1
|
Gu Z, Liu X, Qi Z, Fang Z, Jiang Y, Huang Y, Wang Y, Wu L, Yang Y. An antioxidant nanozyme for targeted cardiac fibrosis therapy post myocardial infarction. J Nanobiotechnology 2024; 22:760. [PMID: 39696342 DOI: 10.1186/s12951-024-03047-6] [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: 07/24/2024] [Accepted: 11/29/2024] [Indexed: 12/20/2024] Open
Abstract
The excessive release of reactive oxygen species (ROS) after myocardial infarction (MI) disrupts the natural healing process, leading to cardiac fibrosis and compromising patient prognosis. However, the clinical application of many antioxidant drugs for MI treatment is hindered by their poor antioxidant efficacy and inability to specifically target the heart. Here we developed a tannic acid-modified MnO2 nanozyme (named MnO2@TA), which can achieve cardiac targeting to inhibit post-MI fibrosis and enhance cardiac function. Specifically, the MnO2@TA nanozyme, endowed with superoxide dismutase (SOD) and catalase (CAT) activities, effectively scavenges ROS, suppressing fibroblast activation and mitigating cardiac fibrosis without affecting cardiac repair. Notably, the incorporation of TA improves the nanozyme's affinity for the elastin and collagen-rich extracellular matrix in cardiac tissues, significantly increasing its retention and uptake within the heart and thereby enhancing its anti-fibrotic efficacy. In a murine myocardial infarction model, MnO2@TA demonstrates remarkable cardiac protection and safety, significantly improving cardiac function while attenuating cardiac fibrosis. This study presents a valuable reference for clinical research aimed at inhibiting cardiac fibrosis and advancing myocardial infarction treatments.
Collapse
Affiliation(s)
- Ziyi Gu
- Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Department of Cardiovascular Surgery, School of Medicine, Renji Hospital, Institute of Molecular Medicine (IMM), Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Xueliang Liu
- Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Department of Cardiovascular Surgery, School of Medicine, Renji Hospital, Institute of Molecular Medicine (IMM), Shanghai Jiao Tong University, Shanghai, 200240, China
- Punan Branch of Renji Hospital, Shanhai Jiaotong University School of Medicine, Shanghai, 200125, China
| | - Zhen Qi
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Zhou Fang
- Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Department of Cardiovascular Surgery, School of Medicine, Renji Hospital, Institute of Molecular Medicine (IMM), Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Yiting Jiang
- Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Department of Cardiovascular Surgery, School of Medicine, Renji Hospital, Institute of Molecular Medicine (IMM), Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Yuting Huang
- Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Department of Cardiovascular Surgery, School of Medicine, Renji Hospital, Institute of Molecular Medicine (IMM), Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Yongyi Wang
- Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Department of Cardiovascular Surgery, School of Medicine, Renji Hospital, Institute of Molecular Medicine (IMM), Shanghai Jiao Tong University, Shanghai, 200240, China.
| | - Lianming Wu
- Department of Radiology, School of Medicine, Renji Hospital, Shanghai Jiao Tong University, Shanghai, 200240, China.
| | - Yu Yang
- Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Department of Cardiovascular Surgery, School of Medicine, Renji Hospital, Institute of Molecular Medicine (IMM), Shanghai Jiao Tong University, Shanghai, 200240, China.
| |
Collapse
|
2
|
Xie B, Li J, Lou Y, Chen Q, Yang Y, Zhang R, Liu Z, He L, Cheng Y. Reprogramming macrophage metabolism following myocardial infarction: A neglected piece of a therapeutic opportunity. Int Immunopharmacol 2024; 142:113019. [PMID: 39217876 DOI: 10.1016/j.intimp.2024.113019] [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: 05/11/2024] [Revised: 08/15/2024] [Accepted: 08/22/2024] [Indexed: 09/04/2024]
Abstract
Given the global prevalence of myocardial infarction (MI) as the leading cause of mortality, there is an urgent need to devise novel strategies that target reducing infarct size, accelerating cardiac tissue repair, and preventing detrimental left ventricular (LV) remodeling. Macrophages, as a predominant type of innate immune cells, undergo metabolic reprogramming following MI, resulting in alterations in function and phenotype that significantly impact the progression of MI size and LV remodeling. This article aimed to delineate the characteristics of macrophage metabolites during reprogramming in MI and elucidate their targets and functions in cardioprotection. Furthermore, we summarize the currently proposed regulatory mechanisms of macrophage metabolic reprogramming and identify the regulators derived from endogenous products and natural small molecules. Finally, we discussed the challenges of macrophage metabolic reprogramming in the treatment of MI, with the goal of inspiring further fundamental and clinical research into reprogramming macrophage metabolism and validating its potential therapeutic targets for MI.
Collapse
Affiliation(s)
- Baoping Xie
- State Key Laboratory of Traditional Chinese Medicine Syndrome, Key Laboratory for Translational Cancer Research of Chinese Medicine, Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China; Chinese Medicine Guangdong Laboratory, Guangdong, Hengqin, China; Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases of Ministry of Education, Jiangxi Provincial Key Laboratory of Tissue Engineering, Gannan Medical University, Ganzhou 341000, China
| | - Jiahua Li
- State Key Laboratory of Traditional Chinese Medicine Syndrome, Key Laboratory for Translational Cancer Research of Chinese Medicine, Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China; Chinese Medicine Guangdong Laboratory, Guangdong, Hengqin, China
| | - Yanmei Lou
- State Key Laboratory of Traditional Chinese Medicine Syndrome, Key Laboratory for Translational Cancer Research of Chinese Medicine, Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China; Chinese Medicine Guangdong Laboratory, Guangdong, Hengqin, China
| | - Qi Chen
- State Key Laboratory of Traditional Chinese Medicine Syndrome, Key Laboratory for Translational Cancer Research of Chinese Medicine, Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China; Chinese Medicine Guangdong Laboratory, Guangdong, Hengqin, China
| | - Ying Yang
- State Key Laboratory of Traditional Chinese Medicine Syndrome, Key Laboratory for Translational Cancer Research of Chinese Medicine, Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China; Chinese Medicine Guangdong Laboratory, Guangdong, Hengqin, China
| | - Rong Zhang
- State Key Laboratory of Traditional Chinese Medicine Syndrome, Key Laboratory for Translational Cancer Research of Chinese Medicine, Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China; Chinese Medicine Guangdong Laboratory, Guangdong, Hengqin, China
| | - Zhongqiu Liu
- State Key Laboratory of Traditional Chinese Medicine Syndrome, Key Laboratory for Translational Cancer Research of Chinese Medicine, Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China; Chinese Medicine Guangdong Laboratory, Guangdong, Hengqin, China.
| | - Liu He
- Department of Endocrinology, Guangdong Provincial Hospital of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong 510006, China.
| | - Yuanyuan Cheng
- State Key Laboratory of Traditional Chinese Medicine Syndrome, Key Laboratory for Translational Cancer Research of Chinese Medicine, Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China; Chinese Medicine Guangdong Laboratory, Guangdong, Hengqin, China.
| |
Collapse
|
3
|
Kong C, Guo Z, Teng T, Yao Q, Yu J, Wang M, Ma Y, Wang P, Tang Q. Electroactive Nanomaterials for the Prevention and Treatment of Heart Failure: From Materials and Mechanisms to Applications. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2406206. [PMID: 39268781 DOI: 10.1002/smll.202406206] [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: 07/31/2024] [Revised: 09/02/2024] [Indexed: 09/15/2024]
Abstract
Heart failure (HF) represents a cardiovascular disease that significantly threatens global well-being and quality of life. Electroactive nanomaterials, characterized by their distinctive physical and chemical properties, emerge as promising candidates for HF prevention and management. This review comprehensively examines electroactive nanomaterials and their applications in HF intervention. It presents the definition, classification, and intrinsic characteristics of conductive, piezoelectric, and triboelectric nanomaterials, emphasizing their mechanical robustness, electrical conductivity, and piezoelectric coefficients. The review elucidates their applications and mechanisms: 1) early detection and diagnosis, employing nanomaterial-based sensors for real-time cardiac health monitoring; 2) cardiac tissue repair and regeneration, providing mechanical, chemical, and electrical stimuli for tissue restoration; 3) localized administration of bioactive biomolecules, genes, or pharmacotherapeutic agents, using nanomaterials as advanced drug delivery systems; and 4) electrical stimulation therapies, leveraging their properties for innovative pacemaker and neurostimulation technologies. Challenges in clinical translation, such as biocompatibility, stability, and scalability, are discussed, along with future prospects and potential innovations, including multifunctional and stimuli-responsive nanomaterials for precise HF therapies. This review encapsulates current research and future directions concerning the use of electroactive nanomaterials in HF prevention and management, highlighting their potential to innovating in cardiovascular medicine.
Collapse
Affiliation(s)
- Chunyan Kong
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, P. R. China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, P. R. China
| | - Zhen Guo
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, P. R. China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, P. R. China
| | - Teng Teng
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, P. R. China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, P. R. China
| | - Qi Yao
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, P. R. China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, P. R. China
| | - Jiabin Yu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, P. R. China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, P. R. China
| | - Mingyu Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, P. R. China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, P. R. China
| | - Yulan Ma
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, P. R. China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, P. R. China
| | - Pan Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, P. R. China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, P. R. China
| | - Qizhu Tang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, P. R. China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, P. R. China
| |
Collapse
|
4
|
Li S, Li F, Wang Y, Li W, Wu J, Hu X, Tang T, Liu X. Multiple delivery strategies of nanocarriers for myocardial ischemia-reperfusion injury: current strategies and future prospective. Drug Deliv 2024; 31:2298514. [PMID: 38147501 PMCID: PMC10763895 DOI: 10.1080/10717544.2023.2298514] [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: 08/02/2023] [Accepted: 11/30/2023] [Indexed: 12/28/2023] Open
Abstract
Acute myocardial infarction, characterized by high morbidity and mortality, has now become a serious health hazard for human beings. Conventional surgical interventions to restore blood flow can rapidly relieve acute myocardial ischemia, but the ensuing myocardial ischemia-reperfusion injury (MI/RI) and subsequent heart failure have become medical challenges that researchers have been trying to overcome. The pathogenesis of MI/RI involves several mechanisms, including overproduction of reactive oxygen species, abnormal mitochondrial function, calcium overload, and other factors that induce cell death and inflammatory responses. These mechanisms have led to the exploration of antioxidant and inflammation-modulating therapies, as well as the development of myocardial protective factors and stem cell therapies. However, the short half-life, low bioavailability, and lack of targeting of these drugs that modulate these pathological mechanisms, combined with liver and spleen sequestration and continuous washout of blood flow from myocardial sites, severely compromise the expected efficacy of clinical drugs. To address these issues, employing conventional nanocarriers and integrating them with contemporary biomimetic nanocarriers, which rely on passive targeting and active targeting through precise modifications, can effectively prolong the duration of therapeutic agents within the body, enhance their bioavailability, and augment their retention at the injured myocardium. Consequently, these approaches significantly enhance therapeutic effectiveness while minimizing toxic side effects. This article reviews current drug delivery systems used for MI/RI, aiming to offer a fresh perspective on treating this disease.
Collapse
Affiliation(s)
- Shengnan Li
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Institution of Clinical Pharmacy, Central South University, Changsha, China
| | - Fengmei Li
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Institution of Clinical Pharmacy, Central South University, Changsha, China
| | - Yan Wang
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Institution of Clinical Pharmacy, Central South University, Changsha, China
| | - Wenqun Li
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Institution of Clinical Pharmacy, Central South University, Changsha, China
| | - Junyong Wu
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Institution of Clinical Pharmacy, Central South University, Changsha, China
| | - Xiongbin Hu
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Institution of Clinical Pharmacy, Central South University, Changsha, China
| | - Tiantian Tang
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Institution of Clinical Pharmacy, Central South University, Changsha, China
| | - Xinyi Liu
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Institution of Clinical Pharmacy, Central South University, Changsha, China
| |
Collapse
|
5
|
Liu J, Xi Z, Fan C, Mei Y, Zhao J, Jiang Y, Zhao M, Xu L. Hydrogels for Nucleic Acid Drugs Delivery. Adv Healthc Mater 2024; 13:e2401895. [PMID: 39152918 DOI: 10.1002/adhm.202401895] [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: 07/05/2024] [Indexed: 08/19/2024]
Abstract
Nucleic acid drugs are one of the hot spots in the field of biomedicine in recent years, and play a crucial role in the treatment of many diseases. However, its low stability and difficulty in target drug delivery are the bottlenecks restricting its application. Hydrogels are proven to be promising for improving the stability of nucleic acid drugs, reducing the adverse effects of rapid degradation, sudden release, and unnecessary diffusion of nucleic acid drugs. In this review, the strategies of loading nucleic acid drugs in hydrogels are summarized for various biomedical research, and classify the mechanism principles of these strategies, including electrostatic binding, hydrogen bond based binding, hydrophobic binding, covalent bond based binding and indirect binding using various carriers. In addition, this review also describes the release strategies of nucleic acid drugs, including photostimulation-based release, enzyme-responsive release, pH-responsive release, and temperature-responsive release. Finally, the applications and future research directions of hydrogels for delivering nucleic acid drugs in the field of medicine are discussed.
