1
|
Xue J, Zhuang J, Wang X, Meng T, Wu J, Zhang X, Zhang G. Mechanisms and Therapeutic Strategies for Myocardial Ischemia-Reperfusion Injury in Diabetic States. ACS Pharmacol Transl Sci 2024; 7:3691-3717. [PMID: 39698288 PMCID: PMC11651189 DOI: 10.1021/acsptsci.4c00272] [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: 05/08/2024] [Revised: 10/14/2024] [Accepted: 10/23/2024] [Indexed: 12/20/2024]
Abstract
In patients with myocardial infarction, one of the complications that may occur after revascularization is myocardial ischemia-reperfusion injury (IRI), characterized by a depleted myocardial oxygen supply and absence of blood flow recovery after reperfusion, leading to expansion of myocardial infarction, poor healing of myocardial infarction and reversal of left ventricular remodeling, and an increase in the risk for major adverse cardiovascular events such as heart failure, arrhythmia, and cardiac cell death. As a risk factor for cardiovascular disease, diabetes mellitus increases myocardial susceptibility to myocardial IRI through various mechanisms, increases acute myocardial infarction and myocardial IRI incidence, decreases myocardial responsiveness to protective strategies and efficacy of myocardial IRI protective methods, and increases diabetes mellitus mortality through myocardial infarction. This Review summarizes the mechanisms, existing therapeutic strategies, and potential therapeutic targets of myocardial IRI in diabetic states, which has very compelling clinical significance.
Collapse
Affiliation(s)
- Jing Xue
- Department
of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Jialu Zhuang
- Department
of Endocrinology, First Affiliated Hospital
of Anhui Medical University, Hefei 230031, China
| | - Xinyue Wang
- Department
of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Tao Meng
- Department
of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Jin Wu
- Department
of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Xiaoqian Zhang
- Department
of Endocrinology, First Affiliated Hospital
of Anhui Medical University, Hefei 230031, China
| | - Guiyang Zhang
- Department
of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| |
Collapse
|
2
|
Li Y, Li Z, Ren Y, Lei Y, Yang S, Shi Y, Peng H, Yang W, Guo T, Yu Y, Xiong Y. Mitochondrial-derived peptides in cardiovascular disease: Novel insights and therapeutic opportunities. J Adv Res 2024; 64:99-115. [PMID: 38008175 PMCID: PMC11464474 DOI: 10.1016/j.jare.2023.11.018] [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: 09/20/2023] [Revised: 11/14/2023] [Accepted: 11/17/2023] [Indexed: 11/28/2023] Open
Abstract
BACKGROUND Mitochondria-derived peptides (MDPs) represent a recently discovered family of peptides encoded by short open reading frames (ORFs) found within mitochondrial genes. This group includes notable members including humanin (HN), mitochondrial ORF of the 12S rDNA type-c (MOTS-c), and small humanin-like peptides 1-6 (SHLP1-6). MDPs assume pivotal roles in the regulation of diverse cellular processes, encompassing apoptosis, inflammation, and oxidative stress, which are all essential for sustaining cellular viability and normal physiological functions. Their emerging significance extends beyond this, prompting a deeper exploration into their multifaceted roles and potential applications. AIM OF REVIEW This review aims to comprehensively explore the biogenesis, various types, and diverse functions of MDPs. It seeks to elucidate the central roles and underlying mechanisms by which MDPs participate in the onset and development of cardiovascular diseases (CVDs), bridging the connections between cell apoptosis, inflammation, and oxidative stress. Furthermore, the review highlights recent advancements in clinical research related to the utilization of MDPs in CVD diagnosis and treatment. KEY SCIENTIFIC CONCEPTS OF REVIEW MDPs levels are diminished with aging and in the presence of CVDs, rendering them potential new indicators for the diagnosis of CVDs. Also, MDPs may represent a novel and promising strategy for CVD therapy. In this review, we delve into the biogenesis, various types, and diverse functions of MDPs. We aim to shed light on the pivotal roles and the underlying mechanisms through which MDPs contribute to the onset and advancement of CVDs connecting cell apoptosis, inflammation, and oxidative stress. We also provide insights into the current advancements in clinical research related to the utilization of MDPs in the treatment of CVDs. This review may provide valuable information with MDPs for CVD diagnosis and treatment.
Collapse
Affiliation(s)
- Yang Li
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi'an 710069, Shaanxi, PR China
| | - Zhuozhuo Li
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi'an 710069, Shaanxi, PR China
| | - Yuanyuan Ren
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi'an 710069, Shaanxi, PR China
| | - Ying Lei
- School of Medicine, Northwest University, Xi'an 710069, Shaanxi, PR China
| | - Silong Yang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi'an 710069, Shaanxi, PR China
| | - Yuqi Shi
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi'an 710069, Shaanxi, PR China
| | - Han Peng
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi'an 710069, Shaanxi, PR China
| | - Weijie Yang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi'an 710069, Shaanxi, PR China
| | - Tiantian Guo
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi'an 710069, Shaanxi, PR China
| | - Yi Yu
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi'an 710069, Shaanxi, PR China; School of Medicine, Northwest University, Xi'an 710069, Shaanxi, PR China.
| | - Yuyan Xiong
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi'an 710069, Shaanxi, PR China; Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, The Affiliated Hospital of Northwest University, 710018 Xi'an, Shaanxi, PR China.
| |
Collapse
|
3
|
Williams LE, Frishman WH. FDY-5301: An Innovative Approach to The Treatment of Revascularization Coronary Injury. Cardiol Rev 2024; 32:429-432. [PMID: 36883828 DOI: 10.1097/crd.0000000000000517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/09/2023]
Abstract
After experiencing an acute ST-segment elevation myocardial infarction (STEMI), percutaneous coronary intervention (PCI) is a preferred method of restoring blood flow to the heart. While this reperfusion has long-term benefits, it can result in reperfusion injury in the short term, which involves the formation of reactive oxygen species (ROS) and neutrophil recruitment. FDY-5301 is a sodium iodide-based drug that acts as a catalyst in the conversion of hydrogen peroxide to water and oxygen. FDY-5301 is designed to be administered as an intravenous bolus following a STEMI, before reperfusion with PCI, to reduce the damage associated with reperfusion injury. Clinical trials have shown FDY-5301 administration to be safe, feasible, and fast-acting in its ability to increase plasma iodide concentration, and the results are favorable in demonstrating potential efficacy. FDY-5301 shows potential in its use to reduce the effects of reperfusion injury, and ongoing Phase 3 trials will allow for continued evaluation of its performance.
