1
|
Yavuz E, Cengiz IZ, Arslan A, Eser C. Analysis of the protective effect of hydrogen sulfide over time in ischemic rat skin flaps. ANN CHIR PLAST ESTH 2025:S0294-1260(25)00008-1. [PMID: 39863446 DOI: 10.1016/j.anplas.2025.01.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 01/09/2025] [Accepted: 01/14/2025] [Indexed: 01/27/2025]
Abstract
BACKGROUND Hydrogen sulfide (H2S) is a widely studied gasotransmitter, and its protective effect against ischemia-reperfusion damage has been explored in several studies. Therefore, a requirement exists for a comprehensive study about H2S effects on ischemia-reperfusion damage in flap surgery. The aim of this study is to examine the effect of hydrogen sulfide by creating ischemia-reperfusion injury in the vascular-stemmed island flap prepared from the rat groin area. MATERIALS AND METHODS "Wistar albino" rats weighing between 250 and 300 grams were divided into 4 groups (group 1, group 2, group 3, group 4). Each group was divided into 2 subgroups: subgroup A (control) and subgroup B (H2S). In each group, skin flaps were elevated as an island flap with a superficial epigastric artery pedicle, 6 × 4cm from the groin area. In subgroup B (H2S), liquid hydrogen sulfide was injected through the tail vein 20minutes before ischemia at a final concentration of 10μM. Femoral artery and vein blood flows were stopped with separate microclips and left in ischemia, according to the planned ischemia hours of the flaps: group 1 as 1 hour, group 2 as 2hours, group 3 as 3hours, and group 4 as 6hours. Later, microclips were removed, and blood flow restored again. After 12hours of reperfusion, the rats were sacrificed by cervical dislocation, and tissue samples were taken. From the samples taken, neutrophil count in ischemic tissue, MDA (malondialdehyde) measurement, and damage in the tissue were evaluated by electron microscopy. RESULTS On electron microscopy inspection at all hours (1, 2, 3, and 6), hydrogen sulfide was found to provide protection against ischemia, reperfusion damage, and apoptosis at the cellular level. There was a statistically significant (P=0.035) decrease in the tissue neutrophil count at the 1st, 2nd, and 3rd hours. In the tissue MDA measurement, a statistically significant (P=0.026) decrease in hydrogen sulfide was detected at the first hour. There was no statistically significant difference in the 6th hour tissue neutrophil count and 2nd, 3rd, and 6th hour tissue MDA measurement. CONCLUSION Electron microscopy results in this study showed that hydrogen sulfide had antiapoptotic effects on reperfusion damage in skin flaps at all hours. However, the neutrophil counts showed it had cytoprotective and anti-inflammatory properties during the 1st, 2nd, and 3rd hours following ischemia, but not during the 6th hour. Tissue MDA levels indicate that H2S mitigates significant I/R injury during the 1st hour but not in the subsequent 2nd, 3rd, and 6th hours. These results led to the hypothesis that, in order to offer a strong enough protective effect against I/R damage, H2S should be administered repeatedly or at varying concentrations. After more research on how H2S affects skin flaps, we believe that it can be used in plastic surgery practices.
Collapse
Affiliation(s)
- E Yavuz
- Department of Plastic, Reconstructive, and Aesthetic Surgery, Faculty of Medicine, Çukurova University, Adana, Turkey.
| | - I Z Cengiz
- Department of Plastic, Reconstructive, and Aesthetic Surgery, Faculty of Medicine, Istanbul Atlas University, Istanbul, Turkey
| | - A Arslan
- Department of Plastic, Reconstructive, and Aesthetic Surgery, Faculty of Medicine, Çukurova University, Adana, Turkey
| | - C Eser
- Department of Plastic, Reconstructive, and Aesthetic Surgery, Faculty of Medicine, Çukurova University, Adana, Turkey
| |
Collapse
|
2
|
Li L, Cao C, Guo H, Lin L, Li L, Zhang Y, Xin G, Liu Z, Xu S, Han X, Zhang Q, Fu J. Protective mechanism of safflower yellow injection on myocardial ischemia-reperfusion injury in rats by activating NLRP3 inflammasome. BMC Complement Med Ther 2025; 25:9. [PMID: 39789550 PMCID: PMC11715333 DOI: 10.1186/s12906-025-04747-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Accepted: 01/06/2025] [Indexed: 01/12/2025] Open
Abstract
OBJECTIVES This study intended to explore whether the protective effect safflower yellow injection (SYI) on myocardial ischemia-reperfusion (I/R) injury in rats mediated of the NLRP3 inflammasome signaling. METHODS The I/R model was prepared by ligating the left anterior descending coronary artery for 45 min and then releasing the blood flow for 150 min. 96 male Wistar rats were randomly divided into sham group, I/R group, Hebeishuang group (HBS), SYI high-dose group (I/R + SYI-H), SYI medium-dose group (I/R + SYI-M) and SYI low-dose group (I/R + SYI-L). Cell experiments were divided into normal control group (NC), Oxygen glucose deprivation/reoxygenation group (OGD/R), OGD/R + SYI group, OGD/R + SYI + Chloroquine group (OGD/R + SYI + CQ). The area of myocardial ischemia infarction and pathological changes were observed by the Tetrazolium method (TTC) and HE staining. Myocardial enzymes such as aspartate aminotransferase (AST), lactate dehydrogenase (LDH) and creatine kinase (CK) were measured by chemiluminescence (CL) method. The inflammatory factors levels of TNF-α, IL-1β, MCP-1, and IL-6 were detected by ELISA. The expressions of inflammatory-related proteins (Caspase-1, NLRP3, TLR4, NF-κB), autophagosome-related proteins (LC3-I, LC3-II,LC3-II/LC3-I), apoptosis-related proteins (Bax, Bcl-2, Caspase-3, Bcl-2/Bax) and autophagy-related proteins (p62/SQSTM1, PI3K, p-Akt, mTOR) were detected by Western-Blot. Cell morphology and cell viability were detected by transmission electron microscopy and CCK-8. RESULTS In vivo, compared with sham group, the percentage of myocardial infarction area was increased and myocardial tissue arrangement was disordered in I/R group. In addition, the activities of myocardial enzymes, the contents of inflammatory factors, the expressions of inflammatory-related proteins, autophagy-related proteins, autophagosome-related proteins, Bax and Caspase-3 were increased, while Bcl-2 and Bcl-2/Bax were decreased. SYI treatment reversed these trends, except for the expression of autophagosome-related proteins. In vitro, SYI decreased the contents of inflammatory factors and the expressions of inflammatory-related proteins, autophagy-related proteins and autophagosome-related proteins caused by OGD/R. However, the contents of inflammatory factors and the expression of inflammatory-related proteins, p62/SQSTM1 and mTOR were increased, while PI3K, p-AKT, LC3-II/LC3-I were significantly decreased in OGD/R + SYI + CQ group. CONCLUSIONS SYI can promote myocardial tissue autophagy by regulating NLRP3, thereby attenuating the myocardial inflammatory response and protecting damaged myocardium in I/R rats.
Collapse
Affiliation(s)
- Lingmei Li
- Kunshan Hospital of Chinese Medicine, Kunshan, China
| | - Ce Cao
- Institute of Basic Medical Sciences of Xiyuan Hospital, Beijing Key Laboratory of Chinese Materia Pharmacology, China Academy of Chinese Medical Sciences, National Clinical Research Center of Traditional Chinese Medicine for Cardiovascular Diseases, Beijing, China
| | - Hao Guo
- Institute of Basic Medical Sciences of Xiyuan Hospital, Beijing Key Laboratory of Chinese Materia Pharmacology, China Academy of Chinese Medical Sciences, National Clinical Research Center of Traditional Chinese Medicine for Cardiovascular Diseases, Beijing, China
| | - Li Lin
- Institute of Basic Medical Sciences of Xiyuan Hospital, Beijing Key Laboratory of Chinese Materia Pharmacology, China Academy of Chinese Medical Sciences, National Clinical Research Center of Traditional Chinese Medicine for Cardiovascular Diseases, Beijing, China
| | - Lei Li
- Institute of Basic Medical Sciences of Xiyuan Hospital, Beijing Key Laboratory of Chinese Materia Pharmacology, China Academy of Chinese Medical Sciences, National Clinical Research Center of Traditional Chinese Medicine for Cardiovascular Diseases, Beijing, China
| | - Yehao Zhang
- Institute of Basic Medical Sciences of Xiyuan Hospital, Beijing Key Laboratory of Chinese Materia Pharmacology, China Academy of Chinese Medical Sciences, National Clinical Research Center of Traditional Chinese Medicine for Cardiovascular Diseases, Beijing, China
| | - Gaojie Xin
- Institute of Basic Medical Sciences of Xiyuan Hospital, Beijing Key Laboratory of Chinese Materia Pharmacology, China Academy of Chinese Medical Sciences, National Clinical Research Center of Traditional Chinese Medicine for Cardiovascular Diseases, Beijing, China
| | - Zixin Liu
- Institute of Basic Medical Sciences of Xiyuan Hospital, Beijing Key Laboratory of Chinese Materia Pharmacology, China Academy of Chinese Medical Sciences, National Clinical Research Center of Traditional Chinese Medicine for Cardiovascular Diseases, Beijing, China
| | - Shujuan Xu
- Institute of Basic Medical Sciences of Xiyuan Hospital, Beijing Key Laboratory of Chinese Materia Pharmacology, China Academy of Chinese Medical Sciences, National Clinical Research Center of Traditional Chinese Medicine for Cardiovascular Diseases, Beijing, China
| | - Xiao Han
- Institute of Basic Medical Sciences of Xiyuan Hospital, Beijing Key Laboratory of Chinese Materia Pharmacology, China Academy of Chinese Medical Sciences, National Clinical Research Center of Traditional Chinese Medicine for Cardiovascular Diseases, Beijing, China.