Collapse
Affiliation(s)
- Jiaping Liu
- School of Pharmacy, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang, 110016, P. R. China
| | - Ziyue Xi
- School of Pharmacy, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang, 110016, P. R. China
| | - Chuanyong Fan
- School of Pharmacy, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang, 110016, P. R. China
| | - Yihua Mei
- School of Pharmacy, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang, 110016, P. R. China
| | - Jiale Zhao
- School of Pharmacy, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang, 110016, P. R. China
| | - Yingying Jiang
- School of Pharmacy, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang, 110016, P. R. China
| | - Ming Zhao
- School of Pharmacy, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang, 110016, P. R. China
| | - Lu Xu
- School of Pharmacy, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang, 110016, P. R. China
| |
Collapse
|
6
|
Athmuri DN, Bhattacharyya J, Bhatnagar N, Shiekh PA. Alleviating hypoxia and oxidative stress for treatment of cardiovascular diseases: a biomaterials perspective. J Mater Chem B 2024; 12:10490-10515. [PMID: 39302443 DOI: 10.1039/d4tb01126k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/22/2024]
Abstract
A state of hypoxia (lack of oxygen) persists in the initial and later phases of healing in cardiovascular diseases, which can alter the tissue's repair or regeneration, ultimately affecting the structure and functionality of the related organ. Consequently, this results in a cascade of events, leading to metabolic stress and the production of reactive oxygen species (ROS) and autophagy. This unwanted situation not only limits the oxygen supply to the needy tissues but also creates an inflammatory state, limiting the exchange of nutrients and other supplements. Consequently, biomaterials have gained considerable attention to alleviate hypoxia and oxidative stress in cardiovascular diseases. Numerous oxygen releasing and antioxidant biomaterials have been developed and proven to alleviate hypoxia and oxidative stress. This review article summarizes the mechanisms involved in cardiovascular pathologies due to hypoxia and oxidative stress, as well as the treatment modalities currently in practice. The applications, benefits and possible shortcomings of these approaches have been discussed. Additionally, the review explores the role of novel biomaterials in combating the limitations of existing approaches, primarily focusing on the development of oxygen-releasing and antioxidant biomaterials for cardiac repair and regeneration. It also directs attention to various other potential applications with critical insights for further advancement in this area.
Collapse
Affiliation(s)
- Durga Nandini Athmuri
- SMART Lab, Centre for Biomedical Engineering, Indian Institute of Technology Delhi, Hauz Khas, New Delhi-110016, India.
| | - Jayanta Bhattacharyya
- Bio-therapeutics Lab, Centre for Biomedical Engineering, Indian Institute of Technology Delhi, Hauz Khas, New Delhi-110016, India
| | - Naresh Bhatnagar
- Department of Mechanical Engineering, Indian Institute of Technology Delhi, Hauz Khas, New Delhi-110016, India
| | - Parvaiz Ahmad Shiekh
- SMART Lab, Centre for Biomedical Engineering, Indian Institute of Technology Delhi, Hauz Khas, New Delhi-110016, India.
| |
Collapse
|
7
|
Tian M, Dong B, Li W, Wang L, Yu H. Applications of Novel Microscale and Nanoscale Materials for Theranostics: From Design to Clinical Translation. Pharmaceutics 2024; 16:1339. [PMID: 39458667 PMCID: PMC11511338 DOI: 10.3390/pharmaceutics16101339] [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: 08/22/2024] [Revised: 10/12/2024] [Accepted: 10/17/2024] [Indexed: 10/28/2024] Open
Abstract
The growing global prevalence of chronic diseases has highlighted the limitations of conventional drug delivery methods, which often suffer from non-specific distribution, systemic toxicity, and poor bioavailability. Microscale and nanoscale materials have emerged as innovative solutions, offering enhanced targeting, controlled release, and the convergence of therapeutic and diagnostic functions, referred to as theranostics. This review explores the design principles, mechanisms of action, and clinical applications of various novel micro- and nanomaterials in diseases such as cancer, cardiovascular disorders, and infectious diseases. These materials enable real-time monitoring of therapeutic responses and facilitate precision medicine approaches. Additionally, this paper addresses the significant challenges hindering clinical translation, including biocompatibility, potential toxicity, and regulatory issues. Ongoing clinical trials demonstrate the potential of nanomaterials in theranostic applications, but further research is needed to overcome the barriers to widespread clinical adoption. This work aims to contribute to the acceleration of integrating nanomedicine into clinical practice, ultimately enhancing the efficacy and safety of therapeutic interventions.
Collapse
Affiliation(s)
- Mengxiang Tian
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310058, China; (M.T.); (B.D.); (W.L.)
- Provincial Key Laboratory of Precise Diagnosis and Treatment of Abdominal Infection, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, China
| | - Bingzhi Dong
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310058, China; (M.T.); (B.D.); (W.L.)
- Provincial Key Laboratory of Precise Diagnosis and Treatment of Abdominal Infection, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, China
| | - Weiqi Li
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310058, China; (M.T.); (B.D.); (W.L.)
- Provincial Key Laboratory of Precise Diagnosis and Treatment of Abdominal Infection, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, China
| | - Liying Wang
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310058, China; (M.T.); (B.D.); (W.L.)
- Provincial Key Laboratory of Precise Diagnosis and Treatment of Abdominal Infection, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, China
| | - Hong Yu
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310058, China; (M.T.); (B.D.); (W.L.)
- Provincial Key Laboratory of Precise Diagnosis and Treatment of Abdominal Infection, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, China
| |
Collapse
|
8
|
Naziri Z, Rahimlou M, Rezaei M, Tabrizi R, Nasr M, Motazedian M, Kardeh S. High dietary antioxidant intake linked to lower risk of myocardial infarction: a nested case-control study. BMC Cardiovasc Disord 2024; 24:485. [PMID: 39261811 PMCID: PMC11391677 DOI: 10.1186/s12872-024-04158-6] [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: 06/30/2024] [Accepted: 09/02/2024] [Indexed: 09/13/2024] Open
Abstract
BACKGROUND In developing nations, myocardial infarction (MI) remains a significant contributor to deaths from sudden cardiac arrest, with diet playing a key role in its incidence through oxidative stress mechanisms. Although the connection between the Dietary Antioxidant Index (DAI) and cardiovascular diseases has been demonstrated in some studies, the relationship between DAI and MI has not been extensively explored. Therefore, this research aims to investigate this association. METHODS We conducted a nested case-control study involving 156 MI cases and 312 healthy controls, utilizing data from the Fasa Adults Cohort Study (FACS), a population-based study of individuals aged 35-70 residing in Fasa, Iran, with 11,097 participants included at baseline. The DAI was determined by normalizing the intake values of six dietary vitamins and minerals, adjusting by subtracting the global mean, and then dividing by the global standard deviation. MI diagnosis was established by an experienced cardiologist using electronic medical records. Conditional logistic regression was employed to examine the association between DAI and MI. RESULTS There were no significant differences between the case and control groups in terms of age (P = 0.96), gender distribution (P = 0.98), and education level (P = 0.38). In a multiple conditional logistic regression analysis, after adjusting for key variables-including body mass index (BMI), smoking status, education level, and serum levels of triglycerides (TG), low-density lipoprotein (LDL), high-density lipoprotein (HDL), total cholesterol (TC), fasting blood sugar (FBS), saturated fatty acids (SFA), and polyunsaturated fatty acids (PUFA)-an inverse association was found between DAI and the risk of myocardial infarction (MI) [adjusted Odds Ratio (Adj OR) = 0.88, 95% Confidence Interval (CI): 0.85-0.92; P < 0.001]. CONCLUSIONS This study highlights the crucial role of the DAI in reducing the risk of myocardial infarction. Promoting diets rich in antioxidants presents a straightforward and effective strategy for MI prevention and the promotion of cardiovascular health, underscoring the novelty and significance of this research in dietary approaches to disease prevention.
Collapse
Affiliation(s)
- Zahra Naziri
- Student Research Committee, Fasa University of Medical Sciences, Fasa, Iran
- USERN Office, Fasa University of Medical Sciences, Fasa, Iran
| | - Mehran Rahimlou
- Department of Nutrition, School of Public Health, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Mehdi Rezaei
- Department of Cardiology, Fars Society of Internal Medicine, Fars-Iranian Heart Association, Shiraz, Iran
| | - Reza Tabrizi
- Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran.
| | | | - Mohammadreza Motazedian
- Clinical Research Development Unit, Valiasr Hospital, Fasa University of Medical Sciences, Fasa, Iran
| | - Sina Kardeh
- Department of Medicine, University of British Columbia, Vancouver, BC, V6T1Z3, Canada
| |
Collapse
|
9
|
Hong X, Tian G, Dai B, Zhou X, Gao Y, Zhu L, Liu H, Zhu Q, Zhang L, Zhu Y, Ren D, Guo C, Nan J, Liu X, Wang J, Ren T. Copper-loaded Milk-Protein Derived Microgel Preserves Cardiac Metabolic Homeostasis After Myocardial Infarction. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2401527. [PMID: 39007192 PMCID: PMC11425262 DOI: 10.1002/advs.202401527] [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: 02/12/2024] [Revised: 06/11/2024] [Indexed: 07/16/2024]
Abstract
Myocardial Infarction (MI) is a leading cause of death worldwide. Metabolic modulation is a promising therapeutic approach to prevent adverse remodeling after MI. However, whether material-derived cues can treat MI through metabolic regulation is mainly unexplored. Herein, a Cu2+ loaded casein microgel (CuCMG) aiming to rescue the pathological intramyocardial metabolism for MI amelioration is developed. Cu2+ is an important ion factor involved in metabolic pathways, and intracardiac copper drain is observed after MI. It is thus speculated that intramyocardial supplementation of Cu2+ can rescue myocardial metabolism. Casein, a milk-derived protein, is screened out as Cu2+ carrier through molecular-docking based on Cu2+ loading capacity and accessibility. CuCMGs notably attenuate MI-induced cardiac dysfunction and maladaptive remodeling, accompanied by increased angiogenesis. The results from unbiased transcriptome profiling and oxidative phosphorylation analyses support the hypothesis that CuCMG prominently rescued the metabolic homeostasis of myocardium after MI. These findings enhance the understanding of the design and application of metabolic-modulating biomaterials for ischemic cardiomyopathy therapy.