Collapse
Affiliation(s)
- Lauren E Williams
- Department of Medicine, New York Medical College/Westchester Medical Center, Valhalla, New York
| | - William H Frishman
- Departments of Medicine and Cardiology, New York Medical College/Westchester Medical Center, Valhalla, New York
| |
Collapse
|
4
|
Gajardo Cortez AIJ, Lillo-Moya J, San-Martín-Martinez D, Pozo-Martinez J, Morales P, Prieto JC, Aguayo R, Puentes Á, Ramos C, Silva S, Catalán M, Ramos K, Olea-Azar C, Rodrigo R. Safety and Pharmacokinetics of a Combined Antioxidant Therapy against Myocardial Reperfusion Injury: A Phase 1 Randomized Clinical Trial in Healthy Humans. Clin Pharmacol Drug Dev 2024; 13:1051-1060. [PMID: 38973337 DOI: 10.1002/cpdd.1443] [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: 03/03/2024] [Accepted: 06/03/2024] [Indexed: 07/09/2024]
Abstract
Myocardial reperfusion injury (MRI) accounts for up to 50% of the final size in acute myocardial infarction and other conditions associated with ischemia-reperfusion. Currently, there is still no therapy to prevent MRI, but it is well known that oxidative stress has a key role in its mechanism. We previously reduced MRI in rats through a combined antioxidant therapy (CAT) of ascorbic acid, N-acetylcysteine, and deferoxamine. This study determines the safety and pharmacokinetics of CAT in a Phase I clinical trial. Healthy subjects (n = 18) were randomized 2:1 to CAT or placebo (NaCl 0.9% i.v.). Two different doses/infusion rates of CATs were tested in a single 90-minute intravenous infusion. Blood samples were collected at specific times for 180 minutes to measure plasma drug concentrations (ascorbic acid, N-acetylcysteine, and deferoxamine) and oxidative stress biomarkers. Adverse events were registered during infusion and followed for 30 days. Both CAT1 and CAT2 significantly increased the CAT drug concentrations compared to placebo (P < .05). Most of the pharmacokinetic parameters were similar between CAT1 and CAT2. In total, 6 adverse events were reported, all nonserious and observed in CAT1. The ferric-reducing ability of plasma (an antioxidant biomarker) increased in both CAT groups compared to placebo (P < .001). The CAT is safe in humans and a potential treatment for patients with acute myocardial infarction undergoing reperfusion therapy.
Collapse
Affiliation(s)
- Abraham I J Gajardo Cortez
- Pathophysiology Program, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile
- Intensive Care Unit, Department of Medicine, Hospital Clínico Universidad de Chile, University of Chile, Santiago, Chile
| | - José Lillo-Moya
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Daniel San-Martín-Martinez
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Josue Pozo-Martinez
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile
- Laboratorio de Química Médica, Facultad de Ciencia y Tecnología, Universidad del Azuay, Cuenca, Ecuador
| | - Pablo Morales
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Juan C Prieto
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile
- Cardiovascular Department, Hospital Clínico Universidad de Chile, University of Chile, Santiago, Chile
| | - Rubén Aguayo
- Cardiology Unit, Department of Medicine, Occident Division, San Juan de Dios Hospital, Santiago, Chile
| | - Ángel Puentes
- Cardiology Unit, Department of Medicine, Occident Division, San Juan de Dios Hospital, Santiago, Chile
| | - Cristobal Ramos
- Department of Radiology, Hospital Clínico Universidad de Chile, Campus Norte, Santiago, Chile
| | - Solange Silva
- Department of Radiology, Hospital Clínico Universidad de Chile, Campus Norte, Santiago, Chile
| | - Mabel Catalán
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Karla Ramos
- Intensive Medicine Unit, Faculty of Medicine, Pontifical Catholic University of Chile, Santiago, Chile
| | - Claudio Olea-Azar
- Department of Inorganic and Analytical Chemistry, Faculty of Chemical and Pharmaceutical Sciences, University of Chile, Santiago, Chile
| | - Ramón Rodrigo
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile
| |
Collapse
|
5
|
Lv Q, Lin J, Huang H, Ma B, Li W, Chen J, Wang M, Wang X, Fu G, Xiao Y. Nanosponge for Iron Chelation and Efflux: A Ferroptosis-Inhibiting Approach for Myocardial Infarction Therapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2305895. [PMID: 38671590 PMCID: PMC11220697 DOI: 10.1002/advs.202305895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 04/11/2024] [Indexed: 04/28/2024]
Abstract
Myocardial infarction (MI), a consequence of coronary artery occlusion, triggers the degradation of ferritin, resulting in elevated levels of free iron in the heart and thereby inducing ferroptosis. Targeting myocardial ferroptosis through the chelation of excess iron has therapeutic potential for MI treatment. However, iron chelation in post ischemic injury areas using conventional iron-specific chelators is hindered by ineffective myocardial intracellular chelation, rapid clearance, and high systemic toxicity. A chitosan-desferrioxamine nanosponge (CDNS) is designed by co-crosslinking chitosan and deferoxamine through noncovalent gelation to address these challenges. This architecture facilitates direct iron chelation regardless of deferoxamine (DFO) release due to its sponge-like porous hydrogel structure. Upon cellular internalization, CDNS can effectively chelate cellular iron and facilitate the efflux of captured iron, thereby inhibiting ferroptosis and associated oxidative stress and lipid peroxidation. In MI mouse models, myocardial injection of CDNS promotes sustainable retention and the suppression of ferroptosis in the infarcted heart. This intervention improves cardiac function and alleviates adverse cardiac remodeling post-MI, leading to decreased oxidative stress and the promotion of angiogenesis due to ferroptosis inhibition by CDNS in the infarcted heart. This study reveals a nanosponge-based nanomedicine targeting myocardial ferroptosis with efficient iron chelation and efflux, offering a promising MI treatment.
Collapse
Affiliation(s)
- Qingbo Lv
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang ProvinceDepartment of CardiologySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhou310016China
| | - Jun Lin
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang ProvinceDepartment of CardiologySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhou310016China
- Department of Cardiovascular SurgerySun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhou510120China
| | - He Huang
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang ProvinceDepartment of CardiologySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhou310016China
| | - Boxuan Ma
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang ProvinceDepartment of CardiologySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhou310016China
| | - Wujiao Li
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang ProvinceDepartment of CardiologySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhou310016China
| | - Jiawen Chen
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang ProvinceDepartment of CardiologySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhou310016China
| | - Meihui Wang
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang ProvinceDepartment of CardiologySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhou310016China
| | - Xiaoyu Wang
- Qiushi Academy for Advanced StudiesZhejiang UniversityHangzhou310058China
| | - Guosheng Fu
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang ProvinceDepartment of CardiologySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhou310016China
| | - Yun Xiao
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang ProvinceDepartment of CardiologySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhou310016China
| |
Collapse
|
6
|
Shen X, Ma M, Mi R, Zhuang J, Song Y, Yang W, Li H, Lu Y, Yang B, Liu Y, Wu Y, Shen H. EFHD1 promotes osteosarcoma proliferation and drug resistance by inhibiting the opening of the mitochondrial membrane permeability transition pore (mPTP) by binding to ANT3. Cell Mol Life Sci 2024; 81:236. [PMID: 38795203 PMCID: PMC11127909 DOI: 10.1007/s00018-024-05254-8] [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: 08/07/2023] [Revised: 03/19/2024] [Accepted: 04/24/2024] [Indexed: 05/27/2024]
Abstract
Chemoresistance is the main obstacle in the clinical treatment of osteosarcoma (OS). In this study, we investigated the role of EF-hand domain-containing protein 1 (EFHD1) in OS chemotherapy resistance. We found that the expression of EFHD1 was highly correlated with the clinical outcome after chemotherapy. We overexpressed EFHD1 in 143B cells and found that it increased their resistance to cell death after drug treatment. Conversely, knockdown of EFHD1 in 143BR cells (a cisplatin-less-sensitive OS cell line derived from 143B cells) increased their sensitivity to treatment. Mechanistically, EFHD1 bound to adenine nucleotide translocase-3 (ANT3) and inhibited its conformational change, thereby inhibiting the opening of the mitochondrial membrane permeability transition pore (mPTP). This effect could maintain mitochondrial function, thereby favoring OS cell survival. The ANT3 conformational inhibitor carboxyatractyloside (CATR), which can promote mPTP opening, enhanced the chemosensitivity of EFHD1-overexpressing cells when combined with cisplatin. The ANT3 conformational inhibitor bongkrekic acid (BKA), which can inhibit mPTP opening, restored the resistance of EFHD1 knockdown cells. In conclusion, our results suggest that EFHD1-ANT3-mPTP might be a promising target for OS therapy in the future.