| | - Qiong Zhang
- Institute of Basic Medical Sciences of Xiyuan Hospital, Beijing Key Laboratory of Chinese Materia Pharmacology, China Academy of Chinese Medical Sciences, National Clinical Research Center of Traditional Chinese Medicine for Cardiovascular Diseases, Beijing, China.
| | - Jianhua Fu
- Institute of Basic Medical Sciences of Xiyuan Hospital, Beijing Key Laboratory of Chinese Materia Pharmacology, China Academy of Chinese Medical Sciences, National Clinical Research Center of Traditional Chinese Medicine for Cardiovascular Diseases, Beijing, China.
| |
Collapse
|
3
|
Zheng XB, Wang C, Zhang M, Yao BQ, Wu HY, Hou SX. Exogenous H 2S targeting PI3K/AKT/mTOR pathway alleviates chronic intermittent hypoxia-induced myocardial damage through inhibiting oxidative stress and enhancing autophagy. Sleep Breath 2024; 29:43. [PMID: 39627628 DOI: 10.1007/s11325-024-03216-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Revised: 11/16/2024] [Accepted: 11/22/2024] [Indexed: 03/26/2025]
Abstract
AIMS Hydrogen sulfide (H2S) is a novel gas signaling molecule that has been researched in several physiological and pathological conditions, indicating that strategies targeting H2S may provide clinical benefits in diseases such as chronic cardiomyopathy. Here, we reveal the effect of H2S on chronic intermittent hypoxia (CIH)-related myocardial damage and its mechanistic relevance to phosphoinositol-3 kinase (PI3K). MATERIALS Mice were subjected to a 4-week CIH process to induce myocardial damage, which was accompanied by daily administration of NaHS (a H2S donor) and LY294002 (an inhibitor of PI3K). Changes in heart function were evaluated via echocardiography. Histological examination was applied to assess heart tissue lesions. Myocardial apoptosis was detected by TUNEL staining and apoptosis-associated protein expression. Furthermore, the effects of NaHS on autophagy and the PI3K/AKT/mTOR pathway were investigated. Finally, the level of inflammation is also affected by related proteins. KEY FINDINGS The CIH group presented increased myocardial dysfunction and heart tissue lesions. Echocardiography and histological analysis revealed that, compared with control mice, CIH-treated mice presented significantly more severe left ventricular remodeling and decreased myocardial contractile function. In addition, the apoptosis index and oxidative markers were significantly elevated in the CIH group compared with those in the control group. The autophagy marker Beclin-1 was decreased, while p62 was elevated by CIH treatment. H2S supplementation with NaHS significantly improved cardiac function and alleviated fibrosis, oxidative stress, and apoptosis but upregulated autophagy in CIH mice, and these effects were also observed in animals that underwent only PI3K blockade. Furthermore, PI3K/AKT pathway-mediated inhibition of the mammalian target of rapamycin (mTOR) pathway, the Nrf2/HO-1 pathway and proinflammatory NF-κB activity were shown to play a role in the therapeutic effect of NaHS after CIH stimulation.
Collapse
Affiliation(s)
- Xiao-Bin Zheng
- Department of Cardiology, Shanxi Cardiovascular Hospital, Taiyuan City, 030000, China.
| | - Chao Wang
- Department of Cardiology, Shanxi Cardiovascular Hospital, Taiyuan City, 030000, China
| | - Ming Zhang
- Department of Cardiology, Shanxi Cardiovascular Hospital, Taiyuan City, 030000, China
| | - Bing-Qi Yao
- Department of Cardiology, Shanxi Cardiovascular Hospital, Taiyuan City, 030000, China
| | - Hai-Yan Wu
- Department of Cardiology, Shanxi Cardiovascular Hospital, Taiyuan City, 030000, China
| | - Shu-Xian Hou
- Department of Cardiology, Shanxi Cardiovascular Hospital, Taiyuan City, 030000, China
| |
Collapse
|
4
|
Yu J, Mu X, Guan C, Wang Y, Li H. Tyrobp deficiency blocks NLRP3-mediated inflammation and pyroptosis to alleviate myocardial ischemia-reperfusion injury through regulating Syk. Tissue Cell 2024; 91:102555. [PMID: 39276487 DOI: 10.1016/j.tice.2024.102555] [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: 06/14/2024] [Revised: 08/23/2024] [Accepted: 09/05/2024] [Indexed: 09/17/2024]
Abstract
PURPOSE The present study aims to investigate the biological function of Tyrobp in myocardial ischemia-reperfusion injury (MIRI) and to clarify its potential reaction mechanisms. METHODS AC16 cells were induced by oxygen-glucose deprivation/reoxygenation (OGD/R) to simulate the MIRI in vitro. The cell transfection technology was used to downregulate Tyrobp, followed by assessment of cell damage, apoptosis and cytokines production via Cell Counting Kit (CCK)-8 assay, lactate dehydrogenase (LDH) release assay, TUNEL and ELISA assays, respectively. Immunofluorescence assay was performed to assess GSDMD. Corresponding proteins were detected via western blotting, and Co-immunoprecipitation (Co-IP) assay was used to validate proteins interaction. RESULTS Tyrobp was upregulated in OGD/R-exposed AC16 cells, and Tyrobp deficiency significantly alleviated OGD/R-caused cell viability loss, LDH release and cell apoptosis in AC16 cells. Meanwhile, Tyrobp deficiency inhibited the activation of NLRP3 inflammasome, reduced the production of cytokines and inhibited GSDMD intensity and GSDMD-N expression. Additionally, Tyrobp could interact with Syk and regulate Syk/NF-κB signaling. The rescue experiments showed that the above effects of Tyrobp deficiency on OGD/R-exposed AC16 cells were partly weakened by Syk overexpression. CONCLUSION Tyrobp deficiency alleviated MIRI by inhibiting NLRP3-mediated inflammation and pyroptosis through regulating Syk, providing a novel target for the treatment of MIRI.
Collapse
Affiliation(s)
- Jie Yu
- Cardiovascular Care Unit, The First Hospital of Hebei Medical University, Shijiazhuang 050000, China
| | - Xiu'e Mu
- Department of Anesthesiology, The First Hospital of Hebei Medical University, Shijiazhuang 050000, China
| | - Chang Guan
- Cardiovascular Care Unit, The First Hospital of Hebei Medical University, Shijiazhuang 050000, China
| | - Yaqin Wang
- Cardiovascular Care Unit, The First Hospital of Hebei Medical University, Shijiazhuang 050000, China
| | - Hongying Li
- Department of Cardiovascular Surgery, The First Hospital of Hebei Medical University, Shijiazhuang 050000, China.
| |
Collapse
|
5
|
Zhang Q, Zhang Y, Guo S, Wang X, Wang H. Hydrogen sulfide plays an important role by regulating microRNA in different ischemia-reperfusion injury. Biochem Pharmacol 2024; 229:116503. [PMID: 39179120 DOI: 10.1016/j.bcp.2024.116503] [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: 04/25/2024] [Revised: 07/28/2024] [Accepted: 08/20/2024] [Indexed: 08/26/2024]
Abstract
MicroRNAs (miRNAs) are the short endogenous non-coding RNAs that regulate the expression of the target gene at posttranscriptional level through degrading or inhibiting the specific target messenger RNAs (mRNAs). MiRNAs regulate the expression of approximately one-third of protein coding genes, and in most cases inhibit gene expression. MiRNAs have been reported to regulate various biological processes, such as cell proliferation, apoptosis and differentiation. Therefore, miRNAs participate in multiple diseases, including ischemia-reperfusion (I/R) injury. Hydrogen sulfide (H2S) was once considered as a colorless, toxic and harmful gas with foul smelling. However, in recent years, it has been discovered that it is the third gas signaling molecule after carbon monoxide (CO) and nitric oxide (NO), with multiple important biological functions. Increasing evidence indicates that H2S plays a vital role in I/R injury through regulating miRNA, however, the mechanism has not been fully understood. In this review, we summarized the current knowledge about the role of H2S in I/R injury by regulating miRNAs, and analyzed its mechanism in detail.
