte apoptosis when compared with IV delivered non-targeted EVs at 28-day post-MI. Systemic administration of CMP-EVs improved cardiac function and reduced remote zone cardiomyocyte apoptosis compared with IV-administered unmodified EVs, demonstrating a strategy to optimize therapeutic EV delivery post-MI.


Subject(s)
Extracellular Vesicles , Myocardial Infarction , Myocytes, Cardiac , Animals , Myocardial Infarction/therapy , Extracellular Vesicles/metabolism , Mice , Myocytes, Cardiac/metabolism , Male , Mice, Inbred C57BL , Ventricular Remodeling
3.
J Cell Mol Med ; 28(11): e18466, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38847482

ABSTRACT

Heart failure with preserved ejection fraction (HFpEF) is a clinical syndrome characterized by pulmonary and systemic congestion resulting from left ventricular diastolic dysfunction and increased filling pressure. Currently, however, there is no evidence on effective pharmacotherapy for HFpEF. In this study, we aimed to investigate the therapeutic effect of total xanthones extracted from Gentianella acuta (TXG) on HFpEF by establishing an high-fat diet (HFD) + L-NAME-induced mouse model. Echocardiography was employed to assess the impact of TXG on the cardiac function in HFpEF mice. Haematoxylin and eosin staining, wheat germ agglutinin staining, and Masson's trichrome staining were utilized to observe the histopathological changes following TXG treatment. The results demonstrated that TXG alleviated HFpEF by reducing the expressions of genes associated with myocardial hypertrophy, fibrosis and apoptosis. Furthermore, TXG improved cardiomyocyte apoptosis by inhibiting the expression of apoptosis-related proteins. Mechanistic investigations revealed that TXG could activate the inositol-requiring enzyme 1α (IRE1α)/X-box-binding protein 1 (Xbp1s) signalling pathway, but the knockdown of IRE1α using the IRE1α inhibitor STF083010 or siRNA-IRE1α impaired the ability of TXG to ameliorate cardiac remodelling in HFpEF models. In conclusion, TXG alleviates myocardial hypertrophy, fibrosis and apoptosis through the activation of the IRE1α/Xbp1s signalling pathway, suggesting its potential beneficial effects on HFpEF patients.


Subject(s)
Apoptosis , Endoribonucleases , Heart Failure , Protein Serine-Threonine Kinases , Signal Transduction , X-Box Binding Protein 1 , Xanthones , Animals , Endoribonucleases/metabolism , Endoribonucleases/genetics , Heart Failure/drug therapy , Heart Failure/metabolism , X-Box Binding Protein 1/metabolism , X-Box Binding Protein 1/genetics , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Signal Transduction/drug effects , Mice , Male , Xanthones/pharmacology , Xanthones/isolation & purification , Apoptosis/drug effects , Disease Models, Animal , Mice, Inbred C57BL , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Diet, High-Fat/adverse effects , Fibrosis , Stroke Volume/drug effects
4.
BMC Cardiovasc Disord ; 24(1): 470, 2024 Sep 03.
Article in English | MEDLINE | ID: mdl-39223509

ABSTRACT

BACKGROUND: Glucose fluctuations may be involved in the pathophysiological process of cardiomyocyte apoptosis, but the exact mechanism remains elusive. This study focused on exploring the mechanisms related to glucose fluctuation-induced cardiomyocyte apoptosis. METHODS: Diabetic rats established via an injection of streptozotocin were randomized to five groups: the controlled diabetic (CD) group, the uncontrolled diabetic (UD) group, the glucose fluctuated diabetic (GFD) group, the GFD group rats with the injection of 0.9% sodium chloride (NaCl) (GFD + NaCl) and the GFD group rats with the injection of N-acetyl-L-cysteine (NAC) (GFD + NAC). Twelve weeks later, cardiac function and apoptosis related protein expressions were tested. Proteomic analysis was performed to further analyze the differential protein expression pattern of CD and GFD. RESULTS: The left ventricular ejection fraction levels and fractional shortening levels were decreased in the GFD group, compared with those in the CD and UD groups. Positive cells tested by DAB-TUNEL were increased in the GFD group, compared with those in the CD group. The expression of Bcl-2 was decreased, but the expressions of Bax, cleaved caspase-3 and cleaved caspase-9 were increased in response to glucose fluctuations. Compared with CD, there were 527 upregulated and 152 downregulated proteins in GFD group. Txnip was one of the differentially expressed proteins related to oxidative stress response. The Txnip expression was increased in the GFD group, while the Akt phosphorylation level was decreased. The interaction between Txnip and Akt was enhanced when blood glucose fluctuated. Moreover, the application of NAC partially reversed glucose fluctuations-induced cardiomyocyte apoptosis. CONCLUSIONS: Glucose fluctuations lead to cardiomyocyte apoptosis by up-regulating Txnip expression and enhancing Txnip-Akt interaction.