Collapse
Affiliation(s)
- Xin Shen
- Department of Orthopedics, The Eighth Affiliated Hospital of Sun Yat-sen University, No. 3025 Shennan Zhong Road, Shenzhen, 518033, Guangdong, China
| | - Mengjun Ma
- Department of Orthopedics, The Eighth Affiliated Hospital of Sun Yat-sen University, No. 3025 Shennan Zhong Road, Shenzhen, 518033, Guangdong, China
| | - Rujia Mi
- Center for Biotherapy, The Eighth Affiliated Hospital of Sun Yat-sen University, No. 3025 Shennan Zhong Road, Shenzhen, 518033, Guangdong, China
| | - Jiahao Zhuang
- Department of Orthopedics, The Eighth Affiliated Hospital of Sun Yat-sen University, No. 3025 Shennan Zhong Road, Shenzhen, 518033, Guangdong, China
| | - Yihui Song
- Department of Orthopedics, The Eighth Affiliated Hospital of Sun Yat-sen University, No. 3025 Shennan Zhong Road, Shenzhen, 518033, Guangdong, China
| | - Wen Yang
- Department of Orthopedics, The Eighth Affiliated Hospital of Sun Yat-sen University, No. 3025 Shennan Zhong Road, Shenzhen, 518033, Guangdong, China
| | - Hongyu Li
- Department of Orthopedics, The Eighth Affiliated Hospital of Sun Yat-sen University, No. 3025 Shennan Zhong Road, Shenzhen, 518033, Guangdong, China
| | - Yixuan Lu
- Center for Biotherapy, The Eighth Affiliated Hospital of Sun Yat-sen University, No. 3025 Shennan Zhong Road, Shenzhen, 518033, Guangdong, China
| | - Biao Yang
- Department of Orthopedics, The Eighth Affiliated Hospital of Sun Yat-sen University, No. 3025 Shennan Zhong Road, Shenzhen, 518033, Guangdong, China
| | - Yinliang Liu
- Department of Orthopedics, The Eighth Affiliated Hospital of Sun Yat-sen University, No. 3025 Shennan Zhong Road, Shenzhen, 518033, Guangdong, China
| | - Yanfeng Wu
- Center for Biotherapy, The Eighth Affiliated Hospital of Sun Yat-sen University, No. 3025 Shennan Zhong Road, Shenzhen, 518033, Guangdong, China.
| | - Huiyong Shen
- Department of Orthopedics, The Eighth Affiliated Hospital of Sun Yat-sen University, No. 3025 Shennan Zhong Road, Shenzhen, 518033, Guangdong, China.
| |
Collapse
|
7
|
Elmahdy A, Shekka Espinosa A, Kakaei Y, Pylova T, Jha A, Zulfaj E, Krasnikova M, Al-Awar A, Sheybani Z, Sevastianova V, Berger E, Nejat A, Molander L, Andersson EA, Omerovic E, Hussain S, Redfors B. Ischemic preconditioning affects phosphosites and accentuates myocardial stunning while reducing infarction size in rats. Front Cardiovasc Med 2024; 11:1376367. [PMID: 38559672 PMCID: PMC10978780 DOI: 10.3389/fcvm.2024.1376367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 02/29/2024] [Indexed: 04/04/2024] Open
Abstract
Background and aims Ischemic preconditioning (IPC), i.e., brief periods of ischemia, protect the heart from subsequent prolonged ischemic injury, and reduces infarction size. Myocardial stunning refers to transient loss of contractility in the heart after myocardial ischemia that recovers without permanent damage. The relationship between IPC and myocardial stunning remains incompletely understood. This study aimed primarily to examine the effects of IPC on the relationship between ischemia duration, stunning, and infarct size in an ischemia-reperfusion injury model. Secondarily, this study aimed to examine to which extent the phosphoproteomic changes induced by IPC relate to myocardial contractile function. Methods and results Rats were subjected to different durations of left anterior descending artery (LAD) occlusion, with or without preceding IPC. Echocardiograms were acquired to assess cardiac contraction in the affected myocardial segment. Infarction size was evaluated using triphenyl tetrazolium chloride staining. Phosphoproteomic analysis was performed in heart tissue from preconditioned and non-preconditioned animals. In contrast to rats without IPC, reversible akinesia was observed in a majority of the rats that were subjected to IPC and subsequently exposed to ischemia of 13.5 or 15 min of ischemia. Phosphoproteomic analysis revealed significant differential regulation of 786 phosphopeptides between IPC and non-IPC groups, with significant associations with the sarcomere, Z-disc, and actin binding. Conclusion IPC induces changes in phosphosites of proteins involved in myocardial contraction; and both accentuates post-ischemic myocardial stunning and reduces infarct size.
Collapse
Affiliation(s)
- Ahmed Elmahdy
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Aaron Shekka Espinosa
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Yalda Kakaei
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Tetiana Pylova
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Abhishek Jha
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Ermir Zulfaj
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Maryna Krasnikova
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Amin Al-Awar
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Zahra Sheybani
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Valentyna Sevastianova
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Evelin Berger
- Proteomics Core Facility, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Amirali Nejat
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Linnea Molander
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Erik Axel Andersson
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Elmir Omerovic
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Cardiology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Shafaat Hussain
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Björn Redfors
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
- Department of Cardiology, Sahlgrenska University Hospital, Gothenburg, Sweden
| |
Collapse
|
8
|
Verma VK, Mutneja E, Malik S, Sahu AK, Prajapati V, Bhardwaj P, Ray R, Nag TC, Bhatia J, Arya DS. Abatacept: A Promising Repurposed Solution for Myocardial Infarction-Induced Inflammation in Rat Models. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2024; 2024:3534104. [PMID: 38957586 PMCID: PMC11219209 DOI: 10.1155/2024/3534104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Revised: 02/12/2024] [Accepted: 02/28/2024] [Indexed: 07/04/2024]
Abstract
Myocardial infarction (MI) is irreversible damage to the myocardial tissue caused by prolonged ischemia/hypoxia, subsequently leading to loss of contractile function and myocardial damage. However, after a perilous period, ischemia-reperfusion (IR) itself causes the generation of oxygen free radicals, disturbance in cation homeostasis, depletion of cellular energy stores, and activation of innate and adaptive immune responses. The present study employed Abatacept (ABT), which is an anti-inflammatory drug, originally used as an antirheumatic response agent. To investigate the cardioprotective potential of ABT, primarily, the dose was optimized in a chemically induced model of myocardial necrosis. Thereafter, ABT optimized the dose of 5 mg/kg s.c. OD was investigated for its cardioprotective potential in a surgical model of myocardial IR injury, where animals (n = 30) were randomized into five groups: Sham, IR-C, Telmi10 + IR (Telmisartan, 10 mg/kg oral OD), ABT5 + IR, ABT perse. ABT and telmisartan were administered for 21 days. On the 21st day, animals were subjected to LAD coronary artery occlusion for 60 min, followed by reperfusion for 45 min. Further, the cardioprotective potential was assessed through hemodynamic parameters, oxidant-antioxidant biochemical enzymatic parameters, cardiac injury, inflammatory markers, histopathological analysis, TUNEL assay, and immunohistochemical evaluation, followed by immunoblotting to explore signaling pathways. The statistics were performed by one-way analysis of variance, followed by the Tukey comparison post hoc tests. Noteworthy, 21 days of ABT pretreatment amended the hemodynamic and ventricular functions in the rat models of MI. The cardioprotective potential of ABT is accompanied by inhibiting MAP kinase signaling and modulating Nrf-2/HO-1 proteins downstream signaling cascade. Overall, the present work bolsters the previously known anti-inflammatory role of ABT in MI and contributes a mechanistic insight and application of clinically approved drugs in averting the activation of inflammatory response.