Collapse
Affiliation(s)
- Xiaoqian Hong
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
- State Key Laboratory of Transvascular Implantation Devices, Heart Regeneration and Repair Key Laboratory Zhejiang Province, Hangzhou, 310009, China
| | - Geer Tian
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
- State Key Laboratory of Transvascular Implantation Devices, Heart Regeneration and Repair Key Laboratory Zhejiang Province, Hangzhou, 310009, China
- Binjiang Institute of Zhejiang University, Hangzhou, 310053, China
| | - Binyao Dai
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Xuhao Zhou
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
- State Key Laboratory of Transvascular Implantation Devices, Heart Regeneration and Repair Key Laboratory Zhejiang Province, Hangzhou, 310009, China
| | - Ying Gao
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
- State Key Laboratory of Transvascular Implantation Devices, Heart Regeneration and Repair Key Laboratory Zhejiang Province, Hangzhou, 310009, China
| | - Lianlian Zhu
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
- State Key Laboratory of Transvascular Implantation Devices, Heart Regeneration and Repair Key Laboratory Zhejiang Province, Hangzhou, 310009, China
| | - Haoran Liu
- School of Engineering, Westlake University, Hangzhou, 310023, China
| | - Qinchao Zhu
- Institute of Dairy Science, College of Animal Sciences, Zhejiang University, Hangzhou, 310027, China
| | - Liwen Zhang
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Yang Zhu
- State Key Laboratory of Transvascular Implantation Devices, Heart Regeneration and Repair Key Laboratory Zhejiang Province, Hangzhou, 310009, China
- Binjiang Institute of Zhejiang University, Hangzhou, 310053, China
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Daxi Ren
- Institute of Dairy Science, College of Animal Sciences, Zhejiang University, Hangzhou, 310027, China
| | - Chengchen Guo
- School of Engineering, Westlake University, Hangzhou, 310023, China
| | - Jinliang Nan
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
- State Key Laboratory of Transvascular Implantation Devices, Heart Regeneration and Repair Key Laboratory Zhejiang Province, Hangzhou, 310009, China
| | - Xianbao Liu
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
- State Key Laboratory of Transvascular Implantation Devices, Heart Regeneration and Repair Key Laboratory Zhejiang Province, Hangzhou, 310009, China
| | - Jian'an Wang
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
- State Key Laboratory of Transvascular Implantation Devices, Heart Regeneration and Repair Key Laboratory Zhejiang Province, Hangzhou, 310009, China
| | - Tanchen Ren
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
- State Key Laboratory of Transvascular Implantation Devices, Heart Regeneration and Repair Key Laboratory Zhejiang Province, Hangzhou, 310009, China
| |
Collapse
|
10
|
Sai Priya T, Ramalingam V, Suresh Babu K. Natural products: A potential immunomodulators against inflammatory-related diseases. Inflammopharmacology 2024:10.1007/s10787-024-01562-4. [PMID: 39196458 DOI: 10.1007/s10787-024-01562-4] [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: 08/12/2024] [Accepted: 08/14/2024] [Indexed: 08/29/2024]
Abstract
The incidence and prevalence of inflammatory-related diseases (IRDs) are increasing worldwide. Current approved treatments for IRDs in the clinic are combat against inhibiting the pro-inflammatory cytokines. Though significant development in the treatment in the IRDs has been achieved, the severe side effects and inefficiency of currently practicing treatments are endless challenge. Drug discovery from natural sources is efficacious over a resurgence and also natural products are leading than the synthetic molecules in both clinical trials and market. The use of natural products against IRDs is a conventional therapeutic approach since it is a reservoir of unique structural chemistry, accessibility and bioactivities with reduced side effects and low toxicity. In this review, we discuss the cause of IRDs, treatment of options for IRDs and the impact and adverse effects of currently practicing clinical drugs. As well, the significant role of natural products against various IRDs, the limitations in the clinical development of natural products and thus pave the way for development of natural products as immunomodulators against IRDs are also discussed.
Collapse
Affiliation(s)
- Telukuntla Sai Priya
- Department of Natural Products & Medicinal Chemistry, CSIR-Indian Institute of Chemical Technology, Hyderabad, 500 007, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Vaikundamoorthy Ramalingam
- Department of Natural Products & Medicinal Chemistry, CSIR-Indian Institute of Chemical Technology, Hyderabad, 500 007, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Katragadda Suresh Babu
- Department of Natural Products & Medicinal Chemistry, CSIR-Indian Institute of Chemical Technology, Hyderabad, 500 007, India.
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India.
| |
Collapse
|
11
|
Han D, Wang F, Shen D. Nanomedicines as Guardians of the Heart: Unleashing the Power of Antioxidants to Alleviate Myocardial Ischemic Injury. Theranostics 2024; 14:5336-5370. [PMID: 39267789 PMCID: PMC11388064 DOI: 10.7150/thno.99961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Accepted: 08/16/2024] [Indexed: 09/15/2024] Open
Abstract
Ischemic heart disease (IHD) is increasingly recognized as a significant cardiovascular disease with a growing global incidence. Interventions targeting the oxidative microenvironment have long been pivotal in therapeutic strategies. However, many antioxidant drugs face limitations due to pharmacokinetic and delivery challenges, such as short half-life, poor stability, low bioavailability, and significant side effects. Fortunately, nanotherapies exhibit considerable potential in addressing IHD. Nanomedicines offer advantages such as passive/active targeting, prolonged circulation time, enhanced bioavailability, and diverse carrier options. This comprehensive review explores the advancements in nanomedicines for mitigating IHD through oxidative stress regulation, providing an extensive overview for researchers in the field of antioxidant nanomedicines. By inspiring further research, this study aims to accelerate the development of novel therapies for myocardial injury.
Collapse
Affiliation(s)
- Dongjian Han
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
- Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, China
| | - Fuhang Wang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
- Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, China
| | - Deliang Shen
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
- Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, China
| |
Collapse
|
12
|
Song L, Yuan X, Zhao Z, Wang P, Wu W, Wang J. A Biomimetic Nanocarrier Strategy Targets Ferroptosis and Efferocytosis During Myocardial Infarction. Int J Nanomedicine 2024; 19:8253-8270. [PMID: 39157734 PMCID: PMC11330256 DOI: 10.2147/ijn.s461212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 08/10/2024] [Indexed: 08/20/2024] Open
Abstract
Background Myocardial infarction (MI) is characterized by irreversible cardiomyocyte death resulting from an inadequate supply of oxygenated blood to the myocardium. Recent studies have indicated that ferroptosis, a form of regulated cell death, exacerbates myocardial injury during MI. Concurrently, the upregulation of CD47 on the surface of damaged myocardium following MI impairs the clearance of dead cells by macrophages, thereby hindering efferocytosis. In this context, simultaneously inhibiting ferroptosis and enhancing efferocytosis may represent a promising strategy to mitigate myocardial damage post-MI. Methods In this study, we engineered platelet membrane-coated hollow mesoporous silicon nanoparticles (HMSN) to serve as a drug delivery system, encapsulating ferroptosis inhibitor, Ferrostatin-1, along with an anti-CD47 antibody. We aimed to assess the potential of these nanoparticles (designated as Fer-aCD47@PHMSN) to specifically target the site of MI and evaluate their efficacy in reducing cardiomyocyte death and inflammation. Results The platelet membrane coating on the nanoparticles significantly enhanced their ability to successfully target the site of myocardial infarction (MI). Our findings demonstrate that treatment with Fer-aCD47@PHMSN resulted in a 38.5% reduction in cardiomyocyte ferroptosis under hypoxia, indicated by decreased lipid peroxidation and increased in vitro. Additionally, Fer-aCD47@PHMSN improved cardiomyocyte efferocytosis by approximately 15% in vitro. In MI mice treated with Fer-aCD47@PHMSN, we observed a substantial reduction in cardiomyocyte death (nearly 30%), decreased inflammation, and significant improvement in cardiac function. Conclusion Our results demonstrated that the cooperation between the two agents induced anti-ferroptosis effects and enhanced dead cardiomyocyte clearance by macrophage as well as anti-inflammation effects. Thus, our nanoparticle Fer-aCD47@PHMSN provides a new therapeutic strategy for targeted therapy of MI.
Collapse
Affiliation(s)
- Lin Song
- School of Basic Medicine, Qingdao University, Qingdao, 266071, People’s Republic of China
| | - Xiaosu Yuan
- School of Basic Medicine, Qingdao University, Qingdao, 266071, People’s Republic of China
| | - Zhonghao Zhao
- School of Basic Medicine, Qingdao University, Qingdao, 266071, People’s Republic of China
| | - Peiyan Wang
- School of Basic Medicine, Qingdao University, Qingdao, 266071, People’s Republic of China
| | - Weiwei Wu
- School of Basic Medicine, Qingdao University, Qingdao, 266071, People’s Republic of China
| | - Jianxun Wang
- School of Basic Medicine, Qingdao University, Qingdao, 266071, People’s Republic of China
| |
Collapse
|
13
|
Wu Z, Li W, Wang S, Zheng Z. Role of deubiquitinase USP47 in cardiac function alleviation and anti-inflammatory immunity after myocardial infarction by regulating NLRP3 inflammasome-mediated pyroptotic signal pathways. Int Immunopharmacol 2024; 136:112346. [PMID: 38850785 DOI: 10.1016/j.intimp.2024.112346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 05/22/2024] [Accepted: 05/24/2024] [Indexed: 06/10/2024]
Abstract
Myocardial infarction (MI) is an event of heart attack due to the formation of plaques in the interior walls of the arteries. This study is conducted to explore the role of ubiquitin-specific peptidase 47 (USP47) in cardiac function and inflammatory immunity. MI mouse models were established, followed by an appraisal of cardiac functions, infarct size, pathological changes, and USP47 and NLRP3 levels. MI cell models were established in HL-1 cells using anoxia. Levels of cardiac function-associated proteins, USP7, interferon regulatory factor 1 (IRF1), platelet factor-4 (CXCL4), pyroptotic factors, and neutrophil extracellular traps (NETs) were determined. The bindings of IRF1 to USP47 and the CXCL4 promoter and the ubiquitination of IRF1 were analyzed. USP47 was upregulated in myocardial tissues of MI mice. USP47 inhibition alleviated cardiac functions, and decreased infarct size, pro-inflammatory cytokines, NETs, NLRP3, and pyroptosis. The ubiquitination and expression levels of IRF1 were increased by silencing USP47, and IRF1 bound to the CXCL4 promoter to promote CXCL4. Overexpression of IRF1 or CXCL4 in vitro and injection of Nigericin in vivo reversed the effect of silencing USP47 on alleviating pyroptosis and cardiac functions. Collectively, USP47 stabilized IRF1 and promoted CXCL4, further promoting pyroptosis, impairing cardiac functions, and aggravating immune inflammation through NLRP3 pathways.
Collapse
Affiliation(s)
- Zheng Wu
- Center for Coronary Artery Disease, Beijing Anzhen Hospital, Capital Medical University, Beijing, China.
| | - Wenzheng Li
- Center for Coronary Artery Disease, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Shaoping Wang
- Center for Coronary Artery Disease, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Ze Zheng
- Center for Coronary Artery Disease, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
14
|
Singhal R, Sarangi MK, Rath G. Injectable Hydrogels: A Paradigm Tailored with Design, Characterization, and Multifaceted Approaches. Macromol Biosci 2024; 24:e2400049. [PMID: 38577905 DOI: 10.1002/mabi.202400049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 03/22/2024] [Indexed: 04/06/2024]
Abstract
Biomaterials denoting self-healing and versatile structural integrity are highly curious in the biomedicine segment. The injectable and/or printable 3D printing technology is explored in a few decades back, which can alter their dimensions temporarily under shear stress, showing potential healing/recovery tendency with patient-specific intervention toward the development of personalized medicine. Thus, self-healing injectable hydrogels (IHs) are stunning toward developing a paradigm for tissue regeneration. This review comprises the designing of IHs, rheological characterization and stability, several benchmark consequences for self-healing IHs, their translation into tissue regeneration of specific types, applications of IHs in biomedical such as anticancer and immunomodulation, wound healing and tissue/bone regeneration, antimicrobial potentials, drugs, gene and vaccine delivery, ocular delivery, 3D printing, cosmeceuticals, and photothermal therapy as well as in other allied avenues like agriculture, aerospace, electronic/electrical industries, coating approaches, patents associated with therapeutic/nontherapeutic avenues, and numerous futuristic challenges and solutions.
Collapse
Affiliation(s)
- Rishika Singhal
- Department of Pharmaceutics, Amity Institute of Pharmacy, Amity University, Malhaur Railway Station Road, Gomti Nagar, Lucknow, Uttar Pradesh, 201313, India
| | - Manoj Kumar Sarangi
- Department of Pharmaceutics, Amity Institute of Pharmacy, Amity University, Malhaur Railway Station Road, Gomti Nagar, Lucknow, Uttar Pradesh, 201313, India
| | - Goutam Rath
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Siksha 'O' Anusandhan University, Bhubaneswar, Odisha, 751030, India
| |
Collapse
|
15
|
Song L, Jia K, Yang F, Wang J. Advanced Nanomedicine Approaches for Myocardial Infarction Treatment. Int J Nanomedicine 2024; 19:6399-6425. [PMID: 38952676 PMCID: PMC11215519 DOI: 10.2147/ijn.s467219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 06/04/2024] [Indexed: 07/03/2024] Open
Abstract
Myocardial infarction, usually caused by the rupture of atherosclerotic plaque, leads to irreversible ischemic cardiomyocyte death within hours followed by impaired cardiac performance or even heart failure. Current interventional reperfusion strategies for myocardial infarction still face high mortality with the development of heart failure. Nanomaterial-based therapy has made great progress in reducing infarct size and promoting cardiac repair after MI, although most studies are preclinical trials. This review focuses primarily on recent progress (2016-now) in the development of various nanomedicines in the treatment of myocardial infarction. We summarize these applications with the strategy of mechanism including anti-cardiomyocyte death strategy, activation of neovascularization, antioxidants strategy, immunomodulation, anti-cardiac remodeling, and cardiac repair.