Subject(s)
Apoptosis Regulatory Proteins , Apoptosis , Blood Glucose , Carrier Proteins , Diabetes Mellitus, Experimental , Myocytes, Cardiac , Proto-Oncogene Proteins c-akt , Rats, Sprague-Dawley , Signal Transduction , Animals , Myocytes, Cardiac/pathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/drug effects , Apoptosis/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Diabetes Mellitus, Experimental/metabolism , Male , Carrier Proteins/metabolism , Blood Glucose/metabolism , Apoptosis Regulatory Proteins/metabolism , Phosphorylation , Ventricular Function, Left/drug effects , Thioredoxins/metabolism , Diabetic Cardiomyopathies/metabolism , Diabetic Cardiomyopathies/pathology , Diabetic Cardiomyopathies/physiopathology , Diabetic Cardiomyopathies/etiology , Proteomics , Rats , Protein Interaction Maps , Cell Cycle Proteins
5.
Int J Med Sci ; 21(12): 2324-2333, 2024.
Article in English | MEDLINE | ID: mdl-39310254

ABSTRACT

Diabetic cardiomyopathy (DCM) triggers a detrimental shift in mitochondrial dynamics, characterized by increased fission and decreased fusion, contributing to cardiomyocyte apoptosis and cardiac dysfunction. This study investigated the impact of modulating mitochondrial dynamics on DCM outcomes and underlying mechanisms in a mouse model. DCM induction led to upregulation of fission genes (Drp1, Mff, Fis1) and downregulation of fusion genes (Mfn1, Mfn2, Opa1). Inhibiting fission with Mdivi-1 or promoting fusion with Ginsenoside Rg1 preserved cardiac function, as evidenced by improved left ventricular ejection fraction (LVEF), fractional shortening (FS), and E/A ratio. Both treatments also reduced infarct size and attenuated cardiomyocyte apoptosis, indicated by decreased caspase-3 activity. Mechanistically, Mdivi-1 enhanced mitochondrial function by improving mitochondrial membrane potential, reducing reactive oxygen species (ROS) production, and increasing ATP generation. Ginsenoside Rg1 also preserved mitochondrial integrity and function under hypoxic conditions in HL-1 cardiomyocytes. These findings suggest that restoring the balance of mitochondrial dynamics through pharmacological interventions targeting either fission or fusion may offer a promising therapeutic strategy for mitigating MI-induced cardiac injury and improving patient outcomes.


Subject(s)
Apoptosis , Diabetic Cardiomyopathies , Ginsenosides , Mitochondrial Dynamics , Myocytes, Cardiac , Ventricular Dysfunction, Left , Animals , Mitochondrial Dynamics/drug effects , Diabetic Cardiomyopathies/drug therapy , Diabetic Cardiomyopathies/pathology , Diabetic Cardiomyopathies/metabolism , Mice , Ginsenosides/pharmacology , Ginsenosides/therapeutic use , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Ventricular Dysfunction, Left/drug therapy , Apoptosis/drug effects , Humans , Quinazolinones/pharmacology , Quinazolinones/therapeutic use , Reactive Oxygen Species/metabolism , Disease Models, Animal , Male , Membrane Potential, Mitochondrial/drug effects
6.
Sleep Breath ; 28(1): 41-51, 2024 Mar.
Article in English | MEDLINE | ID: mdl-37548920

ABSTRACT

Obstructive sleep apnea (OSA) is characterized by intermittent hypoxia (IH) and is strongly associated with adverse cardiovascular outcomes. Myocardial injury and dysfunction have been commonly observed in clinical practice, particularly in patients with severe OSA. However, the underlying mechanisms remain obscure. In this review, we summarized the molecular mechanisms by which IH impact on myocardial injury and dysfunction. In brief, IH-induced cardiomyocyte death proceeds through the regulation of multiple biological processes, including differentially expressed transcription factors, alternative epigenetic programs, and altered post-translational modification. Besides cell death, various cardiomyocyte injuries, such as endoplasmic reticulum stress, occurs with IH. In addition to the direct effects on cardiomyocytes, IH has been found to deteriorate myocardial blood and energy supply by affecting the microvascular structure and disrupting glucose and lipid metabolism. For better diagnosis and treatment of OSA, further studies on the molecular mechanisms of IH-induced myocardial injury and dysfunction are essential.


Subject(s)
Cardiovascular System , Sleep Apnea, Obstructive , Humans , Hypoxia , Myocytes, Cardiac/metabolism
7.
Int Heart J ; 65(4): 713-722, 2024.
Article in English | MEDLINE | ID: mdl-39085110

ABSTRACT

Heart failure (HF) is a clinical syndrome caused by the progression of various cardiac diseases to severe stages, and exercise training plays a positive role in the development of HF. This study aimed to investigate the impact of different intensities of exercise training on HF rats.In this study, we established two HF rat models by intraperitoneal injection of isoproterenol at 2.5 mg/kg/day and abdominal aortic coarctation. After exercise training for 4 weeks, the heart weight/body weight ratio and echocardiography results were measured. Moreover, the regulatory effect of different exercise intensities on myocardial function in HF model rats was verified using tissue staining, western blotting, and reagent kits.Exercise training had a bidirectional adjust effect on HF. A running training program of 20 minutes/time had the most significant effect on improving myocardial function in HF rats, whereas exercise intensity of 40 minutes/time or 50 minutes/time did not significantly improve myocardial function in HF rats. Moreover, exercise intensities of 20 minutes/time and 30 minutes/time could reduce the expression levels of the HF markers NT-proBNP and BNP in rats, but the effect was more significant at a duration of 20 minutes/time. We also found that compared with other exercise intensities, 20 minutes/time exercise intensity could significantly improve myocardial fibrosis, promote cardiomyocyte autophagy, and reduce apoptosis in combating HF.Furthermore, an exercise intensity of 20 minutes/time can significantly ameliorate the progression of HF. However, the degree of significance of increasing exercise intensity in improving HF progression is weakened or has no significant effect.