Collapse
Affiliation(s)
- Vipin Kumar Verma
- Cardiovascular Research Laboratory, Department of Pharmacology, All India Institute of Medical Sciences, New Delhi-110029, India
| | - Ekta Mutneja
- Cardiovascular Research Laboratory, Department of Pharmacology, All India Institute of Medical Sciences, New Delhi-110029, India
| | - Salma Malik
- Cardiovascular Research Laboratory, Department of Pharmacology, All India Institute of Medical Sciences, New Delhi-110029, India
| | - Anil Kumar Sahu
- Cardiovascular Research Laboratory, Department of Pharmacology, All India Institute of Medical Sciences, New Delhi-110029, India
| | - Vaishali Prajapati
- Cardiovascular Research Laboratory, Department of Pharmacology, All India Institute of Medical Sciences, New Delhi-110029, India
| | - Priya Bhardwaj
- Cardiovascular Research Laboratory, Department of Pharmacology, All India Institute of Medical Sciences, New Delhi-110029, India
| | - Ruma Ray
- Cardiac Pathology Laboratory, Department of Pathology, All India Institute of Medical Sciences, New Delhi-110029, India
| | - Tapas Chandra Nag
- Department of Anatomy, All India Institute of Medical Sciences, New Delhi-110029, India
| | - Jagriti Bhatia
- Cardiovascular Research Laboratory, Department of Pharmacology, All India Institute of Medical Sciences, New Delhi-110029, India
| | - Dharamvir Singh Arya
- Cardiovascular Research Laboratory, Department of Pharmacology, All India Institute of Medical Sciences, New Delhi-110029, India
| |
Collapse
|
9
|
Ajami M, Sotoudeheian M, Houshiar-Rad A, Esmaili M, Naeini F, Mohammadi Nasrabadi F, Doaei S, Milani-Bonab A. Quercetin may reduce the risk of developing the symptoms of COVID-19. AVICENNA JOURNAL OF PHYTOMEDICINE 2024; 14:189-201. [PMID: 38966631 PMCID: PMC11221767 DOI: 10.22038/ajp.2023.22920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 09/10/2022] [Accepted: 09/11/2022] [Indexed: 07/06/2024]
Abstract
Objective Recent evidence reported that some dietary compounds like quercetin and apigenin as the most well-known flavonoids with anti-inflammatory effects may inhibit SARS-CoV-2 main protease. The hypothesis of the promising effects and possible mechanisms of action of quercetin against COVID-19 were assessed in this article. Materials and Methods Related papers on the inhibitory effects of quercetin against COVID-19 were collected using the following search strategy: "corona or coronavirus or COVID or COVID-19 or viral or virus" AND "nutrient or flavonoid or Quercetin". Results The findings indicated that quercetin can be considered an effective agent against COVID-19 because of its SARS-CoV-2 main protease and RNA-dependent RNA polymerase inhibitory effects. In addition, quercetin may attenuate angiotensin-converting enzyme-2 (ACE-2) receptors leading to a reduction of SARS-CoV-2 ability to enter host cells. Moreover, the antiviral, anti-inflammatory, and immunomodulatory activities of quercetin have been frequently reported. Conclusion Quercetin may be an effective agent for managing the complications of COVID-19. Further longitudinal human studies are warranted.
Collapse
Affiliation(s)
- Marjan Ajami
- Department of Food and Nutrition Policy and Planning Research, Faculty of Nutrition Sciences and Food Technology, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Anahita Houshiar-Rad
- Department of Nutrition Research, Faculty of Nutrition Sciences and Food Technology, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mina Esmaili
- Department of Nutrition Research, Faculty of Nutrition Sciences and Food Technology, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemeh Naeini
- Department of Clinical Nutrition, School of Nutritional Sciences and Dietetics, Tehran university of Medical Science, Tehran, Iran
| | - Fatemeh Mohammadi Nasrabadi
- Department of Food and Nutrition Policy and Planning Research, Faculty of Nutrition Sciences and Food Technology, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Saied Doaei
- Department of Community Nutrition, Faculty of Nutrition and Food Technology, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ali Milani-Bonab
- Department of Food and Nutrition Policy and Planning Research, Faculty of Nutrition Sciences and Food Technology, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
10
|
Jiang L, Zhou X, Zhao X, Wang Z, Huang A, Huang Y, Sun H, Guan F, Jiang W. Tetrandrine downregulates TRPV2 expression to ameliorate myocardial ischemia/reperfusion injury in rats via regulation of cardiomyocyte apoptosis, calcium homeostasis and mitochondrial function. Eur J Pharmacol 2024; 964:176246. [PMID: 38061472 DOI: 10.1016/j.ejphar.2023.176246] [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: 05/30/2023] [Revised: 11/29/2023] [Accepted: 11/30/2023] [Indexed: 12/30/2023]
Abstract
Our previous study has indicated that tetrandrine (TET) can target miR-202-5p to repress the activation of transient receptor potential vanilloid type 2 (TRPV2), eventually ameliorating the progression of myocardial ischemia/reperfusion injury (MI/RI). This study is aimed to further ascertain the detailed mechanisms between TET and TRPV2 in MI/RI pathogenesis. Here, a myocardial I/R injury rat model and a hypoxia-reoxygenation (H/R) model in rat myocardial cell line (H9C2 cells) were established. We reported that pronounced upregulation of TRPV2 was observed in I/R rats and H/R-induced H9C2 cells. Silencing of TRPV2 could improve cardiac function and myocardial injury, reduced infarction size, and promoted cardiomyocyte proliferation in I/R rats. In I/R rats or H/R-induced H9C2 cells, cardiomyocyte apoptosis was inhibited by knocking-down TRPV2. Meanwhile, the silenced TRPV2 or TET treatment ameliorated the damaged mitochondrial structure, mitigated ROS generation, restored the impaired ΔΨM, inhibited mPTP opening and alleviated Ca2+ overload in H/R-induced H9C2 cells. The results obtained from the overexpression of TRPV2 were contrary to those depicted above. Moreover, TET could downregulate TRPV2 expression, while the overexpression of TRPV2 could reverse the above protective effects of TET in H/R-induced H9C2 cells. The results indicated that TET may function as a TRPV2 blocking agent, thereby attenuating the progression of MI/RI through modulation of cardiomyocyte apoptosis, calcium homeostasis and mitochondrial function. These findings offer a theoretical foundation for potential clinical application of TET therapy in patients with MI/RI.
