1.Cardiomyocyte pyroptosis inhibited by dental pulp-derived mesenchymal stem cells via the miR-19a-3p/IRF-8/MAPK pathway in ischemia-reperfusion.
Yi LI ; Xiang WANG ; Sixian WENG ; Chenxi XIA ; Xuyang MENG ; Chenguang YANG ; Ying GUO ; Zuowei PEI ; Haiyang GAO ; Fang WANG
Chinese Medical Journal 2025;138(18):2336-2346
BACKGROUND:
The protective effect of mesenchymal stem cells (MSCs) on cardiac ischemia-reperfusion (I/R) injury has been widely reported. Dental pulp-derived mesenchymal stem cells (DP-MSCs) have therapeutic effects on various diseases, including diabetes and cirrhosis. This study aimed to determine the therapeutic effects of DP-MSCs on I/R injury and elucidate the underlying mechanism.
METHODS:
Myocardial I/R injury model mice were treated with DP-MSCs or a miR-19a-3p mimic. The infarct volume, fibrotic area, pyroptosis, inflammation level, and cardiac function were measured. Cardiomyocytes exposed to hypoxia-reoxygenation were transfected with the miR-19a-3p mimic, miR-19a-3p inhibitor, or negative control. Pyroptosis and protein expression in the interferon regulatory factor 8/mitogen-activated protein kinase (IRF-8/MAPK) pathway were measured.
RESULTS:
DP-MSCs protected cardiac function in cardiac I/R-injured mice and inhibited cardiomyocyte pyroptosis. The upregulation of miR-19a-3p protected cardiac function, inhibited cardiomyocyte pyroptosis, and inhibited IRF-8/MAPK signaling in cardiac I/R-injured mice. DP-MSCs inhibited cardiomyocyte pyroptosis and the IRF-8/MAPK signaling by upregulating the miR-19a-3p levels in cardiomyocytes injured by I/R.
CONCLUSION
DP-MSCs protected cardiac function by inhibiting cardiomyocyte pyroptosis through miR-19a-3p under I/R conditions.
Animals
;
MicroRNAs/metabolism*
;
Pyroptosis/genetics*
;
Mesenchymal Stem Cells/metabolism*
;
Myocytes, Cardiac/cytology*
;
Mice
;
Male
;
Mice, Inbred C57BL
;
Dental Pulp/cytology*
;
Myocardial Reperfusion Injury/therapy*
;
MAP Kinase Signaling System/physiology*
2.The regulation and mechanism of apolipoprotein A5 on myocardial lipid deposition.
Xiao-Jie YANG ; Jiang LI ; Jing-Yuan CHEN ; Teng-Teng ZHU ; Yu-Si CHEN ; Hai-Hua QIU ; Wen-Jie CHEN ; Xiao-Qin LUO ; Jun LUO
Acta Physiologica Sinica 2025;77(1):35-46
The current study aimed to clarify the roles of apolipoprotein A5 (ApoA5) and milk fat globule-epidermal growth factor 8 (Mfge8) in regulating myocardial lipid deposition and the regulatory relationship between them. The serum levels of ApoA5 and Mfge8 in obese and healthy people were compared, and the obesity mouse model induced by the high-fat diet (HFD) was established. In addition, primary cardiomyocytes were purified and identified from the hearts of suckling mice. The 0.8 mmol/L sodium palmitate treatment was used to establish the lipid deposition cardiomyocyte model in vitro. ApoA5-overexpressing adenovirus was used to observe its effects on cardiac function and lipids. The expressions of the fatty acid uptake-related molecules and Mfge8 on transcription or translation levels were detected. Co-immunoprecipitation was used to verify the interaction between ApoA5 and Mfge8 proteins. Immunofluorescence was used to observe the co-localization of Mfge8 protein with ApoA5 or lysosome-associated membrane protein 2 (LAMP2). Recombinant rMfge8 was added to cardiomyocytes to investigate the regulatory mechanism of ApoA5 on Mfge8. The results showed that participants in the simple obesity group had a significant decrease in serum ApoA5 levels (P < 0.05) and a significant increase in Mfge8 levels (P < 0.05) in comparison with the healthy control group. The adenovirus treatment successfully overexpressed ApoA5 in HFD-fed obese mice and palmitic acid-induced lipid deposition cardiomyocytes, respectively. ApoA5 reduced the weight of