Subject(s)
Disease Models, Animal , Heart Failure , Physical Conditioning, Animal , Rats, Sprague-Dawley , Animals , Heart Failure/physiopathology , Heart Failure/therapy , Heart Failure/metabolism , Rats , Physical Conditioning, Animal/physiology , Male , Apoptosis , Natriuretic Peptide, Brain/metabolism , Natriuretic Peptide, Brain/blood , Echocardiography , Myocytes, Cardiac/metabolism , Isoproterenol/pharmacology , Myocardium/metabolism , Myocardium/pathology , Autophagy/physiology
8.
J Cell Mol Med ; 27(17): 2495-2506, 2023 09.
Article in English | MEDLINE | ID: mdl-37395157

ABSTRACT

To explore the underlying mechanism of lncRNA MALAT1 in the pathogenesis of diabetic cardiomyopathy (DCM). DCM models were confirmed in db/db mice. MiRNAs in myocardium were detected by miRNA sequencing. The interactions of miR-185-5p with MALAT1 and RhoA were validated by dual-luciferase reporter assays. Primary neonatal cardiomyocytes were cultured with 5.5 or 30 mmol/L D-glucose (HG) in the presence or absence of MALAT1-shRNA and fasudil, a ROCK inhibitor. MALAT1 and miR-185-5p expression were determined by real-time quantitative PCR. The apoptotic cardiomyocytes were evaluated using flow cytometry and TUNEL staining. SOD activity and MDA contents were measured. The ROCK activity, phosphorylation of Drp1S616 , mitofusin 2 and apoptosis-related proteins were analysed by Western blotting. Mitochondrial membrane potential was examined by JC-1. MALAT1 was significantly up-regulated while miR-185-5p was down-regulated in myocardium of db/db mice and HG-induced cardiomyocytes. MALAT1 regulated RhoA/ROCK pathway via sponging miR-185-5p in cardiomyocytes in HG. Knockdown of MALAT1 and fasudil all inhibited HG-induced oxidative stress, and alleviated imbalance of mitochondrial dynamics and mitochondrial dysfunction, accompanied by reduced cardiomyocyte apoptosis. MALAT1 activated the RhoA/ROCK pathway via sponging miR-185-5p and mediated HG-induced oxidative stress, mitochondrial damage and apoptosis of cardiomyocytes in mice.


Subject(s)
MicroRNAs , RNA, Long Noncoding , Mice , Animals , Myocytes, Cardiac/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Apoptosis/genetics , Oxidative Stress , Glucose/toxicity , Glucose/metabolism , Mitochondria/metabolism
9.
Int J Exp Pathol ; 104(1): 23-32, 2023 02.
Article in English | MEDLINE | ID: mdl-36583453

ABSTRACT

Sepsis remains a worldwide public health problem. This study aims to explore the role and mechanism of transcriptional factors (TFs) in sepsis-induced myocardial injury. Firstly, TF KLF13 was selected to explore its role in sepsis-induced myocardial injury. The caecal ligation and puncture (CLP) -induced sepsis mouse model was established and the septic mice were examined using standard histopathological methods. KLF13 expression was detected in the septic mouse heart and was also seen in a lipoploysaccharide (LPS) -induced cellular inflammation model. To explore this further both pro-apoptotic cleaved-caspase3/caspase3 and Bax levels and anti-apoptotic Bcl2 levels were examined, also in both models, In addition inflammatory cytokine (IL-1ß, TNF-α, IL-8 and MCP-1) production and IκB-α protein level and p65 phosphorylation were examined in both septic mice and LPS-induced cells. Thus three parameters - cardiomyocyte apoptosis, inflammatory response and NF-κB pathway activation were evaluated under similar conditions. The septic mice showed significant oedema, disordered myofilament arrangement and degradation and necrosis to varying degrees in the myocardial cells. KLF13 was downregulated in both the septic mouse heart and the LPS-induced cellular inflammation model. Furthermore, both models showed abnormally increased cardiomyocyte apoptosis (increased cleaved-caspase3/caspase and Bax protein levels and decreased Bcl2 level), elevated inflammation (increased production of inflammatory cytokines) and the activated NF-κB pathway (increased p65 phosphorylation and decreased IκB-α protein level). KLF13 overexpression notably ameliorated sepsis-induced myocardial injury in vivo and in vitro. KLF13 overexpression protected against sepsis-induced myocardial injury and LPS-induced cellular inflammation and apoptosis via inhibiting the inflammatory pathways (especially NF-κB signalling) and cardiomyocyte apoptosis.