Collapse
Affiliation(s)
- Lelin Jiang
- The Second Clinical College of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China.
| | - Xue Zhou
- Department of Cardiology, The Dingli Clinical College of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China.
| | - Xiaoli Zhao
- Department of Cardiology, The Dingli Clinical College of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China.
| | - Zhaolin Wang
- The Medical College of Shanghai University, Shanghai, 200000, China.
| | - Anwu Huang
- Department of Cardiology, The Dingli Clinical College of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China.
| | - Yiwei Huang
- Department of Cardiology, The Dingli Clinical College of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China.
| | - Huanghui Sun
- Department of Heart Function Examination, The Dingli Clinical College of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China.
| | - Fanlu Guan
- Department of Cardiology, The Third Clinical College of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China.
| | - Wenbing Jiang
- Department of Cardiology, The Dingli Clinical College of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China.
| |
Collapse
|
11
|
Jia Y, Pan J. CKLF1, transcriptionally activated by FOXC1, promotes hypoxia/reoxygenation‑induced oxidative stress and inflammation in H9c2 cells by NLRP3 inflammasome activation. Exp Ther Med 2024; 27:59. [PMID: 38234613 PMCID: PMC10790169 DOI: 10.3892/etm.2023.12347] [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: 07/12/2023] [Accepted: 08/11/2023] [Indexed: 01/19/2024] Open
Abstract
Myocardial ischemia/reperfusion (I/R) injury is a clinical challenge in the treatment of ischemic heart disease. The present study aimed to establish a hypoxia/reoxygenation (H/R)-induced H9c2 cell model to explore the role and mechanism of chemokine-like factor 1 (CKLF1) in myocardial I/R injury. First, CKLF1 expression was measured in H/R-induced H9c2 cells by reverse transcription-quantitative PCR and western blotting. Subsequently, after CKLF1 silencing, cell viability and apoptosis were evaluated by Cell Counting Kit-8 assay and flow cytometry. In addition, 2,7-dichlorodihydrofluorescein diacetate staining was used to assess the levels of cellular reactive oxygen species. Additionally, the levels of superoxide dismutase, glutathione peroxidase and malondialdehyde, and the contents of inflammatory factors IL-6, IL-1β and TNF-α were detected using corresponding commercially available kits. Western blotting was used to examine the expression levels of proteins involved in the NOD-like receptor family, pyrin domain containing 3 (NLRP3) inflammasome. The JASPAR database predicted that forkhead box protein C1 (FOXC1) would bind to the CKLF1 promoter region, and dual luciferase and chromatin immunoprecipitation assays were performed to verify it. Subsequently, FOXC1 overexpression and CKLF1 silencing were used to clarify the regulatory mechanism of FOXC1 on CKLF1 in H/R-induced H9c2 cells. The results revealed that CKLF1 expression was markedly enhanced in H/R-stimulated H9c2 cells. CKLF1 knockdown enhanced the viability and inhibited the apoptosis of H9c2 cells exposed to H/R. Moreover, the oxidative stress and inflammation induced by H/R were alleviated following CKLF1 silencing. CKLF1 knockdown also inhibited NLRP3 inflammasome activation. Furthermore, FOXC1 bound to the CKLF1 promoter region to upregulate CKLF1 expression, and FOXC1 overexpression alleviated the effects of CKLF1 knockdown on H9c2 cell damage induced by H/R via activation of the NLRP3 inflammasome. In conclusion, CKLF1 transcriptionally activated by FOXC1 may promote H/R-induced oxidative stress and inflammation in H9c2 cells via NLRP3 inflammasome activation.
Collapse
Affiliation(s)
- Yinfeng Jia
- Department of Cardiovascular Medicine, The Second People's Hospital of Yueqing, Wenzhou, Zhejiang 325608, P.R. China
| | - Jiansheng Pan
- Department of Cardiovascular Medicine, The Second People's Hospital of Yueqing, Wenzhou, Zhejiang 325608, P.R. China
| |
Collapse
|
12
|
Hussein MA, Shahidul Islam M, Ali AA, Mansour MS, Bondok M, Salem MA, Amein AS, ElGizawy HA. Malva parviflora seed oil; Isolation, gas chromatographic profiling and its cardioprotective activity against myocardial infraction in animal model. JOURNAL OF KING SAUD UNIVERSITY - SCIENCE 2024; 36:103060. [DOI: 10.1016/j.jksus.2023.103060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/25/2024]
|
13
|
Liu Z, Liu G, Wang Y, Zheng C, Guo Y. Association between skeletal muscle and left ventricular mass in patients with hyperthyroidism. Front Endocrinol (Lausanne) 2024; 15:1301529. [PMID: 38356960 PMCID: PMC10864587 DOI: 10.3389/fendo.2024.1301529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 01/08/2024] [Indexed: 02/16/2024] Open
Abstract
Objective This study aims to investigate the relationship between skeletal muscle and left ventricular mass (LVM) in patients with hyperthyroidism, providing theoretical and data-based foundations for further research on the interaction between secondary muscle atrophy and cardiac remodeling. Methods A retrospective data collection was conducted, including 136 patients with hyperthyroidism (Study group) and 50 healthy participants (control group). The Study group was further divided into Group A (high LVM) and Group B (low LVM) based on LVM size. Multiple linear regression analysis was performed to examine the correlation between skeletal muscle and LVM, with model evaluation. Based on the results, further nonlinear regression analysis was conducted to explore the detailed relationship between skeletal muscle and LVM. Results Compared to the control group, the Study group exhibited significantly lower LVM, skeletal muscle mass index (SMI), and skeletal muscle mass (SMM) (P<0.05). Within the subgroups, Group A had significantly higher SMI, SMM, and hand grip strength compared to Group B (P<0.05). The results of the multiple linear regression showed a certain correlation between SMI (β=0.60, P=0.042, 95% CI=0.02~1.17) and hand grip strength (β=0.34, P=0.045, 95% CI=0.01~0.67) with LVM. However, the residuals of the multiple regression did not follow a normal distribution (K-S=2.50, P<0.01). Further results from a generalized linear model and structural equation modeling regression also demonstrated a correlation between SMI (β=0.60, P=0.040, 95% CI=0.03~1.17) (β=0.60, P=0.042, 95% CI=0.02~1.17) and hand grip strength (β=0.34, P=0.043, 95% CI=0.01~0.67) (β=0.34, P=0.045, 95% CI=0.01~0.67) with LVM. Conclusion Patients with hyperthyroidism may exhibit simultaneous decreases in LVM, SMM, and SMI. The LVM in patients is correlated with SMM and hand grip strength, highlighting the need for further exploration of the causal relationship and underlying mechanisms. These findings provide a basis for the prevention and treatment of secondary sarcopenia and cardiac pathology in patients with hyperthyroidism.