Collapse
Affiliation(s)
- Qi Zhang
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, Henan 475004, China
| | - Yanting Zhang
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, Henan 475004, China
| | - Shiyun Guo
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, Henan 475004, China
| | - Xiao Wang
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, Henan 475004, China
| | - Honggang Wang
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, Henan 475004, China.
| |
Collapse
|
6
|
Cao L, Wang XL, Chu T, Wang YW, Fan YQ, Chen YH, Zhu YW, Zhang J, Ji XY, Wu DD. Role of gasotransmitters in necroptosis. Exp Cell Res 2024; 442:114233. [PMID: 39216662 DOI: 10.1016/j.yexcr.2024.114233] [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: 07/04/2024] [Revised: 08/28/2024] [Accepted: 08/29/2024] [Indexed: 09/04/2024]
Abstract
Gasotransmitters are endogenous gaseous signaling molecules that can freely pass through cell membranes and transmit signals between cells, playing multiple roles in cell signal transduction. Due to extensive and ongoing research in this field, we have successfully identified many gasotransmitters so far, among which nitric oxide, carbon monoxide, and hydrogen sulfide are best studied. Gasotransmitters are implicated in various diseases related to necroptosis, such as cardiovascular diseases, inflammation, ischemia-reperfusion, infectious diseases, and neurological diseases. However, the mechanisms of their effects on necroptosis are not fully understood. This review focuses on endogenous gasotransmitter synthesis and metabolism and discusses their roles in necroptosis, aiming to offer new insights for the therapeutic approaches to necroptosis-associated diseases.
Collapse
Affiliation(s)
- Lei Cao
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, School of Stomatology, Henan University, Kaifeng, Henan, 475004, China
| | - Xue-Li Wang
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, School of Stomatology, Henan University, Kaifeng, Henan, 475004, China
| | - Ti Chu
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, School of Stomatology, Henan University, Kaifeng, Henan, 475004, China
| | - Yan-Wen Wang
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, School of Stomatology, Henan University, Kaifeng, Henan, 475004, China
| | - Yong-Qi Fan
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, School of Stomatology, Henan University, Kaifeng, Henan, 475004, China
| | - Yu-Hang Chen
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, School of Stomatology, Henan University, Kaifeng, Henan, 475004, China
| | - Yi-Wen Zhu
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, School of Stomatology, Henan University, Kaifeng, Henan, 475004, China
| | - Jing Zhang
- Department of Stomatology, The First Affiliated Hospital of Henan University, Kaifeng, Henan, 475001, China.
| | - Xin-Ying Ji
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, School of Stomatology, Henan University, Kaifeng, Henan, 475004, China; Faculty of Basic Medical Subjects, Shu-Qing Medical College of Zhengzhou, Zhengzhou, Henan, 450064, China.
| | - Dong-Dong Wu
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, School of Stomatology, Henan University, Kaifeng, Henan, 475004, China; Department of Stomatology, Huaihe Hospital of Henan University, School of Stomatology, Kaifeng, Henan, 475000, China; Kaifeng Key Laboratory of Periodontal Tissue Engineering, School of Stomatology, Henan University, Kaifeng, Henan, 475004, China.
| |
Collapse
|
7
|
Liu L, Yao Y, Liu Y, Hong B, Li Z, Chen X, Zhang Y, Fu H, Yang D, Yang C. Targeted H 2S-Mediated Gas Therapy with pH-Sensitive Release Property for Myocardial Ischemia-Reperfusion Injury by Platelet Membrane. Biomater Res 2024; 28:0061. [PMID: 39161346 PMCID: PMC11330987 DOI: 10.34133/bmr.0061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 07/02/2024] [Indexed: 08/21/2024] Open
Abstract
Management of myocardial ischemia-reperfusion injury (MIRI) in reperfusion therapy remains a major obstacle in the field of cardiovascular disease, but current available therapies have not yet been achieved in mitigating myocardial injury due to the complex pathological mechanisms of MIRI. Exogenous delivery of hydrogen sulfide (H2S) to the injured myocardium can be an effective strategy for treating MIRI due to the multiple physiologic functions of H2S, including anti-inflammatory, anti-apoptotic, and mitochondrial protective effects. Here, to realize the precise delivery and release of H2S, we proposed the targeted H2S-mediated gas therapy with pH-sensitive release property mediated by platelet membranes (PMs). In this study, a biomimetic functional poly(lactic-co-ethanolic acid) nanoparticle (RAPA/JK-1-PLGA@PM) was fabricated by loading rapamycin (RAPA; mTOR inhibitor) and JK-1 (H2S donor) and then coated with PM. In vitro observations were conducted including pharmaceutical evaluation, H2S release behaviors, hemolysis analysis, serum stability, cellular uptake, cytotoxicity, inhibition of myocardial apoptosis, and anti-inflammation. In vivo examinations were performed including targeting ability, restoration of cardiac function, inhibition of pathological remodeling, and anti-inflammation. RAPA/JK-1-PLGA@PM was successfully prepared with good size distribution and stability. Utilizing the natural infarct-homing ability of PM, RAPA/JK-1-PLGA@PM could be effectively targeted to the damaged myocardium. RAPA/JK-1-PLGA@PM continuously released H2S triggered by inflammatory microenvironment, which could inhibit cardiomyocyte apoptosis, realize the transition of pro-inflammation, and alleviate myocardial injury demonstrated in hypoxia/reoxygenation myocardial cell in vitro. Precise delivery and release of H2S attenuated inflammatory response and cardiac damage, promoted cardiac repair, and ameliorated cardiac function proven in MIRI mouse model in vivo. This research outlined the novel nanoplatform that combined immunosuppressant agents and H2S donor with the pH-sensitive release property, offering a promising therapeutic for MIRI treatment that leveraged the synergistic effects of gas therapy.
Collapse
Affiliation(s)
- Lin Liu
- Department of Pharmacy,
The Second Affiliated Hospital of Shantou University Medical College, Shantou 515041, China
- Department of Pharmacy,
Shantou University Medical College, Shantou 515041, China
| | - Yucen Yao
- Department of Pharmacy,
Shantou University Medical College, Shantou 515041, China
- College of Pharmacy, Jiamusi University, Jiamusi 154007, China
| | - Yang Liu
- Department of Pharmacy,
The Second Affiliated Hospital of Shantou University Medical College, Shantou 515041, China
- Department of Pharmacy,
Shantou University Medical College, Shantou 515041, China
| | - Bingrong Hong
- Department of Pharmacy,
The Second Affiliated Hospital of Shantou University Medical College, Shantou 515041, China
- Department of Pharmacy,
Shantou University Medical College, Shantou 515041, China
| | - Ziqing Li
- Department of Pharmacy,
The Second Affiliated Hospital of Shantou University Medical College, Shantou 515041, China
- Department of Pharmacy,
Shantou University Medical College, Shantou 515041, China
| | - Xuejun Chen
- Department of Pharmacy,
The Second Affiliated Hospital of Shantou University Medical College, Shantou 515041, China
- Department of Pharmacy,
Shantou University Medical College, Shantou 515041, China
| | - Yaofeng Zhang
- Department of Pharmacy,
The Second Affiliated Hospital of Shantou University Medical College, Shantou 515041, China
| | - Hongbo Fu
- Department of Pharmacy,
The Second Affiliated Hospital of Shantou University Medical College, Shantou 515041, China
| | - Degong Yang
- Department of Pharmacy,
Shantou University Medical College, Shantou 515041, China
- Department of Pharmacy, Department of Dermatology,
The First Affiliated Hospital of Shantou University Medical College, Shantou 515041, China
| | - Chunrong Yang
- Department of Pharmacy,
The Second Affiliated Hospital of Shantou University Medical College, Shantou 515041, China
- Department of Pharmacy,
Shantou University Medical College, Shantou 515041, China
| |
Collapse
|
8
|
Sun HJ, Lu QB, Zhu XX, Ni ZR, Su JB, Fu X, Chen G, Zheng GL, Nie XW, Bian JS. Pharmacology of Hydrogen Sulfide and Its Donors in Cardiometabolic Diseases. Pharmacol Rev 2024; 76:846-895. [PMID: 38866561 DOI: 10.1124/pharmrev.123.000928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 04/13/2024] [Accepted: 06/10/2024] [Indexed: 06/14/2024] Open
Abstract
Cardiometabolic diseases (CMDs) are major contributors to global mortality, emphasizing the critical need for novel therapeutic interventions. Hydrogen sulfide (H2S) has garnered enormous attention as a significant gasotransmitter with various physiological, pathophysiological, and pharmacological impacts within mammalian cardiometabolic systems. In addition to its roles in attenuating oxidative stress and inflammatory response, burgeoning research emphasizes the significance of H2S in regulating proteins via persulfidation, a well known modification intricately associated with the pathogenesis of CMDs. This review seeks to investigate recent updates on the physiological actions of endogenous H2S and the pharmacological roles of various H2S donors in addressing diverse aspects of CMDs across cellular, animal, and clinical studies. Of note, advanced methodologies, including multiomics, intestinal microflora analysis, organoid, and single-cell sequencing techniques, are gaining traction due to their ability to offer comprehensive insights into biomedical research. These emerging approaches hold promise in characterizing the pharmacological roles of H2S in health and diseases. We will critically assess the current literature to clarify the roles of H2S in diseases while also delineating the opportunities and challenges they present in H2S-based pharmacotherapy for CMDs. SIGNIFICANCE STATEMENT: This comprehensive review covers recent developments in H2S biology and pharmacology in cardiometabolic diseases CMDs. Endogenous H2S and its donors show great promise for the management of CMDs by regulating numerous proteins and signaling pathways. The emergence of new technologies will considerably advance the pharmacological research and clinical translation of H2S.