Subject(s)
Apoptosis , Kruppel-Like Transcription Factors , Myocardium , NF-kappa B , Sepsis , Animals , Mice , Inflammation/pathology , Lipopolysaccharides , NF-kappa B/metabolism , NF-KappaB Inhibitor alpha , Sepsis/complications , Kruppel-Like Transcription Factors/genetics , Myocardium/pathology
10.
Phytother Res ; 37(12): 5854-5870, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37655750

ABSTRACT

Doxorubicin (DOX) has aroused contradiction between its potent anti-tumor capacity and severe cardiotoxicity. Galangin (Gal) possesses antioxidant, anti-inflammatory, and antiapoptotic activities. We aimed to explore the role and underlying mechanisms of Gal on DOX-induced cardiotoxicity. Mice were intraperitoneally injected with DOX (3 mg/kg, every 2 days for 2 weeks) to generate cardiotoxicity model and Gal (15 mg/kg, 2 weeks) was co-administered via gavage daily. Nuclear factor erythroid 2-related factor 2 (Nrf2) specific inhibitor, ML385, was employed to explore the underlying mechanisms. Compared to DOX-insulted mice, Gal effectively improved cardiac dysfunction and ameliorated myocardial damage. DOX-induced increase of reactive oxygen species, malondialdehyde, and NADPH oxidase activity and downregulation of superoxide dismutase (SOD) activity were blunted by Gal. Gal also markedly blocked increase of IL-1ß, IL-6, and TNF-α in DOX-insulted heart. Mechanistically, Gal reversed DOX-induced downregulation of Nrf2, HO-1, and promoted nuclear translocation of Nrf2. ML385 markedly blunted the cardioprotective effects of Gal, as well as inhibitive effects on oxidative stress and inflammation. Gal ameliorates DOX-induced cardiotoxicity by suppressing oxidative stress and inflammation via activating Nrf2/HO-1 signaling pathway. Gal may serve as a promising cardioprotective agent for DOX-induced cardiotoxicity.


Subject(s)
Cardiotoxicity , Heme Oxygenase-1 , Mice , Animals , Cardiotoxicity/drug therapy , Heme Oxygenase-1/metabolism , NF-E2-Related Factor 2/metabolism , Apoptosis , Oxidative Stress , Doxorubicin/adverse effects , Signal Transduction , Inflammation/metabolism , Myocytes, Cardiac
11.
Int Arch Allergy Immunol ; 183(5): 552-565, 2022.
Article in English | MEDLINE | ID: mdl-34915498

ABSTRACT

OBJECTIVE: Sepsis is a predominant reason for the growing morbidity and mortality in the world. The role of circular RNAs (CircRNAs) is actively researched in sepsis. In this study, we attempt to find out the effect of CircRNA protein tyrosine kinase 2 (circPTK2) on cardiomyocyte apoptosis in septic mice. METHODS: Septic mouse model was established by cecal ligation and puncture. Then circPTK2 expression was detected and the role of circPTK2 in myocardial damage was assessed after circPTK2 expression was silenced using Ad-sh-circHIPK3. The subcellular localization of circPTK2 was analyzed. Besides, the binding relation between circPTK2 and microRNA (miR)-29b-3p and between miR-29b-3p and BCL2 antagonist/killer 1 (BAK1) was verified. The expression of miR-29b-3p and BAK1 in the myocardium was detected. Functional rescue was conducted to evaluate the role of miR-29b-3p and BAK1 in cardiomyocyte apoptosis in septic mice. RESULTS: CircPTK2 was highly expressed in the myocardium of septic mice, while circPTK2 silencing relieved the cardiac function and reduced inflammatory reaction and cardiomyocyte apoptosis of septic mice. Mechanically, circPTK2 competitively bound to miR-29b-3p to upregulate BAK1 mRNA level. Inhibition of miR-29b-3p and BAK1 overexpression could counteract the protective role of circPTK2 silencing in the myocardium of septic mice. CONCLUSION: CircPTK2 is overexpressed in the myocardium of septic mice. CircPTK2 competitively bound to miR-29b-3p to upregulate BAK1 mRNA level, to promote cardiomyocyte apoptosis, inflammatory response, and myocardial damage of the myocardium of septic mice.


Subject(s)
MicroRNAs , Sepsis , Animals , Apoptosis/genetics , Mice , MicroRNAs/genetics , MicroRNAs/metabolism , Myocytes, Cardiac/metabolism , RNA, Circular/genetics , RNA, Messenger/metabolism , Sepsis/genetics , Sepsis/metabolism , bcl-2 Homologous Antagonist-Killer Protein/genetics , bcl-2 Homologous Antagonist-Killer Protein/metabolism
12.
Acta Pharmacol Sin ; 43(8): 1970-1978, 2022 Aug.
Article in English | MEDLINE | ID: mdl-34931018