HFD-fed obese mice (P < 0.05), shortened left ventricular isovolumic relaxation time (IVRT), increased left ventricular ejection fraction (LVEF), and significantly reduced plasma levels of triglycerides (TG) and cholesterol (CHOL) (P < 0.05). In myocardial tissue and cardiomyocytes, the overexpression of ApoA5 significantly reduced the deposition of TG (P < 0.05), transcription of fatty acid translocase (FAT/CD36) (P < 0.05), fatty acid-binding protein (FABP) (P < 0.05), and fatty acid transport protein (FATP) (P < 0.05), and protein expression of Mfge8 (P < 0.05), while the transcription levels of Mfge8 were not significantly altered (P > 0.05). In vitro, the Mfge8 protein was captured using ApoA5 as bait protein, indicating a direct interaction between them. Overexpression of ApoA5 led to an increase in co-localization of Mfge8 with ApoA5 or LAMP2 in cardiomyocytes under lipid deposition status. On this basis, exogenous added recombinant rMfge8 counteracted the improvement of lipid deposition in cardiomyocytes by ApoA5. The above results indicate that the overexpression of ApoA5 can reduce fatty acid uptake in myocardial cells under lipid deposition status by regulating the content and cellular localization of Mfge8 protein, thereby significantly reducing myocardial lipid deposition and improving cardiac diastolic and systolic function.
Animals
;
Humans
;
Mice
;
Myocytes, Cardiac/metabolism*
;
Obesity/physiopathology*
;
Male
;
Apolipoprotein A-V/blood*
;
Lipid Metabolism/physiology*
;
Milk Proteins/blood*
;
Myocardium/metabolism*
;
Diet, High-Fat
;
Antigens, Surface/physiology*
;
Mice, Inbred C57BL
;
Cells, Cultured
;
Female
3.Disulfiram alleviates cardiac hypertrophic injury by inhibiting TAK1-mediated PANoptosis.
Wei-Dong LI ; Xuan-Yang SHEN ; Xiao-Lu JIANG ; Hong-Fu WEN ; Yuan SHEN ; Mei-Qi ZHANG ; Wen-Tao TAN
Acta Physiologica Sinica 2025;77(2):222-230
The study aims to examine the effects and potential mechanisms of disulfiram (DSF) on cardiac hypertrophic injury, focusing on the role of transforming growth factor-β-activated kinase 1 (TAK1)-mediated pan-apoptosis (PANoptosis). H9C2 cardiomyocytes were treated with angiotensin II (Ang II, 1 µmol/L) to establish an in vitro model of myocardial hypertrophy. DSF (40 µmol/L) was used to treat cardiomyocyte hypertrophic injury models, either along or in combination with the TAK1 inhibitor, 5z-7-oxozeaenol (5z-7, 0.1 µmol/L). We assessed cell damage using propidium iodide (PI) staining, measured cell viability with CCK8 assay, quantified inflammatory factor levels in cell culture media via ELISA, detected TAK1 and RIPK1 binding rates using immunoprecipitation, and analyzed the protein expression levels of key proteins in the TAK1-mediated PANoptosis pathway using Western blot. In addition, the surface area of cardiomyocytes was measured with Phalloidin staining. The results showed that Ang II significantly reduced the cellular viability of H9C2 cardiomyocytes and the binding rate of TAK1 and RIPK1, significantly increased the surface area of H9C2 cardiomyocytes, PI staining positive rate, levels of inflammatory factors [interleukin-1β (IL-1β), IL-18, and tumor necrosis factor α (TNF-α)] in cell culture media and p-TAK1/TAK1 ratio, and significantly up-regulated key proteins in the PANoptosis pathway [pyroptosis-related proteins NLRP3, Caspase-1 (p20), and GSDMD-N (p30), apoptosis-related proteins Caspase-3 (p17), Caspase-7 (p20), and Caspase-8 (p18), as well as necroptosis-related proteins p-MLKL, RIPK1, and RIPK3]. DSF significantly reversed the above changes induced by Ang II. Both 5z-7 and exogenous IL-1β weakened these cardioprotective effects of DSF. These results suggest that DSF may alleviate cardiac hypertrophic injury by inhibiting TAK1-mediated PANoptosis.