Collapse
Affiliation(s)
- Lin Song
- School of Basic Medicine, Qingdao University, Qingdao, People’s Republic of China
| | - Kangwei Jia
- School of Basic Medicine, Qingdao University, Qingdao, People’s Republic of China
| | - Fuqing Yang
- School of Basic Medicine, Qingdao University, Qingdao, People’s Republic of China
| | - Jianxun Wang
- School of Basic Medicine, Qingdao University, Qingdao, People’s Republic of China
| |
Collapse
|
16
|
Zhang YS, Liu ZY, Liu ZY, Lin LC, Chen Q, Zhao JY, Tao H. m6A epitranscriptomic modification of inflammation in cardiovascular disease. Int Immunopharmacol 2024; 134:112222. [PMID: 38728881 DOI: 10.1016/j.intimp.2024.112222] [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: 04/07/2024] [Revised: 04/28/2024] [Accepted: 05/05/2024] [Indexed: 05/12/2024]
Abstract
Cardiovascular disease is currently the number one cause of death endangering human health. There is currently a large body of research showing that the development of cardiovascular disease and its complications is often accompanied by inflammatory processes. In recent years, epitranscriptional modifications have been shown to be involved in regulating the pathophysiological development of inflammation in cardiovascular diseases, with 6-methyladenine being one of the most common RNA transcriptional modifications. In this review, we link different cardiovascular diseases, including atherosclerosis, heart failure, myocardial infarction, and myocardial ischemia-reperfusion, with inflammation and describe the regulatory processes involved in RNA methylation. Advances in RNA methylation research have revealed the close relationship between the regulation of transcriptome modifications and inflammation in cardiovascular diseases and brought potential therapeutic targets for disease diagnosis and treatment. At the same time, we also discussed different cell aspects. In addition, in the article we also describe the different application aspects and clinical pathways of RNA methylation therapy. In summary, this article reviews the mechanism, regulation and disease treatment effects of m6A modification on inflammation and inflammatory cells in cardiovascular diseases in recent years. We will discuss issues facing the field and new opportunities that may be the focus of future research.
Collapse
Affiliation(s)
- Yun-Sen Zhang
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, PR China
| | - Zhi-Yan Liu
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, PR China
| | - Zhen-Yu Liu
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, PR China
| | - Li-Chan Lin
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, PR China
| | - Qi Chen
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, PR China.
| | - Jian-Yuan Zhao
- Institute for Developmental and Regenerative Cardiovascular Medicine, MOE-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China.
| | - Hui Tao
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, PR China; Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, PR China.
| |
Collapse
|
17
|
Chen R, Zhang H, Tang B, Luo Y, Yang Y, Zhong X, Chen S, Xu X, Huang S, Liu C. Macrophages in cardiovascular diseases: molecular mechanisms and therapeutic targets. Signal Transduct Target Ther 2024; 9:130. [PMID: 38816371 PMCID: PMC11139930 DOI: 10.1038/s41392-024-01840-1] [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: 12/23/2023] [Revised: 04/02/2024] [Accepted: 04/21/2024] [Indexed: 06/01/2024] Open
Abstract
The immune response holds a pivotal role in cardiovascular disease development. As multifunctional cells of the innate immune system, macrophages play an essential role in initial inflammatory response that occurs following cardiovascular injury, thereby inducing subsequent damage while also facilitating recovery. Meanwhile, the diverse phenotypes and phenotypic alterations of macrophages strongly associate with distinct types and severity of cardiovascular diseases, including coronary heart disease, valvular disease, myocarditis, cardiomyopathy, heart failure, atherosclerosis and aneurysm, which underscores the importance of investigating macrophage regulatory mechanisms within the context of specific diseases. Besides, recent strides in single-cell sequencing technologies have revealed macrophage heterogeneity, cell-cell interactions, and downstream mechanisms of therapeutic targets at a higher resolution, which brings new perspectives into macrophage-mediated mechanisms and potential therapeutic targets in cardiovascular diseases. Remarkably, myocardial fibrosis, a prevalent characteristic in most cardiac diseases, remains a formidable clinical challenge, necessitating a profound investigation into the impact of macrophages on myocardial fibrosis within the context of cardiac diseases. In this review, we systematically summarize the diverse phenotypic and functional plasticity of macrophages in regulatory mechanisms of cardiovascular diseases and unprecedented insights introduced by single-cell sequencing technologies, with a focus on different causes and characteristics of diseases, especially the relationship between inflammation and fibrosis in cardiac diseases (myocardial infarction, pressure overload, myocarditis, dilated cardiomyopathy, diabetic cardiomyopathy and cardiac aging) and the relationship between inflammation and vascular injury in vascular diseases (atherosclerosis and aneurysm). Finally, we also highlight the preclinical/clinical macrophage targeting strategies and translational implications.
Collapse
Affiliation(s)
- Runkai Chen
- Department of Cardiology, Laboratory of Heart Center, Heart Center, Translational Medicine Research Center, Zhujiang Hospital, Southern Medical University, 253 Industrial Avenue, Guangzhou, 510280, China
| | - Hongrui Zhang
- Department of Cardiology, Laboratory of Heart Center, Heart Center, Translational Medicine Research Center, Zhujiang Hospital, Southern Medical University, 253 Industrial Avenue, Guangzhou, 510280, China
| | - Botao Tang
- Department of Cardiology, Laboratory of Heart Center, Heart Center, Translational Medicine Research Center, Zhujiang Hospital, Southern Medical University, 253 Industrial Avenue, Guangzhou, 510280, China
| | - Yukun Luo
- Department of Cardiology, Laboratory of Heart Center, Heart Center, Translational Medicine Research Center, Zhujiang Hospital, Southern Medical University, 253 Industrial Avenue, Guangzhou, 510280, China
| | - Yufei Yang
- Department of Cardiology, Laboratory of Heart Center, Heart Center, Translational Medicine Research Center, Zhujiang Hospital, Southern Medical University, 253 Industrial Avenue, Guangzhou, 510280, China
| | - Xin Zhong
- Department of Cardiology, Laboratory of Heart Center, Heart Center, Translational Medicine Research Center, Zhujiang Hospital, Southern Medical University, 253 Industrial Avenue, Guangzhou, 510280, China
| | - Sifei Chen
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Xinjie Xu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China.
| | - Shengkang Huang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China.
| | - Canzhao Liu
- Department of Cardiology, Laboratory of Heart Center, Heart Center, Translational Medicine Research Center, Zhujiang Hospital, Southern Medical University, 253 Industrial Avenue, Guangzhou, 510280, China.
| |
Collapse
|
18
|
Chen Y, Zhang J. A bibliography of smart nanomaterials biological application in myocardial infarction research. Medicine (Baltimore) 2024; 103:e37672. [PMID: 38579096 PMCID: PMC10994481 DOI: 10.1097/md.0000000000037672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 02/29/2024] [Indexed: 04/07/2024] Open
Abstract
Myocardial infarction has been considered the top cause of mortality globally. Numerous studies investigated the biological application of smart nanomaterials in myocardial infarction. Our study aimed to provide an overview of this area through bibliography research. Literature related to the biological application of nanomaterials was retrieved from the web of science core collection database. Bibliography analysis was performed using Microsoft Excel, VOSviewer, Citespace, and the R package "bibliometrix." A total of 1226 publications were included. The USA, China, and India carried out the most of studies. Harvard University is the most productive institution. Matthias Nahrendorf ranked first in article volume and also owned the highest impact. Keyword burst analysis indicated the frontiers and hotspots to be gold nanoparticles and iron oxide nanoparticles. This bibliography analysis provides a comprehensive overview of uncovered current research trends and emerging hotspots of nanomaterials' biological application in myocardial infarction, thus inspiring further investigations.
Collapse
Affiliation(s)
- Yi Chen
- Department of Emergency Medicine, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Institute of Disaster Medicine, Sichuan University, Nursing Key Laboratory of Sichuan Province, Chengdu, China
| | - Jianna Zhang
- Department of Emergency Medicine, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Institute of Disaster Medicine, Sichuan University, Nursing Key Laboratory of Sichuan Province, Chengdu, China
| |
Collapse
|
19
|
Shalini V, Shanmugam R, Manigandan P. Cytoplasmic Leakage and Protein Leakage Analysis of Ocimum Gratissimum Stem Extract-Mediated Silver Nanoparticles Against Wound Pathogens. JOURNAL OF PHARMACY AND BIOALLIED SCIENCES 2024; 16:S1354-S1359. [PMID: 38882859 PMCID: PMC11174165 DOI: 10.4103/jpbs.jpbs_578_23] [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: 08/12/2023] [Revised: 11/15/2023] [Accepted: 11/17/2023] [Indexed: 06/18/2024] Open
Abstract
Aim The current work intends to examine the antibacterial activity of silver nanoparticles (AgNPs) mediated by Ocimum gratissimum stem extract against wound infections. Materials and Methods To evaluate the membrane damage brought on by AgNPs, analyses of cytoplasmic leakage and protein leakage assays were performed. Results The outcomes demonstrated that all of the tested bacterial strains were significantly resistant to the AgNPs' antibacterial activity. AgNPs damaged membranes and caused cellular contents to leak in the target pathogens, according to an examination of protein and cytoplasmic leakage. Conclusion According to the current investigation, AgNPs mediated by Ocimum gratissimum stem extract may be effective antibacterial agents against microorganisms that cause wounds.
Collapse
Affiliation(s)
- V Shalini
- Nanobiomedicine Lab, Centre for Global Health Research, Saveetha Medical College and Hospital, Saveetha Institute of Medical And Technical Sciences, Saveetha University, Chennai, Tamil Nadu, India
| | - Rajeshkumar Shanmugam
- Nanobiomedicine Lab, Centre for Global Health Research, Saveetha Medical College and Hospital, Saveetha Institute of Medical And Technical Sciences, Saveetha University, Chennai, Tamil Nadu, India
| | - Pradeep Manigandan
- Nanobiomedicine Lab, Centre for Global Health Research, Saveetha Medical College and Hospital, Saveetha Institute of Medical And Technical Sciences, Saveetha University, Chennai, Tamil Nadu, India
| |
Collapse
|
20
|
Liu F, Chen Y, Qin D, Qian C. Interleukin-22 inhibits cardiac fibrosis by regulating fibroblast metabolic reprogramming in myocardial infarction. Pathol Res Pract 2024; 256:155256. [PMID: 38492359 DOI: 10.1016/j.prp.2024.155256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 01/09/2024] [Accepted: 03/06/2024] [Indexed: 03/18/2024]
Abstract
Cardiac fibrosis, a significant characteristic of cardiovascular diseases, leads to ventricular remodeling and impaired cardiac function. In this study, we aimed to investigate the role of Interleukin-22 (IL-22) in myocardial fibrosis following myocardial infarction (MI) and to explore the underlying metabolic mechanisms. Here we analyzed the single-cell sequencing data and found that the level of aerobic glycolysis was significantly higher in cardiac fibrosis in MI patient, which we validated through in vivo experiments. Utilizing MI mouse model, these experiments revealed decreased serum IL-22 levels and increased levels of AngII and TGF-β1. However, treatment with exogenous IL-22 reversed these changes, reduced infarct size, and fibrosis. In vitro experiments demonstrated that IL-22 inhibited AngII-induced fibroblast-to-myofibroblast transition (FMT) by suppressing the expression of α-SMA, Cola1, and Cola3. Metabolic analysis indicated that IL-22 decreased the expression of glycolytic enzymes and reduced lactate production in cardiac fibroblasts. Further in vivo experiments confirmed the inhibitory effect of IL-22 on Pyruvate kinase isoform M2 (PKM2) levels in heart tissue. Additionally, IL-22 activated the c-Jun N-terminal kinase (JNK) pathway, while inhibition of JNK partially reversed IL-22's effect on PKM2 activity. These findings suggest that IL-22 mitigates cardiac fibrosis and FMT by inhibiting aerobic glycolysis by activating the JNK/PKM2 pathway. Our study highlights IL-22 as a potential therapeutic target for myocardial fibrosis and cardiovascular diseases, providing insights into its role in regulating fibrosis and glycolysis. These findings pave the way for developing targeted therapies and investigating additional metabolic pathways for improved treatment outcomes in the field of cardiovascular diseases.
Collapse
Affiliation(s)
- Fang Liu
- Department of Vascular Surgery, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China; International Genome Center, Jiangsu University, Zhenjiang 212013, China.
| | - Yueqi Chen
- International Genome Center, Jiangsu University, Zhenjiang 212013, China
| | - Demeng Qin
- International Genome Center, Jiangsu University, Zhenjiang 212013, China
| | - Cheng Qian
- International Genome Center, Jiangsu University, Zhenjiang 212013, China
| |
Collapse
|
21
|
Fang W, Xie S, Deng W. Ferroptosis mechanisms and regulations in cardiovascular diseases in the past, present, and future. Cell Biol Toxicol 2024; 40:17. [PMID: 38509409 PMCID: PMC10955039 DOI: 10.1007/s10565-024-09853-w] [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: 11/28/2023] [Accepted: 02/27/2024] [Indexed: 03/22/2024]
Abstract
Cardiovascular diseases (CVDs) are the main diseases that endanger human health, and their risk factors contribute to high morbidity and a high rate of hospitalization. Cell death is the most important pathophysiology in CVDs. As one of the cell death mechanisms, ferroptosis is a new form of regulated cell death (RCD) that broadly participates in CVDs (such as myocardial infarction, heart transplantation, atherosclerosis, heart failure, ischaemia/reperfusion (I/R) injury, atrial fibrillation, cardiomyopathy (radiation-induced cardiomyopathy, diabetes cardiomyopathy, sepsis-induced cardiac injury, doxorubicin-induced cardiac injury, iron overload cardiomyopathy, and hypertrophic cardiomyopathy), and pulmonary arterial hypertension), involving in iron regulation, metabolic mechanism and lipid peroxidation. This article reviews recent research on the mechanism and regulation of ferroptosis and its relationship with the occurrence and treatment of CVDs, aiming to provide new ideas and treatment targets for the clinical diagnosis and treatment of CVDs by clarifying the latest progress in CVDs research.
Collapse
Affiliation(s)
- Wenxi Fang
- Department of Cardiology, Renmin Hospital of Wuhan University, Jiefang Road 238, Wuhan, 430060, People's Republic of China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, People's Republic of China
| | - Saiyang Xie
- Department of Cardiology, Renmin Hospital of Wuhan University, Jiefang Road 238, Wuhan, 430060, People's Republic of China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, People's Republic of China
| | - Wei Deng
- Department of Cardiology, Renmin Hospital of Wuhan University, Jiefang Road 238, Wuhan, 430060, People's Republic of China.
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, People's Republic of China.
| |
Collapse
|
22
|
Zheng S, Liu T, Chen M, Sun F, Fei Y, Chen Y, Tian X, Wu Z, Zhu Z, Zheng W, Wang Y, Wang W. Morroniside induces cardiomyocyte cell cycle activity and promotes cardiac repair after myocardial infarction in adult rats. Front Pharmacol 2024; 14:1260674. [PMID: 38273822 PMCID: PMC10808748 DOI: 10.3389/fphar.2023.1260674] [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: 08/07/2023] [Accepted: 12/18/2023] [Indexed: 01/27/2024] Open
Abstract
Introduction: Acute myocardial infarction (AMI) is characterized by the loss of cardiomyocytes, which impairs cardiac function and eventually leads to heart failure. The induction of cardiomyocyte cell cycle activity provides a new treatment strategy for the repair of heart damage. Our previous study demonstrated that morroniside exerts cardioprotective effects. This study investigated the effects and underlying mechanisms of action of morroniside on cardiomyocyte cell cycle activity and cardiac repair following AMI. Methods: Neonatal rat cardiomyocytes (NRCMs) were isolated and exposed to oxygen-glucose deprivation (OGD) in vitro. A rat model of AMI was established by ligation of the left anterior descending coronary artery (LAD) in vivo. Immunofluorescence staining was performed to detect newly generated cardiomyocytes. Western blotting was performed to assess the expression of cell cycle-related proteins. Electrocardiography (ECG) was used to examine pathological Q waves. Masson's trichrome and wheat germ agglutinin (WGA) staining assessed myocardial fibrosis and hypertrophy. Results: The results showed that morroniside induced cardiomyocyte cell cycle activity and increased the levels of cell cycle proteins, including cyclin D1, CDK4, cyclin A2, and cyclin B1, both in vitro and in vivo. Moreover, morroniside reduced myocardial fibrosis and remodeling. Discussion: In conclusion, our study demonstrated that morroniside stimulates cardiomyocyte cell cycle activity and cardiac repair in adult rats, and that these effects may be related to the upregulation of cell cycle proteins.
Collapse
Affiliation(s)
- Songyang Zheng
- Department of Experimental Animal Laboratory, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Tingting Liu
- Department of Experimental Animal Laboratory, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Mengqi Chen
- Department of Experimental Animal Laboratory, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Fangling Sun
- Department of Experimental Animal Laboratory, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Yihuan Fei
- School of Chemical and Pharmaceutical Engineering, Hebei University of Science and Technology, Shijiazhuang, Hebei, China
| | - Yanxi Chen
- School of Chemical and Pharmaceutical Engineering, Hebei University of Science and Technology, Shijiazhuang, Hebei, China
| | - Xin Tian
- Department of Experimental Animal Laboratory, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Zheng Wu
- Department of Functional Neurosurgery, Xuanwu Hospital Capital Medical University, Beijing, China
| | - Zixin Zhu
- Department of Experimental Animal Laboratory, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Wenrong Zheng
- Department of Experimental Animal Laboratory, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Yufeng Wang
- Department of Experimental Animal Laboratory, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Wen Wang
- Department of Experimental Animal Laboratory, Xuanwu Hospital of Capital Medical University, Beijing, China
- Beijing Institute for Brain Disorders, Beijing, China
| |
Collapse
|
23
|
Li L, Li L, Cao C, Guo F, Wang A, Lin L, Liu Z, Meng H, Zhang P, Xin G, Liu J, Ren J, Fu J. Investigation of the active ingredients of Shuangshen Ningxin Fomula and the mechanism underlying their protective effects against myocardial ischemia-reperfusion injury by mass spectrometric imaging. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 123:155184. [PMID: 37951149 DOI: 10.1016/j.phymed.2023.155184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 10/20/2023] [Accepted: 11/02/2023] [Indexed: 11/13/2023]
Abstract
BACKGROUND Traditional Chinese medicine, particularly Shuangshen Ningxin Capsule (SSNX), has been studied intensely. SSNX includes total ginseng saponins (from Panax ginseng Meyer), total phenolic acids from Salvia miltiorrhiza Bunge, and total alkaloids from Corydalis yanhusuo W. T. Wang. It has been suggested to protect against myocardial ischemia by a mechanism that has not been fully elucidated. METHODS The composition and content of SSNX were determined by UHPLC-Q-TOFQ-TOF / MS. Then, a rat model of myocardial ischemia-reperfusion injury was established, and the protective effect of SSNX was measured. The protective mechanism was investigated using spatial metabolomics. RESULTS We found that SSNX significantly improved left ventricular function and ameliorated pathological damages in rats with myocardial ischemia-reperfusion injury. Using matrix-assisted laser desorption ionization time-of-flight mass spectrometry (MALDI-TOF MS), the protective mechanism of SSNX was examined by comparing the monomer components of drugs targeted in myocardial tissue with the distribution of myocardial energy metabolism-related molecules and phospholipids. Interestingly, some lipids display inconsistent content distribution in the myocardial ischemia risk and non-risk zones. These discrepancies reflect the degree of myocardial injury in different regions. CONCLUSION These findings suggest that SSNX protects against myocardial ischemia-reperfusion injury by correcting abnormal myocardial energy metabolism, changing the levels and distribution patterns of phospholipids, and stabilizing the structure of the myocardial cell membrane. MALDI-TOF MS can detect the spatial distribution of small molecule metabolites in the myocardium and can be used in pharmacological research.
Collapse
Affiliation(s)
- Lingmei Li
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, Beijing Key Laboratory of Chinese Materia Pharmacology, China Academy of Chinese Medical Sciences, Beijing 100091, China; Kunshan Hospital of Traditional Chinese Medicine, Jiangsu 215300, China
| | - Lei Li
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, Beijing Key Laboratory of Chinese Materia Pharmacology, China Academy of Chinese Medical Sciences, Beijing 100091, China
| | - Ce Cao
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, Beijing Key Laboratory of Chinese Materia Pharmacology, China Academy of Chinese Medical Sciences, Beijing 100091, China
| | - Fan Guo
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, Beijing Key Laboratory of Chinese Materia Pharmacology, China Academy of Chinese Medical Sciences, Beijing 100091, China
| | - Aoao Wang
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, Beijing Key Laboratory of Chinese Materia Pharmacology, China Academy of Chinese Medical Sciences, Beijing 100091, China
| | - Li Lin
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, Beijing Key Laboratory of Chinese Materia Pharmacology, China Academy of Chinese Medical Sciences, Beijing 100091, China
| | - Zixin Liu
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, Beijing Key Laboratory of Chinese Materia Pharmacology, China Academy of Chinese Medical Sciences, Beijing 100091, China
| | - Hongxu Meng
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, Beijing Key Laboratory of Chinese Materia Pharmacology, China Academy of Chinese Medical Sciences, Beijing 100091, China
| | - Peng Zhang
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, Beijing Key Laboratory of Chinese Materia Pharmacology, China Academy of Chinese Medical Sciences, Beijing 100091, China
| | - Gaojie Xin
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, Beijing Key Laboratory of Chinese Materia Pharmacology, China Academy of Chinese Medical Sciences, Beijing 100091, China
| | - Jianxun Liu
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, Beijing Key Laboratory of Chinese Materia Pharmacology, China Academy of Chinese Medical Sciences, Beijing 100091, China.
| | - Junguo Ren
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, Beijing Key Laboratory of Chinese Materia Pharmacology, China Academy of Chinese Medical Sciences, Beijing 100091, China.
| | - Jianhua Fu
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, Beijing Key Laboratory of Chinese Materia Pharmacology, China Academy of Chinese Medical Sciences, Beijing 100091, China.
| |
Collapse
|
24
|
Lisboa ES, Serafim C, Santana W, Dos Santos VLS, de Albuquerque-Junior RLC, Chaud MV, Cardoso JC, Jain S, Severino P, Souto EB. Nanomaterials-combined methacrylated gelatin hydrogels (GelMA) for cardiac tissue constructs. J Control Release 2024; 365:617-639. [PMID: 38043727 DOI: 10.1016/j.jconrel.2023.11.056] [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: 08/03/2023] [Revised: 11/21/2023] [Accepted: 11/28/2023] [Indexed: 12/05/2023]
Abstract
Among non-communicable diseases, cardiovascular diseases are the most prevalent, accounting for approximately 17 million deaths per year. Despite conventional treatment, cardiac tissue engineering emerges as a potential alternative for the advancement and treatment of these patients, using biomaterials to replace or repair cardiac tissues. Among these materials, gelatin in its methacrylated form (GelMA) is a biodegradable and biocompatible polymer with adjustable biophysical properties. Furthermore, gelatin has the ability to replace and perform collagen-like functions for cell development in vitro. The interest in using GelMA hydrogels combined with nanomaterials is increasingly growing to promote the responsiveness to external stimuli and improve certain properties of these hydrogels by exploring the incorporation of nanomaterials into these hydrogels to serve as electrical signaling conductive elements. This review highlights the applications of electrically conductive nanomaterials associated with GelMA hydrogels for the development of structures for cardiac tissue engineering, by focusing on studies that report the combination of GelMA with nanomaterials, such as gold and carbon derivatives (carbon nanotubes and graphene), in addition to the possibility of applying these materials in 3D tissue engineering, developing new possibilities for cardiac studies.
Collapse
Affiliation(s)
- Erika S Lisboa
- University of Tiradentes (Unit) and Institute of Technology and Research (ITP), Av. Murilo Dantas, 300, 49010-390 Aracaju, Brazil
| | - Carine Serafim
- University of Tiradentes (Unit) and Institute of Technology and Research (ITP), Av. Murilo Dantas, 300, 49010-390 Aracaju, Brazil
| | - Wanessa Santana
- University of Tiradentes (Unit) and Institute of Technology and Research (ITP), Av. Murilo Dantas, 300, 49010-390 Aracaju, Brazil
| | - Victoria L S Dos Santos
- University of Tiradentes (Unit) and Institute of Technology and Research (ITP), Av. Murilo Dantas, 300, 49010-390 Aracaju, Brazil
| | - Ricardo L C de Albuquerque-Junior
- Post-Graduate Program in Dentistry, Department of Dentistry, Federal University of Santa Catarina, Florianópolis 88040-370, Brazil; Department of Pathology, Health Sciences Center, Federal University of Santa Catarina, Florianópolis 88040-370, Brazil
| | - Marco V Chaud
- Laboratory of Biomaterials and Nanotechnology of UNISO (LaBNUS), University of Sorocaba, Sorocaba, São Paulo, Brazil
| | - Juliana C Cardoso
- University of Tiradentes (Unit) and Institute of Technology and Research (ITP), Av. Murilo Dantas, 300, 49010-390 Aracaju, Brazil
| | - Sona Jain
- University of Tiradentes (Unit) and Institute of Technology and Research (ITP), Av. Murilo Dantas, 300, 49010-390 Aracaju, Brazil
| | - Patrícia Severino
- University of Tiradentes (Unit) and Institute of Technology and Research (ITP), Av. Murilo Dantas, 300, 49010-390 Aracaju, Brazil.
| | - Eliana B Souto
- Laboratory of Pharmaceutical Technology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; UCIBIO - Applied Molecular Biosciences Unit, MEDTECH, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; Associate Laboratory i4HB - Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal.
| |
Collapse
|
25
|
Chen F, Xue Q, He N, Zhang X, Li S, Zhao C. The association and application of sonodynamic therapy and autophagy in diseases. Life Sci 2023; 334:122215. [PMID: 37907152 DOI: 10.1016/j.lfs.2023.122215] [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: 08/30/2023] [Revised: 10/09/2023] [Accepted: 10/25/2023] [Indexed: 11/02/2023]
Abstract
Sonodynamic therapy (SDT) is a new non-invasive treatment method proposed based on photodynamic therapy (PDT). It has advantages such as high precision, strong tissue penetration, minimal side effects, and good patient compliance. With the maturation of nanomedicine, the application of nanosonosensitizers has further propelled the development of SDT. In recent years, people have developed many new types of sonosensitizers and explored the mechanisms of SDT. Among them, the studies about the relationship between autophagy and SDT have attracted increasing attention. After the SDT, cells usually undergo autophagy as a self-protective mechanism to resist external stimuli and reduce cell damage, which is beneficial for the treatment of atherosclerosis (AS), diabetes, and myocardial infarction but counterproductive in cancer treatment. However, under certain treatment conditions, excessive upregulation of autophagy can also promote cell death, which is beneficial for cancer treatment. This article reviews the latest research progress on the relationship between SDT and autophagy in cancers, AS, diabetes, and myocardial infarction. We also discuss and propose the challenges and prospects in enhancing SDT efficacy by regulating autophagy, with the hope of promoting the development of this promising therapeutic approach.
Collapse
Affiliation(s)
- Fang Chen
- Department of Abdominal Ultrasound, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Qingwen Xue
- Department of Abdominal Ultrasound, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Ningning He
- School of Basic Medicine, Qingdao Medical College, Qingdao University, Qingdao, China
| | - Xuehui Zhang
- Department of Abdominal Ultrasound, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Shangyong Li
- School of Basic Medicine, Qingdao Medical College, Qingdao University, Qingdao, China.
| | - Cheng Zhao
- Department of Abdominal Ultrasound, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China.
| |
Collapse
|
26
|
Li S, Pei H, He S, Liang H, Guo R, Liu N, Mo Z. Chiral Carbon Dots and Chiral Carbon Dots with Circularly Polarized Luminescence: Synthesis, Mechanistic Investigation and Applications. Chem Asian J 2023; 18:e202300770. [PMID: 37819766 DOI: 10.1002/asia.202300770] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Revised: 10/07/2023] [Accepted: 10/10/2023] [Indexed: 10/13/2023]
Abstract
Chiral carbon dots (CCDs) can be widely used in various fields such as chiral recognition, chiral catalysis and biomedicine because of their unique optical properties, low toxicity and good biocompatibility. In addition, CCDs with circularly polarized luminescence (CPL) can be synthesized, thus broadening the prospects of CCDs applications. Since the research on CCDs is still in its infancy, this paper reviews the chiral origin, formation mechanism, chiral evolution, synthesis and emerging applications of CCDs, with a special focus on CCDs with CPL activity. It is hoped that it will provide some reference to solve the current problems faced by CCDs. Finally, the opportunities and challenges of the current research on CCDs are described, and their future development trends have also been prospected.
Collapse
Affiliation(s)
- Shijing Li
- Research Center of Gansu Military and Civilian Integration Advanced Structural Materials, Key Laboratory of Eco-Environment-Related Polymer Materials, Ministry of Education of China, Key Laboratory of Polymer Materials of Gansu Province, College of Chemistry and Chemical Engineering, Northwest Normal University, Lanzhou, 730070, China
| | - Hebing Pei
- Research Center of Gansu Military and Civilian Integration Advanced Structural Materials, Key Laboratory of Eco-Environment-Related Polymer Materials, Ministry of Education of China, Key Laboratory of Polymer Materials of Gansu Province, College of Chemistry and Chemical Engineering, Northwest Normal University, Lanzhou, 730070, China
| | - Simin He
- Research Center of Gansu Military and Civilian Integration Advanced Structural Materials, Key Laboratory of Eco-Environment-Related Polymer Materials, Ministry of Education of China, Key Laboratory of Polymer Materials of Gansu Province, College of Chemistry and Chemical Engineering, Northwest Normal University, Lanzhou, 730070, China
| | - Hao Liang
- Research Center of Gansu Military and Civilian Integration Advanced Structural Materials, Key Laboratory of Eco-Environment-Related Polymer Materials, Ministry of Education of China, Key Laboratory of Polymer Materials of Gansu Province, College of Chemistry and Chemical Engineering, Northwest Normal University, Lanzhou, 730070, China
| | - Ruibin Guo
- Research Center of Gansu Military and Civilian Integration Advanced Structural Materials, Key Laboratory of Eco-Environment-Related Polymer Materials, Ministry of Education of China, Key Laboratory of Polymer Materials of Gansu Province, College of Chemistry and Chemical Engineering, Northwest Normal University, Lanzhou, 730070, China
| | - Nijuan Liu
- Research Center of Gansu Military and Civilian Integration Advanced Structural Materials, Key Laboratory of Eco-Environment-Related Polymer Materials, Ministry of Education of China, Key Laboratory of Polymer Materials of Gansu Province, College of Chemistry and Chemical Engineering, Northwest Normal University, Lanzhou, 730070, China
| | - Zunli Mo
- Research Center of Gansu Military and Civilian Integration Advanced Structural Materials, Key Laboratory of Eco-Environment-Related Polymer Materials, Ministry of Education of China, Key Laboratory of Polymer Materials of Gansu Province, College of Chemistry and Chemical Engineering, Northwest Normal University, Lanzhou, 730070, China
| |
Collapse
|
27
|
Hong X, Tian G, Zhu Y, Ren T. Exogeneous metal ions as therapeutic agents in cardiovascular disease and their delivery strategies. Regen Biomater 2023; 11:rbad103. [PMID: 38173776 PMCID: PMC10761210 DOI: 10.1093/rb/rbad103] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 10/26/2023] [Accepted: 11/11/2023] [Indexed: 01/05/2024] Open
Abstract
Metal ions participate in many metabolic processes in the human body, and their homeostasis is crucial for life. In cardiovascular diseases (CVDs), the equilibriums of metal ions are frequently interrupted, which are related to a variety of disturbances of physiological processes leading to abnormal cardiac functions. Exogenous supplement of metal ions has the potential to work as therapeutic strategies for the treatment of CVDs. Compared with other therapeutic drugs, metal ions possess broad availability, good stability and safety and diverse drug delivery strategies. The delivery strategies of metal ions are important to exert their therapeutic effects and reduce the potential toxic side effects for cardiovascular applications, which are also receiving increasing attention. Controllable local delivery strategies for metal ions based on various biomaterials are constantly being designed. In this review, we comprehensively summarized the positive roles of metal ions in the treatment of CVDs from three aspects: protecting cells from oxidative stress, inducing angiogenesis, and adjusting the functions of ion channels. In addition, we introduced the transferability of metal ions in vascular reconstruction and cardiac tissue repair, as well as the currently available engineered strategies for the precise delivery of metal ions, such as integrated with nanoparticles, hydrogels and scaffolds.
Collapse
Affiliation(s)
- Xiaoqian Hong
- Department of Cardiology of the Second Affiliated Hospital and State Key Laboratory of Transvascular Implantation Devices, Cardiovascular Key Laboratory of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Geer Tian
- Department of Cardiology of the Second Affiliated Hospital and State Key Laboratory of Transvascular Implantation Devices, Cardiovascular Key Laboratory of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou 310009, China
- Binjiang Institute of Zhejiang University, Hangzhou 310053, China
| | - Yang Zhu
- Binjiang Institute of Zhejiang University, Hangzhou 310053, China
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
| | - Tanchen Ren
- Department of Cardiology of the Second Affiliated Hospital and State Key Laboratory of Transvascular Implantation Devices, Cardiovascular Key Laboratory of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou 310009, China
| |
Collapse
|
28
|
Omidian H, Babanejad N, Cubeddu LX. Nanosystems in Cardiovascular Medicine: Advancements, Applications, and Future Perspectives. Pharmaceutics 2023; 15:1935. [PMID: 37514121 PMCID: PMC10386572 DOI: 10.3390/pharmaceutics15071935] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 07/02/2023] [Accepted: 07/07/2023] [Indexed: 07/30/2023] Open
Abstract
Cardiovascular diseases (CVDs) remain a leading cause of morbidity and mortality globally. Despite significant advancements in the development of pharmacological therapies, the challenges of targeted drug delivery to the cardiovascular system persist. Innovative drug-delivery systems have been developed to address these challenges and improve therapeutic outcomes in CVDs. This comprehensive review examines various drug delivery strategies and their efficacy in addressing CVDs. Polymeric nanoparticles, liposomes, microparticles, and dendrimers are among the drug-delivery systems investigated in preclinical and clinical studies. Specific strategies for targeted drug delivery, such as magnetic nanoparticles and porous stent surfaces, are also discussed. This review highlights the potential of innovative drug-delivery systems as effective strategies for the treatment of CVDs.
Collapse
Affiliation(s)
- Hossein Omidian
- Barry and Judy Silverman College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL 33328, USA
| | - Niloofar Babanejad
- Barry and Judy Silverman College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL 33328, USA
| | - Luigi X Cubeddu
- Barry and Judy Silverman College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL 33328, USA
| |
Collapse
|
29
|
Xu G, Duan Z, Luo K, Yang J. Nanotherapeutics for solid organ transplantation: new kid on the block. EBioMedicine 2023; 92:104624. [PMID: 37209534 PMCID: PMC10209123 DOI: 10.1016/j.ebiom.2023.104624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Accepted: 05/07/2023] [Indexed: 05/22/2023] Open
Affiliation(s)
- Gang Xu
- Liver Transplant Center, Organ Transplant Center, Department of Radiology, Huaxi MR Research Center (HMRRC), National Clinical Research Center for Geriatrics, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu 610041, China; Laboratory of Liver Transplantation, NHC Key Laboratory of Transplant Engineering and Immunology, Chengdu, 610041, China
| | - Zhenyu Duan
- Liver Transplant Center, Organ Transplant Center, Department of Radiology, Huaxi MR Research Center (HMRRC), National Clinical Research Center for Geriatrics, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Kui Luo
- Liver Transplant Center, Organ Transplant Center, Department of Radiology, Huaxi MR Research Center (HMRRC), National Clinical Research Center for Geriatrics, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu 610041, China.
| | - Jiayin Yang
- Liver Transplant Center, Organ Transplant Center, Department of Radiology, Huaxi MR Research Center (HMRRC), National Clinical Research Center for Geriatrics, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu 610041, China; Laboratory of Liver Transplantation, NHC Key Laboratory of Transplant Engineering and Immunology, Chengdu, 610041, China.
| |
Collapse
|
30
|
Zhu Q, Luo Y, Wen Y, Wang D, Li J, Fan Z. Semaglutide inhibits ischemia/reperfusion-induced cardiomyocyte apoptosis through activating PKG/PKCε/ERK1/2 pathway. Biochem Biophys Res Commun 2023; 647:1-8. [PMID: 36706596 DOI: 10.1016/j.bbrc.2023.01.049] [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: 12/20/2022] [Revised: 01/03/2023] [Accepted: 01/16/2023] [Indexed: 01/19/2023]
Abstract
Apoptosis is a major pathophysiological change following myocardial ischemia/reperfusion (I/R) injury. Glucagon-like peptide 1 (GLP-1) and its receptor GLP-1R are widely expressed in the cardiovascular system and GLP-1/GLP-1R activates the protein kinase G (PKG)-related signaling pathway. Therefore, this study tested whether semaglutide, a new GLP-1 analog, inhibits I/R injury-induced cardiomyocyte apoptosis by activating the PKG/PKCε/ERK1/2 pathway. We induced myocardial I/R injury in rats and hypoxia/reoxygenation (H/R) injury in H9C2 cells and detected the effects of semaglutide, a PKG analog (8-Br-cGMP), and a PKG inhibitor (KT-5823) on the PKG/PKCε/ERK1/2 pathway and cardiomyocyte apoptosis. We found that semaglutide upregulated GLP-1R levels, and both semaglutide and 8-Br-cGMP activated the PKG/PKCε/ERK1/2 pathway, inhibited myocardial infarction (MI), decreased hs-cTNT levels, increased NT-proBNP levels, and suppressed cardiomyocyte apoptosis in I/R rats and H/R H9C2 cells. However, KT-5823 exerted contrasting effects with semaglutide and 8-Br-cGMP, and KT-5823 weakened the cardioprotective effects of semaglutide. In conclusion, semaglutide inhibits I/R injury-induced cardiomyocyte apoptosis by activating the PKG/PKCε/ERK1/2 pathway. The beneficial effect of GLP-1/GLP-1R, involved in the activation of the PKG/PKCε/ERK1/2 pathway, may provide a novel treatment method for myocardial I/R injury.
Collapse
Affiliation(s)
- Qiuxia Zhu
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Number 25, Taiping Street. Jiangyang District, 400042, Luzhou, Sichuan, China
| | - Yong Luo
- Department of Cardiology, Chongqing University Jiangjin Hospital, School of Medicine, Chongqing University, Number 725, Jiangzhou Avenue, Jiangjin District, 402260, Chongqing, China
| | - Yuetao Wen
- Department of Neurosurgery, Chongqing University Jiangjin Hospital, School of Medicine, Chongqing University, Number 725, Jiangzhou Avenue, Jiangjin District, 402260, Chongqing, China
| | - Ding Wang
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Number 25, Taiping Street. Jiangyang District, 400042, Luzhou, Sichuan, China
| | - Jing Li
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Number 25, Taiping Street. Jiangyang District, 400042, Luzhou, Sichuan, China
| | - Zhongcai Fan
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Number 25, Taiping Street. Jiangyang District, 400042, Luzhou, Sichuan, China.
| |
Collapse
|
31
|
Xu Y, Dong Z, Zhang R, Wang Z, Shi Y, Liu M, Yang J, Yang T, Zhang R, Wang T, Zhang J, Zhang Y, Xiang F, Han Y, Wu J, Miao Z, Chen Q, Li Q, Wang Z, Tian Y, Guo Y. Sonodynamic therapy reduces cardiomyocyte apoptosis through autophagy activated by reactive oxygen species in myocardial infarction. Free Radic Biol Med 2023; 195:36-46. [PMID: 36529292 DOI: 10.1016/j.freeradbiomed.2022.12.080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 12/14/2022] [Accepted: 12/14/2022] [Indexed: 12/23/2022]
Abstract
Myocardial infarction (MI) is lethal to patients because of acute ischemia and hypoxia leading to cardiac tissue apoptosis. Autophagy played a key role in MI through affecting the survival of cardiomyocytes. LncRNA-MHRT (myosin heavy-chain-associated RNA transcripts) was specific to the heart and cardiomyocytes, and inhibition of lncRNA-MHRT transcription under pathological stimuli could cause cardiac hypertrophy and even heart failure (HF). Sonodynamic therapy (SDT) is a new and developing medical technique that utilizes low-intensity ultrasound to locally activate a preloaded sonosensitizer. Our group previously reported that SDT could regulate autophagy. In this study, we investigated whether SDT could reduce MI-induced cardiomyocyte apoptosis via activating autophagy pathway. SDT improved cardiac function and suppresses MI-induced cardiomyocyte apoptosis. SDT alleviated MI-induced cardiomyocyte apoptosis by improving autophagy. MHRT mediated the inhibiting effect of SDT on cardiomyocyte apoptosis via activating autophagy pathway. Our data reveal a novel effect that SDT protects against MI and confirm that SDT reduces MI-induced cardiomyocyte apoptosis via activating MHRT-mediated-autophagy. Thus, our findings also indicate that SDT may be used as a potential method for treatment of post-myocardial infarction heart failure.
Collapse
Affiliation(s)
- Yingjie Xu
- Department of Cardiology, The First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Zengxiang Dong
- The Key Laboratory of Cardiovascular Disease Acousto-Optic Electromagnetic Diagnosis and Treatment in Heilongjiang Province, The First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Rongzhen Zhang
- Department of Cardiology, The First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Zeng Wang
- Department of Cardiology, The First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Yuanqi Shi
- The Key Laboratory of Cardiovascular Disease Acousto-Optic Electromagnetic Diagnosis and Treatment in Heilongjiang Province, The First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Mingyu Liu
- The Key Laboratory of Cardiovascular Disease Acousto-Optic Electromagnetic Diagnosis and Treatment in Heilongjiang Province, The First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Jiemei Yang
- Department of Cardiology, The First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Tao Yang
- Department of Cardiology, The First Affiliated Hospital, Harbin Medical University, Harbin, China
| | | | - Tengyu Wang
- Department of Cardiology, The First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Jingyu Zhang
- Department of Geriatrics, The First Affiliated Hospital of Harbin Medical University, Harbin, China; Shenzhen Qianhai Henkou Free Trade Zone Hospital, Shenzhen, China
| | - Yu Zhang
- Department of Cardiology, The First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Fei Xiang
- Department of Cardiology, The First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Yingjun Han
- Department of Cardiology, The First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Jiawen Wu
- Department of Cardiology, The First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Zhihan Miao
- Department of Cardiology, The First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Qiuyu Chen
- Department of Cardiology, The First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Qi Li
- Department of Cardiology, The First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Zeyao Wang
- Department of Cardiology, The First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Ye Tian
- Department of Cardiology, The First Affiliated Hospital, Harbin Medical University, Harbin, China; The Key Laboratory of Cardiovascular Disease Acousto-Optic Electromagnetic Diagnosis and Treatment in Heilongjiang Province, The First Affiliated Hospital, Harbin Medical University, Harbin, China; Department of Pathophysiology and Key Laboratory of Cardiovascular Pathophysiology, Harbin Medical University, Key Laboratory of Cardiovascular Medicine Research, Harbin Medical University, Ministry of Education, Harbin, China.
| | - Yuanyuan Guo
- Department of Cardiology, The First Affiliated Hospital, Harbin Medical University, Harbin, China; Department of Geriatrics, The First Affiliated Hospital of Harbin Medical University, Harbin, China.
| |
Collapse
|
32
|
Li P, Wang K, Yin J, Qi L, Hu H, Yang P, Shi Y, Li Y, Feng M, Lyu H, Ge W, Li X, Yan S. lncRNA LOC100911717-targeting GAP43-mediated sympathetic remodeling after myocardial infarction in rats. Front Cardiovasc Med 2023; 9:1019435. [PMID: 36684596 PMCID: PMC9859628 DOI: 10.3389/fcvm.2022.1019435] [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: 08/15/2022] [Accepted: 12/01/2022] [Indexed: 01/08/2023] Open
Abstract
Objective Sympathetic remodeling after myocardial infarction (MI) is the primary cause of ventricular arrhythmias (VAs), leading to sudden cardiac death (SCD). M1-type macrophages are closely associated with inflammation and sympathetic remodeling after MI. Long noncoding RNAs (lncRNAs) are critical for the regulation of cardiovascular disease development. Therefore, this study aimed to identify the lncRNAs involved in MI and reveal a possible regulatory mechanism. Methods and results M0- and M1-type macrophages were selected for sequencing and screened for differentially expressed lncRNAs. The data revealed that lncRNA LOC100911717 was upregulated in M1-type macrophages but not in M0-type macrophages. In addition, the lncRNA LOC100911717 was upregulated in heart tissues after MI. Furthermore, an RNA pull-down assay revealed that lncRNA LOC100911717 could interact with growth-associated protein 43 (GAP43). Essentially, immunofluorescence assays and programmed electrical stimulation demonstrated that GAP43 expression was suppressed and VA incidence was reduced after lncRNA LOC100911717 knockdown in rat hearts using an adeno-associated virus. Conclusions We observed a novel relationship between lncRNA LOC100911717 and GAP43. After MI, lncRNA LOC100911717 was upregulated and GAP43 expression was enhanced, thus increasing the extent of sympathetic remodeling and the frequency of VA events. Consequently, silencing lncRNA LOC100911717 could reduce sympathetic remodeling and VAs.
Collapse
Affiliation(s)
- Pingjiang Li
- Department of Cardiology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Shandong Medicine and Health Key Laboratory of Cardiac Electrophysiology and Arrhythmia, Jinan, China,Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Kang Wang
- Department of Cardiology, Cheeloo College of Medicine, Shandong Qianfoshan Hospital, Shandong University, Jinan, China
| | - Jie Yin
- Department of Cardiology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Shandong Medicine and Health Key Laboratory of Cardiac Electrophysiology and Arrhythmia, Jinan, China
| | - Lei Qi
- Department of Cardiology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Shandong Medicine and Health Key Laboratory of Cardiac Electrophysiology and Arrhythmia, Jinan, China,Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Hesheng Hu
- Department of Cardiology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Shandong Medicine and Health Key Laboratory of Cardiac Electrophysiology and Arrhythmia, Jinan, China
| | - Peijin Yang
- Department of Cardiology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Shandong Medicine and Health Key Laboratory of Cardiac Electrophysiology and Arrhythmia, Jinan, China,Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Yugen Shi
- Department of Cardiology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Shandong Medicine and Health Key Laboratory of Cardiac Electrophysiology and Arrhythmia, Jinan, China
| | - Yan Li
- Shandong Medicine and Health Key Laboratory of Cardiac Electrophysiology and Arrhythmia, Medical Research Center, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Meng Feng
- Department of Cardiology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Shandong Medicine and Health Key Laboratory of Cardiac Electrophysiology and Arrhythmia, Jinan, China,Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Hangji Lyu
- Department of Cardiology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Shandong Medicine and Health Key Laboratory of Cardiac Electrophysiology and Arrhythmia, Jinan, China,Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Weili Ge
- Department of Cardiology, Taizhou Hospital, Wenzhou Medical University, Taizhou, Zhejiang, China
| | - Xiaolu Li
- Department of Cardiology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Shandong Medicine and Health Key Laboratory of Cardiac Electrophysiology and Arrhythmia, Jinan, China
| | - Suhua Yan
- Department of Cardiology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Shandong Medicine and Health Key Laboratory of Cardiac Electrophysiology and Arrhythmia, Jinan, China,*Correspondence: Suhua Yan ✉
| |
Collapse
|
33
|
Yang Y, Xia Z, Xu C, Zhai C, Yu X, Li S. Ciprofol attenuates the isoproterenol-induced oxidative damage, inflammatory response and cardiomyocyte apoptosis. Front Pharmacol 2022; 13:1037151. [PMID: 36483733 PMCID: PMC9723392 DOI: 10.3389/fphar.2022.1037151] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 10/28/2022] [Indexed: 01/25/2023] Open
Abstract
Background and Purpose: Ciprofol (HSK3486), a novel 2,6-disubstituted phenol derivative, is a new intravenous anesthetic compound with a similar chemical structure to propofol. Animal studies have also shown that propofol plays a protective role in a variety of cardiovascular diseases, including myocardial infarction, myocardial ischemia-reperfusion injury and takotsubo syndrome. However, whether ciprofol exerts cardioprotective effects on myocardial infarction remains unclear. Thus, the aim of this work was to explore the potential cardioprotective mechanism of ciprofol on isoproterenol (ISO)-induced myocardial infarction. Experimental Approach: In the present study, male C57BL/6 mice were subjected to subcutaneous injection of ISO (100 mg/kg) for 2 consecutive days to induce experimental myocardial infarction. Herein, we found that ciprofol could inhibit the abnormal increase in myocardial injury enzymes, the area of myocardial infarction and cardiac dysfunction in ISO-treated mice. Ciprofol administration increased the activity of superoxide dismutase and reduced the levels of NADPH oxidase and malondialdehyde in ISO-treated hearts. Additionally, ciprofol administration markedly reduced the expression of pro-inflammatory cytokines and cardiomyocyte apoptosis. In an in vitro model, the results also confirmed that ciprofol could inhibit ISO-induced oxidative damage, the inflammatory response and cardiomyocyte apoptosis. Moreover, ciprofol can activate the sirtuin1 (Sirt1)/nuclear factor erythroid 2-related factor 2 (Nrf2) pathway and Sirt1 and Nrf2 inhibition almost abolished ciprofol-mediated cardioprotective effects. Interpretation: Ciprofol protects the heart against ISO-induced myocardial infarction by reducing cardiac oxidative stress, the inflammatory response and cardiomyocyte apoptosis.
Collapse
|
34
|
Huang Q, Zhang C, Tang S, Wu X, Peng X. Network Pharmacology Analyses of the Pharmacological Targets and Therapeutic Mechanisms of Salvianolic Acid A in Myocardial Infarction. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2022; 2022:8954035. [PMID: 36248430 PMCID: PMC9556248 DOI: 10.1155/2022/8954035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 06/25/2022] [Accepted: 06/30/2022] [Indexed: 11/05/2022]
Abstract
Objective Salvianolic acid A, a natural polyphenolic ingredient extracted from traditional Chinese medicine, possesses an excellent pharmacological activity against cardiovascular diseases. Herein, therapeutic mechanisms of salvianolic acid A in myocardial infarction were explored through systematic and comprehensive network pharmacology analyses. Methods The chemical structure of salvianolic acid A was retrieved from PubChem database. Targets of salvianolic acid A were estimated through SwissTargetPrediction, HERB, and TargetNet databases. Additionally, by GeneCards, OMIM, DisGeNET, and TTD online tools, myocardial infarction-relevant targets were predicted. Following intersection, therapeutic targets were determined. The interaction of their products was evaluated with STRING database, and hub therapeutic targets were selected. GO and KEGG enrichment analyses of therapeutic targets were then implemented. H9C2 cells were exposed to oxygen-glucose deprivation/reoxygenation (OGD/R) to mimic myocardial infarction and administrated with salvianolic acid A. Cellular proliferation was assayed via CCK-8 assay, and hub therapeutic targets were verified with RT-qPCR. Results In total, 120 therapeutic targets of salvianolic acid A in myocardial infarction were identified. There were close interactions between their products. Ten hub therapeutic targets were determined, covering SRC, CTNNB1, PIK3CA, AKT1, RELA, EGFR, FYN, ITGB1, MAPK8, and NFKB1. Therapeutic targets were significantly correlated to myocardial infarction-relevant pathways, especially PI3K-Akt signaling pathway. Salvianolic acid A administration remarkably ameliorated the viability of OGD/R-induced H9C2 cells, and altered the expression of hub therapeutic targets. Conclusion Our work uncovers therapeutic mechanisms of salvianolic acid A for the treatment of myocardial infarction, providing a new insight into further research on salvianolic acid A.
Collapse
Affiliation(s)
- Qing Huang
- Department of Cardiology, Wuhan Fourth Hospital, Wuhan, Hubei, China
| | - Chao Zhang
- Heart Function Testing Center of Cardiovascular Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Shaoyong Tang
- Department of Cardiology, Wuhan Fourth Hospital, Wuhan, Hubei, China
| | - Xiaoyan Wu
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Xiong Peng
- Department of Cardiology, Wuhan Fourth Hospital, Wuhan, Hubei, China
| |
Collapse
|
35
|
Iravani S, Varma RS. Advanced Drug Delivery Micro- and Nanosystems for Cardiovascular Diseases. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27185843. [PMID: 36144581 PMCID: PMC9506137 DOI: 10.3390/molecules27185843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 08/29/2022] [Accepted: 09/07/2022] [Indexed: 12/03/2022]
Abstract
Advanced drug delivery micro- and nanosystems have been widely explored due to their appealing specificity/selectivity, biodegradability, biocompatibility, and low toxicity. They can be applied for the targeted delivery of pharmaceuticals, with the benefits of good biocompatibility/stability, non-immunogenicity, large surface area, high drug loading capacity, and low leakage of drugs. Cardiovascular diseases, as one of the primary mortalities cause worldwide with significant impacts on the quality of patients’ life, comprise a variety of heart and circulatory system pathologies, such as peripheral vascular diseases, myocardial infarction, heart failure, and coronary artery diseases. Designing novel micro- and nanosystems with suitable targeting properties and smart release behaviors can help circumvent crucial challenges of the tolerability, low stability, high toxicity, and possible side- and off-target effects of conventional drug delivery routes. To overcome different challenging issues, namely physiological barriers, low efficiency of drugs, and possible adverse side effects, various biomaterials-mediated drug delivery systems have been formulated with reduced toxicity, improved pharmacokinetics, high bioavailability, sustained release behavior, and enhanced therapeutic efficacy for targeted therapy of cardiovascular diseases. Despite the existing drug delivery systems encompassing a variety of biomaterials for treating cardiovascular diseases, the number of formulations currently approved for clinical use is limited due to the regulatory and experimental obstacles. Herein, the most recent advancements in drug delivery micro- and nanosystems designed from different biomaterials for the treatment of cardiovascular diseases are deliberated, with a focus on the important challenges and future perspectives.
Collapse
Affiliation(s)
- Siavash Iravani
- Faculty of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan 81746-73461, Iran
- Correspondence: (S.I.); (R.S.V.)
| | - Rajender S. Varma
- Regional Centre of Advanced Technologies and Materials, Czech Advanced Technology and Research Institute, Palacký University in Olomouc, Šlechtitelů 27, Olomouc 78371, Czech Republic
- Correspondence: (S.I.); (R.S.V.)
| |
Collapse
|
36
|
Zhang Q, Guo Y, Zhang B, Liu H, Peng Y, Wang D, Zhang D. Identification of hub biomarkers of myocardial infarction by single-cell sequencing, bioinformatics, and machine learning. Front Cardiovasc Med 2022; 9:939972. [PMID: 35958412 PMCID: PMC9357907 DOI: 10.3389/fcvm.2022.939972] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 07/05/2022] [Indexed: 12/11/2022] Open
Abstract
Background Myocardial infarction (MI) is one of the first cardiovascular diseases endangering human health. Inflammatory response plays a significant role in the pathophysiological process of MI. Messenger RNA (mRNA) has been proven to play a key role in cardiovascular diseases. Single-cell sequencing (SCS) technology is a new technology for high-throughput sequencing analysis of genome, transcriptome, and epigenome at the single-cell level, and it also plays an important role in the diagnosis and treatment of cardiovascular diseases. Machine learning algorithms have a wide scope of utilization in biomedicine and have demonstrated superior efficiency in clinical trials. However, few studies integrate these three methods to investigate the role of mRNA in MI. The aim of this study was to screen the expression of mRNA, investigate the function of mRNA, and provide an underlying scientific basis for the diagnosis of MI. Methods In total, four RNA microarray datasets of MI, namely, GSE66360, GSE97320, GSE60993, and GSE48060, were downloaded from the Gene Expression Omnibus database. The function analysis was carried out by Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), and Disease Ontology (DO) enrichment analysis. At the same time, inflammation-related genes (IRGs) were acquired from the GeneCards database. Then, 52 co-DEGs were acquired from differentially expressed genes (DEGs) in differential analysis, IRGs, and genes from SCS, and they were used to construct a protein-protein interaction (PPI) network. Two machine learning algorithms, namely, (1) least absolute shrinkage and selection operator and (2) support vector machine recursive feature elimination, were used to filter the co-DEGs. Gene set enrichment analysis (GSEA) was performed to screen the hub-modulating signaling pathways associated with the hub genes. The results were validated in GSE97320, GSE60993, and GSE48060 datasets. The CIBERSORT algorithm was used to analyze 22 infiltrating immune cells in the MI and healthy control (CON) groups and to analyze the correlation between these immune cells. The Pymol software was used for molecular docking of hub DEGs and for potential treatment of MI drugs acquired from the COREMINE. Results A total of 126 DEGs were in the MI and CON groups. After screening two machine learning algorithms and key co-DEGs from a PPI network, two hub DEGs (i.e., IL1B and TLR2) were obtained. The diagnostic efficiency of IL1B, TLR2, and IL1B + TLR2 showed good discrimination in the four cohorts. GSEA showed that KEGG enriched by DEGs were mainly related to inflammation-mediated signaling pathways, and GO biological processes enriched by DEGs were linked to biological effects of various inflammatory cells. Immune analysis indicated that IL1B and TLR2 were correlated with various immune cells. Dan shen, san qi, feng mi, yuan can e, can sha, san qi ye, san qi hua, and cha shu gen were identified as the potential traditional Chinese medicine (TCM) for the treatment of MI. 7-hydroxyflavone (HF) had stable combinations with IL1B and TLR2, respectively. Conclusion This study identified two hub DEGs (IL1B and TLR2) and illustrated its potential role in the diagnosis of MI to enhance our knowledge of the underlying molecular mechanism. Infiltrating immune cells played an important role in MI. TCM, especially HF, was a potential drug for the treatment of MI.
Collapse
Affiliation(s)
- Qunhui Zhang
- Research Center for High Altitude Medicine, Key Laboratory of High-Altitude Medicine (Ministry of Education), Key Laboratory of Application and Foundation for High Altitude Medicine Research in Qinghai Province (Qinghai-Utah Joint Research Key Lab for High Altitude Medicine), Qinghai University, Xining, China.,College of Eco-Environmental Engineering, Qinghai University, Xining, China
| | - Yang Guo
- Research Center for High Altitude Medicine, Key Laboratory of High-Altitude Medicine (Ministry of Education), Key Laboratory of Application and Foundation for High Altitude Medicine Research in Qinghai Province (Qinghai-Utah Joint Research Key Lab for High Altitude Medicine), Qinghai University, Xining, China.,College of Eco-Environmental Engineering, Qinghai University, Xining, China
| | - Benyin Zhang
- College of Eco-Environmental Engineering, Qinghai University, Xining, China
| | - Hairui Liu
- College of Eco-Environmental Engineering, Qinghai University, Xining, China
| | - Yanfeng Peng
- College of Eco-Environmental Engineering, Qinghai University, Xining, China
| | - Di Wang
- College of Eco-Environmental Engineering, Qinghai University, Xining, China
| | - Dejun Zhang
- Research Center for High Altitude Medicine, Key Laboratory of High-Altitude Medicine (Ministry of Education), Key Laboratory of Application and Foundation for High Altitude Medicine Research in Qinghai Province (Qinghai-Utah Joint Research Key Lab for High Altitude Medicine), Qinghai University, Xining, China.,College of Eco-Environmental Engineering, Qinghai University, Xining, China
| |
Collapse
|