ABSTRACT

Cx43 is the major connexin in ventricular gap junctions, and plays a pivotal role in control of electrical and metabolic communication among adjacent cardiomyocytes. We previously found that Cx43 dephosphorylation at serine 282 (pS282) caused cardiomyocyte apoptosis, which is involved in cardiac ischemia/reperfusion injury. In this study we investigated whether Cx43-S282 hyper-phosphorylation could protect cardiomyocytes against apoptosis. Adenovirus carrying rat full length Cx43 gene (Cx43-wt) or a mutant gene at S282 substituted with aspartic acid (S282D) were transfected into neonatal rat ventricular myocytes (NRVMs) or injected into rat ventricular wall. Rat abdominal aorta constriction model (AAC) was used to assess Cx43-S282 phosphorylation status. We showed that Cx43 phosphorylation at S282 was increased over 2-times compared to Cx43-wt cells at 24 h after transfection, while pS262 and pS368 were unaltered. S282D-transfected cells displayed enhanced gap junctional communication, and increased basal intracellular Ca2+ concentration and spontaneous Ca2+ transients compared to Cx43-wt cells. However, spontaneous apoptosis appeared in NRVMs transfected with S282D for 34 h. Rat ventricular myocardium transfected with S282D in vivo also exhibited apoptotic responses, including increased Bax/Bcl-xL ratio, cytochrome c release as well as caspase-3 and caspase-9 activities, while factor-associated suicide (Fas)/Fas-associated death domain expression and caspase-8 activity remained unaltered. In addition, AAC-induced hypertrophic ventricles had apoptotic injury with Cx43-S282 hyper-phosphorylation compared with Sham ventricles. In conclusion, Cx43 hyper-phosphorylation at S282, as dephosphorylation, also triggers cardiomyocyte apoptosis, but through activation of mitochondrial apoptosis pathway, providing a fine-tuned Cx43-S282 phosphorylation range required for the maintenance of cardiomyocyte function and survival.


Subject(s)
Apoptosis , Connexin 43 , Myocytes, Cardiac , Animals , Connexin 43/genetics , Connexin 43/metabolism , Mitochondria , Myocytes, Cardiac/metabolism , Phosphorylation , Rats , Serine/metabolism
13.
Biochem Genet ; 60(5): 1781-1792, 2022 Oct.
Article in English | MEDLINE | ID: mdl-35092560

ABSTRACT

Although previous studies showed that long non-coding RNAs (lncRNAs) have critical roles in the pathogenesis of acute myocardial infarction (AMI), the underlying molecular mechanism that lncRNAs participate in MI remains unclear. Herein, we explored the expression of lncRNA HOX antisense non-coding RNA (HOTAIR) in the serum of MI patients and mouse model of AMI. Biological functions of HOTAIR in hypoxic H9c2 cells, the in vitro model of MI, were also assessed. RT-qPCR results showed that HOTAIR expression was downregulated in the serum of AMI patients and AMI mice. HOTAIR overexpression promoted H9c2 cell viability and inhibited cell apoptosis under hypoxic conditions. Mechanically, HOTAIR was regulated by miR-206 and FN1 was the direct target of miR-206. More importantly, miR-206 overexpression or FN1 knockdown reversed the effect of HOTAIR overexpression on H9c2 cell viability and apoptosis under hypoxic conditions. Therefore, targeting the HOTAIR/miR-206/FN1 axis may be a promising therapeutic method for MI.


Subject(s)
MicroRNAs , Myocardial Infarction , RNA, Long Noncoding , Animals , Apoptosis/genetics , Cell Line , Cell Survival/genetics , Fibronectins/genetics , Humans , Mice , MicroRNAs/genetics , Myocardial Infarction/genetics , RNA, Long Noncoding/genetics , Rats
14.
Mol Med ; 27(1): 13, 2021 02 10.
Article in English | MEDLINE | ID: mdl-33568044

ABSTRACT

BACKGROUND: Gestational diabetes mellitus is a risk factor for congenital heart defects. The article aimed to investigate the expression and roles of MST1, YAP1, Last1/2 and Survivin in modulating HG-induced cardiomyocyte apoptosis and maternal diabetes-induced heart abnormality. METHODS: Diabetes mellitus was induced in rats using streptozotocin. The protein expression and phosphorylation analysis in fetal heart tissue was assessed by western blot and immunohistochemical staining. Hoechst 33342 staining assay was performed to explore H9C2 apoptosis. The gene and protein expression in H9C2 cells was assessed by quantitative PCR and western blot. Knockdown of gene expression was assessed by RNA interference. RESULTS: Our results revealed that increased MST1 protein levels in the heart tissues of the offspring of diabetic rats in vivo and in H9C2 cardiomyocytes under HG treatment in vitro, respectively. Knockdown and overexpression experiments showed that MST1 played a key role in mediating HG-induced apoptosis in cardiomyocytes. Downregulation of YAP1 was associated with HG-induced, MST1-mediated cardiomyocyte apoptosis. Further study showed that MST1 downregulated the protein level of YAP1 through mediation of YAP1 phosphorylation on Ser127 and Ser397; this process also required LATS1/2 participation. MST1 overexpression increased the phosphorylation levels of LATS1/2, which were also shown to be increased in the heart tissues of diabetic offspring. We also found that YAP1 mediated the expression of Survivin during HG-induced apoptosis, and the Survivin-inhibitor YM155 partially inhibited the role of YAP1 in suppressing apoptosis induced by HG in cardiomyocytes. CONCLUSION: These findings reveal a regulatory mechanism of MST1/YAP1/Survivin signaling in modulating cardiomyocyte apoptosis in vitro and maternal diabetes-induced congenital heart defects in vivo.