Collapse
Affiliation(s)
- Zhenchao Liu
- Institute of Integrative Medicine, Qingdao University, Qingdao, Shandong, China
| | - Guang Liu
- Shandong Provincial Sports Center, Shandong Administration of Sports, Jinan, Shandong, China
| | - Yanzhi Wang
- Academic Affairs Office, Binzhou Medical University, Yantai, Shandong, China
| | - Chongwen Zheng
- Department of Neurology, The 2nd Affiliated Hospital of Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
| | - Yunliang Guo
- Institute of Integrative Medicine, Qingdao University, Qingdao, Shandong, China
| |
Collapse
|
14
|
Lin Y, Yang X, Li Y, Huang DJ, Sun ZQ. A newly synthesized flavone avoids COMT-catalyzed methylation and mitigates myocardial ischemia/reperfusion injury in H9C2 cells via JNK and P38 pathways. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2024; 27:492-499. [PMID: 38419895 PMCID: PMC10897552 DOI: 10.22038/ijbms.2023.74358.16149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 11/08/2023] [Indexed: 03/02/2024]
Abstract
Objectives Luteolin is a flavone that provides defense against myocardial ischemia/reperfusion (I/R) injury. However, this compound is subjected to methylation mediated by catechol-O-methyltransferase (COMT), thus influencing its pharmacological effect. To synthesize a new flavone from luteolin that avoids COMT-catalyzed methylation and find out the protective mechanism of LUA in myocardial I/R injury. Materials and Methods Luteolin and 2,2'-azobis (2-amidinopropane) dihydrochloride (AAPH) were used to synthesize the new flavone known as LUAAPH-1 (LUA). Then, the myocardial ischemia/reperfusion injury cell model was established using H9c2 cells to detect the effect in myocardial ischemia/reperfusion regulation and to identify the underlying mechanism. Results Pretreatment with LUA (20 μmol/l) substantially increased cell viability while reducing cell apoptosis rate and caspase-3 expression induced by I/R, and the protective effect of LUA on cell viability was stronger than diosmetin, which is the major methylated metabolite of luteolin. In addition, intracellular reactive oxygen species (ROS) production and calcium accumulation were both inhibited by LUA. Furthermore, we identified that LUA markedly relieved the promotive effects of I/R stimulation upon JNK and p38 phosphorylation. Conclusion LUT pretreatment conveys significant cardioprotective effects after myocardial I/R injury, and JNK and p38 MAPK signaling pathway may be involved.
Collapse
Affiliation(s)
- Ye Lin
- School of Medical and Health Engineering, Changzhou University, Changzhou 213164, P.R. China
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Xin Yang
- Food Science and Technology Program, Department of Chemistry, Faculty of Science, National University of Singapore, Singapore 117597, Singapore
| | - Yan Li
- School of Medical and Health Engineering, Changzhou University, Changzhou 213164, P.R. China
| | - De-Jian Huang
- Food Science and Technology Program, Department of Chemistry, Faculty of Science, National University of Singapore, Singapore 117597, Singapore
- National University of Singapore (Suzhou) Research Institute, Suzhou, Jiangsu 215123, P.R. China
| | - Zhi-Qin Sun
- Changzhou Second People's Hospital, Changzhou 213000, P.R. China
- School of Medical and Health Engineering, Changzhou University, Changzhou 213164, P.R. China
| |
Collapse
|
15
|
Rabinovich-Nikitin I, Kirshenbaum LA. Circadian regulated control of myocardial ischemia-reperfusion injury. Trends Cardiovasc Med 2024; 34:1-7. [PMID: 36150629 DOI: 10.1016/j.tcm.2022.09.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 09/15/2022] [Accepted: 09/16/2022] [Indexed: 01/14/2023]
Abstract
Circadian mechanisms have been associated with the pathogenesis of a variety of cardiovascular diseases, including myocardial ischemia-reperfusion injury (I-R). Myocardial ischemia resulting from impaired oxygen delivery to cardiac muscle sets into motion a cascade of cellular events that paradoxically triggers greater cardiac dysfunction upon reinstitution of coronary blood supply, a phenomenon known as I-R. I-R injury has been attributed to a number of cellular defects including increased reactive oxygen species (ROS), increased intracellular calcium and impaired mitochondrial bioenergetics that ultimately lead to cardiac cell death, ventricular remodeling and heart failure. Emerging evidence has identified a strong correlation between cellular defects that underlie I-R and the disrupted circadian. In fact, recent studies have shown that circadian dysfunction exacerbates cardiac injury following MI from impaired cellular quality control mechanisms such as autophagy, which are vital in the clearance of damaged cellular proteins and organelles such as mitochondria from the cell. The accumulation of cellular debris is posited as the central underlying cause of excessive cardiac cell death and ventricular dysfunction following MI. The complexities that govern the interplay between circadian biology and I-R injury following MI is at its infancy and understanding how circadian misalignment, such as in shift workers impacts I-R injury is of great scientific and clinical importance toward development of new therapeutic strategies using chronotherapy and circadian regulation to mitigate cardiac injury and improve cardiac outcomes after injury. In this review, we highlight recent advances in circadian biology and adaptive cellular quality control mechanisms that influence cardiac injury in response to MI injury with a specific focus on how circadian biology can be utilized to further cardiovascular medicine and patient care.
Collapse
Affiliation(s)
- Inna Rabinovich-Nikitin
- Department of Physiology and Pathophysiology, The Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Canada
| | - Lorrie A Kirshenbaum
- Department of Physiology and Pathophysiology, The Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Canada; Department of Pharmacology and Therapeutics Rady College of Medicine, Max Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba R2H 2H6, Canada.
| |
Collapse
|
16
|
Khan SU, Khan SU, Suleman M, Khan MU, Khan MS, Arbi FM, Hussain T, Mohammed Alsuhaibani A, S Refat M. Natural Allies for Heart Health: Nrf2 Activation and Cardiovascular Disease Management. Curr Probl Cardiol 2024; 49:102084. [PMID: 37714318 DOI: 10.1016/j.cpcardiol.2023.102084] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 09/11/2023] [Indexed: 09/17/2023]
Abstract
The term "cardiovascular diseases" (CVD) refers to various ailments that affect the heart and blood vessels, including myocardial ischemia, congenital heart defects, heart failure, rheumatic heart disease, hypertension, peripheral artery disease, atherosclerosis, and cardiomyopathies. Despite significant breakthroughs in preventative measures and treatment choices, CVDs significantly contribute to morbidity and mortality, imposing a considerable financial burden. Oxidative stress (OS) is a fundamental contributor to the development and progression of CVDs, resulting from an inherent disparity in generating reactive oxygen species. The disparity above significantly contributes to the aberrant operation of the cardiovascular system. To tackle this issue, therapeutic intervention primarily emphasizes the nuclear erythroid 2-related factor 2 (Nrf2), a transcription factor crucial in regulating endogenous antioxidant defense systems against OS. The Nrf2 exhibits potential as a promising target for effectively managing CVDs. Significantly, an emerging field of study is around the utilization of natural substances to stimulate the activation of Nrf2, hence facilitating the promotion of cardioprotection. This technique introduces a new pathway for treating CVD. The substances above elicit their advantageous effects by mitigating the impact of OS via initiating Nrf2 signaling. The primary objective of our study is to provide significant insights that can contribute to advancing treatment methods, including natural products. These strategies aim to tackle the obstacles associated with CVDs.
Collapse
Affiliation(s)
- Safir Ullah Khan
- Hefei National Laboratory for Physical Sciences at the Microscale, School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Shahid Ullah Khan
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City and South west University, College of Agronomy and Biotechnology, Southwest University, Chongqing, China; Department of Biochemistry, Women Medical and Dental College, Khyber Medical University, Abbottabad, Khyber Pakhtunkhwa, Pakistan.
| | - Muhammad Suleman
- Center for Biotechnology and Microbiology, University of Swat, Swat, Pakistan; Laboratory of Animal Research Center (LARC), Qatar University, Doha, Qatar
| | - Munir Ullah Khan
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, International Research Center for X Polymers, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, China
| | | | | | - Talib Hussain
- Women Dental College Abbottabad, Khyber Pakhtunkhwa, Pakistan
| | - Amnah Mohammed Alsuhaibani
- Department of Physical Sport Science, College of Education, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Moamen S Refat
- Department of Chemistry, College of Science, Taif University, Taif, Saudi Arabia
| |
Collapse
|
17
|
Yu CJ, Xia F, Ruan L, Hu SP, Zhu WJ, Yang K. Circ_0004771 Promotes Hypoxia/Reoxygenation Induced Cardiomyocyte Injury via Activation of Mitogen-Activated Protein Kinase Signaling Pathway. Int Heart J 2023; 64:1125-1132. [PMID: 37967979 DOI: 10.1536/ihj.23-333] [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] [Indexed: 11/17/2023]
Abstract
This study aimed to observe the mechanism and effect of circ_0004771 on cardiomyocyte injury in acute myocardial infarction (AMI). The differences in circ_0004771 expression in the blood of AMI patients and healthy volunteers were observed by Real-Time Quantitative Reverse Transcription-Polymerase Chain Reaction. AMI cell models were constructed by hypoxia/reoxygenation (H/R)-induced injury in human cardiomyocytes (AC16 cells). The changes of circ_0004771 expression in AMI cells were observed. After transfection with the knockdown or overexpression of circ_0004771 vector in AMI cells, Cell Counting Kit-8 (CCK-8) assay and propidium iodide/FITC-Annexin V staining were performed to detect cell proliferation and apoptosis levels, extracellular lactate dehydrogenase (LDH) activity, malondialdehyde (MDA) concentration, and superoxide dismutase (SOD) activity. Expression levels of Mitogen-activated protein kinase (MAPK) signaling pathway-related proteins (p-MEK1/2, MEK1/2, p-ERK1/2, ERK1/2), and endoplasmic reticulum (ER) stress proteins (GRP78 and CHOP-1) were observed in each group of cells by western blot method. The expression level of circ_0004771 was significantly reduced in both clinical samples and cells of AMI. When circ_0004771 was knocked down in AMI cells, it resulted in a decrease in cell proliferation level and significant increase in apoptosis level. The inhibition of circ_0004771 expression caused leakage of LDH in AMI cells, accumulation of intracellular MDA, and inhibition of SOD activity. In addition, the knockdown of circ_0004771 significantly increased the levels of p-MEK1/2, p-ERK1/2, GRP78, and CHOP-1 in H/R-induced AC16 cells. However, the overexpression of circ_0004771 resulted in the opposite result as when circ_0004771 was knocked down. A low level of circ_0004771 in AMI activates the MAPK signaling pathway in cardiomyocytes as well as encourages intracellular oxidative stress and ER stress, thereby inhibiting cell proliferation and promoting apoptosis.
Collapse
Affiliation(s)
- Chun-Jun Yu
- Department of Cardiovascular Surgery, Wuhan Asia General Hospital
| | - Feng Xia
- Department of Cardiovascular Surgery, Wuhan Asia General Hospital
| | - Lin Ruan
- Department of Cardiovascular Surgery, Wuhan Asia General Hospital
| | - Sheng-Peng Hu
- Department of Cardiovascular Surgery, Wuhan Asia General Hospital
| | - Wen-Jie Zhu
- Department of Cardiovascular Surgery, Wuhan Asia General Hospital
| | - Kai Yang
- Department of Cardiovascular Surgery, Wuhan Asia General Hospital
| |
Collapse
|
18
|
Xu YP, Lu XY, Song ZQ, Lin H, Chen YH. The protective effect of vagus nerve stimulation against myocardial ischemia/reperfusion injury: pooled review from preclinical studies. Front Pharmacol 2023; 14:1270787. [PMID: 38034997 PMCID: PMC10682444 DOI: 10.3389/fphar.2023.1270787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 10/31/2023] [Indexed: 12/02/2023] Open
Abstract
Aims: Myocardial ischemia-reperfusion (I/R) injury markedly undermines the protective benefits of revascularization, contributing to ventricular dysfunction and mortality. Due to complex mechanisms, no efficient ways exist to prevent cardiomyocyte reperfusion damage. Vagus nerve stimulation (VNS) appears as a potential therapeutic intervention to alleviate myocardial I/R injury. Hence, this meta-analysis intends to elucidate the potential cellular and molecular mechanisms underpinning the beneficial impact of VNS, along with its prospective clinical implications. Methods and Results: A literature search of MEDLINE, PubMed, Embase, and Cochrane Database yielded 10 articles that satisfied the inclusion criteria. VNS was significantly correlated with a reduced infarct size following myocardial I/R injury [Weighed mean difference (WMD): 25.24, 95% confidence interval (CI): 32.24 to 18.23, p < 0.001] when compared to the control group. Despite high heterogeneity (I2 = 95.3%, p < 0.001), sensitivity and subgroup analyses corroborated the robust efficacy of VNS in limiting infarct expansion. Moreover, meta-regression failed to identify significant influences of pre-specified covariates (i.e., stimulation type or site, VNS duration, condition, and species) on the primary estimates. Notably, VNS considerably impeded ventricular remodeling and cardiac dysfunction, as evidenced by improved left ventricular ejection fraction (LVEF) (WMD: 10.12, 95% CI: 4.28; 15.97, p = 0.001) and end-diastolic pressure (EDP) (WMD: 5.79, 95% CI: 9.84; -1.74, p = 0.005) during the reperfusion phase. Conclusion: VNS offers a protective role against myocardial I/R injury and emerges as a promising therapeutic strategy for future clinical application.