Animals
;
MAP Kinase Kinase Kinases/physiology*
;
Rats
;
Myocytes, Cardiac/pathology*
;
Disulfiram/pharmacology*
;
Cardiomegaly
;
Apoptosis/drug effects*
;
Cell Line
;
Angiotensin II
;
Necroptosis/drug effects*
;
Interleukin-1beta/metabolism*
;
Receptor-Interacting Protein Serine-Threonine Kinases/metabolism*
;
Lactones
;
Resorcinols
;
Zearalenone/administration & dosage*
4.Cardiac β-adrenergic receptor regulation of mitochondrial function in heart failure.
Ai-Ming LIU ; Wen-Li XU ; Han XIAO ; Er-Dan DONG
Acta Physiologica Sinica 2024;76(6):865-880
Heart failure is characterized by abnormal β-adrenergic receptor (β-AR) activation and mitochondrial dysfunction. In heart failure, overactivation of β-AR mediates key pathological processes in cardiomyocytes, including oxidative stress, calcium overload and metabolic abnormalities, which subsequently lead to inflammation, myocardial apoptosis and necrosis. Mitochondria are the core organelles for energy metabolism, and also play a vital role in calcium homeostasis, redox balance and signaling transduction. Moderate β-AR activation is conducive to maintaining mitochondrial homeostasis and physiological cardiomyocyte function. However, β-AR overactivation in heart failure disrupts mitochondrial function through multiple mechanisms. Therefore, our review aims to elucidate how β-AR regulates mitochondrial function, particularly under sympathetic stress, impacting oxidative stress, apoptosis, necrosis, and metabolic imbalance. By describing these mechanisms, we seek to propose new insights and therapeutic targets for the prevention and treatment of heart failure.
Heart Failure/physiopathology*
;
Humans
;
Receptors, Adrenergic, beta/physiology*
;
Mitochondria, Heart/physiology*
;
Animals
;
Oxidative Stress/physiology*
;
Myocytes, Cardiac/physiology*
;
Apoptosis/physiology*
;
Signal Transduction/physiology*
5.Single-cell analysis reveals an Angpt4-initiated EPDC-EC-CM cellular coordination cascade during heart regeneration.
Zekai WU ; Yuan SHI ; Yueli CUI ; Xin XING ; Liya ZHANG ; Da LIU ; Yutian ZHANG ; Ji DONG ; Li JIN ; Meijun PANG ; Rui-Ping XIAO ; Zuoyan ZHU ; Jing-Wei XIONG ; Xiangjun TONG ; Yan ZHANG ; Shiqiang WANG ; Fuchou TANG ; Bo ZHANG
Protein & Cell 2023;14(5):350-368
Mammals exhibit limited heart regeneration ability, which can lead to heart failure after myocardial infarction. In contrast, zebrafish exhibit remarkable cardiac regeneration capacity. Several cell types and signaling pathways have been reported to participate in this process. However, a comprehensive analysis of how different cells and signals interact and coordinate to regulate cardiac regeneration is unavailable. We collected major cardiac cell types from zebrafish and performed high-precision single-cell transcriptome analyses during both development and post-injury regeneration. We revealed the cellular heterogeneity as well as the molecular progress of cardiomyocytes during these processes, and identified a subtype of atrial cardiomyocyte exhibiting a stem-like state which may transdifferentiate into ventricular cardiomyocytes during regeneration. Furthermore, we identified a regeneration-induced cell (RIC) population in the epicardium-derived cells (EPDC), and demonstrated Angiopoietin 4 (Angpt4) as a specific regulator of heart regeneration. angpt4 expression is specifically and transiently activated in RIC, which initiates a signaling cascade from EPDC to endocardium through the Tie2-MAPK pathway, and further induces activation of cathepsin K in cardiomyocytes through RA signaling. Loss of angpt4 leads to defects in scar tissue resolution and cardiomyocyte proliferation, while overexpression of angpt4 accelerates regeneration. Furthermore, we found that ANGPT4 could enhance proliferation of neonatal rat cardiomyocytes, and promote cardiac repair in mice after myocardial infarction, indicating that the function of Angpt4 is conserved in mammals. Our study provides a mechanistic understanding of heart regeneration at single-cell precision, identifies Angpt4 as a key regulator of cardiomyocyte proliferation and regeneration, and offers a novel therapeutic target for improved recovery after human heart injuries.
Humans
;
Mice
;
Rats
;
Cell Proliferation
;
Heart/physiology*
;
Mammals
;
Myocardial Infarction/metabolism*
;
Myocytes, Cardiac/metabolism*
;
Pericardium/metabolism*
;
Single-Cell Analysis
;
Zebrafish/metabolism*
6.Progress in the Role of Mechanical Stimulus in Cardiac Development.
Ming-Hui XIE ; Wei-Hua QIAO ; Hong CAO ; Jia-Wei SHI ; Nian-Guo DONG
Acta Academiae Medicinae Sinicae 2022;44(1):164-172
Mechanical stimulus is critical to cardiovascular development during embryogenesis period.The mechanoreceptors of endocardial cells and cardiac myocytes may sense mechanical signals and initiate signal transduction that induce gene expression at a cellular level,and then translate molecular-level events into tissue-level deformations,thus guiding embryo development.This review summarizes the regulatory roles of mechanical signals in the early cardiac development including the formation of heart tube,looping,valve and septal morphogenesis,ventricular development and maturation.Further,we discuss the potential mechanical transduction mechanisms of platelet endothelial cell adhesion molecule 1-vascular endothelial-cadherin-vascular endothelial growth factor receptor 2 complex,primary cilia,ion channels,and other mechanical sensors that affect some cardiac malformations.
Animals
;
Heart/embryology*
;
Humans
;
Mechanotransduction, Cellular
;
Myocytes, Cardiac/physiology*
;
Vascular Endothelial Growth Factor A/metabolism*
7.Effects of interval training on calcium transient and contractile function of single ventricular myocyte in myocardial infarction adult rats.
Wen Yan BO ; Da Gang LI ; Zhen Jun TIAN
Chinese Journal of Applied Physiology 2019;35(2):121-125
OBJECTIVE:
To investigate the effects of interval training on calcium transient and contractile function in ischemic ventricular myocytes of rats with myocardial infarction and their synchronization.
METHODS:
Twenty-four male sprague-dawley rats in three years old, were randomly divided into three groups (n=8): sham-operated group(S), sedentary MI group(MI) and MI with interval training group (ME). The MI model was established by ligation of the left anterior descending coronary artery. The rats in ME group started training 1 week after MI operation. The S model was established by threading only without ligation. ME model took one week adaptive training, 10 m/min and 30 min/d, then took subsequently 8-week aerobic interval training: 10 min×10 m/min, then reran the rats with 2 intensities 15 m/min×6 min and 25 m/min×4 min, 1 h/d, 5 d/week. After training 24 hours, the cardiomyocytes of all groups were isolated by using the Langendorff fusion system. The contractile function and calcium transient of single ventricular myocyte in myocardial infarction adult rats were detected by IonOptix. Calcium transients were measured as [Ca] amplitude, departure velocity, ratio, TTB50%, TTP and TTP50%, return velocity and ratio amplitude. PTA, SL, ±dl/dtmax and SL shortening% were tested to evaluate contractility.
RESULTS:
Compared with S, the levels of [Ca] amplitude, departure velocity, ratio amplitude and return velocity, SL shortening%, PTA and ±dl/dtmax of MI were decreased(P<0.01), the levels of TTP, TTP50% and TTB50% of MI were increased(P<0.01); Compared with MI, the levels of departure velocity, ratio amplitude, return velocity and [Ca] amplitude of ME were increased(P<0.01), the levels of TTB50%, TTP and TTP50% of ME were decreased(P<0.01, P<0.05). The levels of SL shortening%, PTA and ±dl/dtmax of ME were increased(P<0.01, P<0.05).
CONCLUSION
Interval training can improve calcium transient and contractile function of single ventricular myocyte in myocardial infarction adult rats.
Animals
;
Calcium
;
physiology
;
Male
;
Myocardial Contraction
;
Myocardial Infarction
;
pathology
;
Myocytes, Cardiac
;
physiology
;
Physical Conditioning, Animal
;
Random Allocation
;
Rats
;
Rats, Sprague-Dawley
8.Salvianolic Acid A Protects Neonatal Cardiomyocytes Against Hypoxia/Reoxygenation-Induced Injury by Preserving Mitochondrial Function and Activating Akt/GSK-3β Signals.
Xue-Li LI ; Ji-Ping FAN ; Jian-Xun LIU ; Li-Na LIANG
Chinese journal of integrative medicine 2019;25(1):23-30
OBJECTIVE:
To investigate the effects of salvianolic acid A (SAA) on cardiomyocyte apoptosis and mitochondrial dysfunction in response to hypoxia/reoxygenation (H/R) injury and to determine whether the Akt signaling pathway might play a role.
METHODS:
An in vitro model of H/R injury was used to study outcomes on primary cultured neonatal rat cardiomyocytes. The cardiomyocytes were treated with 12.5, 25, 50 μg/mL SAA at the beginning of hypoxia and reoxygenation, respectively. Adenosine triphospate (ATP) and reactive oxygen species (ROS) levels were assayed. Cell apoptosis was evaluated by flow cytometry and the expression of cleaved-caspase 3, Bax and Bcl-2 were detected by Western blotting. The effects of SAA on mitochondrial dysfunction were examined by determining the mitochondrial membrane potential (△Ψm) and mitochondrial permeability transition pore (mPTP), followed by the phosphorylation of Akt (p-Akt) and GSK-3β (p-GSK-3β), which were measured by Western blotting.
RESULTS:
SAA significantly preserved ATP levels and reduced ROS production. Importantly, SAA markedly reduced the number of apoptotic cells and decreased cleaved-caspase 3 expression levels, while also reducing the ratio of Bax/Bcl-2. Furthermore, SAA prevented the loss of △Ψm and inhibited the activation of mPTP. Western blotting experiments further revealed that SAA significantly increased the expression of p-Akt and p-GSK-3β, and the increase in p-GSK-3β expression was attenuated after inhibition of the Akt signaling pathway with LY294002.
CONCLUSION
SAA has a protective effect on cardiomyocyte H/R injury; the underlying mechanism may be related to the preservation of mitochondrial function and the activation of the Akt/GSK-3β signaling pathway.
Adenosine Triphosphate
;
analysis
;
Animals
;
Animals, Newborn
;
Caffeic Acids
;
pharmacology
;
Cell Hypoxia
;
Cells, Cultured
;
Glycogen Synthase Kinase 3 beta
;
physiology
;
Lactates
;
pharmacology
;
Mitochondria, Heart
;
drug effects
;
physiology
;
Mitochondrial Membrane Transport Proteins
;
drug effects
;
Myocytes, Cardiac
;
drug effects
;
Proto-Oncogene Proteins c-akt
;
physiology
;
Rats
;
Rats, Sprague-Dawley
;
Reactive Oxygen Species
;
metabolism
;
Signal Transduction
;
physiology
9.Rictor regulates mitochondrial calcium signaling in mouse embryo stem cell-derived cardiomyocytes.
Ying SHAO ; Jiadan WANG ; Danyan ZHU
Journal of Zhejiang University. Medical sciences 2019;48(1):65-74
OBJECTIVE:
To explore the expression, localization and regulatory effect on mitochondrial calcium signaling of Rictor in embryonic stem cell-derived cardiomyocytes (ESC-CMs).
METHODS:
Classical embryonic stem cell cardiomyogenesis model was used for differentiation of mouse embryonic stem cells into cardiomyocytes. The location of Rictor in ESC-CMs was investigated by immunofluorescence and Western blot. The expression of Rictor in mouse embryonic stem cells was interfered with lentiviral technology, then the superposition of mitochondria and endoplasmic reticulum (ER) in ESC-CMs was detected with immunofluorescence method; the cellular ultrastructure of ESC-CMs was observed by transmission electron microscope; the mitochondrial calcium transients of ESC-CMs was detected by living cell workstation;immunoprecipitation was used to detect the interaction between 1,5,5-trisphosphate receptor (IP3 receptor, IP3R), glucose-regulated protein 75 (Grp75) and voltage-dependent anion channel 1 (VDAC1) in mitochondrial outer membrane; the expression of mitochondrial fusion protein (mitonusin-2, Mfn2) was detected by Western blot.