Subject(s)
Diabetes Mellitus, Experimental/metabolism , Glucose/adverse effects , Intracellular Signaling Peptides and Proteins/metabolism , Myocytes, Cardiac/cytology , Protein Serine-Threonine Kinases/metabolism , Survivin/metabolism , Animals , Apoptosis/drug effects , Cell Line , Diabetes Mellitus, Experimental/chemically induced , Disease Models, Animal , Down-Regulation , Imidazoles/pharmacology , Intracellular Signaling Peptides and Proteins/chemistry , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Naphthoquinones/pharmacology , Phosphorylation , Rats , Streptozocin , YAP-Signaling Proteins
15.
Exp Mol Pathol ; 118: 104573, 2021 02.
Article in English | MEDLINE | ID: mdl-33212125

ABSTRACT

Epidemiological studies have shown an increased risk of cardiovascular diseases in children born to mothers who smoked during pregnancy. The cardiovascular risk in the offspring associated with in utero nicotine exposure is further exaggerated by maternal obesity. The consumption of electronic cigarettes (e-cigarettes) is alarmingly increasing among adolescents and young adults without the knowledge of their harmful health effects. There has also been a substantial increase in e-cigarette use by women of reproductive age. This study investigates the detrimental effects of gestational exposure of e-cigarette and a high-fat diet (HFD) on neonatal hearts. Time-mated pregnant mice were fed a HFD and exposed to saline or e-cigarette aerosol with 2.4% nicotine from embryonic day 4 (E4) to E20. We demonstrated that in utero exposure of e-cigarettes and HFD from E4 to E20 triggers cardiomyocyte (CM) apoptosis in the offspring at postnatal day1 (PND1), PND3, and PND14. Induction of CM apoptosis following gestational exposure of e-cigarettes and HFD was associated with inactivation of AMP-activated protein kinase (AMPK), increased cardiac oxidative stress coupled with perturbation of cardiac BAX/BCL-2 ratio and activation of caspase 3 at PND 14. Electron microscopy further revealed that left ventricles of pups at PND14 after e-cigarette exposure exhibited apoptotic nuclei, convoluted nuclear membranes, myofibrillar derangement, and enlarged mitochondria occasionally showing signs of crystolysis, indicative of cardiomyopathy and cardiac dysfunction. Our results show profound adverse effects of prenatal exposure of e-cigarette plus HFD in neonatal hearts that may lead to long-term adverse cardiac consequences in the adult.


Subject(s)
Apoptosis , Diet, High-Fat/adverse effects , Electronic Nicotine Delivery Systems/statistics & numerical data , Myocytes, Cardiac/pathology , Nicotine/toxicity , Prenatal Exposure Delayed Effects/pathology , AMP-Activated Protein Kinases/metabolism , Animals , Animals, Newborn , Female , Male , Mice , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Nicotine/analysis , Oxidative Stress , Pregnancy , Prenatal Exposure Delayed Effects/chemically induced , Prenatal Exposure Delayed Effects/metabolism
16.
BMC Cardiovasc Disord ; 21(1): 249, 2021 05 21.
Article in English | MEDLINE | ID: mdl-34020615

ABSTRACT

OBJECTIVE: Proinflammatory cytokine interleukin 17 (IL-17) is involved in ventricular remodeling, mainly of the left ventricle. This study was designed to explore the role of IL-17 played in the pathogenesis of right ventricular hypertrophy (RVH), aiming to provide a novel treatment target or diagnostic biomarker options for improving the care of RVH patients. METHODS: C57BL/6 mice were maintained in 10% O2 chamber or room air for four weeks. Right ventricular hypertrophy index (RVHI), RV/body weight ratio, pulmonary arteriolar remodeling determined by percent media thickness (%MT), and the cardiomyocyte diameter of RV were evaluated. Mice were treated with exogenous recombinant mouse IL-17 (rmIL-17, 1 µg per dose twice a week) for four weeks. H9c2 cardiomyocytes were cultured and treated with IL-17 (10 ng/mL) and STAT3 inhibitor (10 ng/mL) either under normoxia (21% O2, 5% CO2, 74% N2) or under hypoxia (3% O2, 5% CO2, 92% N2). Cardiomyocyte viability was assessed by Cell counting kit 8 (CCK-8) assay. The mRNA level was detected by RT-PCR, where as the protein expression was measured by Western blot, immunohistochemistry, and immunofluorescent analyses. RESULTS: In vivo experiments showed that IL-17 did not affect the pulmonary artery under normoxia, after treatment with rmIL-17, %MT was not changed, while RVHI and the RV/body weight ratio were increased, indicating that IL-17 directly induced right ventricular hypertrophy. In a time-course study, the mice were exposed to hypoxia for 0, 1, 2, 3, 4 weeks, respectively. We found that the expression of IL-17 was gradually upregulated in RV tissue in a time-dependent manner after one week of hypoxia exposure, especially at the third and fourth week. Cardiomyocyte hypertrophy and apoptosis were observed after the exposure of the mice to hypoxia for four weeks, rmIL-17 further aggravated the hypoxia-induced cardiomyocyte hypertrophy and apoptosis. The expression of p-STAT3 in the IL-17-deficient mice was lower than in the wild-type mice. In vitro, IL-17 inhibited cardiomyocyte viability and induced cardiomyocyte apoptosis via STAT3 under both normoxic and hypoxic conditions. CONCLUSIONS: These findings support a role for IL-17 as a mediator in the pathogenesis RVH, which might be considered as a potential novel anti-inflammation therapeutic strategy or diagnostic biomarker for RVH.