Collapse
Affiliation(s)
- Yu-Peng Xu
- The First Clinical Medical College, Wenzhou Medical University, Wenzhou, China
| | - Xin-Yu Lu
- The First Clinical Medical College, Wenzhou Medical University, Wenzhou, China
| | - Zheng-Qi Song
- The First Clinical Medical College, Wenzhou Medical University, Wenzhou, China
| | - Hui Lin
- Department of Respiratory, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yi-He Chen
- Department of Cardiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
19
|
Ma N, Xu H, Zhang W, Sun X, Guo R, Liu D, Zhang L, Liu Y, Zhang J, Qiao C, Chen D, Luo A, Bai J. Genome-wide analysis revealed the dysregulation of RNA binding protein-correlated alternative splicing events in myocardial ischemia reperfusion injury. BMC Med Genomics 2023; 16:251. [PMID: 37858115 PMCID: PMC10585833 DOI: 10.1186/s12920-023-01706-5] [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: 01/10/2023] [Accepted: 10/16/2023] [Indexed: 10/21/2023] Open
Abstract
BACKGROUND Myocardial ischemia reperfusion injury (MIRI), the tissue damage which is caused by the returning of blood supply to tissue after a period of ischemia, greatly reduces the therapeutic effect of treatment of myocardial infarction. But the underlying functional mechanisms of MIRI are still unclear. METHODS We constructed mouse models of MIRI, extracted injured and healthy myocardial tissues, and performed transcriptome sequencing experiments (RNA-seq) to systematically investigate the dysregulated transcriptome of MIRI, especially the alternative splicing (AS) regulation and RNA binding proteins (RBPs). Selected RBPs and MIRI-associated AS events were then validated by RT-qPCR experiments. RESULTS The differentially expressed gene (DEG) analyses indicated that transcriptome profiles were changed by MIRI and that DEGs' enriched functions were consistent with MIRI's dysregulated pathways. Furthermore, the AS profile was synergistically regulated and showed clear differences between the mouse model and the healthy samples. The exon skipping events significantly increased in MIRI model samples, while the opposite cassette exon events significantly decreased. According to the functional analysis, regulated alternative splicing genes (RASGs) were enriched in protein transport, cell division /cell cycle, RNA splicing, and endocytosis pathways, which were associated with the development of MIRI. Meanwhile, 493 differentially expressed RBPs (DE RBPs) were detected, most of which were correlated with the changed ratios of AS events. In addition, nine DE RBP genes were validated, including Eif5, Pdia6, Tagln2, Vasp, Zfp36l2, Grsf1, Idh2, Ndrg2, and Uqcrc1. These nine DE RBPs were correlated with RASGs enriched in translation process, cell growth and division, and endocytosis pathways, highly consistent with the functions of all RASGs. Finally, we validated the AS ratio changes of five regulated alternative splicing events (RASEs) derived from important regulatory genes, including Mtmr3, Cdc42, Cd47, Fbln2, Vegfa, and Fhl2. CONCLUSION Our study emphasized the critical roles of the dysregulated AS profiles in MIRI development, investigated the potential functions of MIRI-associated RASGs, and identified regulatory RBPs involved in AS regulation. We propose that the identified RASEs and RBPs could serve as important regulators and potential therapeutic targets in MIRI treatment in the future.
Collapse
Affiliation(s)
- Ning Ma
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, P.R. China
| | - Hao Xu
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, P.R. China
| | - Weihua Zhang
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, P.R. China
| | - Xiaoke Sun
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, P.R. China
| | - Ruiming Guo
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, P.R. China
| | - Donghai Liu
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, P.R. China
| | - Liang Zhang
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, P.R. China
| | - Yang Liu
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, P.R. China
| | - Jian Zhang
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, P.R. China
| | - Chenhui Qiao
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, P.R. China
| | - Dong Chen
- Wuhan Ruixing Biotechnology Co., Ltd, Wuhan, 430206, Hubei, P.R. China
| | - Ailing Luo
- Wuhan Ruixing Biotechnology Co., Ltd, Wuhan, 430206, Hubei, P.R. China
| | - Jingyun Bai
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, P.R. China.
| |
Collapse
|
20
|
Orellana-Urzúa S, Briones-Valdivieso C, Chichiarelli S, Saso L, Rodrigo R. Potential Role of Natural Antioxidants in Countering Reperfusion Injury in Acute Myocardial Infarction and Ischemic Stroke. Antioxidants (Basel) 2023; 12:1760. [PMID: 37760064 PMCID: PMC10525378 DOI: 10.3390/antiox12091760] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 09/07/2023] [Accepted: 09/11/2023] [Indexed: 09/29/2023] Open
Abstract
Stroke and acute myocardial infarction are leading causes of mortality worldwide. The latter accounts for approximately 9 million deaths annually. In turn, ischemic stroke is a significant contributor to adult physical disability globally. While reperfusion is crucial for tissue recovery, it can paradoxically exacerbate damage through oxidative stress (OS), inflammation, and cell death. Therefore, it is imperative to explore diverse approaches aimed at minimizing ischemia/reperfusion injury to enhance clinical outcomes. OS primarily arises from an excessive generation of reactive oxygen species (ROS) and/or decreased endogenous antioxidant potential. Natural antioxidant compounds can counteract the injury mechanisms linked to ROS. While promising preclinical results, based on monotherapies, account for protective effects against tissue injury by ROS, translating these models into human applications has yielded controversial evidence. However, since the wide spectrum of antioxidants having diverse chemical characteristics offers varied biological actions on cell signaling pathways, multitherapy has emerged as a valuable therapeutic resource. Moreover, the combination of antioxidants in multitherapy holds significant potential for synergistic effects. This study was designed with the aim of providing an updated overview of natural antioxidants suitable for preventing myocardial and cerebral ischemia/reperfusion injuries.
Collapse
Affiliation(s)
- Sofía Orellana-Urzúa
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago 8380000, Chile;
| | | | - Silvia Chichiarelli
- Department of Biochemical Sciences “A. Rossi-Fanelli”, Sapienza University of Rome, 00185 Rome, Italy;
| | - Luciano Saso
- Department of Physiology and Pharmacology “Vittorio Erspamer”, Faculty of Pharmacy and Medicine, Sapienza University, P.le Aldo Moro 5, 00185 Rome, Italy;
| | - Ramón Rodrigo
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago 8380000, Chile;
| |
Collapse
|
21
|
Li W, Luo Y, Huang Z, Shen S, Dai C, Shen S, Qi X, Liang G, Luo W. Costunolide Protects Myocardium From Ischemia Reperfusion Injury by Inhibiting Oxidative Stress Through Nrf2/Keap1 Pathway Activation. J Cardiovasc Pharmacol 2023; 82:117-127. [PMID: 37000981 DOI: 10.1097/fjc.0000000000001422] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Accepted: 03/16/2023] [Indexed: 06/19/2023]
Abstract
ABSTRACT Costunolide (Cos) is a naturally occurring sesquiterpene lactone that exhibits antioxidative properties. In this study, we demonstrate the protective mechanism of Cos against ischemia/reperfusion (I/R)-induced myocardial injury. Cos significantly decreased levels of reactive oxygen species and ameliorated apoptosis of I/R cardiomyocytes both in vitro and in vivo. Further investigation revealed that Cos increased expression of the antioxidant proteins HO-1 and NQO-1 and decreased the Bax/Bcl-2 ratio, thus protecting cardiac cells. NF-E2-related factor 2 (Nrf2) silencing significantly attenuated the protective effects of Cos in tert-butyl hydroperoxide (TBHP)-treated H9C2 cells. Additionally, Cos significantly intensified the I/R- or TBHP-induced dissociation of the Kelch-like ECH-associated protein 1 (Keap1)/Nrf2 complex both in vitro and in vivo. These results suggest that activation of Nrf2/Keap1 using Cos may be a therapeutic strategy for myocardial I/R injury.
Collapse
Affiliation(s)
- Weixin Li
- Medical Research Center, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
- Department of Cardiology, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yue Luo
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zhuqi Huang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
- Department of Cardiology, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Siyuan Shen
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
- Department of Cardiology, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Chengyi Dai
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
- Department of Cardiology, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Sirui Shen
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
- Department of Cardiology, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xiaoxiao Qi
- Department of Pharmacy, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; and
| | - Guang Liang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
- School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Wu Luo
- Medical Research Center, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|