RESULTS:
Rictor was mainly localized in the endoplasmic reticulum and mitochondrial-endoplasmic reticulum membrane (MAM) in ESC-CMs. Immunofluorescence results showed that Rictor was highly overlapped with ER and mitochondria in ESC-CMs. After mitochondrial and ER were labeled with Mito-Tracker Red and ER-Tracker Green, it was demonstrated that the mitochondria of the myocardial cells in the Rictor group were scattered, and the superimposition rate of mitochondria and ER was lower than that of the negative control group (<0.01). The MAM structures were decreased in ESC-CMs after knockdown of Rictor. The results of the living cell workstation showed that the amplitude of mitochondrial calcium transients by ATP stimulation in ESC-CMs was decreased after knockdown of Rictor (<0.01). The results of co-immunoprecipitation showed that the interaction between IP3R, Grp75 and VDAC1 in the MAM structure of the cardiomyocytes in the Rictor group was significantly attenuated (<0.01); the results of Western blot showed that the expression of Mfn2 protein was significantly decreased (<0.01).
CONCLUSIONS
Using lentiviral technology to interfere Rictor expression in mouse embryonic stem cells, the release of calcium from the endoplasmic reticulum to mitochondria in ESC-CMs decreases, which may be affected by reducing the interaction of IP3R, Grp75, VDAC1 and decreasing the expression of Mfn2, leading to the damage of MAM structure.
Animals
;
Calcium Signaling
;
genetics
;
Gene Expression Regulation
;
genetics
;
Gene Knockdown Techniques
;
Mice
;
Mitochondria
;
physiology
;
Mouse Embryonic Stem Cells
;
Myocytes, Cardiac
;
physiology
;
Protein Transport
;
Rapamycin-Insensitive Companion of mTOR Protein
;
genetics
;
metabolism
10.miR-494-3p reduces insulin sensitivity in diabetic cardiomyocytes by down-regulation of insulin receptor substrate 1.
Jie WU ; Xing-Hua QIN ; Zuo-Xu HOU ; Zi-Hao FU ; Guo-Hua LI ; Hong-Yan YANG ; Xing ZHANG ; Feng GAO
Acta Physiologica Sinica 2019;71(2):271-278
More and more evidence suggests that microRNA is widely involved in the regulation of cardiovascular function. Our preliminary experiment showed that miR-494-3p was increased in heart of diabetic rats, and miR-494-3p was reported to be related to metabolism such as obesity and exercise. Therefore, this study was aimed to explore the role of miR-494-3p in diabetic myocardial insulin sensitivity and the related mechanism. The diabetic rat model was induced by high fat diet (45 kcal% fat, 12 weeks) combined with streptozotocin (STZ, 30 mg/kg), and cardiac tissue RNA was extracted for qPCR. The results showed that the level of miR-494-3p was significantly up-regulated in the myocardium of diabetic rats compared with the control (P < 0.05). The level of miR-494-3p in H9c2 cells cultured in high glucose and high fat medium (HGHF) was significantly increased (P < 0.01) with the increase of sodium palmitate concentration, whereas down-regulation of miR-494-3p in HGHF treated cells led to an increase in insulin-stimulated glucose uptake (P < 0.01) and the ratio of p-Akt/Akt (P < 0.05). Over-expression of miR-494-3p in H9c2 cell line significantly inhibited insulin-stimulated glucose uptake and phosphorylation of Akt (P < 0.01). Bioinformatics combined with Western blotting experiments confirmed insulin receptor substrate 1 (IRS1) as a target molecule of miR-494-3p. These results suggest that miR-494-3p reduces insulin sensitivity in diabetic cardiomyocytes by down-regulating IRS1.
Animals
;
Diabetes Mellitus, Experimental
;
physiopathology
;
Down-Regulation
;
Insulin
;
Insulin Receptor Substrate Proteins
;
physiology
;
Insulin Resistance
;
MicroRNAs
;
genetics
;
Myocytes, Cardiac
;
physiology
;
Rats

Result Analysis
Print
Save
E-mail