1
|
Abdel-Hamid HA, Marey H, Fouli Gaber Ibrahim M. Hydrogen sulfide protects the endometrium in a rat model of type 1 diabetes via modulation of PPARγ/mTOR and Nrf-2/NF-κb pathways. Arch Physiol Biochem 2024; 130:909-920. [PMID: 38685691 DOI: 10.1080/13813455.2024.2347239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Revised: 02/25/2024] [Accepted: 04/19/2024] [Indexed: 05/02/2024]
Abstract
Diabetes is one of the leading causes of endometrial diseases in women. No study has addressed the influence of hydrogen sulphide (H2S) donors on endometrial injury on top of type 1 diabetes. This research was conducted to study either the effect of sodium hydrosulphide (NaHS), the H2S donor, or DL-propargylglycine (PAG), the inhibitor of endogenous H2S production, on the endometrium of diabetic rats. A total of 40 female Wistar rats were separated into control group, diabetic group, diabetic group treated with NaHS and diabetic group treated with PAG. Serum levels of insulin, glucose, total cholesterol (TC) and triglycerides (TG) were assessed. Uterine tissue markers of oxidative stress, inflammation, apoptosis and cell proliferation were analysed. Diabetes-induced endometrial overgrowth associated with oxidative stress, inflammation and inhibition of apoptosis. NaHS administration reversed the previous conditions while PAG administration got them worse. We concluded that H2S prevented endometrial overgrowth in a rat model of type 1 diabetes through modulation of PPARγ/mTOR and Nrf-2/NF-κB pathways.
Collapse
Affiliation(s)
- Heba A Abdel-Hamid
- Department of Medical Physiology, Faculty of Medicine, Minia University, Minia, Egypt
- Department of Medical Physiology, Faculty of Medicine, Al-Baha University, KSA
| | - Heba Marey
- Department of Medical Biochemistry, Faculty of Medicine, Minia University, Minia, Egypt
| | | |
Collapse
|
2
|
Kieronska-Rudek A, Ascencao K, Chlopicki S, Szabo C. Increased hydrogen sulfide turnover serves a cytoprotective role during the development of replicative senescence. Biochem Pharmacol 2024; 230:116595. [PMID: 39454733 DOI: 10.1016/j.bcp.2024.116595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 09/15/2024] [Accepted: 10/22/2024] [Indexed: 10/28/2024]
Abstract
The mammalian gasotransmitter hydrogen sulfide (H2S) is produced by enzymes such as cystathionine β-synthase (CBS), cystathionine γ-lyase (CSE), 3-mercaptopyruvate sulfurtransferase (3-MST). Prior studies suggest that H2S may have cytoprotective and anti-aging effects. This project explores the regulation and role of endogenous H2S in a murine model of replicative senescence. H2S and polysulfide levels in RAW 264.7 murine macrophages (control cells: passage 5-10; senescent cells: passage 30-40) were measured using fluorescent probes. The expression of H2S-related enzymes and the activity of senescence marker beta-galactosidase (SA-β-Gal) were also analyzed. CBS, CSE, and 3-MST were inhibited using selective pharmacological inhibitors. Senescence led to a moderate upregulation of CBS and in a significant increase in CSE and 3-MST. H2S degradation enzymes were also elevated in senescence. Inhibition of H2S-producing enzymes reduced H2S levels but increased polysulfides. Inhibition of H2S production during senescence suppressed cell proliferation, and elevated SA-β-Gal and p21 levels. Comparing young and old mice spleens revealed downregulation of CBS and ETHE1 and upregulation of rhodanese and SUOX in older mice. The results demonstrate that increased reactive sulfur turnover occurs in senescent macrophages and that reactive sulfur species support cell proliferation and regulate cellular senescence.
Collapse
Affiliation(s)
- Anna Kieronska-Rudek
- Chair of Pharmacology, Department of Science and Medicine, University of Fribourg, Fribourg, Switzerland; Jagiellonian University, Jagiellonian Centre for Experimental Therapeutics (JCET), Cracow, Poland
| | - Kelly Ascencao
- Chair of Pharmacology, Department of Science and Medicine, University of Fribourg, Fribourg, Switzerland
| | - Stefan Chlopicki
- Jagiellonian University, Jagiellonian Centre for Experimental Therapeutics (JCET), Cracow, Poland; Jagiellonian University Medical College, Chair of Pharmacology, Faculty of Medicine, Cracow, Poland
| | - Csaba Szabo
- Chair of Pharmacology, Department of Science and Medicine, University of Fribourg, Fribourg, Switzerland.
| |
Collapse
|
3
|
Ye Z, Lin J, He C, Yu P, Cao G, Shen Q, Wang C. Polydatin protects against articular cartilage degeneration by regulating autophagy mediated by the AMPK/mTOR signaling pathway. Histol Histopathol 2024; 39:1505-1515. [PMID: 38606875 DOI: 10.14670/hh-18-739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/13/2024]
Abstract
BACKGROUND Knee osteoarthritis (KOA) is one of the leading causes of disability. Polydatin has a potential effect on KOA treatment but the therapeutic mechanism is not clear. This study aims to investigate the therapeutic action of polydatin in KOA and its mechanism in activating autophagy via the AMP-activated protein kinase (AMPK)/mTOR signaling pathway. METHODS After a KOA rat model was established by anterior cruciate ligament transection surgery, model rats were treated with polydatin 40 mg/kg for 30 days. Subsequently, cartilage tissues were collected, and hematoxylin-eosin (HE), Safranin-O, and TUNEL staining, and western blotting were performed to evaluate the pathological damage and autophagy-related protein expression. Then, human chondrocyte C28/I2 cells were stimulated with lipopolysaccharide (LPS), and the effects of polydatin on C28/I2 cell viability, apoptosis, and autophagy-related protein expression were detected by MTT, Flow Cytometry, and western blot. In addition, an AMPK inhibitor (Dorsomorphin 2HCl) was used to probe the cell proliferation and apoptosis of polydatin-administered C28/I2 cells. RESULTS Polydatin ameliorated the pathological damage in rat cartilage tissues and inhibited cell apoptosis in KOA rats. Meanwhile, in C28/I2 cells, polydatin promoted viability and reduced apoptosis. In addition, the protein expression of collagen II, LC3II/LC3I, Beclin-1, and p-AMPK/AMPK were upregulated, and p62 and p-mTOR/mTOR were downregulated by polydatin treatment. Interestingly, relative results showed that the protective effect of polydatin in LPS-stimulated-C28/I2 cells was blocked by the AMPK/mTOR inhibitor, dorsomorphin 2HCl. CONCLUSION Our research showed that polydatin reduced apoptosis and activated autophagy both in vivo and in vitro by the AMPK/mTOR signaling pathway to protect against KOA, which provided the basis for further investigation into the potential therapeutic impact of polydatin on KOA.
Collapse
Affiliation(s)
- Zhengcong Ye
- Department of Orthopedics, Hangzhou Xiaoshan District Hospital of Traditional Chinese Medicine, Hangzhou, PR China,
| | - Jian Lin
- Department of Orthopedics, Hangzhou Xiaoshan District Hospital of Traditional Chinese Medicine, Hangzhou, PR China
| | - Chun He
- Department of Orthopedics, Hangzhou Xiaoshan District Hospital of Traditional Chinese Medicine, Hangzhou, PR China
| | - Pengzheng Yu
- Department of Orthopedics, Hangzhou Xiaoshan District Orthopedics and Traumatology Hospital, Hangzhou, PR China
| | - Guoping Cao
- Department of Orthopedics, Hangzhou Xiaoshan District Hospital of Traditional Chinese Medicine, Hangzhou, PR China
| | - Qinrong Shen
- Department of Orthopedics, Shaoxing Hospital of Traditional Chinese Medicine, Shaoxing, Zhejiang, PR China
| | - Canfeng Wang
- Department of Orthopedics, Hangzhou Xiaoshan District Hospital of Traditional Chinese Medicine, Hangzhou, PR China.
| |
Collapse
|
4
|
Onalan E, Erbay B, Buran İK, Erol D, Tektemur A, Kuloglu T, Ozercan IH. Effects and Mechanism of AP39 on Ovarian Functions in Rats Exposed to Cisplatin and Chronic Immobilization Stress. J Menopausal Med 2024; 30:104-119. [PMID: 39315502 PMCID: PMC11439572 DOI: 10.6118/jmm.23015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 08/02/2024] [Accepted: 08/03/2024] [Indexed: 09/25/2024] Open
Abstract
OBJECTIVES Premature ovarian failure (POF) rat models are essential for elucidating the hormonal and ovarian molecular mechanisms of human POF diseases and developing new therapeutic agents. This study aimed to compare the applicability of chronic immobilization stress (CIS) as a POF model with that of cisplatin and to examine the impact of AP39, a mitochondrial protective agent, on ovarian function in rats treated with cisplatin and CIS. METHODS Sixty Sprague-Dawley female rats were divided equally into six groups (10 per group): Control, Cisplatin, AP39, Cisplatin + AP39, CIS, and CIS + AP39. Ovarian dysfunction was induced with cisplatin (3 mg/kg) or CIS. Forced swim test, hormone concentrations, estrous cyclicity, histopathology, follicle counts, and molecular alterations in the ovary and mitochondria were analyzed. RESULTS In the CIS and cisplatin groups, mitochondrial biogenesis, egg quality, hormonal profile, estrous cycle, and folliculogenesis significantly declined. Nonetheless, most of the parameters with undesirable results did not normalize after AP39 administration. CONCLUSIONS The cisplatin- and CIS-treated rats exhibited unshared deteriorated hormonal pathways and similarly disrupted gene expression patterns. Our current CIS model did not meet the human POF criteria, which include decreased estradiol levels, despite having advantages in terms of ease of modeling and reproducibility and demonstrating pathological changes similar to those observed in human POF. Therefore, rather than using this model as an POF model, using it as a representation of stress-induced ovarian dysfunction would be more appropriate.
Collapse
Affiliation(s)
- Ebru Onalan
- Department of Medical Biology, Faculty of Medicine, Firat University, Elazığ, Türkiye
| | - Bilgi Erbay
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| | - İlay Kavuran Buran
- Department of Medical Biology, Faculty of Medicine, Firat University, Elazığ, Türkiye.
| | - Deniz Erol
- Department of Medical Genetics, Faculty of Medicine, Firat University, Elazığ, Türkiye
| | - Ahmet Tektemur
- Department of Medical Biology, Faculty of Medicine, Firat University, Elazığ, Türkiye
| | - Tuncay Kuloglu
- Department of Histology and Embryology, Faculty of Medicine, Firat University, Elazığ, Türkiye
| | | |
Collapse
|
5
|
Lu Y, Li Y, Xie Y, Bu J, Yuan R, Zhang X. Exploring Sirtuins: New Frontiers in Managing Heart Failure with Preserved Ejection Fraction. Int J Mol Sci 2024; 25:7740. [PMID: 39062982 PMCID: PMC11277469 DOI: 10.3390/ijms25147740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Revised: 07/05/2024] [Accepted: 07/11/2024] [Indexed: 07/28/2024] Open
Abstract
With increasing research, the sirtuin (SIRT) protein family has become increasingly understood. Studies have demonstrated that SIRTs can aid in metabolism and affect various physiological processes, such as atherosclerosis, heart failure (HF), hypertension, type 2 diabetes, and other related disorders. Although the pathogenesis of HF with preserved ejection fraction (HFpEF) has not yet been clarified, SIRTs have a role in its development. Therefore, SIRTs may offer a fresh approach to the diagnosis, treatment, and prevention of HFpEF as a novel therapeutic intervention target.
Collapse
Affiliation(s)
- Ying Lu
- Department of Cardiology, Lanzhou University Second Hospital, Lanzhou 730031, China; (Y.L.); (Y.X.); (J.B.); (R.Y.)
| | - Yongnan Li
- Department of Cardiac Surgery, Lanzhou University Second Hospital, Lanzhou 730031, China;
| | - Yixin Xie
- Department of Cardiology, Lanzhou University Second Hospital, Lanzhou 730031, China; (Y.L.); (Y.X.); (J.B.); (R.Y.)
| | - Jiale Bu
- Department of Cardiology, Lanzhou University Second Hospital, Lanzhou 730031, China; (Y.L.); (Y.X.); (J.B.); (R.Y.)
| | - Ruowen Yuan
- Department of Cardiology, Lanzhou University Second Hospital, Lanzhou 730031, China; (Y.L.); (Y.X.); (J.B.); (R.Y.)
| | - Xiaowei Zhang
- Department of Cardiology, Lanzhou University Second Hospital, Lanzhou 730031, China; (Y.L.); (Y.X.); (J.B.); (R.Y.)
| |
Collapse
|
6
|
Chu J, Wang K, Lu L, Zhao H, Hu J, Xiao W, Wu Q. Advances of Iron and Ferroptosis in Diabetic Kidney Disease. Kidney Int Rep 2024; 9:1972-1985. [PMID: 39081773 PMCID: PMC11284386 DOI: 10.1016/j.ekir.2024.04.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Revised: 03/29/2024] [Accepted: 04/01/2024] [Indexed: 08/02/2024] Open
Abstract
Diabetes mellitus presents a significant threat to human health because it disrupts energy metabolism and gives rise to various complications, including diabetic kidney disease (DKD). Metabolic adaptations occurring in the kidney in response to diabetes contribute to the pathogenesis of DKD. Iron metabolism and ferroptosis, a recently defined form of cell death resulting from iron-dependent excessive accumulation of lipid peroxides, have emerged as crucial players in the progression of DKD. In this comprehensive review, we highlight the profound impact of adaptive and maladaptive responses regulating iron metabolism on the progression of kidney damage in diabetes. We summarize the current understanding of iron homeostasis and ferroptosis in DKD. Finally, we propose that precise manipulation of iron metabolism and ferroptosis may serve as potential strategies for kidney management in diabetes.
Collapse
Affiliation(s)
- Jiayi Chu
- Department of Radiology, Center of Regenerative and Aging Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Zhejiang, China
| | - Kewu Wang
- Department of Radiology, Center of Regenerative and Aging Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Zhejiang, China
| | - Lulu Lu
- Department of Nutrition and Toxicology, Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines of Zhejiang Province, School of Public Health, Hangzhou Normal University, Hangzhou, China
| | - Hui Zhao
- Department of Radiology, Center of Regenerative and Aging Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Zhejiang, China
| | - Jibo Hu
- Department of Radiology, Center of Regenerative and Aging Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Zhejiang, China
| | - Wenbo Xiao
- Department of Radiology, the First Affiliated Hospital, College of Medicine, Zhejiang University, Zhejiang, China
| | - Qian Wu
- Department of Radiology, Center of Regenerative and Aging Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Zhejiang, China
| |
Collapse
|
7
|
He K, Zhang H, Tan B, Song C, Liang Z, Zhang L, Tian D, Xiao L, Xue H, Guo Q, Teng X, Jin S, An C, Wu Y. Hydrogen Sulfide Ameliorates Heart Aging by Downregulating Matrix Metalloproteinase-9. Cardiovasc Drugs Ther 2024:10.1007/s10557-024-07586-w. [PMID: 38884920 DOI: 10.1007/s10557-024-07586-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/21/2024] [Indexed: 06/18/2024]
Abstract
PURPOSE Aging contributes significantly to cardiovascular diseases and cardiac dysfunction, leading to the upregulation of matrix metalloproteinase-9 (MMP-9) in the heart and a significant decrease in hydrogen sulfide (H2S) content, coupled with impaired cardiac diastolic function. This study explores whether supplementing exogenous hydrogen sulfide during aging ameliorates the decline in H2S concentration in the heart, suppresses MMP-9 expression, and improves the age-associated impairment in cardiac morphology and function. METHODS We collected plasma from healthy individuals of different ages to determine the relationship between aging and H2S and MMP-9 levels through Elisa detection and liquid chromatography-tandem mass spectrometry (LC/MC) detection of plasma H2S content. Three-month-old mice were selected as the young group, while 18-month-old mice were selected as the old group, and sodium hydrosulfide (NaHS) was injected intraperitoneally from 15 months old until 18 months old as the old + NaHS group. Plasma MMP-9 content was detected using Elisa, plasma H2S content, cardiac H2S content, and cystathionine gamma-lyase (CSE) activity were detected using LC/MC, and cardiac function was detected using echocardiography. Heart structure was assessed using hematoxylin and eosin staining, Masone staining was used to detect the degree of cardiac fibrosis, while western blot was used to detect the expression of MMP-9, CSE, and aging marker proteins. Knockdown of MMP-9 and CSE in H9c2 cells using small interfering RNA was carried out to determine the upstream-downstream relationship between MMP-9 and CSE. RESULTS H2S content in the plasma of healthy individuals decreases with escalating age, whereas MMP-9 level rises with age progression. Aging leads to a decrease in H2S levels in the heart and plasma of mice, severe impairment of cardiac diastolic function, interstitial relaxation, and fibrosis of the heart. Supplementing with exogenous H2S can improve these phenomena. CONCLUSION H2S maintains the structure and function of the heart by inhibiting the expression of MMP-9 during the aging process.
Collapse
Affiliation(s)
- Kaichuan He
- Department of Physiology, Hebei Medical University, 050017, Hebei, China
| | - Huaxing Zhang
- Core Facilities and Centers, Hebei Medical University, 050017, Hebei, China
| | - Bo Tan
- Clinical Pharmacokinetic Laboratory, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, 201203, Shanghai, China
| | - Chengqing Song
- Department of Physiology, Hebei Medical University, 050017, Hebei, China
| | - Zihui Liang
- Clinical Practice Teaching Department, Hebei Medical University, 050017, Hebei, China
| | - Lixia Zhang
- Department of Medical Laboratory, Hebei Children's Hospital, 050017, Hebei, China
| | - Danyang Tian
- Department of Physiology, Hebei Medical University, 050017, Hebei, China
| | - Lin Xiao
- Department of Physiology, Hebei Medical University, 050017, Hebei, China
| | - Hongmei Xue
- Department of Physiology, Hebei Medical University, 050017, Hebei, China
| | - Qi Guo
- Department of Physiology, Hebei Medical University, 050017, Hebei, China
| | - Xu Teng
- Department of Physiology, Hebei Medical University, 050017, Hebei, China
| | - Sheng Jin
- Department of Physiology, Hebei Medical University, 050017, Hebei, China
| | - Cuixia An
- Department of Psychiatry, the First Hospital of Hebei Medical University, 050031, Hebei, China.
| | - Yuming Wu
- Department of Physiology, Hebei Medical University, 050017, Hebei, China.
- Hebei Collaborative Innovation Center for Cardio-Cerebrovascular Disease, 050017, Hebei, China.
- Hebei Key Laboratory of Cardiovascular Homeostasis and Aging, 050017, Hebei, China.
- The Key Laboratory of Neural and Vascular Biology, Ministry of Education, 050017, Hebei, China.
| |
Collapse
|
8
|
Liu M, Zeng C, Zhang Y, Xin Y, Deng S, Hu X. Protective role of hydrogen sulfide against diabetic cardiomyopathy by inhibiting pyroptosis and myocardial fibrosis. Biomed Pharmacother 2024; 175:116613. [PMID: 38657502 DOI: 10.1016/j.biopha.2024.116613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 04/10/2024] [Accepted: 04/17/2024] [Indexed: 04/26/2024] Open
Abstract
Diabetic cardiomyopathy (DCM) contributes significantly to the heightened mortality rate observed among diabetic patients, with myocardial fibrosis (MF) being a pivotal element in the disease's progression. Hydrogen sulfide (H2S) has been shown to mitigate MF, but the specific underlying mechanisms have yet to be thoroughly understood. A connection has been established between the evolution of DCM and the incidence of cardiomyocyte pyroptosis. Our research offers insights into H2S protective impact and its probable mode of action against DCM, analyzed through the lens of MF. In this study, a diabetic rat model was developed using intraperitoneal injections of streptozotocin (STZ), and hyperglycemia-stimulated cardiomyocytes were employed to replicate the cellular environment of DCM. There was a marked decline in the expression of cystathionine γ-lyase (CSE), a catalyst for H2S synthesis, in both the STZ-induced diabetic rats and hyperglycemia-stimulated cardiomyocytes. Experimental results in vivo indicated that H2S ameliorates MF and enhances cardiac functionality in diabetic rats by mitigating cardiomyocyte pyroptosis. In vitro assessments highlighted the induction of cardiomyocyte pyroptosis and the subsequent decline in cell viability under hyperglycemic conditions. However, the administration of sodium hydrosulfide (NaHS) curtailed cardiomyocyte pyroptosis and augmented cell viability. In contrast, propargylglycine (PAG), a CSE inhibitor, reversed the effects rendered by NaHS administration. Additional exploration indicated that the mitigating effect of H2S on cardiomyocyte pyroptosis is modulated through the ROS/NLRP3 pathway. In essence, our findings corroborate the potential of H2S in alleviating MF in diabetic subjects. This therapeutic effect is likely attributable to the regulation of cardiomyocyte pyroptosis via the ROS/NLRP3 pathway. This discovery furnishes a prospective therapeutic target for the amelioration and management of MF associated with diabetes.
Collapse
Affiliation(s)
- Maojun Liu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital,Central South University, 139 Middle Renmin Road, Changsha, Hunan 410011,China
| | - Cheng Zeng
- Department of Cardiovascular Medicine, The Second Xiangya Hospital,Central South University, 139 Middle Renmin Road, Changsha, Hunan 410011,China
| | - Yifeng Zhang
- Department of Cardiovascular Medicine, The Second Xiangya Hospital,Central South University, 139 Middle Renmin Road, Changsha, Hunan 410011,China
| | - Ying Xin
- Department of Cardiovascular Medicine, The Second Xiangya Hospital,Central South University, 139 Middle Renmin Road, Changsha, Hunan 410011,China
| | - Simin Deng
- Department of Cardiovascular Medicine, The Second Xiangya Hospital,Central South University, 139 Middle Renmin Road, Changsha, Hunan 410011,China
| | - Xinqun Hu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital,Central South University, 139 Middle Renmin Road, Changsha, Hunan 410011,China.
| |
Collapse
|
9
|
Zhou Z, Zhao Q, Huang Y, Meng S, Chen X, Zhang G, Chi Y, Xu D, Yin Z, Jiang H, Yu L, Wang H. Berberine ameliorates chronic intermittent hypoxia-induced cardiac remodelling by preserving mitochondrial function, role of SIRT6 signalling. J Cell Mol Med 2024; 28:e18407. [PMID: 38894630 PMCID: PMC11187832 DOI: 10.1111/jcmm.18407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Revised: 04/30/2024] [Accepted: 05/07/2024] [Indexed: 06/21/2024] Open
Abstract
Chronic intermittent hypoxia (CIH) is associated with an increased risk of cardiovascular diseases. Previously, we have shown that berberine (BBR) is a potential cardioprotective agent. However, its effect and mechanism on CIH-induced cardiomyopathy remain uncovered. This study was designed to determine the effects of BBR against CIH-induced cardiac damage and to explore the molecular mechanisms. Mice were exposed to 5 weeks of CIH with or without the treatment of BBR and adeno-associated virus 9 (AAV9) carrying SIRT6 or SIRT6-specific short hairpin RNA. The effect of BBR was evaluated by echocardiography, histological analysis and western blot analysis. CIH caused the inactivation of myocardial SIRT6 and AMPK-FOXO3a signalling. BBR dose-dependently ameliorated cardiac injury in CIH-induced mice, as evidenced by increased cardiac function and decreased fibrosis. Notably, SIRT6 overexpression mimicked these beneficial effects, whereas infection with recombinant AAV9 carrying SIRT6-specific short hairpin RNA abrogated them. Mechanistically, BBR reduced oxidative stress damage and preserved mitochondrial function via activating SIRT6-AMPK-FOXO3a signalling, enhancing mitochondrial biogenesis as well as PINK1-Parkin-mediated mitophagy. Taken together, these data demonstrate that SIRT6 activation protects against the pathogenesis of CIH-induced cardiac dysfunction. BBR attenuates CIH-induced myocardial injury by improving mitochondrial biogenesis and PINK1-Parkin-dependent mitophagy via the SIRT6-AMPK-FOXO3a signalling pathway.
Collapse
Affiliation(s)
- Zijun Zhou
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Department of Cardiovascular SurgeryGeneral Hospital of Northern Theater CommandShenyangLiaoningChina
| | - Qiusheng Zhao
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Department of Cardiovascular SurgeryGeneral Hospital of Northern Theater CommandShenyangLiaoningChina
- Shenyang Joint Logistics Support CenterPharmaceutical Instruments Supervision and Inspection StationShenyangChina
| | - Yuting Huang
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Department of Cardiovascular SurgeryGeneral Hospital of Northern Theater CommandShenyangLiaoningChina
| | - Shan Meng
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Department of Cardiovascular SurgeryGeneral Hospital of Northern Theater CommandShenyangLiaoningChina
- Jinzhou Medical UniversityJinzhouLiaoningChina
| | - Xin Chen
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Department of Cardiovascular SurgeryGeneral Hospital of Northern Theater CommandShenyangLiaoningChina
- Jinzhou Medical UniversityJinzhouLiaoningChina
| | - Guoxin Zhang
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Department of Cardiovascular SurgeryGeneral Hospital of Northern Theater CommandShenyangLiaoningChina
| | - Yanbang Chi
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Department of Cardiovascular SurgeryGeneral Hospital of Northern Theater CommandShenyangLiaoningChina
| | - Dengyue Xu
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Department of Cardiovascular SurgeryGeneral Hospital of Northern Theater CommandShenyangLiaoningChina
- School of Biomedical Engineering, Faculty of MedicineDalian University of TechnologyDalianLiaoningChina
| | - Zongtao Yin
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Department of Cardiovascular SurgeryGeneral Hospital of Northern Theater CommandShenyangLiaoningChina
| | - Hui Jiang
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Department of Cardiovascular SurgeryGeneral Hospital of Northern Theater CommandShenyangLiaoningChina
| | - Liming Yu
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Department of Cardiovascular SurgeryGeneral Hospital of Northern Theater CommandShenyangLiaoningChina
| | - Huishan Wang
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Department of Cardiovascular SurgeryGeneral Hospital of Northern Theater CommandShenyangLiaoningChina
| |
Collapse
|
10
|
Zhao J, Yang T, Yi J, Hu H, Lai Q, Nie L, Liu M, Chu C, Yang J. AP39 through AMPK-ULK1-FUNDC1 pathway regulates mitophagy, inhibits pyroptosis, and improves doxorubicin-induced myocardial fibrosis. iScience 2024; 27:109321. [PMID: 38558936 PMCID: PMC10981016 DOI: 10.1016/j.isci.2024.109321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 01/10/2024] [Accepted: 02/20/2024] [Indexed: 04/04/2024] Open
Abstract
Doxorubicin induces myocardial injury and fibrosis. Still, no effective interventions are available. AP39 is an H2S donor that explicitly targets mitochondria. This study investigated whether AP39 could improve doxorubicin-induced myocardial fibrosis. Doxorubicin induced significant myocardial fibrosis while suppressing mitophagy-related proteins and elevating pyroptosis-related proteins. Conversely, AP39 reverses these effects, enhancing mitophagy and inhibiting pyroptosis. In vitro experiments revealed that AP39 inhibited H9c2 cardiomyocyte pyroptosis, improved doxorubicin-induced impairment of mitophagy, reduced ROS levels, ameliorated the mitochondrial membrane potential, and upregulated AMPK-ULK1-FUNDC1 expression. In contrast, AMPK inhibitor (dorsomorphin) and ULK1 inhibitor (SBI-0206965) reversed AP39 antagonism of doxorubicin-induced FUNDC1-mediated impairment of mitophagy and secondary cardiomyocyte pyroptosis. These results suggest that mitochondria-targeted H2S can antagonize doxorubicin-induced pyroptosis and impaired mitophagy in cardiomyocytes via AMPK-ULK1-FUNDC1 and ameliorated myocardial fibrosis and remodeling.
Collapse
Affiliation(s)
- Junxiong Zhao
- Department of Cardiology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421000, China
| | - Ting Yang
- School of Pharmaceutical Science of University of South China, Hengyang 421000, China
- Department of Pharmacy, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421000, China
| | - Jiali Yi
- Department of Cardiology, Hunan University of Medicine General Hospital, Huaihua 418000, China
| | - Hongmin Hu
- Department of Cardiology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421000, China
| | - Qi Lai
- School of Pharmaceutical Science of University of South China, Hengyang 421000, China
- Department of Pharmacy, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421000, China
| | - Liangui Nie
- Department of Cardiology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421000, China
| | - Maojun Liu
- Department of Cardiology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421000, China
| | - Chun Chu
- Department of Pharmacy, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421000, China
| | - Jun Yang
- Department of Cardiology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421000, China
| |
Collapse
|
11
|
Lv L, Chen Q, Lu J, Zhao Q, Wang H, Li J, Yuan K, Dong Z. Potential regulatory role of epigenetic modifications in aging-related heart failure. Int J Cardiol 2024; 401:131858. [PMID: 38360101 DOI: 10.1016/j.ijcard.2024.131858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Revised: 12/27/2023] [Accepted: 02/10/2024] [Indexed: 02/17/2024]
Abstract
Heart failure (HF) is a serious clinical syndrome and a serious development or advanced stage of various heart diseases. Aging is an independent factor that causes pathological damage in cardiomyopathy and participates in the occurrence of HF at the molecular level by affecting mechanisms such as telomere shortening and mitochondrial dysfunction. Epigenetic changes have a significant impact on the aging process, and there is increasing evidence that genetic and epigenetic changes are key features of aging and aging-related diseases. Epigenetic modifications can affect genetic information by changing the chromatin state without changing the DNA sequence. Most of the genetic loci that are highly associated with cardiovascular diseases (CVD) are located in non-coding regions of the genome; therefore, the epigenetic mechanism of CVD has attracted much attention. In this review, we focus on the molecular mechanisms of HF during aging and epigenetic modifications mediating aging-related HF, emphasizing that epigenetic mechanisms play an important role in the pathogenesis of aging-related CVD and can be used as potential diagnostic and prognostic biomarkers, as well as therapeutic targets.
Collapse
Affiliation(s)
- Lin Lv
- Department of Pharmacy, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - QiuYu Chen
- The Key Laboratory of Cardiovascular Disease Acousto-Optic Electromagnetic Diagnosis and Treatment in Heilongjiang Province, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jing Lu
- Department of Pharmacy, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Qi Zhao
- Department of Pharmacy, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - HongYan Wang
- Department of Pharmacy, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - JiaHao Li
- Department of Pharmacy, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - KeYing Yuan
- Department of Pharmacy, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - ZengXiang Dong
- The Key Laboratory of Cardiovascular Disease Acousto-Optic Electromagnetic Diagnosis and Treatment in Heilongjiang Province, First Affiliated Hospital of Harbin Medical University, Harbin, China; NHC Key Laboratory of Cell Transplantation, First Affiliated Hospital of Harbin Medical University, Harbin, China.
| |
Collapse
|
12
|
Zhang B, Li Y, Liu N, Liu B. AP39, a novel mitochondria-targeted hydrogen sulfide donor ameliorates doxorubicin-induced cardiotoxicity by regulating the AMPK/UCP2 pathway. PLoS One 2024; 19:e0300261. [PMID: 38568919 PMCID: PMC10990198 DOI: 10.1371/journal.pone.0300261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Accepted: 02/25/2024] [Indexed: 04/05/2024] Open
Abstract
Doxorubicin (DOX) is a broad-spectrum, highly effective antitumor agent; however, its cardiotoxicity has greatly limited its use. Hydrogen sulfide (H2S) is an endogenous gaseous transmitter that exerts cardioprotective effects via the regulation of oxidative stress and apoptosis and maintenance of mitochondrial function, among other mechanisms. AP39 is a novel mitochondria-targeted H2S donor that, at appropriate concentrations, attenuates intracellular oxidative stress damage, maintains mitochondrial function, and ameliorates cardiomyocyte injury. In this study, DOX-induced cardiotoxicity models were established using H9c2 cells and Sprague-Dawley rats to evaluate the protective effect of AP39 and its mechanisms of action. Both in vivo and in vitro experiments showed that DOX induces oxidative stress injury, apoptosis, and mitochondrial damage in cardiomyocytes and decreases the expression of p-AMPK/AMPK and UCP2. All DOX-induced changes were attenuated by AP39 treatment. Furthermore, the protective effect of AP39 was significantly attenuated by the inhibition of AMPK and UCP2. The results suggest that AP39 ameliorates DOX-induced cardiotoxicity by regulating the expression of AMPK/UCP2.
Collapse
Affiliation(s)
- Bin Zhang
- The Second Hospital of Jilin University, Nanguan District, Changchun City, Jilin Province, China
| | - Yangxue Li
- The Second Hospital of Jilin University, Nanguan District, Changchun City, Jilin Province, China
| | - Ning Liu
- The Second Hospital of Jilin University, Nanguan District, Changchun City, Jilin Province, China
| | - Bin Liu
- The Second Hospital of Jilin University, Nanguan District, Changchun City, Jilin Province, China
| |
Collapse
|
13
|
Shi X, Li H, Guo F, Li D, Xu F. Novel ray of hope for diabetic wound healing: Hydrogen sulfide and its releasing agents. J Adv Res 2024; 58:105-115. [PMID: 37245638 PMCID: PMC10982866 DOI: 10.1016/j.jare.2023.05.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Revised: 05/16/2023] [Accepted: 05/20/2023] [Indexed: 05/30/2023] Open
Abstract
BACKGROUND Diabetes mellitus (DM) is a long-term metabolic disease accompanied by difficulties in wound healing placing a severe financial and physical burden on patients. As one of the important signal transduction molecules, both endogenous and exogenous hydrogen sulfide (H2S) was found to promote diabetic wound healing in recent studies. H2S at physiological concentrations can not only promote cell migration and adhesion functions, but also resist inflammation, oxidative stress and inappropriate remodeling of the extracellular matrix. AIM OF REVIEW The purpose of this review is to summarize current research on the function of H2S in diabetic wound healing at all stages, and propose future directions. KEY SCIENTIFIC CONCEPTS OF REVIEW In this review, first, the various factors affecting wound healing under diabetic pathological conditions and the in vivo H2S generation pathway are briefly introduced. Second, how H2S may improve diabetic wound healing is categorized and described. Finally, we discuss the relevant H2S donors and new dosage forms, analyze and reveal the characteristics of many typical H2S donors, which may provide new ideas for the development of H2S-released agents to improve diabetic wound healing.
Collapse
Affiliation(s)
- Xinyi Shi
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Haonan Li
- Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, and School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Fengrui Guo
- Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, and School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Dahong Li
- Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, and School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang 110016, PR China.
| | - Fanxing Xu
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, PR China.
| |
Collapse
|
14
|
Wei W, Li T, Chen J, Fan Z, Gao F, Yu Z, Jiang Y. SIRT3/6: an amazing challenge and opportunity in the fight against fibrosis and aging. Cell Mol Life Sci 2024; 81:69. [PMID: 38294557 PMCID: PMC10830597 DOI: 10.1007/s00018-023-05093-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 11/29/2023] [Accepted: 12/09/2023] [Indexed: 02/01/2024]
Abstract
Fibrosis is a typical aging-related pathological process involving almost all organs, including the heart, kidney, liver, lung, and skin. Fibrogenesis is a highly orchestrated process defined by sequences of cellular response and molecular signals mechanisms underlying the disease. In pathophysiologic conditions associated with organ fibrosis, a variety of injurious stimuli such as metabolic disorders, epigenetic changes, and aging may induce the progression of fibrosis. Sirtuins protein is a kind of deacetylase which can regulate cell metabolism and participate in a variety of cell physiological functions. In this review, we outline our current understanding of common principles of fibrogenic mechanisms and the functional role of SIRT3/6 in aging-related fibrosis. In addition, sequences of novel protective strategies have been identified directly or indirectly according to these mechanisms. Here, we highlight the role and biological function of SIRT3/6 focus on aging fibrosis, as well as their inhibitors and activators as novel preventative or therapeutic interventions for aging-related tissue fibrosis.
Collapse
Affiliation(s)
- Wenxin Wei
- School of Queen Mary, Nanchang University, Nanchang, 330031, China
| | - Tian Li
- School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China.
| | - Jinlong Chen
- School of Chemistry and Chemical Engineering, Nangchang University, 999 Xuefu Rd, Nanchang, 330031, China
| | - Zhen Fan
- The Hospital Affiliated to Shanxi University of Chinese Medicine, Xianyang, 712000, China.
| | - Feng Gao
- Shanxi University of Chinese Medicine, Xianyang, 712046, China
| | - Zhibiao Yu
- School of Chemistry and Chemical Engineering, Nangchang University, 999 Xuefu Rd, Nanchang, 330031, China
| | - Yihao Jiang
- School of Chemistry and Chemical Engineering, Nangchang University, 999 Xuefu Rd, Nanchang, 330031, China.
| |
Collapse
|
15
|
Yang F, Zhong W, Pan S, Wang Y, Xiao Q, Gao X. Recent advances in the mechanism of hydrogen sulfide in wound healing in diabetes. Biochem Biophys Res Commun 2024; 692:149343. [PMID: 38065000 DOI: 10.1016/j.bbrc.2023.149343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 11/13/2023] [Accepted: 11/24/2023] [Indexed: 01/06/2024]
Abstract
Wound healing difficulties in diabetes continue to be a clinical challenge, posing a considerable burden to patients and society. Recently, exploration of the mechanism of wound healing and associated treatment options in diabetes has become topical. Of note, the positive role of hydrogen sulfide in promoting wound healing has been demonstrated in recent studies. Hydrogen sulfide is a confirmed gas transmitter in mammals, playing an essential role in pathology and physiology. This review describes the mechanism underlying the role of hydrogen sulfide in the promotion of diabetic wound healing and the potential for hydrogen sulfide supplementation as a therapeutic application.
Collapse
Affiliation(s)
- Fengze Yang
- College of Stomatology, Chongqing Medical University, Chongqing, 401147, China; Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, 401147, China; Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, 401147, China.
| | - Wenjie Zhong
- College of Stomatology, Chongqing Medical University, Chongqing, 401147, China; Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, 401147, China; Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, 401147, China.
| | - Shengyuan Pan
- College of Stomatology, Chongqing Medical University, Chongqing, 401147, China; Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, 401147, China; Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, 401147, China.
| | - Yue Wang
- College of Stomatology, Chongqing Medical University, Chongqing, 401147, China; Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, 401147, China; Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, 401147, China.
| | - Qingyue Xiao
- College of Stomatology, Chongqing Medical University, Chongqing, 401147, China; Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, 401147, China; Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, 401147, China.
| | - Xiang Gao
- College of Stomatology, Chongqing Medical University, Chongqing, 401147, China; Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, 401147, China; Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, 401147, China.
| |
Collapse
|
16
|
Zhang P, Liu X, Yu X, Zhuo Y, Li D, Yang L, Lu Y. Protective Effects of Liriodendrin on Myocardial Infarction-Induced Fibrosis in Rats via the PI3K/Akt Autophagy Pathway: A Network Pharmacology Study. Comb Chem High Throughput Screen 2024; 27:1566-1575. [PMID: 37461344 DOI: 10.2174/1386207326666230717155641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 05/31/2023] [Accepted: 05/31/2023] [Indexed: 07/04/2024]
Abstract
BACKGROUND Liriodendrin (LIR) has been reported to improve cardiac function in rats following myocardial infarction. However, its role and mechanism in reparative myocardial fibrosis remain unclear. METHODS In this study, a rat model of myocardial fibrosis was established via left anterior descending artery ligation and randomly divided into three groups (n = 6 per group): sham-operated, myocardial infarction, and LIR intervention (100 mg/kg/day) groups. The pharmacological effects of LIR were assessed using echocardiography, hematoxylin, and eosin (H&E) staining, and Masson staining. Network pharmacology and bioinformatics were utilized to identify potential mechanisms of LIR, which were further validated via western blot analysis. RESULTS Our findings demonstrated that LIR improved cardiac function, histology scores, and col lagen volume fraction. Moreover, LIR downregulated the expression of Beclin-1, LC3-II/LC3-I while upregulating the expression of p62, indicating LIR-inhibited autophagy in the heart after myocardial infarction. Further analysis revealed that the PI3K/Akt signaling pathway was significantly enriched and validated by western blot. This analysis suggested that the ratios of p- PI3K/PI3K, p-Akt/Akt, and p-mTOR/mTOR were significantly increased. CONCLUSION LIR may attenuate myocardial infarction-induced fibrosis in rats by inhibiting excessive myocardial autophagy, with the potential mechanism involving the activation of the PI3K/Akt/mTOR pathway.
Collapse
Affiliation(s)
- Ping Zhang
- Department of Cardiology, Tianjin Nankai Hospital, Tianjin, 300100, China
- Tianjin Key Laboratory of Acute Abdomen Disease Associated Organ Injury and ITCWM Repair, Tianjin Nankai Hospital, Tianjin, 300100, China
| | - Xuanming Liu
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Xin Yu
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Yuzhen Zhuo
- Tianjin Key Laboratory of Acute Abdomen Disease Associated Organ Injury and ITCWM Repair, Tianjin Nankai Hospital, Tianjin, 300100, China
| | - Dihua Li
- Tianjin Key Laboratory of Acute Abdomen Disease Associated Organ Injury and ITCWM Repair, Tianjin Nankai Hospital, Tianjin, 300100, China
| | - Lei Yang
- Tianjin Key Laboratory of Acute Abdomen Disease Associated Organ Injury and ITCWM Repair, Tianjin Nankai Hospital, Tianjin, 300100, China
| | - Yanmin Lu
- Tianjin Key Laboratory of Acute Abdomen Disease Associated Organ Injury and ITCWM Repair, Tianjin Nankai Hospital, Tianjin, 300100, China
| |
Collapse
|
17
|
Yang T, Yang Q, Lai Q, Zhao J, Nie L, Liu S, Yang J, Chu C. AP39 inhibits ferroptosis by inhibiting mitochondrial autophagy through the PINK1/parkin pathway to improve myocardial fibrosis with myocardial infarction. Biomed Pharmacother 2023; 165:115195. [PMID: 37516015 DOI: 10.1016/j.biopha.2023.115195] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 07/10/2023] [Accepted: 07/18/2023] [Indexed: 07/31/2023] Open
Abstract
BACKGROUND AND PURPOSE Research has revealed the involvement of mitochondrial autophagy and iron death in the pathogenesis of myocardial fibrosis. The objective of this study is to investigate whether the mitochondrial-targeted H2S donor AP39 inhibits mitochondrial autophagy and antagonizes myocardial cell iron death through the PINK1/Parkin pathway, thereby improving myocardial fibrosis in rats with myocardial infarction. EXPERIMENTAL APPROACH A rat model of myocardial infarction was created by intraperitoneal injection of a high dose of isoproterenol, and H9c2 myocardial cells were subjected to hypoxic injury induced by CoCl2. Western blot, RT-PCR, transmission electron microscopy, immunohistochemistry, as well as echocardiography, and studies on isolated hearts were employed. KEY RESULTS In the hearts of rats with myocardial infarction, there was a significant accumulation of interstitial collagen fibers, accompanied by downregulation of CSE protein expression, activation of the PINK1/Parkin signaling pathway, and activation of mitochondrial autophagy. Intervention with AP39 resulted in a significant improvement of the aforementioned changes, which could be reversed by the addition of PAG. Similar results were observed in vitro experiments. Furthermore, the addition of CCCP reversed the antagonistic effect of AP39 on myocardial cell iron death, while the addition of RSL3 reversed the inhibitory effect of AP39 on collagen production in myocardial cells. CONCLUSION AND IMPLICATIONS The mitochondrial-targeted H2S donor AP39 can inhibit mitochondrial autophagy through the PINK1/Parkin pathway, antagonize myocardial cell iron death, and improve myocardial fibrosis in rats with myocardial infarction.
Collapse
Affiliation(s)
- Ting Yang
- Department of Pharmacy, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421000, Hunan Province, China; School of Pharmaceutical Science of University of South China, Hengyang 421000, Hunan Province, China
| | - Qi Yang
- Department of Cardiology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421000, Hunan Province, China
| | - Qi Lai
- Department of Pharmacy, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421000, Hunan Province, China; School of Pharmaceutical Science of University of South China, Hengyang 421000, Hunan Province, China
| | - Junxiong Zhao
- Department of Cardiology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421000, Hunan Province, China
| | - Liangui Nie
- Department of Cardiology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421000, Hunan Province, China
| | - Shengquan Liu
- Department of Cardiology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421000, Hunan Province, China
| | - Jun Yang
- Department of Cardiology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421000, Hunan Province, China.
| | - Chun Chu
- Department of Pharmacy, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421000, Hunan Province, China.
| |
Collapse
|
18
|
Wu T, Qu Y, Xu S, Wang Y, Liu X, Ma D. SIRT6: A potential therapeutic target for diabetic cardiomyopathy. FASEB J 2023; 37:e23099. [PMID: 37462453 DOI: 10.1096/fj.202301012r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Revised: 06/26/2023] [Accepted: 07/05/2023] [Indexed: 07/21/2023]
Abstract
The abnormal lipid metabolism in diabetic cardiomyopathy can cause myocardial mitochondrial dysfunction, lipotoxicity, abnormal death of myocardial cells, and myocardial remodeling. Mitochondrial homeostasis and normal lipid metabolism can effectively slow down the development of diabetic cardiomyopathy. Recent studies have shown that SIRT6 may play an important role in the pathological changes of diabetic cardiomyopathy such as myocardial cell death, myocardial hypertrophy, and myocardial fibrosis by regulating mitochondrial oxidative stress and glucose and lipid metabolism. Therefore, understanding the function of SIRT6 and its role in the pathogenesis of diabetic cardiomyopathy is of great significance for exploring and developing new targets and drugs for the treatment of diabetic cardiomyopathy. This article reviews the latest findings of SIRT6 in the pathogenesis of diabetic cardiomyopathy, focusing on the regulation of mitochondria and lipid metabolism by SIRT6 to explore potential clinical treatments.
Collapse
Affiliation(s)
- Tao Wu
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yiwei Qu
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Shengjie Xu
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yong Wang
- Department of Cardiology, Shandong University of Traditional Chinese Medicine Affiliated Hospital, Jinan, China
| | - Xue Liu
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Dufang Ma
- Department of Cardiology, Shandong University of Traditional Chinese Medicine Affiliated Hospital, Jinan, China
| |
Collapse
|
19
|
Tyagi R, Chakraborty S, Tripathi SJ, Jung IR, Kim SF, Snyder SH, Paul BD. Inositol polyphosphate multikinase modulates redox signaling through nuclear factor erythroid 2-related factor 2 and glutathione metabolism. iScience 2023; 26:107199. [PMID: 37456841 PMCID: PMC10345128 DOI: 10.1016/j.isci.2023.107199] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 03/24/2023] [Accepted: 06/20/2023] [Indexed: 07/18/2023] Open
Abstract
Maintenance of redox balance plays central roles in a plethora of signaling processes. Although physiological levels of reactive oxygen and nitrogen species are crucial for functioning of certain signaling pathways, excessive production of free radicals and oxidants can damage cell components. The nuclear factor erythroid 2-related factor 2 (Nrf2) signaling cascade is the key pathway that mediates cellular response to oxidative stress. It is controlled at multiple levels, which serve to maintain redox homeostasis within cells. We show here that inositol polyphosphate multikinase (IPMK) is a modulator of Nrf2 signaling. IPMK binds Nrf2 and attenuates activation and expression of Nrf2 target genes. Furthermore, depletion of IPMK leads to elevated glutathione and cysteine levels, resulting in increased resistance to oxidants. Accordingly, targeting IPMK may restore redox balance under conditions of cysteine and glutathione insufficiency.
Collapse
Affiliation(s)
- Richa Tyagi
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Suwarna Chakraborty
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Sunil Jamuna Tripathi
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Ik-Rak Jung
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Johns Hopkins University, Baltimore, MD 21224, USA
| | - Sangwon F. Kim
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Johns Hopkins University, Baltimore, MD 21224, USA
| | - Solomon H. Snyder
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Bindu D. Paul
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Lieber Institute for Brain Development, Baltimore, MD 21205, USA
| |
Collapse
|
20
|
Yang J, Kim W, Kim DR. Autophagy in Cell Survival and Death. Int J Mol Sci 2023; 24:ijms24054744. [PMID: 36902171 PMCID: PMC10002575 DOI: 10.3390/ijms24054744] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 02/27/2023] [Indexed: 03/05/2023] Open
Abstract
Autophagy is a degradative process to remove damaged or unnecessary cellular components, and it has been implicated in many biological processes during cell survival and death [...].
Collapse
|
21
|
Zhao J, Wu Q, Yang T, Nie L, Liu S, Zhou J, Chen J, Jiang Z, Xiao T, Yang J, Chu C. Gaseous signal molecule SO 2 regulates autophagy through PI3K/AKT pathway inhibits cardiomyocyte apoptosis and improves myocardial fibrosis in rats with type II diabetes. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2022; 26:541-556. [PMID: 36302628 PMCID: PMC9614393 DOI: 10.4196/kjpp.2022.26.6.541] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 09/08/2022] [Accepted: 09/13/2022] [Indexed: 11/06/2022]
Abstract
Myocardial fibrosis is a key link in the occurrence and development of diabetic cardiomyopathy. Its etiology is complex, and the effect of drugs is not good. Cardiomyocyte apoptosis is an important cause of myocardial fibrosis. The purpose of this study was to investigate the effect of gaseous signal molecule sulfur dioxide (SO2) on diabetic myocardial fibrosis and its internal regulatory mechanism. Masson and TUNEL staining, Western-blot, transmission electron microscopy, RT-qPCR, immunofluorescence staining, and flow cytometry were used in the study, and the interstitial collagen deposition, autophagy, apoptosis, and changes in phosphatidylinositol 3-kinase (PI3K)/AKT pathways were evaluated from in vivo and in vitro experiments. The results showed that diabetic myocardial fibrosis was accompanied by cardiomyocyte apoptosis and down-regulation of endogenous SO2-producing enzyme aspartate aminotransferase (AAT)1/2. However, exogenous SO2 donors could up-regulate AAT1/2, reduce apoptosis of cardiomyocytes induced by diabetic rats or high glucose, inhibit phosphorylation of PI3K/AKT protein, up-regulate autophagy, and reduce interstitial collagen deposition. In conclusion, the results of this study suggest that the gaseous signal molecule SO2 can inhibit the PI3K/AKT pathway to promote cytoprotective autophagy and inhibit cardiomyocyte apoptosis to improve myocardial fibrosis in diabetic rats. The results of this study are expected to provide new targets and intervention strategies for the prevention and treatment of diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Junxiong Zhao
- Department of Pharmacy, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421000, China,Department of Cardiology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421000, China
| | - Qian Wu
- Department of General Practice, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421000, China
| | - Ting Yang
- Department of Pharmacy, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421000, China,School of Pharmaceutical Science of University of South China, Hengyang 421000, China
| | - Liangui Nie
- Department of Cardiology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421000, China
| | - Shengquan Liu
- Department of Cardiology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421000, China
| | - Jia Zhou
- Department of Ultrasound Medicine, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421000, China
| | - Jian Chen
- Department of Critical Care Medicine, The Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang 421000, China
| | - Zhentao Jiang
- Department of Cardiology, The Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang 421000, China
| | - Ting Xiao
- Department of Cardiology, Shenzhen Longhua District Central Hospital, Longhua Central Hospital Affiliated Guang-dong Medical University, Shenzhen 518000, China,Ting Xiao, E-mail:
| | - Jun Yang
- Department of Cardiology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421000, China,Jun Yang, E-mail:
| | - Chun Chu
- Department of Pharmacy, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421000, China,Correspondence Chun Chu, E-mail:
| |
Collapse
|
22
|
hUMSCs Transplantation Regulates AMPK/NR4A1 Signaling Axis to Inhibit Ovarian Fibrosis in POI Rats. Stem Cell Rev Rep 2022:10.1007/s12015-022-10469-y. [PMID: 36307672 DOI: 10.1007/s12015-022-10469-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/17/2022] [Indexed: 11/26/2022]
Abstract
BACKGROUND The mechanism of human Umbilical Cord Mesenchymal Stem Cells (hUMSCs) transplantation to improve ovarian function in the rats with Premature Ovarian Insufficiency (POI) is still unclear. The aim of this study is to investigate the signal axis mechanism that is involved in the ovarian function recovery of POI rats following hUMSCs transplantation. METHODS The rat model with POI was established by intraperitoneal injection of cisplatin. The hUMSCs were transplanted by caudal vein injection into POI rats. Hematoxylin-eosin (H&E) staining was performed to examine the morphology of rat ovarian tissue. Masson staining, Sirus red staining and immunofluorescence were used to observe the fibrosis extent of ovarian tissue. The levels of serum sex hormones and the expression of fibrosis related markers in ovarian tissues were measured by enzyme-linked immunosorbent assay (ELISA). The expression of NR4A1, Phospho-NR4A1 and AMP-activated protein kinase (AMPK) signaling in rat ovarian tissues was measured by immunohistochemistry and immunofluorescence. The role of AMPK/NR4A1 signaling axis in the regulation of ovarian function recovery in POI rats following hUMSCs transplantation was further investigated by adenovirus and siRNA intervention in isolated stromal cells. RESULTS The results showed that the hUMSCs transplantation significantly inhibited ovarian tissue fibrosis and restored the ovarian function in POI rats. The level of NR4A1 and AMPK expression in ovarian tissue of POI rats after hUMSCs transplantation was significantly increased compared with the control group. In the cultured ovarian stromal cells, the similar results were obtained on the expression of NR4A1 and its regulation on fibrosis related molecular markers in Cisplatin (CDDP) damaged stromal cells following hUMSCs supernatant treatment. Both hUMSCs supernatant treatment and the addition of AMPK inhibitors increased NR4A1 expression in stromal cells. And after NR4A1 molecular intervention, fibrosis-related indicators in stromal cells changed. The data suggests that the AMPK/NR4A1 signaling axis is involved in the ovarian function changes in POI rats following hUMSCs transplantation. CONCLUSION The data from this study indicate that the inhibition of tissue fibrosis and recovery of ovarian function is regulated by AMPK/NR4A1 signaling axis in POI rats following hUMSCs transplantation.
Collapse
|
23
|
Polydatin Attenuates Cisplatin-Induced Acute Kidney Injury via SIRT6-Mediated Autophagy Activation. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:9035547. [PMID: 36160707 PMCID: PMC9507782 DOI: 10.1155/2022/9035547] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 07/17/2022] [Accepted: 08/26/2022] [Indexed: 11/18/2022]
Abstract
In the treatment of malignant tumors, the effectiveness of cisplatin (CP) is limited by its nephrotoxicity, leading to cisplatin-induced acute kidney injury (CP-AKI). Polydatin (PD) has been demonstrated to regulate autophagy in tumors, sepsis, and diabetes. We have recently confirmed that PD attenuated CP-AKI by inhibiting ferroptosis, but it is not clear whether PD can regulate autophagy to protect from CP-AKI. The purpose of this study was to investigate the effect of PD on autophagy in CP-treated HK-2 cells and CP-AKI mouse models, exploring the role of sirtuin 6 (SIRT6) upregulated by PD. In this study, the blocking of autophagy flux was observed in both CP-treated HK-2 cells in vitro and CP-AKI mouse models in vivo, whereas this blocking was reversed by PD, which was characterized by the increase of autophagy microtubule-associated protein light chain 3 II expression and autophagolysosome/autophagosome ratio and the decrease of p62 expression. Furthermore, PD also significantly increased the expression of SIRT6 in vivo and in vitro. The protective effect of PD manifested by the stimulating of autophagy flux, with the reducing of inflammatory response and oxidative stress, which included downregulation of tumor necrosis factor-α and interleukin-1β, decreased activity of myeloperoxidase and content of malondialdehyde, and increased activity of superoxide dismutase and level of glutathione, both in vivo and in vitro, was reversed by either inhibition of autophagy flux by chloroquine or downregulation of SIRT6 by OSS-128167. Taken together, the present findings provide the first evidence demonstrating that PD exhibited nephroprotective effects on CP-AKI by restoring SIRT6-mediated autophagy flux mechanisms.
Collapse
|
24
|
Li X, Li W, Zhang Z, Wang W, Huang H. SIRT6 overexpression retards renal interstitial fibrosis through targeting HIPK2 in chronic kidney disease. Front Pharmacol 2022; 13:1007168. [PMID: 36172184 PMCID: PMC9510922 DOI: 10.3389/fphar.2022.1007168] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Accepted: 08/22/2022] [Indexed: 11/27/2022] Open
Abstract
Introduction: Renal interstitial fibrosis is a common pathophysiological change in the chronic kidney disease (CKD). Nicotinamide adenine dinucleotide (NAD)-dependent deacetylase sirtuin 6 (SIRT6) is demonstrated to protect against kidney injury. Vitamin B3 is the mostly used form of NAD precursors. However, the role of SIRT6 overexpression in renal interstitial fibrosis of CKD and the association between dietary vitamin B3 intake and renal function remain to be elucidated. Methods: Wild-type (WT) and SIRT6-transgene (SIRT6-Tg) mice were given with high-adenine diets to establish CKD model. HK2 cells were exposed to transforming growth factor β1 (TGF-β1) in vitro to explore related mechanism. Population data from Multi-Ethnic Study of Atherosclerosis (MESA) was used to examine the association between dietary vitamin B3 intake and renal function decline. Results: Compared to WT mice, SIRT6-Tg mice exhibited alleviated renal interstitial fibrosis as evidenced by reduced collagen deposit, collagen I and α-smooth muscle actin expression. Renal function was also improved in SIRT6-Tg mice. Homeodomain interacting protein kinase 2 (HIPK2) was induced during the fibrogenesis in CKD, while HIPK2 was downregulated after SIRT6 overexpression. Further assay in vitro confirmed that SIRT6 depletion exacerbated epithelial-to-mesenchymal transition of HK2 cells, which might be linked with HIPK2 upregulation. HIPK2 was inhibited by SIRT6 in the post-transcriptional level. Population study indicated that higher dietary vitamin B3 intake was independently correlated with a lower risk of estimate glomerular filtration rate decline in those ≥65 years old during follow-up. Conclusion: SIRT6/HIPK2 axis serves as a promising target of renal interstitial fibrosis in CKD. Dietary vitamin B3 intake is beneficial for renal function in the old people.
Collapse
Affiliation(s)
- Xiaoxue Li
- Department of Cardiology, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Wenxin Li
- Department of Cardiology, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Zhengzhipeng Zhang
- Department of Cardiology, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Weidong Wang
- Department of Pathophysiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Hui Huang
- Department of Cardiology, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
- *Correspondence: Hui Huang,
| |
Collapse
|
25
|
Li X, Liu L, Jiang W, Liu M, Wang Y, Ma H, Mu N, Wang H. SIRT6 Protects Against Myocardial Ischemia-Reperfusion Injury by Attenuating Aging-Related CHMP2B Accumulation. J Cardiovasc Transl Res 2022; 15:740-753. [PMID: 35235147 DOI: 10.1007/s12265-021-10184-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Accepted: 11/03/2021] [Indexed: 12/15/2022]
Abstract
Impaired autophagic flux induces aging-related ischemia vulnerability, which is the hallmark pathology in cardiac aging. Our previous work has confirmed that the accumulation of charged multivesicular body protein 2B (CHMP2B), a subunit of the ESCRT-III complex, in the heart can impair autophagy flux. However, whether CHMP2B accumulation contributes to aging-related intolerance to ischemia/reperfusion (I/R) injury and the regulatory mechanism for CHMP2B in aged heart remain elusive. The cardiac CHMP2B level was significantly higher in aged human myocardium than that in young myocardium. Increased CHMP2B were shown to inhibit autophagic flux leading to the deterioration of MI/R injury in aged mice hearts. Interestingly, a negative correlation was observed between SIRT6 and CHMP2B expression in human heart samples. Specific activation of SIRT6 suppressed CHMP2B accumulation and ameliorated autophagy flux in aged hearts. Using myocardial-specific SIRT6 heterozygous knockout mice and recovery experiments confirmed that SIRT6 regulated myocardial CHMP2B levels. Finally, activation of SIRT6 decreased acetylation of FoxO1 to promote its transcriptional function on Atrogin-1, a muscle-specific ubiquitin ligase, which subsequently enhanced the degradation of CHMP2B by Atrogin-1. This is a novel mechanism for SIRT6 against aging-related myocardial ischemia vulnerability, particularly by preventing excessive accumulation of autophagy key factors.
Collapse
Affiliation(s)
- Xiaokang Li
- Department of Cardiology, Tangdu Hospital, Fourth Military Medical University, No. 1 Xinsi Rd, Xi'an, China
| | - Lin Liu
- Department of Dermatology, Tangdu Hospital, Fourth Military Medical University, No. 1 Xinsi Rd, Xi'an, 710032, China
| | - Wenhua Jiang
- Institute of Medical Research, Northwestern Polytechnical University, Xi'an, China
| | - Manling Liu
- Department of Physiology and Pathophysiology, School of Basic Medicine, Fourth Military Medical University, No. 169 Changle West Rd, Xi'an, China
| | - Yishi Wang
- Department of Physiology and Pathophysiology, School of Basic Medicine, Fourth Military Medical University, No. 169 Changle West Rd, Xi'an, China
| | - Heng Ma
- Department of Physiology and Pathophysiology, School of Basic Medicine, Fourth Military Medical University, No. 169 Changle West Rd, Xi'an, China.
| | - Nan Mu
- Department of Physiology and Pathophysiology, School of Basic Medicine, Fourth Military Medical University, No. 169 Changle West Rd, Xi'an, China.
| | - Haiyan Wang
- Department of Cardiology, Tangdu Hospital, Fourth Military Medical University, No. 1 Xinsi Rd, Xi'an, China.
| |
Collapse
|
26
|
Yeon Park S, Cho W, Abd El-Aty A, Hacimuftuoglu A, Hoon Jeong J, Woo Jung T. Valdecoxib attenuates lipid-induced hepatic steatosis through autophagy-mediated suppression of endoplasmic reticulum stress. Biochem Pharmacol 2022; 199:115022. [DOI: 10.1016/j.bcp.2022.115022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Revised: 03/22/2022] [Accepted: 03/23/2022] [Indexed: 02/09/2023]
|
27
|
Barrow K, Wang Y, Yu R, Zhu J, Yang G. H 2S protects from oxidative stress-driven ACE2 expression and cardiac aging. Mol Cell Biochem 2022; 477:1393-1403. [PMID: 35147902 PMCID: PMC8831182 DOI: 10.1007/s11010-022-04386-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Accepted: 02/02/2022] [Indexed: 11/10/2022]
Abstract
Cystathionine gamma-lyase (CSE)-derived hydrogen sulfide (H2S) plays an essential role in preserving cardiac functions. Angiotensin-converting enzyme 2 (ACE2) acts as the negative regulator of the renin-angiotensin system, exerting anti-oxidative stress and anti-inflammatory properties within the body. The interplays of CSE/H2S signaling and ACE2 in cardiac aging are unclear. In this study, the regulatory roles of H2S on ACE2 expression in mouse heart tissue and rat cardiomyocytes under different stress conditions were investigated. It was found that ACE2 protein level was lower in heart tissues from old mice (56-week-old) than young mice (8-week-old), and the knockout of CSE (CSE KO) induced moderate oxidative stress and further inhibited ACE2 protein level in mouse hearts at both young and old age. Incubation of rat cardiac cells (H9C2) with a low dose of H2O2 (50 µM) suppressed ACE2 protein level and induced cellular senescence, which was completely reversed by co-incubation with 30 µM NaHS (a H2S donor). Prolonged nutrient excess is an increased risk of heart disorders by causing metabolic dysfunction and cardiac remodeling. We further found high-fat diet feeding stimulated ACE2 expression and induced severe oxidative stress in CSE KO heart in comparison with wild-type heart. Lipid overload in H9C2 cells to mimic a status of nutrient excess also enhanced the expression of ACE2 protein and induced severe oxidative stress and cell senescence, which were significantly attenuated by the supplementation of exogenous H2S. Furthermore, the manipulation of ACE2 expression partially abolished the protective role of H2S against cellular senescence. These results demonstrate the dynamic roles of H2S in the maintenance of ACE2 levels under different levels of oxidative stress, pointing to the potential implications in targeting the CSE/H2S system for the interruption of aging and diabetes-related heart disorders.
Collapse
Affiliation(s)
- Kalem Barrow
- School of Natural Sciences, Laurentian University, Sudbury, Canada.,Cardiovascular and Metabolic Research Unit, Laurentian University, Sudbury, Canada
| | - Yuehong Wang
- School of Natural Sciences, Laurentian University, Sudbury, Canada.,Cardiovascular and Metabolic Research Unit, Laurentian University, Sudbury, Canada
| | - Ruihuan Yu
- School of Natural Sciences, Laurentian University, Sudbury, Canada.,Cardiovascular and Metabolic Research Unit, Laurentian University, Sudbury, Canada
| | - Jiechun Zhu
- School of Natural Sciences, Laurentian University, Sudbury, Canada.,Cardiovascular and Metabolic Research Unit, Laurentian University, Sudbury, Canada
| | - Guangdong Yang
- School of Natural Sciences, Laurentian University, Sudbury, Canada. .,Cardiovascular and Metabolic Research Unit, Laurentian University, Sudbury, Canada.
| |
Collapse
|
28
|
Cui T, Liu W, Yu C, Ren J, Li Y, Shi X, Li Q, Zhang J. Protective Effects of Allicin on Acute Myocardial Infarction in Rats via Hydrogen Sulfide-mediated Regulation of Coronary Arterial Vasomotor Function and Myocardial Calcium Transport. Front Pharmacol 2022; 12:752244. [PMID: 35046802 PMCID: PMC8762278 DOI: 10.3389/fphar.2021.752244] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 11/30/2021] [Indexed: 12/12/2022] Open
Abstract
Acute myocardial infarction (AMI) is a condition with high morbidity and mortality, for which effective treatments are lacking. Allicin has been reported to exert therapeutic effects on AMI, but the underlying mechanisms of its action have not been fully elucidated. To investigate this, a rat model of AMI was generated by ligating the left anterior descending branch of the coronary artery. DL-propargylglycine (PAG), a specific hydrogen sulfide (H2S) synthetase inhibitor, was used to examine the effects of allicin on H2S production. Isolated coronary arteries and cardiomyocytes were assessed for vascular reactivity and cellular Ca2+ transport using a multiwire myography system and a cell-contraction-ion detection system, respectively. Allicin administration improved cardiac function and myocardial pathology, reduced myocardial enzyme levels, and increased H2S and H2S synthetase levels. Allicin administration resulted in concentration-dependent effects on coronary artery dilation, which were mediated by receptor-dependent Ca2+ channels, ATP-sensitive K+ channels, and sarcoplasmic reticulum (SR) Ca2+ release induced by the ryanodine receptor. Allicin administration improved Ca2+ homeostasis in cardiomyocytes by increasing cardiomyocyte contraction, Ca2+ transient amplitude, myofilament sensitivity, and SR Ca2+ content. Allicin also enhanced Ca2+ uptake via SR Ca2+-ATPase and Ca2+ removal via the Na+/Ca2+ exchanger, and it reduced SR Ca2+ leakage. Notably, the protective effects of allicin were partially attenuated by blockade of H2S production with PAG. Our findings provide novel evidence that allicin-induced production of H2S mediates coronary artery dilation and regulation of Ca2+ homeostasis in AMI. Our study presents a novel mechanistic insight into the anti-AMI effects of allicin and highlights the therapeutic potential of this compound.
Collapse
Affiliation(s)
- Tianwei Cui
- Institute of Basic Medical Sciences, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China.,Department of Reproductive Medicine, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Weiyu Liu
- Institute of Basic Medical Sciences, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Chenghao Yu
- Institute of Basic Medical Sciences, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jianxun Ren
- Institute of Basic Medical Sciences, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yikui Li
- Health Prevention Department, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xiaolu Shi
- Beijing Key Laboratory of TCM Basic Research on Prevention and Treatment of Major Disease, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, China
| | - Qiuyan Li
- Department of General Medicine, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jinyan Zhang
- Institute of Basic Medical Sciences, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
29
|
Yang X, Feng J, Liang W, Zhu Z, Chen Z, Hu J, Yang D, Ding G. Roles of SIRT6 in kidney disease: a novel therapeutic target. Cell Mol Life Sci 2021; 79:53. [PMID: 34950960 PMCID: PMC11072764 DOI: 10.1007/s00018-021-04061-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 11/24/2021] [Accepted: 11/25/2021] [Indexed: 12/13/2022]
Abstract
SIRT6 is an NAD+ dependent deacetylase that belongs to the mammalian sirtuin family. SIRT6 is mainly located in the nucleus and regulates chromatin remodeling, genome stability, and gene transcription. SIRT6 extensively participates in various physiological activities such as DNA repair, energy metabolism, oxidative stress, inflammation, and fibrosis. In recent years, the role of epigenetics such as acetylation modification in renal disease has gradually received widespread attention. SIRT6 reduces oxidative stress, inflammation, and renal fibrosis, which is of great importance in maintaining cellular homeostasis and delaying the chronic progression of kidney disease. Here, we review the structure and biological function of SIRT6 and summarize the regulatory mechanisms of SIRT6 in kidney disease. Moreover, the role of SIRT6 as a potential therapeutic target for the progression of kidney disease will be discussed. SIRT6 plays an important role in kidney disease. SIRT6 regulates mitochondrial dynamics and mitochondrial biogenesis, induces G2/M cycle arrest, and plays an antioxidant role in nephrotoxicity, IR, obstructive nephropathy, and sepsis-induced AKI. SIRT6 prevents and delays progressive CKD induced by hyperglycemia, kidney senescence, hypertension, and lipid accumulation by regulating mitochondrial biogenesis, and has antioxidant, anti-inflammatory, and antifibrosis effects. Additionally, hypoxia, inflammation, and fibrosis are the main mechanisms of the AKI-to-CKD transition. SIRT6 plays a critical role in the AKI-to-CKD transition and kidney repair through anti-inflammatory, antifibrotic, and mitochondrial quality control mechanisms. AKI Acute kidney injury, CKD Chronic kidney disease.
Collapse
Affiliation(s)
- Xueyan Yang
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
- Nephrology and Urology Research Institute of Wuhan University, Wuhan, Hubei, China
| | - Jun Feng
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
- Nephrology and Urology Research Institute of Wuhan University, Wuhan, Hubei, China
| | - Wei Liang
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
- Nephrology and Urology Research Institute of Wuhan University, Wuhan, Hubei, China
| | - Zijing Zhu
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
- Nephrology and Urology Research Institute of Wuhan University, Wuhan, Hubei, China
| | - Zhaowei Chen
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
- Nephrology and Urology Research Institute of Wuhan University, Wuhan, Hubei, China
| | - Jijia Hu
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
- Nephrology and Urology Research Institute of Wuhan University, Wuhan, Hubei, China
| | - Dingping Yang
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
- Nephrology and Urology Research Institute of Wuhan University, Wuhan, Hubei, China
| | - Guohua Ding
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China.
- Nephrology and Urology Research Institute of Wuhan University, Wuhan, Hubei, China.
| |
Collapse
|
30
|
Hydrogen Sulfide Attenuates Angiotensin II-Induced Cardiac Fibroblast Proliferation and Transverse Aortic Constriction-Induced Myocardial Fibrosis through Oxidative Stress Inhibition via Sirtuin 3. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:9925771. [PMID: 34603602 PMCID: PMC8486544 DOI: 10.1155/2021/9925771] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 05/30/2021] [Accepted: 08/07/2021] [Indexed: 12/12/2022]
Abstract
Sirtuin 3 (SIRT3) is critical in mitochondrial function and oxidative stress. Our present study investigates whether hydrogen sulfide (H2S) attenuated myocardial fibrosis and explores the possible role of SIRT3 on the protective effects. Neonatal rat cardiac fibroblasts were pretreated with NaHS followed by angiotensin II (Ang II) stimulation. SIRT3 was knocked down with siRNA technology. SIRT3 promoter activity and expression, as well as mitochondrial function, were measured. Male wild-type (WT) and SIRT3 knockout (KO) mice were intraperitoneally injected with NaHS followed by transverse aortic constriction (TAC). Myocardium sections were stained with Sirius red. Hydroxyproline content, collagen I and collagen III, α-smooth muscle actin (α-SMA), and dynamin-related protein 1 (DRP1) expression were measured both in vitro and in vivo. We found that NaHS enhanced SIRT3 promoter activity and increased SIRT3 mRNA expression. NaHS inhibited cell proliferation and hydroxyproline secretion, decreased collagen I, collagen III, α-SMA, and DRP1 expression, alleviated oxidative stress, and improved mitochondrial respiration function and membrane potential in Ang II-stimulated cardiac fibroblasts, which were unavailable after SIRT3 was silenced. In vivo, NaHS reduced hydroxyproline content, ameliorated perivascular and interstitial collagen deposition, and inhibited collagen I, collagen III, and DRP1 expression in the myocardium of WT mice but not SIRT3 KO mice with TAC. Altogether, NaHS attenuated myocardial fibrosis through oxidative stress inhibition via a SIRT3-dependent manner.
Collapse
|
31
|
Lv S, Liu H, Wang H. Exogenous Hydrogen Sulfide Plays an Important Role by Regulating Autophagy in Diabetic-Related Diseases. Int J Mol Sci 2021; 22:ijms22136715. [PMID: 34201520 PMCID: PMC8268438 DOI: 10.3390/ijms22136715] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 06/21/2021] [Accepted: 06/21/2021] [Indexed: 12/12/2022] Open
Abstract
Autophagy is a vital cell mechanism which plays an important role in many physiological processes including clearing long-lived, accumulated and misfolded proteins, removing damaged organelles and regulating growth and aging. Autophagy also participates in a variety of biological functions, such as development, cell differentiation, resistance to pathogens and nutritional hunger. Recently, autophagy has been reported to be involved in diabetes, but the mechanism is not fully understood. Hydrogen sulfide (H2S) is a colorless, water-soluble, flammable gas with the typical odor of rotten eggs, which has been known as a highly toxic gas for many years. However, it has been reported recently that H2S, together with nitric oxide and carbon monoxide, is an important gas signal transduction molecule. H2S has been reported to play a protective role in many diabetes-related diseases, but the mechanism is not fully clear. Recent studies indicate that H2S plays an important role by regulating autophagy in many diseases including cancer, tissue fibrosis diseases and glycometabolic diseases; however, the related mechanism has not been fully studied. In this review, we summarize recent research on the role of H2S in regulating autophagy in diabetic-related diseases to provide references for future related research.
Collapse
|
32
|
Wang YZ, Ngowi EE, Wang D, Qi HW, Jing MR, Zhang YX, Cai CB, He QL, Khattak S, Khan NH, Jiang QY, Ji XY, Wu DD. The Potential of Hydrogen Sulfide Donors in Treating Cardiovascular Diseases. Int J Mol Sci 2021; 22:2194. [PMID: 33672103 PMCID: PMC7927090 DOI: 10.3390/ijms22042194] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 02/05/2021] [Accepted: 02/10/2021] [Indexed: 02/08/2023] Open
Abstract
Hydrogen sulfide (H2S) has long been considered as a toxic gas, but as research progressed, the idea has been updated and it has now been shown to have potent protective effects at reasonable concentrations. H2S is an endogenous gas signaling molecule in mammals and is produced by specific enzymes in different cell types. An increasing number of studies indicate that H2S plays an important role in cardiovascular homeostasis, and in most cases, H2S has been reported to be downregulated in cardiovascular diseases (CVDs). Similarly, in preclinical studies, H2S has been shown to prevent CVDs and improve heart function after heart failure. Recently, many H2S donors have been synthesized and tested in cellular and animal models. Moreover, numerous molecular mechanisms have been proposed to demonstrate the effects of these donors. In this review, we will provide an update on the role of H2S in cardiovascular activities and its involvement in pathological states, with a special focus on the roles of exogenous H2S in cardiac protection.
Collapse
Affiliation(s)
- Yi-Zhen Wang
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China; (Y.-Z.W.); (E.E.N.); (D.W.); (H.-W.Q.); (M.-R.J.); (Y.-X.Z.); (C.-B.C.); (Q.-L.H.); (S.K.); (N.H.K.)
| | - Ebenezeri Erasto Ngowi
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China; (Y.-Z.W.); (E.E.N.); (D.W.); (H.-W.Q.); (M.-R.J.); (Y.-X.Z.); (C.-B.C.); (Q.-L.H.); (S.K.); (N.H.K.)
- Department of Biological Sciences, Faculty of Science, Dar es Salaam University College of Education, Dar es Salaam 2329, Tanzania
| | - Di Wang
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China; (Y.-Z.W.); (E.E.N.); (D.W.); (H.-W.Q.); (M.-R.J.); (Y.-X.Z.); (C.-B.C.); (Q.-L.H.); (S.K.); (N.H.K.)
| | - Hui-Wen Qi
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China; (Y.-Z.W.); (E.E.N.); (D.W.); (H.-W.Q.); (M.-R.J.); (Y.-X.Z.); (C.-B.C.); (Q.-L.H.); (S.K.); (N.H.K.)
| | - Mi-Rong Jing
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China; (Y.-Z.W.); (E.E.N.); (D.W.); (H.-W.Q.); (M.-R.J.); (Y.-X.Z.); (C.-B.C.); (Q.-L.H.); (S.K.); (N.H.K.)
| | - Yan-Xia Zhang
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China; (Y.-Z.W.); (E.E.N.); (D.W.); (H.-W.Q.); (M.-R.J.); (Y.-X.Z.); (C.-B.C.); (Q.-L.H.); (S.K.); (N.H.K.)
| | - Chun-Bo Cai
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China; (Y.-Z.W.); (E.E.N.); (D.W.); (H.-W.Q.); (M.-R.J.); (Y.-X.Z.); (C.-B.C.); (Q.-L.H.); (S.K.); (N.H.K.)
| | - Qing-Lin He
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China; (Y.-Z.W.); (E.E.N.); (D.W.); (H.-W.Q.); (M.-R.J.); (Y.-X.Z.); (C.-B.C.); (Q.-L.H.); (S.K.); (N.H.K.)
- School of Nursing and Health, Henan University, Kaifeng 475004, China
| | - Saadullah Khattak
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China; (Y.-Z.W.); (E.E.N.); (D.W.); (H.-W.Q.); (M.-R.J.); (Y.-X.Z.); (C.-B.C.); (Q.-L.H.); (S.K.); (N.H.K.)
- Kaifeng Municipal Key Laboratory of Cell Signal Transduction, Henan Provincial Engineering Centre for Tumor Molecular Medicine, Henan University, Kaifeng 475004, China
- School of Life Sciences, Henan University, Kaifeng 475004, China
| | - Nazeer Hussain Khan
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China; (Y.-Z.W.); (E.E.N.); (D.W.); (H.-W.Q.); (M.-R.J.); (Y.-X.Z.); (C.-B.C.); (Q.-L.H.); (S.K.); (N.H.K.)
- Kaifeng Municipal Key Laboratory of Cell Signal Transduction, Henan Provincial Engineering Centre for Tumor Molecular Medicine, Henan University, Kaifeng 475004, China
- School of Life Sciences, Henan University, Kaifeng 475004, China
| | - Qi-Ying Jiang
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China; (Y.-Z.W.); (E.E.N.); (D.W.); (H.-W.Q.); (M.-R.J.); (Y.-X.Z.); (C.-B.C.); (Q.-L.H.); (S.K.); (N.H.K.)
| | - Xin-Ying Ji
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China; (Y.-Z.W.); (E.E.N.); (D.W.); (H.-W.Q.); (M.-R.J.); (Y.-X.Z.); (C.-B.C.); (Q.-L.H.); (S.K.); (N.H.K.)
- Kaifeng Key Laboratory of Infection and Biological Safety, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China
| | - Dong-Dong Wu
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China; (Y.-Z.W.); (E.E.N.); (D.W.); (H.-W.Q.); (M.-R.J.); (Y.-X.Z.); (C.-B.C.); (Q.-L.H.); (S.K.); (N.H.K.)
- School of Stomatology, Henan University, Kaifeng 475004, China
| |
Collapse
|