Subject(s)
Hypertrophy, Right Ventricular/metabolism , Hypoxia/metabolism , Interleukin-17/metabolism , Myocytes, Cardiac/metabolism , STAT3 Transcription Factor/metabolism , Ventricular Function, Right , Ventricular Remodeling , Animals , Cell Hypoxia , Cell Line , Disease Models, Animal , Hypertrophy, Right Ventricular/pathology , Hypertrophy, Right Ventricular/physiopathology , Hypoxia/pathology , Hypoxia/physiopathology , Interleukin-17/genetics , Interleukin-17/toxicity , Male , Mice, Inbred C57BL , Mice, Knockout , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Phosphorylation , Rats , Signal Transduction , Ventricular Function, Right/drug effects , Ventricular Remodeling/drug effects
17.
Pharm Biol ; 59(1): 335-346, 2021 Dec.
Article in English | MEDLINE | ID: mdl-35086399

ABSTRACT

CONTEXT: Danshen, the dried root and rhizome of Salvia miltiorrhiza Bunge (Labiatae) and honghua, the dried flower of Carthamus tinctorius L. (Compositae) as the herb pair was used to treat cardiovascular diseases (CVD). OBJECTIVE: To study the effects of DHHP on MIRI and mechanisms based on apoptosis and mitochondria. MATERIALS AND METHODS: 36 SD rats (n = 6) were randomly divided into control group (Con), the ischaemia-reperfusion group (IR), positive control (Xinning tablets, XNT, 1 g/kg/d) and DHHP (1.2, 2.4, and 4.8 g/kg/d). Except for Con, the other groups were intragastrically administrated for 5 d, the rat hearts were isolated to establish the MIRI model in vitro for evaluating the effects of DHHP on MIRI. 24 SD rats (n = 6) were randomly divided into Con, IR, DPPH2.4 (2.4 g/kg/d) and DPPH 2.4 + Atractyloside (ATR) (2.4 + 5 mg/kg/d), administered intragastrically for 5 d, then treated with ATR (5 mg/kg/d) by intraperitoneal injection in DPPH2.4 + ATR group, took rat hearts to establish MIRI model in vitro for revealing mechanism. RESULTS: Myocardial infarct sizes were, respectively, 0.35%, 40.09%, 15.84%, 30.13%, concentrations of NAD+ (nmol/gw/w) were 144, 83, 119, and 88, respectively, in Con, IR, DHHP2.4, DHHP2.4 + ATR group. Cleaved caspase-3 were 0.3, 1.6, 0.5 and 1.3% and cleaved caspase-9 were 0.2, 1.1, 0.4 and 0.8%, respectively, in Con, IR, DHHP2.4 and DHHP2.4 + ATR group. The beneficial effects of DHHP on MIRI were reversed by ATR. CONCLUSIONS: The improvement of MIRI by DHHP may be involved in inhibiting MPTP opening, decreasing oxidative damage, alleviating ischaemic injury and inhibiting cardiomyocyte apoptosis.


Subject(s)
Drugs, Chinese Herbal/pharmacology , Myocardial Infarction/prevention & control , Myocardial Reperfusion Injury/drug therapy , Animals , Apoptosis/drug effects , Carthamus tinctorius , Cell Line , Dose-Response Relationship, Drug , Drugs, Chinese Herbal/administration & dosage , Male , Mitochondria/drug effects , Mitochondrial Permeability Transition Pore/metabolism , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Random Allocation , Rats , Rats, Sprague-Dawley , Salvia miltiorrhiza
18.
J Cell Mol Med ; 24(17): 9545-9559, 2020 09.
Article in English | MEDLINE | ID: mdl-32722882

ABSTRACT

RNF4, a poly-SUMO-specific E3 ubiquitin ligase, is associated with protein degradation, DNA damage repair and tumour progression. However, the effect of RNF4 in cardiomyocytes remains to be explored. Here, we identified the alteration of RNF4 from ischaemic hearts and oxidative stress-induced apoptotic cardiomyocytes. Upon myocardial infarction (MI) or H2 O2 /ATO treatment, RNF4 increased rapidly and then decreased gradually. PML SUMOylation and PML nuclear body (PML-NB) formation first enhanced and then degraded upon oxidative stress. Reactive oxygen species (ROS) inhibitor was able to attenuate the elevation of RNF4 expression and PML SUMOylation. PML overexpression and RNF4 knockdown by small interfering RNA (siRNA) enhanced PML SUMOylation, promoted p53 recruitment and activation and exacerbated H2 O2 /ATO-induced cardiomyocyte apoptosis which could be partially reversed by knockdown of p53. In vivo, knockdown of endogenous RNF4 via in vivo adeno-associated virus infection deteriorated post-MI structure remodelling including more extensive interstitial fibrosis and severely fractured and disordered structure. Furthermore, knockdown of RNF4 worsened ischaemia-induced cardiac dysfunction of MI models. Our results reveal a novel myocardial apoptosis regulation model that is composed of RNF4, PML and p53. The modulation of these proteins may provide a new approach to tackling cardiac ischaemia.