Collapse
Affiliation(s)
- Hai-Jian Sun
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China (H.-J.S., X.-X.Z., Z.-R.N., J.-B.S., X.F., G.C., G.-L.Z.); Department of Endocrinology, Affiliated Hospital of Jiangnan University, Jiangnan University, Wuxi, Jiangsu, China (Q.-B.L.); Shenzhen Key Laboratory of Respiratory Diseases, Shenzhen People's Hospital, Shenzhen, Guangdong, China (X.-W.N.); and Department of Pharmacology, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, China (J.-S.B.)
| | - Qing-Bo Lu
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China (H.-J.S., X.-X.Z., Z.-R.N., J.-B.S., X.F., G.C., G.-L.Z.); Department of Endocrinology, Affiliated Hospital of Jiangnan University, Jiangnan University, Wuxi, Jiangsu, China (Q.-B.L.); Shenzhen Key Laboratory of Respiratory Diseases, Shenzhen People's Hospital, Shenzhen, Guangdong, China (X.-W.N.); and Department of Pharmacology, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, China (J.-S.B.)
| | - Xue-Xue Zhu
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China (H.-J.S., X.-X.Z., Z.-R.N., J.-B.S., X.F., G.C., G.-L.Z.); Department of Endocrinology, Affiliated Hospital of Jiangnan University, Jiangnan University, Wuxi, Jiangsu, China (Q.-B.L.); Shenzhen Key Laboratory of Respiratory Diseases, Shenzhen People's Hospital, Shenzhen, Guangdong, China (X.-W.N.); and Department of Pharmacology, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, China (J.-S.B.)
| | - Zhang-Rong Ni
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China (H.-J.S., X.-X.Z., Z.-R.N., J.-B.S., X.F., G.C., G.-L.Z.); Department of Endocrinology, Affiliated Hospital of Jiangnan University, Jiangnan University, Wuxi, Jiangsu, China (Q.-B.L.); Shenzhen Key Laboratory of Respiratory Diseases, Shenzhen People's Hospital, Shenzhen, Guangdong, China (X.-W.N.); and Department of Pharmacology, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, China (J.-S.B.)
| | - Jia-Bao Su
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China (H.-J.S., X.-X.Z., Z.-R.N., J.-B.S., X.F., G.C., G.-L.Z.); Department of Endocrinology, Affiliated Hospital of Jiangnan University, Jiangnan University, Wuxi, Jiangsu, China (Q.-B.L.); Shenzhen Key Laboratory of Respiratory Diseases, Shenzhen People's Hospital, Shenzhen, Guangdong, China (X.-W.N.); and Department of Pharmacology, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, China (J.-S.B.)
| | - Xiao Fu
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China (H.-J.S., X.-X.Z., Z.-R.N., J.-B.S., X.F., G.C., G.-L.Z.); Department of Endocrinology, Affiliated Hospital of Jiangnan University, Jiangnan University, Wuxi, Jiangsu, China (Q.-B.L.); Shenzhen Key Laboratory of Respiratory Diseases, Shenzhen People's Hospital, Shenzhen, Guangdong, China (X.-W.N.); and Department of Pharmacology, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, China (J.-S.B.)
| | - Guo Chen
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China (H.-J.S., X.-X.Z., Z.-R.N., J.-B.S., X.F., G.C., G.-L.Z.); Department of Endocrinology, Affiliated Hospital of Jiangnan University, Jiangnan University, Wuxi, Jiangsu, China (Q.-B.L.); Shenzhen Key Laboratory of Respiratory Diseases, Shenzhen People's Hospital, Shenzhen, Guangdong, China (X.-W.N.); and Department of Pharmacology, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, China (J.-S.B.)
| | - Guan-Li Zheng
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China (H.-J.S., X.-X.Z., Z.-R.N., J.-B.S., X.F., G.C., G.-L.Z.); Department of Endocrinology, Affiliated Hospital of Jiangnan University, Jiangnan University, Wuxi, Jiangsu, China (Q.-B.L.); Shenzhen Key Laboratory of Respiratory Diseases, Shenzhen People's Hospital, Shenzhen, Guangdong, China (X.-W.N.); and Department of Pharmacology, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, China (J.-S.B.)
| | - Xiao-Wei Nie
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China (H.-J.S., X.-X.Z., Z.-R.N., J.-B.S., X.F., G.C., G.-L.Z.); Department of Endocrinology, Affiliated Hospital of Jiangnan University, Jiangnan University, Wuxi, Jiangsu, China (Q.-B.L.); Shenzhen Key Laboratory of Respiratory Diseases, Shenzhen People's Hospital, Shenzhen, Guangdong, China (X.-W.N.); and Department of Pharmacology, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, China (J.-S.B.)
| | - Jin-Song Bian
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China (H.-J.S., X.-X.Z., Z.-R.N., J.-B.S., X.F., G.C., G.-L.Z.); Department of Endocrinology, Affiliated Hospital of Jiangnan University, Jiangnan University, Wuxi, Jiangsu, China (Q.-B.L.); Shenzhen Key Laboratory of Respiratory Diseases, Shenzhen People's Hospital, Shenzhen, Guangdong, China (X.-W.N.); and Department of Pharmacology, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, China (J.-S.B.)
| |
Collapse
|
9
|
Sun X, Wu S, Mao C, Qu Y, Xu Z, Xie Y, Jiang D, Song Y. Therapeutic Potential of Hydrogen Sulfide in Ischemia and Reperfusion Injury. Biomolecules 2024; 14:740. [PMID: 39062455 PMCID: PMC11274451 DOI: 10.3390/biom14070740] [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: 05/22/2024] [Revised: 06/14/2024] [Accepted: 06/19/2024] [Indexed: 07/28/2024] Open
Abstract
Ischemia-reperfusion (I/R) injury, a prevalent pathological condition in medical practice, presents significant treatment challenges. Hydrogen sulfide (H2S), acknowledged as the third gas signaling molecule, profoundly impacts various physiological and pathophysiological processes. Extensive research has demonstrated that H2S can mitigate I/R damage across multiple organs and tissues. This review investigates the protective effects of H2S in preventing I/R damage in the heart, brain, liver, kidney, intestines, lungs, stomach, spinal cord, testes, eyes, and other tissues. H2S provides protection against I/R damage by alleviating inflammation and endoplasmic reticulum stress; inhibiting apoptosis, oxidative stress, and mitochondrial autophagy and dysfunction; and regulating microRNAs. Significant advancements in understanding the mechanisms by which H2S reduces I/R damage have led to the development and synthesis of H2S-releasing agents such as diallyl trisulfide-loaded mesoporous silica nanoparticles (DATS-MSN), AP39, zofenopril, and ATB-344, offering a new therapeutic avenue for I/R injury.
Collapse
Affiliation(s)
- Xutao Sun
- Department of Typhoid, School of Basic Medical Sciences, Heilongjiang University of Chinese Medicine, Harbin 150040, China;
| | - Siyu Wu
- Department of Pharmacology, School of Basic Medical Sciences, Heilongjiang University of Chinese Medicine, Harbin 150040, China; (S.W.); (C.M.); (Y.Q.); (Z.X.)
| | - Caiyun Mao
- Department of Pharmacology, School of Basic Medical Sciences, Heilongjiang University of Chinese Medicine, Harbin 150040, China; (S.W.); (C.M.); (Y.Q.); (Z.X.)
| | - Ying Qu
- Department of Pharmacology, School of Basic Medical Sciences, Heilongjiang University of Chinese Medicine, Harbin 150040, China; (S.W.); (C.M.); (Y.Q.); (Z.X.)
| | - Zihang Xu
- Department of Pharmacology, School of Basic Medical Sciences, Heilongjiang University of Chinese Medicine, Harbin 150040, China; (S.W.); (C.M.); (Y.Q.); (Z.X.)
| | - Ying Xie
- Department of Synopsis of the Golden Chamber, School of Basic Medical Sciences, Heilongjiang University of Chinese Medicine, Harbin 150040, China;
| | - Deyou Jiang
- Department of Synopsis of the Golden Chamber, School of Basic Medical Sciences, Heilongjiang University of Chinese Medicine, Harbin 150040, China;
| | - Yunjia Song
- Department of Pharmacology, School of Basic Medical Sciences, Heilongjiang University of Chinese Medicine, Harbin 150040, China; (S.W.); (C.M.); (Y.Q.); (Z.X.)
| |
Collapse
|
10
|
Tan Q, Dong W, Wang Q, Gao L. Dexmedetomidine alleviates Hypoxia/reoxygenation-induced mitochondrial dysfunction in cardiomyocytes via activation of Sirt3/Prdx3 pathway. Daru 2024; 32:189-196. [PMID: 38407745 PMCID: PMC11087443 DOI: 10.1007/s40199-024-00504-3] [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: 07/25/2023] [Accepted: 01/18/2024] [Indexed: 02/27/2024] Open
Abstract
BACKGROUND Myocardial ischemia/reperfusion injury (MIRI) seriously threatens the health of people. The mitochondrial dysfunction in cardiomyocytes can promote the progression of MIRI. Dexmedetomidine (Dex) could alleviate the myocardial injury, which was known to reverse mitochondrial dysfunction in lung injury. However, the function of Dex in mitochondrial dysfunction during MIRI remains unclear. OBJECTIVE To assess the function of Dex in mitochondrial dysfunction during MIRI. METHODS To investigate the function of Dex in MIRI, H9C2 cells were placed in condition of hypoxia/reoxygenation (H/R). CCK8 assay was performed to test the cell viability, and the mitochondrial membrane potential was evaluated by JC-1 staining. In addition, the binding relationship between Sirt3 and Prdx3 was explored by Co-IP assay. Furthermore, the protein expressions were examined using western blot. RESULTS Dex could abolish H/R-induced mitochondrial dysfunction in H9C2 cells. In addition, H/R treatment significantly inhibited the expression of Sirt3, while Dex partially restored this phenomenon. Knockdown of Sirt3 or Prdx3 obviously reduced the protective effect of Dex on H/R-induced mitochondrial injury. Meanwhile, Sirt3 could enhance the function of Prdx3 via deacetylation of Prdx3. CONCLUSION Dex was found to attenuate H/R-induced mitochondrial dysfunction in cardiomyocytes via activation of Sirt3/Prdx3 pathway. Thus, this study might shed new lights on exploring new strategies for the treatment of MIRI.
Collapse
Affiliation(s)
- Qingyun Tan
- Department of Anesthesiology, The First Affiliated Hospital of Jiamusi University, No.348, Dexiang Street, Xiangyang District, Jiamusi, 154002, Heilongjiang Province, People's Republic of China
| | - Wenming Dong
- Department of Anesthesiology, Shenzhen Baoan Hospital of TCM, Shenzhen, 518133, Guangdong Province, People's Republic of China
| | - Qingdong Wang
- Department of Anesthesiology, The First Affiliated Hospital of Jiamusi University, No.348, Dexiang Street, Xiangyang District, Jiamusi, 154002, Heilongjiang Province, People's Republic of China.
| | - Li Gao
- Department of Anesthesiology, The First Affiliated Hospital of Jiamusi University, No.348, Dexiang Street, Xiangyang District, Jiamusi, 154002, Heilongjiang Province, People's Republic of China.
| |
Collapse
|
11
|
Tánczos B, Vass V, Szabó E, Lovas M, Kattoub RG, Bereczki I, Borbás A, Herczegh P, Tósaki Á. Effects of H 2S-donor ascorbic acid derivative and ischemia/reperfusion-induced injury in isolated rat hearts. Eur J Pharm Sci 2024; 195:106721. [PMID: 38331005 DOI: 10.1016/j.ejps.2024.106721] [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: 11/27/2023] [Revised: 01/30/2024] [Accepted: 02/04/2024] [Indexed: 02/10/2024]
Abstract
Hydrogen sulfide (H2S), a gasotransmitter, plays a crucial role in vasorelaxation, anti-inflammatory processes and mitigating myocardial ischemia/reperfusion-induced injury by regulating various signaling processes. We designed a water soluble H2S-releasing ascorbic acid derivative, BM-164, to combine the beneficial cardiovascular and anti-inflammatory effects of H2S with the excellent water solubility and antioxidant properties of ascorbic acid. DPPH antioxidant assay revealed that the antioxidant activity of BM-164 in the presence of a myocardial tissue homogenate (extract) increased continuously over the 120 min test interval due to the continuous release of H2S from BM-164. The cytotoxicity of BM-164 was tested by MTT assay on H9c2 cells, which resulted in no cytotoxic effect at concentrations of 10 to 30 μM. The possible beneficial effects of BM-164 (30 µM) was examined in isolated 'Langendorff' rat hearts. The incidence of ventricular fibrillation (VF) was significantly reduced from its control value of 79 % to 31 % in the BM-164 treated group, and the infarct size was also diminished from the control value of 28 % to 14 % in the BM-164 treated group. However, coronary flow (CF) and heart rate (HR) values in the BM-164 treated group did not show significantly different levels in comparison with the drug-free control, although a non-significant recovery in both CF and HR was observed at each time point. We attempted to reveal the mechanism of action of BM-164, focusing on the processes of autophagy and apoptosis. The expression of key autophagic and apoptotic markers in isolated rat hearts were detected by Western blot analysis. All the examined autophagy-related proteins showed increased expression levels in the BM-164 treated group in comparison to the drug-free control and/or ascorbic acid treated groups, while the changes in the expression of apoptotic markers were not obvious. In conclusion, the designed water soluble H2S releasing ascorbic acid derivative, BM-164, showed better cardiac protection against ischemia/reperfusion-induced injury compared to the untreated and ascorbic acid treated hearts, respectively.
Collapse
Affiliation(s)
- Bence Tánczos
- Department of Pharmacology, Faculty of Pharmacy, University of Debrecen, Debrecen, Hungary; HUN-REN-DE Pharmamodul Research Group, University of Debrecen, 4032 Debrecen, Nagyerdei krt. 98, Hungary
| | - Virág Vass
- Department of Pharmacology, Faculty of Pharmacy, University of Debrecen, Debrecen, Hungary; HUN-REN-DE Pharmamodul Research Group, University of Debrecen, 4032 Debrecen, Nagyerdei krt. 98, Hungary; Doctoral School of Pharmaceutical Sciences, Faculty of Pharmacy, University of Debrecen, 4032 Debrecen, Nagyerdei krt. 98., Hungary
| | - Erzsébet Szabó
- Department of Pharmacology, Faculty of Pharmacy, University of Debrecen, Debrecen, Hungary; HUN-REN-DE Pharmamodul Research Group, University of Debrecen, 4032 Debrecen, Nagyerdei krt. 98, Hungary
| | - Miklós Lovas
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Debrecen, Debrecen, Hungary
| | - Rasha Ghanem Kattoub
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Debrecen, Debrecen, Hungary; Doctoral School of Pharmaceutical Sciences, Faculty of Pharmacy, University of Debrecen, 4032 Debrecen, Nagyerdei krt. 98., Hungary
| | - Ilona Bereczki
- HUN-REN-DE Pharmamodul Research Group, University of Debrecen, 4032 Debrecen, Nagyerdei krt. 98, Hungary; Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Debrecen, Debrecen, Hungary
| | - Anikó Borbás
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Debrecen, Debrecen, Hungary
| | - Pál Herczegh
- HUN-REN-DE Pharmamodul Research Group, University of Debrecen, 4032 Debrecen, Nagyerdei krt. 98, Hungary; Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Debrecen, Debrecen, Hungary
| | - Árpád Tósaki
- Department of Pharmacology, Faculty of Pharmacy, University of Debrecen, Debrecen, Hungary; HUN-REN-DE Pharmamodul Research Group, University of Debrecen, 4032 Debrecen, Nagyerdei krt. 98, Hungary.
| |
Collapse
|
12
|
Linjacki S, Wang Y, Baath N, Mantle D, Yang G. H 2S Protects from Rotenone-Induced Ferroptosis by Stabilizing Fe-S Clusters in Rat Cardiac Cells. Cells 2024; 13:371. [PMID: 38474335 PMCID: PMC10931451 DOI: 10.3390/cells13050371] [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: 01/05/2024] [Revised: 02/16/2024] [Accepted: 02/19/2024] [Indexed: 03/14/2024] Open
Abstract
Hydrogen sulfide (H2S) has been recently recognized as an important gasotransmitter with cardioprotections, and iron is vital for various cellular activities. This study explored the regulatory role of H2S on iron metabolism and mitochondrial functions in cultured rat cardiac cells. Rotenone, a mitochondrial complex I inhibitor, was used for establishing an in vitro model of ischemic cell damage. It was first found that rotenone induced oxidative stress and lipid peroxidation and decreased mitochondrial membrane potential and ATP generation, eventually causing cell death. The supplement of H2S at a physiologically relevant concentration protected from rotenone-induced ferroptotic cell death by reducing oxidative stress and mitochondrial damage, maintaining GPx4 expression and intracellular iron level. Deferiprone, an iron chelator, would also protect from rotenone-induced ferroptosis. Further studies demonstrated that H2S inhibited ABCB8-mediated iron efflux from mitochondria to cytosol and promoted NFS1-mediated Fe-S cluster biogenesis. It is also found that rotenone stimulated iron-dependent H2S generation. These results indicate that H2S would protect cardiac cells from ischemic damage through preserving mitochondrial functions and intracellular Fe-S cluster homeostasis.
Collapse
Affiliation(s)
- Sara Linjacki
- School of Natural Sciences, Laurentian University, Sudbury, ON P3E 2C6, Canada; (S.L.); (Y.W.); (N.B.); (D.M.)
- Cardiovascular and Metabolic Research Unit, Laurentian University, Sudbury, ON P3E 2C6, Canada
| | - Yuehong Wang
- School of Natural Sciences, Laurentian University, Sudbury, ON P3E 2C6, Canada; (S.L.); (Y.W.); (N.B.); (D.M.)
- Cardiovascular and Metabolic Research Unit, Laurentian University, Sudbury, ON P3E 2C6, Canada
| | - Navjeet Baath
- School of Natural Sciences, Laurentian University, Sudbury, ON P3E 2C6, Canada; (S.L.); (Y.W.); (N.B.); (D.M.)
- Cardiovascular and Metabolic Research Unit, Laurentian University, Sudbury, ON P3E 2C6, Canada
| | - Devin Mantle
- School of Natural Sciences, Laurentian University, Sudbury, ON P3E 2C6, Canada; (S.L.); (Y.W.); (N.B.); (D.M.)
- Cardiovascular and Metabolic Research Unit, Laurentian University, Sudbury, ON P3E 2C6, Canada
| | - Guangdong Yang
- School of Natural Sciences, Laurentian University, Sudbury, ON P3E 2C6, Canada; (S.L.); (Y.W.); (N.B.); (D.M.)
- Cardiovascular and Metabolic Research Unit, Laurentian University, Sudbury, ON P3E 2C6, Canada
| |
Collapse
|
13
|
Wang R, Bai J. Pharmacological interventions targeting the microcirculation following traumatic spinal cord injury. Neural Regen Res 2024; 19:35-42. [PMID: 37488841 PMCID: PMC10479866 DOI: 10.4103/1673-5374.375304] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Revised: 03/08/2023] [Accepted: 04/07/2023] [Indexed: 07/26/2023] Open
Abstract
Traumatic spinal cord injury is a devastating disorder characterized by sensory, motor, and autonomic dysfunction that severely compromises an individual's ability to perform activities of daily living. These adverse outcomes are closely related to the complex mechanism of spinal cord injury, the limited regenerative capacity of central neurons, and the inhibitory environment formed by traumatic injury. Disruption to the microcirculation is an important pathophysiological mechanism of spinal cord injury. A number of therapeutic agents have been shown to improve the injury environment, mitigate secondary damage, and/or promote regeneration and repair. Among them, the spinal cord microcirculation has become an important target for the treatment of spinal cord injury. Drug interventions targeting the microcirculation can improve the microenvironment and promote recovery following spinal cord injury. These drugs target the structure and function of the spinal cord microcirculation and are essential for maintaining the normal function of spinal neurons, axons, and glial cells. This review discusses the pathophysiological role of spinal cord microcirculation in spinal cord injury, including its structure and histopathological changes. Further, it summarizes the progress of drug therapies targeting the spinal cord microcirculation after spinal cord injury.
Collapse
Affiliation(s)
- Rongrong Wang
- Department of Spine and Spinal Cord Surgery, Beijing Bo’ai Hospital, China Rehabilitation Research Center, Beijing, China
- School of Rehabilitation Medicine, Capital Medical University, Beijing, China
| | - Jinzhu Bai
- Department of Spine and Spinal Cord Surgery, Beijing Bo’ai Hospital, China Rehabilitation Research Center, Beijing, China
- School of Rehabilitation Medicine, Capital Medical University, Beijing, China
| |
Collapse
|
14
|
Heusch G, Andreadou I, Bell R, Bertero E, Botker HE, Davidson SM, Downey J, Eaton P, Ferdinandy P, Gersh BJ, Giacca M, Hausenloy DJ, Ibanez B, Krieg T, Maack C, Schulz R, Sellke F, Shah AM, Thiele H, Yellon DM, Di Lisa F. Health position paper and redox perspectives on reactive oxygen species as signals and targets of cardioprotection. Redox Biol 2023; 67:102894. [PMID: 37839355 PMCID: PMC10590874 DOI: 10.1016/j.redox.2023.102894] [Citation(s) in RCA: 46] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 09/04/2023] [Accepted: 09/15/2023] [Indexed: 10/17/2023] Open
Abstract
The present review summarizes the beneficial and detrimental roles of reactive oxygen species in myocardial ischemia/reperfusion injury and cardioprotection. In the first part, the continued need for cardioprotection beyond that by rapid reperfusion of acute myocardial infarction is emphasized. Then, pathomechanisms of myocardial ischemia/reperfusion to the myocardium and the coronary circulation and the different modes of cell death in myocardial infarction are characterized. Different mechanical and pharmacological interventions to protect the ischemic/reperfused myocardium in elective percutaneous coronary interventions and coronary artery bypass grafting, in acute myocardial infarction and in cardiotoxicity from cancer therapy are detailed. The second part keeps the focus on ROS providing a comprehensive overview of molecular and cellular mechanisms involved in ischemia/reperfusion injury. Starting from mitochondria as the main sources and targets of ROS in ischemic/reperfused myocardium, a complex network of cellular and extracellular processes is discussed, including relationships with Ca2+ homeostasis, thiol group redox balance, hydrogen sulfide modulation, cross-talk with NAPDH oxidases, exosomes, cytokines and growth factors. While mechanistic insights are needed to improve our current therapeutic approaches, advancements in knowledge of ROS-mediated processes indicate that detrimental facets of oxidative stress are opposed by ROS requirement for physiological and protective reactions. This inevitable contrast is likely to underlie unsuccessful clinical trials and limits the development of novel cardioprotective interventions simply based upon ROS removal.
Collapse
Affiliation(s)
- Gerd Heusch
- Institute for Pathophysiology, West German Heart and Vascular Center, University of Duisburg-Essen, Essen, Germany.
| | - Ioanna Andreadou
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| | - Robert Bell
- The Hatter Cardiovascular Institute, University College London, London, United Kingdom
| | - Edoardo Bertero
- Chair of Cardiovascular Disease, Department of Internal Medicine and Specialties, University of Genova, Genova, Italy
| | - Hans-Erik Botker
- Department of Cardiology, Institute for Clinical Medicine, Aarhus University, Aarhus N, Denmark
| | - Sean M Davidson
- The Hatter Cardiovascular Institute, University College London, London, United Kingdom
| | - James Downey
- Department of Physiology, University of South Alabama, Mobile, AL, USA
| | - Philip Eaton
- William Harvey Research Institute, Queen Mary University of London, Heart Centre, Charterhouse Square, London, United Kingdom
| | - Peter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary; Pharmahungary Group, Szeged, Hungary
| | - Bernard J Gersh
- Department of Cardiovascular Medicine, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
| | - Mauro Giacca
- School of Cardiovascular and Metabolic Medicine & Sciences, King's College, London, United Kingdom
| | - Derek J Hausenloy
- The Hatter Cardiovascular Institute, University College London, London, United Kingdom; Cardiovascular & Metabolic Disorders Program, Duke-National University of Singapore Medical School, National Heart Research Institute Singapore, National Heart Centre, Yong Loo Lin School of Medicine, National University Singapore, Singapore
| | - Borja Ibanez
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), IIS-Fundación Jiménez Díaz University Hospital, and CIBERCV, Madrid, Spain
| | - Thomas Krieg
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Christoph Maack
- Department of Translational Research, Comprehensive Heart Failure Center, University Clinic Würzburg, Würzburg, Germany
| | - Rainer Schulz
- Institute for Physiology, Justus-Liebig -Universität, Giessen, Germany
| | - Frank Sellke
- Division of Cardiothoracic Surgery, Alpert Medical School of Brown University and Rhode Island Hospital, Providence, RI, USA
| | - Ajay M Shah
- King's College London British Heart Foundation Centre of Excellence, London, United Kingdom
| | - Holger Thiele
- Heart Center Leipzig at University of Leipzig and Leipzig Heart Science, Leipzig, Germany
| | - Derek M Yellon
- The Hatter Cardiovascular Institute, University College London, London, United Kingdom
| | - Fabio Di Lisa
- Dipartimento di Scienze Biomediche, Università degli studi di Padova, Padova, Italy.
| |
Collapse
|
15
|
Kolluru GK, Shackelford RE, Shen X, Dominic P, Kevil CG. Sulfide regulation of cardiovascular function in health and disease. Nat Rev Cardiol 2023; 20:109-125. [PMID: 35931887 PMCID: PMC9362470 DOI: 10.1038/s41569-022-00741-6] [Citation(s) in RCA: 88] [Impact Index Per Article: 44.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/21/2022] [Indexed: 01/21/2023]
Abstract
Hydrogen sulfide (H2S) has emerged as a gaseous signalling molecule with crucial implications for cardiovascular health. H2S is involved in many biological functions, including interactions with nitric oxide, activation of molecular signalling cascades, post-translational modifications and redox regulation. Various preclinical and clinical studies have shown that H2S and its synthesizing enzymes - cystathionine γ-lyase, cystathionine β-synthase and 3-mercaptosulfotransferase - can protect against cardiovascular pathologies, including arrhythmias, atherosclerosis, heart failure, myocardial infarction and ischaemia-reperfusion injury. The bioavailability of H2S and its metabolites, such as hydropersulfides and polysulfides, is substantially reduced in cardiovascular disease and has been associated with single-nucleotide polymorphisms in H2S synthesis enzymes. In this Review, we highlight the role of H2S, its synthesizing enzymes and metabolites, their roles in the cardiovascular system, and their involvement in cardiovascular disease and associated pathologies. We also discuss the latest clinical findings from the field and outline areas for future study.
Collapse
Affiliation(s)
- Gopi K Kolluru
- Department of Pathology, Louisiana State University Health Sciences Center, Shreveport, LA, USA
- Center of Excellence for Cardiovascular Diseases & Sciences, Louisiana State University Health Sciences Center, Shreveport, LA, USA
| | - Rodney E Shackelford
- Department of Pathology, Louisiana State University Health Sciences Center, Shreveport, LA, USA
| | - Xinggui Shen
- Department of Pathology, Louisiana State University Health Sciences Center, Shreveport, LA, USA
- Center of Excellence for Cardiovascular Diseases & Sciences, Louisiana State University Health Sciences Center, Shreveport, LA, USA
| | - Paari Dominic
- Center of Excellence for Cardiovascular Diseases & Sciences, Louisiana State University Health Sciences Center, Shreveport, LA, USA
- Department of Medicine, Louisiana State University Health Sciences Center, Shreveport, LA, USA
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center, Shreveport, LA, USA
| | - Christopher G Kevil
- Department of Pathology, Louisiana State University Health Sciences Center, Shreveport, LA, USA.
- Center of Excellence for Cardiovascular Diseases & Sciences, Louisiana State University Health Sciences Center, Shreveport, LA, USA.
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center, Shreveport, LA, USA.
- Cellular Biology and Anatomy, Louisiana State University Health Sciences Center, Shreveport, LA, USA.
| |
Collapse
|
16
|
Fang F, Sheng J, Guo Y, Wen J, Chen Z. Protection of H 2S against Hypoxia/Reoxygenation Injury in Rat Hippocampal Neurons through Inhibiting Phosphorylation of ROCK 2 at Thr436 and Ser575. Pharmaceuticals (Basel) 2023; 16:218. [PMID: 37259366 PMCID: PMC9966024 DOI: 10.3390/ph16020218] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 01/23/2023] [Accepted: 01/28/2023] [Indexed: 11/19/2023] Open
Abstract
BACKGROUND H2S (hydrogen sulfide) protects cerebral vasodilatation and endothelial cells against oxygen-glucose deprivation/reoxygenation injury via the inhibition of the RhoA-ROCK pathway and ROCK2 expression. However, the inhibitory mechanism of H2S on ROCK2 expression is still unclear. The study aimed to investigate the target and mechanism of H2S in inhibition of ROCK2. METHODS His-ROCK2wild protein was constructed, expressed, and was used for phosphorylation assay in vitro. Liquid chromatography-tandem mass spectrometry (LC-MS/MS) was used to determine the potential phosphorylation sites of ROCK2. Recombinant ROCK2wild-pEGFP-N1, ROCK2T436A-pEGFP-N1, and ROCK2S575F-pEGFP-N1 plasmids were constructed and transfected into rat hippocampal neurons (RHNs). ROCK2 expression, cell viability, the release of lactate dehydrogenase (LDH), nerve-specific enolase (NSE), and Ca2+ were detected to evaluate the neuroprotective mechanism of H2S. RESULTS Phosphorylation at Thr436 and Ser575 of ROCK2 was observed by mass spectrometry when Polo-like kinase 1 (PLK1) and protein kinase A (PKA) were added in vitro, and NaHS significantly inhibited phosphorylation at Thr436 and Ser575. Additionally, NaHS significantly inhibited the expression of ROCK2 and recombinant proteins GFP-ROCK2, GFP-ROCK2T436A, and GFP-ROCK2S575F in transfected RHNs. Compared with empty plasmid, GFP-ROCK2T436A, and GFP-ROCK2S575F groups, NaHS significantly inhibited the release of LDH, NSE, and Ca2+ and promoted ROCK2 activity in the GFP-ROCK2wild group. Thr436 and Ser575 may be dominant sites that mediate NaHS inhibition of ROCK2 protein activity in RHNs. Compared with the empty plasmid, GFP-ROCK2T436A, and the GFP-ROCK2S575F group, NaHS had more significant inhibitory effects on hypoxia/reoxygenation (H/R) injury-induced cell viability reduction and increased LDH and NSE release in the GFP-ROCK2wild group. CONCLUSION Exogenous H2S protected the RHNs against H/R injury via Thr436 and Ser575 of ROCK2. These findings suggested that Thr436 and Ser575 may be the dominant sites that mediated the effect of NaHS on protecting RHNs against H/R injury.
Collapse
Affiliation(s)
| | | | - Yan Guo
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Jiyue Wen
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Zhiwu Chen
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| |
Collapse
|
17
|
He P, Zhang M, Zhao M, Zhang M, Ma B, Lv H, Han Y, Wu D, Zhong Z, Zhao W. A Novel Polysaccharide From Chuanminshen violaceum and Its Protective Effect Against Myocardial Injury. Front Nutr 2022; 9:961182. [PMID: 35911096 PMCID: PMC9330552 DOI: 10.3389/fnut.2022.961182] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Accepted: 06/22/2022] [Indexed: 12/17/2022] Open
Abstract
We isolated and purified a novel polysaccharide from the root of Chuanminshen violaceum, namely, Chuanminshen violaceumis polysaccharide (CVP) and confirmed its structure and molecular weight. Furthermore, in vivo experiment, CVP’s protective effect against myocardial ischemia-reperfusion (I/R) injury in mice was evidenced by significantly reducing I/R-induced myocardial infarction (MI) size, decreasing the secretion of heart damage biomarkers, and improving cardiac function. Then, the myocardial anoxia/reoxygenation (A/R) injury model was established to mimic reperfusion injury. Noticeably, ferroptosis was the major death manner for A/R-damaged H9c2 cells. Meanwhile, CVP significantly inhibited ferroptosis by decreasing intracellular Fe2+ level, enhancing GPX4 expression, and suppressing lipid peroxidation to confront A/R injury. In conclusion, CVP, with a clear structure, ameliorated I/R injury by inhibiting ferroptosis.
Collapse
Affiliation(s)
- Peng He
- School of Basic Medical Sciences, Qingdao University, Qingdao, China
| | - Mi Zhang
- School of Basic Medical Sciences, Qingdao University, Qingdao, China
| | - Meng Zhao
- School of Nursing, Qingdao University, Qingdao, China
| | - Mengyao Zhang
- School of Basic Medical Sciences, Qingdao University, Qingdao, China
| | - Benxu Ma
- The Affiliated Qingdao Central Hospital of Qingdao University, The Second Affiliated Hospital of Medical College of Qingdao University, Qingdao, China
| | - Hongyu Lv
- School of Basic Medical Sciences, Qingdao University, Qingdao, China
| | - Yantao Han
- School of Basic Medical Sciences, Qingdao University, Qingdao, China
| | - Dingtao Wu
- Key Laboratory of Coarse Cereal Processing (Ministry of Agriculture and Rural Affairs), Sichuan Engineering & Technology Research Center of Coarse Cereal Industralization, School of Food and Biological Engineering, Chengdu University, Chengdu, China
| | - Zhangfeng Zhong
- Macau Centre for Research and Development in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Zhuhai, Macao SAR, China
- Zhangfeng Zhong,
| | - Wenwen Zhao
- School of Basic Medical Sciences, Qingdao University, Qingdao, China
- Macau Centre for Research and Development in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Zhuhai, Macao SAR, China
- *Correspondence: Wenwen Zhao,
| |
Collapse
|
18
|
Peng JF, Salami OM, Lei C, Ni D, Habimana O, Yi GH. Targeted mitochondrial drugs for treatment of Myocardial ischemia-reperfusion injury. J Drug Target 2022; 30:833-844. [PMID: 35652502 DOI: 10.1080/1061186x.2022.2085728] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Myocardial ischemia-reperfusion injury (MI/RI) refers to the further damage done to ischemic cardiomyocytes when restoring blood flow. A large body of evidence shows that MI/RI is closely associated with excessive production of mitochondrial reactive oxygen species, mitochondrial calcium overload, disordered mitochondrial energy metabolism, mitophagy, mitochondrial fission, and mitochondrial fusion. According to the way it affects mitochondria, it can be divided into mitochondrial quality abnormalities and mitochondrial quantity abnormalities. Abnormal mitochondrial quality refers to the dysfunction caused by the severe destruction of mitochondria, which then affects the balance of mitochondrial density and number, causing an abnormal mitochondrial quantity. In the past, most of the reports were limited to the study of the mechanism of myocardial ischemia-reperfusion injury, some of which involved mitochondria, but no specific countermeasures were proposed. In this review, we outline the mechanisms for treating myocardial ischemia-reperfusion injury from the direction of mitochondria and focus on targeted interventions and drugs to restore mitochondrial health during abnormal mitochondrial quality control and abnormal mitochondrial quantity control. This is an update in the field of myocardial ischemia-reperfusion injury.
Collapse
Affiliation(s)
- Jin-Fu Peng
- Institute of Pharmacy and Pharmacology, Hunan province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, Hunan, 421001, China.,Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | | | - Cai Lei
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Dan Ni
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Olive Habimana
- International College, University of South China, 28 W Changsheng Road, Hengyang, Hunan 421001, China
| | - Guang-Hui Yi
- Institute of Pharmacy and Pharmacology, Hunan province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, Hunan, 421001, China.,Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| |
Collapse
|
19
|
Impact of the Gastrointestinal Tract Microbiota on Cardiovascular Health and Pathophysiology. J Cardiovasc Pharmacol 2022; 80:13-30. [PMID: 35384898 DOI: 10.1097/fjc.0000000000001273] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 03/25/2022] [Indexed: 11/25/2022]
Abstract
ABSTRACT The microbiota of the gastrointestinal tract (GIT) is an extremely diverse community of microorganisms, and their collective genomes (microbiome) provide a vast arsenal of biological activities, in particular enzymatic ones, which are far from being fully elucidated. The study of the microbiota (and the microbiome) is receiving great interest from the biomedical community as it carries the potential to improve risk-prediction models, refine primary and secondary prevention efforts, and also design more appropriate and personalized therapies, including pharmacological ones. A growing body of evidence, though sometimes impaired by the limited number of subjects involved in the studies, suggests that GIT dysbiosis, i.e. the altered microbial composition, has an important role in causing and/or worsening cardiovascular disease (CVD). Bacterial translocation as well as the alteration of levels of microbe-derived metabolites can thus be important to monitor and modulate, because they may lead to initiation and progression of CVD, as well as to its establishment as chronic state. We hereby aim to provide readers with details on available resources and experimental approaches that are used in this fascinating field of biomedical research, and on some novelties on the impact of GIT microbiota on CVD.
Collapse
|
20
|
Ma W, Zhong T, Chen J, Ke X, Zuo H, Liu Q. Exogenous H2S reverses high glucose-induced endothelial progenitor cells dysfunction via regulating autophagy. Bioengineered 2022; 13:1126-1136. [PMID: 35258406 PMCID: PMC8805971 DOI: 10.1080/21655979.2021.2017695] [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] [Indexed: 12/27/2022] Open
Abstract
This study aims to determine the effect of exogenous hydrogen sulfide (H2S) under high glucose (HG)-induced injury in endothelial progenitor cells (EPCs), and to explore the possible underlying mechanisms. Mononuclear cells were isolated from the peripheral blood of healthy volunteers by density-gradient centrifugation and identified as late EPCs by immunofluorescence and flow cytometry. EPCs were treated with high concentrations of glucose, H2S, Baf-A1, 3-MA or rapamycin. Cell proliferation, cell migration and tube formation were measured using cell counting kit-8, Transwell migration and tube formation assays, respectively. Cellular autophagy flux was detected by RFP-GFP-LC3, and Western blotting was used to examine the protein expression levels of LC3B, P62, and phosphorylated endothelial nitric oxide synthase (eNOS) at Thr495 (p-eNOSThr495). Reactive oxygen species (ROS) levels were measured using a DHE probe. H2S and rapamycin significantly reversed the inhibitory effects of HG on the proliferation, migration, and tube formation of EPCs. Moreover, H2S and rapamycin led to an increase in the number of autophagosomes accompanied by a failure in lysosomal turnover of LC3-II or p62 and p-eNOSThr495 expression and ROS production under the HG condition. However, Baf-A1 and 3-MA reversed the effects of H2S on cell behavior. Collectively, exogenous H2S ameliorated HG-induced EPC dysfunction by promoting autophagic flux and decreasing ROS production by phosphorylating eNOSThr495.
Collapse
Affiliation(s)
- Wenxue Ma
- Department of Cardiology, Huadu District People’s Hospital, Southern Medical University, Guangzhou, China
| | - Tingting Zhong
- Department of Cardiology, Fuwai Hospital, Chinese Academy of Medical Sciences, Shenzhen, (Shenzhen Sun Yat-sen Cardiovascular Hospital), Shenzhen, China
| | - Junqiu Chen
- Department of Cardiology, Fuwai Hospital, Chinese Academy of Medical Sciences, Shenzhen, (Shenzhen Sun Yat-sen Cardiovascular Hospital), Shenzhen, China
| | - Xiao Ke
- Department of Cardiology, Fuwai Hospital, Chinese Academy of Medical Sciences, Shenzhen, (Shenzhen Sun Yat-sen Cardiovascular Hospital), Shenzhen, China
| | - Huihua Zuo
- Department of Cardiology, Fuwai Hospital, Chinese Academy of Medical Sciences, Shenzhen, (Shenzhen Sun Yat-sen Cardiovascular Hospital), Shenzhen, China
| | - Qiang Liu
- Department of Cardiology, Fuwai Hospital, Chinese Academy of Medical Sciences, Shenzhen, (Shenzhen Sun Yat-sen Cardiovascular Hospital), Shenzhen, China
- Department of Cardiology, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, China
| |
Collapse
|
21
|
Sun HJ, Wu ZY, Nie XW, Wang XY, Bian JS. An Updated Insight Into Molecular Mechanism of Hydrogen Sulfide in Cardiomyopathy and Myocardial Ischemia/Reperfusion Injury Under Diabetes. Front Pharmacol 2021; 12:651884. [PMID: 34764865 PMCID: PMC8576408 DOI: 10.3389/fphar.2021.651884] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 09/23/2021] [Indexed: 12/13/2022] Open
Abstract
Cardiovascular diseases are the most common complications of diabetes, and diabetic cardiomyopathy is a major cause of people death in diabetes. Molecular, transcriptional, animal, and clinical studies have discovered numerous therapeutic targets or drugs for diabetic cardiomyopathy. Within this, hydrogen sulfide (H2S), an endogenous gasotransmitter alongside with nitric oxide (NO) and carbon monoxide (CO), is found to play a critical role in diabetic cardiomyopathy. Recently, the protective roles of H2S in diabetic cardiomyopathy have attracted enormous attention. In addition, H2S donors confer favorable effects in myocardial infarction, ischaemia-reperfusion injury, and heart failure under diabetic conditions. Further studies have disclosed that multiplex molecular mechanisms are responsible for the protective effects of H2S against diabetes-elicited cardiac injury, such as anti-oxidative, anti-apoptotic, anti-inflammatory, and anti-necrotic properties. In this review, we will summarize the current findings on H2S biology and pharmacology, especially focusing on the novel mechanisms of H2S-based protection against diabetic cardiomyopathy. Also, the potential roles of H2S in diabetes-aggravated ischaemia-reperfusion injury are discussed.
Collapse
Affiliation(s)
- Hai-Jian Sun
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Zhi-Yuan Wu
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Xiao-Wei Nie
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Xin-Yu Wang
- Department of Endocrinology, The First Affiliated Hospital of Shenzhen University (Shenzhen Second People's Hospital), Shenzhen, China
| | - Jin-Song Bian
- Department of Pharmacology, School of Medicine, Southern University of Science and Technology, Shenzhen, China.,National University of Singapore (Suzhou) Research Institute, Suzhou, China
| |
Collapse
|