Subject(s)
Apoptosis/genetics , Ischemia/genetics , Myocytes, Cardiac/metabolism , Nuclear Proteins/genetics , Transcription Factors/genetics , Animals , Fibrosis/genetics , Male , Mice , Myocardial Infarction/genetics , Oxidative Stress/genetics , RNA, Small Interfering/genetics , Reactive Oxygen Species/metabolism , Sumoylation/genetics , Tumor Suppressor Protein p53/genetics , Ubiquitin-Protein Ligases/genetics
19.
J Cell Physiol ; 235(3): 2710-2721, 2020 03.
Article in English | MEDLINE | ID: mdl-31621911

ABSTRACT

The patients suffering from myocardial infarction (MI) undergo cardiac remodeling with the features of expanded myocardial infarct size and dilated left ventricle. Multiple microRNAs (miRNAs) are emerged as crucial modulators to participate in the remodeling process. This study is mainly intended to clarify the regulatory mechanism of miR-132 in the MI-induced myocardial remodeling. miR-132 low expression, while interleukin-1ß (IL-1ß) high expression was determined in MI by reverse-transcription quantitative polymerase chain reaction and ELISA assays. MI rats showed decreased cardiac function and increased cardiomyocyte apoptosis. Moreover, miR-132 and IL-1ß levels were altered in cardiomyocytes to explore their role in MI, with levels of proapoptotic or antiapoptotic proteins in MI together with cardiac function indexes observed. In addition, upregulation of miR-132, decreased levels of Bax and Cleaved Caspase-3, increased left ventricular ejection fraction, left ventricular fractional shortening, the maximum rate of rise or decrease of left ventricular pressure (±dp/dtmax ), and Bcl-2 level, which could be reversed by overexpressing IL-1ß. All in all, miR-132 inhibits cardiomyocyte apoptosis so as to ameliorate myocardial remodeling in rats with MI through IL-1ß downregulation. Thus, miR-132 is a potential candidate for the MI treatment.


Subject(s)
Interleukin-1beta/metabolism , MicroRNAs/genetics , Myocardial Infarction/pathology , Ventricular Dysfunction, Left/pathology , Ventricular Remodeling/physiology , Adult , Aged , Animals , Apoptosis/genetics , Humans , Male , Middle Aged , Myocardial Infarction/genetics , Myocytes, Cardiac/pathology , Neovascularization, Physiologic/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Rats , Rats, Sprague-Dawley , Stroke Volume/physiology , Ventricular Remodeling/genetics
20.
J Cell Physiol ; 235(11): 8187-8198, 2020 11.
Article in English | MEDLINE | ID: mdl-31967332

ABSTRACT

Cardiomyocyte apoptosis induced by hypoxia and ischemia plays important roles in heart dysfunction after acute myocardial infarction (AMI). However, the mechanism of apoptosis induction remains unclear. A previous study reported that Y-box protein 1 (YB1) is upregulated after myocardial hypoxia/reoxygenation or ischemia/reperfusion (H/R or I/R, respectively) injury; however, whether YB1 is associated with H/R-induced cardiomyocyte apoptosis is completely unknown. In the present study, we investigated the roles of YB1 in H/R-induced cardiomyocyte apoptosis and the possible underlying molecular mechanisms. In vitro, H/R treatment upregulated the YB1 expression in H9C2 cells, whereas YB1 knockdown inhibited H/R-induced cardiomyocyte apoptosis and induced H9C2 cell proliferation via Src homology region 2 domain-containing phosphatase 1 (SHP-1)-mediated activation of signal transducer and activator of transcription 3 (STAT3). In vivo, YB1 knockdown ameliorated AMI, reducing infarct size, cardiomyocyte apoptosis, and oxidative stress, via SHP-1-mediated inactivation of STAT3. Additionally, YB1 knockdown inhibited H/R- or I/R-induced oxidative stress in vitro and in vivo. H/R and I/R increase YB1 expression, and YB1 knockdown ameliorates AMI injury via SHP-1-dependent STAT3 inactivation.


Subject(s)
Apoptosis/physiology , Myocardial Infarction/metabolism , Myocytes, Cardiac/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , STAT3 Transcription Factor/metabolism , Y-Box-Binding Protein 1/metabolism , Animals , Cell Hypoxia , Cell Line , Female , Male , Myocardial Infarction/complications , Myocardial Infarction/pathology , Myocardial Reperfusion Injury/etiology , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Myocytes, Cardiac/pathology , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL