1
|
Date S, Bhatt LK. Targeting high-mobility-group-box-1-mediated inflammation: a promising therapeutic approach for myocardial infarction. Inflammopharmacology 2024:10.1007/s10787-024-01586-w. [PMID: 39487941 DOI: 10.1007/s10787-024-01586-w] [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: 08/30/2024] [Accepted: 10/11/2024] [Indexed: 11/04/2024]
Abstract
Myocardial ischemia, resulting from coronary artery blockage, precipitates cardiac arrhythmias, myocardial structural changes, and heart failure. The pathophysiology of MI is mainly based on inflammation and cell death, which are essential in aggravating myocardial ischemia and reperfusion injury. Emerging research highlights the functionality of high mobility group box-1, a non-histone nucleoprotein functioning as a chromosomal stabilizer and inflammatory mediator. HMGB1's release into the extracellular compartment during ischemia acts as damage-associated molecular pattern, triggering immune reaction by pattern recognition receptors and exacerbating tissue inflammation. Its involvement in signaling pathways like PI3K/Akt, TLR4/NF-κB, and RAGE/HMGB1 underscores its significance in promoting angiogenesis, apoptosis, and reducing inflammation, which is crucial for MI treatment strategies. This review highlights the complex function of HMGB1 in the pathogenesis of myocardial infarction by summarizing novel findings on the protein in ischemic situations. Understanding the mechanisms underlying HMGB1 could widen the way to specific treatments that minimize the severity of MI and enhance patient outcomes.
Collapse
Affiliation(s)
- Shrutika Date
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Vile Parle (W), Mumbai, India
| | - Lokesh Kumar Bhatt
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Vile Parle (W), Mumbai, India.
| |
Collapse
|
2
|
Shi J, Guan B, Gong M, He X. Kirenol Alleviates Inflammation and Oxidative Stress to Improve Myocardial Ischemia/Reperfusion Injury in Rats. J Cardiovasc Pharmacol 2024; 84:539-544. [PMID: 39186590 DOI: 10.1097/fjc.0000000000001626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 08/09/2024] [Indexed: 08/28/2024]
Abstract
ABSTRACT Ischemic heart disease gravely threatens human health and even results in death. Kirenol is predominantly derived from the Herba Siegesbeckiae plant species and possesses a wide range of biological effects (such as antibacterial, anti-inflammatory, anticancer, and cardioprotective). However, the regulatory effects and associated mechanisms of kirenol in myocardial ischemia/reperfusion injury (MI/RI) remain unclear. In this study, first, the MI/RI rat model was established. It was demonstrated that kirenol protected against the aggravation of cardiac function in MI/RI rats. In addition, the inflammation was induced by ischemia reperfusion (IR), which was likewise affected by kirenol (5 or 10 mg/kg). Moreover, IR enhanced oxidative stress, a process that was counteracted by kirenol. Next, cell apoptosis was discovered to be heightened after IR, but this effect was neutralized by kirenol. Finally, it was revealed that kirenol has the ability to block the activation of the NF-κB pathway. In conclusion, it was disclosed that kirenol alleviated inflammation and oxidative stress through modulating the NF-κB pathway to improve MI/RI in rats. This work may offer novel insights for searching useful drugs for treating MI/RI.
Collapse
Affiliation(s)
- Jinlong Shi
- Department of Cardio-Thoracic Surgery, The First Affiliated Hospital of Yangtze University, Jingzhou, China
| | - Bingfeng Guan
- Department of Cardio-Thoracic Surgery, The First Affiliated Hospital of Yangtze University, Jingzhou, China
| | - Minghui Gong
- Department of Intensive Care Unit, The First Affiliated Hospital of Yangtze University, Jingzhou, China ; and
| | - Xinyi He
- Major of Stomatology, Medical College of Soochow University, Suzhou, China
| |
Collapse
|
3
|
Liu Y, Wu H, Zhou G, Zhang D, Yang Q, Li Y, Yang X, Sun J. Role of M6a Methylation in Myocardial Ischemia-Reperfusion Injury and Doxorubicin-Induced Cardiotoxicity. Cardiovasc Toxicol 2024; 24:918-928. [PMID: 39026038 DOI: 10.1007/s12012-024-09898-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 07/11/2024] [Indexed: 07/20/2024]
Abstract
Cardiovascular disease remains the leading cause of death worldwide, with acute myocardial infarction and anticancer drug-induced cardiotoxicity being the significant factors. The most effective treatment for acute myocardial infarction is rapid restoration of coronary blood flow by thrombolytic therapy or percutaneous coronary intervention. However, myocardial ischemia-reperfusion injury (MI/RI) after reperfusion therapy is common in acute myocardial infarction, thus affecting the prognosis of patients with acute myocardial infarction. There is no effective treatment for MI/RI. Anthracyclines such as Doxorubicin (DOX) have limited clinical use due to their cardiotoxicity, and the mechanism of DOX-induced cardiac injury is complex and not yet fully understood. N6-methyladenosine (m6A) plays a crucial role in many biological processes. Emerging evidence suggests that m6A methylation plays a critical regulatory role in MI/RI and DOX-induced cardiotoxicity (DIC), suggesting that m6A may serve as a novel biomarker and therapeutic target for MI/RI and DIC. M6A methylation may mediate the pathophysiological processes of MI/RI and DIC by regulating cellular autophagy, apoptosis, oxidative stress, and inflammatory response. In this paper, we first focus on the relationship between m6A methylation and MI/RI, then further elucidate that m6A methylation may mediate the pathophysiological process of MI/RI through the regulation of cellular autophagy, apoptosis, oxidative stress, and inflammatory response. Finally, briefly outline the roles played by m6A in DIC, which will provide a new methodology and direction for the research and treatment of MI/RI and DIC.
Collapse
Affiliation(s)
- Yanfang Liu
- Institute of Cardiovascular Diseases, China Three Gorges University, Hubei, China
- Department of Cardiology, Yichang Central People's Hospital, Yichang, 443003, China
- Hubei Key Laboratory of Ischemic Cardiovascular Disease, Yichang, China
- HuBei Clinical Research Center for Ischemic Cardiovascular Disease, Yichang, China
| | - Hui Wu
- Institute of Cardiovascular Diseases, China Three Gorges University, Hubei, China.
- Department of Cardiology, Yichang Central People's Hospital, Yichang, 443003, China.
- Hubei Key Laboratory of Ischemic Cardiovascular Disease, Yichang, China.
- HuBei Clinical Research Center for Ischemic Cardiovascular Disease, Yichang, China.
| | - Gang Zhou
- Institute of Cardiovascular Diseases, China Three Gorges University, Hubei, China
- Department of Cardiology, Yichang Central People's Hospital, Yichang, 443003, China
- Hubei Key Laboratory of Ischemic Cardiovascular Disease, Yichang, China
- HuBei Clinical Research Center for Ischemic Cardiovascular Disease, Yichang, China
| | - Dong Zhang
- Institute of Cardiovascular Diseases, China Three Gorges University, Hubei, China
- Department of Cardiology, Yichang Central People's Hospital, Yichang, 443003, China
- Hubei Key Laboratory of Ischemic Cardiovascular Disease, Yichang, China
- HuBei Clinical Research Center for Ischemic Cardiovascular Disease, Yichang, China
| | - Qingzhuo Yang
- Institute of Cardiovascular Diseases, China Three Gorges University, Hubei, China
- Department of Cardiology, Yichang Central People's Hospital, Yichang, 443003, China
- Hubei Key Laboratory of Ischemic Cardiovascular Disease, Yichang, China
- HuBei Clinical Research Center for Ischemic Cardiovascular Disease, Yichang, China
| | - Yi Li
- Institute of Cardiovascular Diseases, China Three Gorges University, Hubei, China
- Department of Cardiology, Yichang Central People's Hospital, Yichang, 443003, China
- Hubei Key Laboratory of Ischemic Cardiovascular Disease, Yichang, China
- HuBei Clinical Research Center for Ischemic Cardiovascular Disease, Yichang, China
| | - Xiaoting Yang
- Institute of Cardiovascular Diseases, China Three Gorges University, Hubei, China
- Department of Cardiology, Yichang Central People's Hospital, Yichang, 443003, China
- Hubei Key Laboratory of Ischemic Cardiovascular Disease, Yichang, China
- HuBei Clinical Research Center for Ischemic Cardiovascular Disease, Yichang, China
| | - Jianfeng Sun
- Department of Vascular Surgery, The First College of Medical Science, Yichang Central People's Hospital, China Three Gorges University, Hubei, 443000, China
| |
Collapse
|
4
|
He DW, Liu DZ, Luo XZ, Chen CB, Lu CH, Na N, Huang F. HMGB1-RAGE axis contributes to myocardial ischemia/reperfusion injury via regulation of cardiomyocyte autophagy and apoptosis in diabetic mice. Biol Chem 2024; 405:167-176. [PMID: 37768929 DOI: 10.1515/hsz-2023-0134] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 08/22/2023] [Indexed: 09/30/2023]
Abstract
Patients with acute myocardial infarction complicated with diabetes are more likely to develop myocardial ischemia/reperfusion (I/R) injury (MI/RI) during reperfusion therapy. Both HMGB1 and RAGE play important roles in MI/RI. However, the specific mechanisms of HMGB1 associated with RAGE are not fully clarified in diabetic MI/RI. This study aimed to investigate whether the HMGB1-RAGE axis induces diabetic MI/RI via regulating autophagy and apoptosis. A db/db mouse model of MI/RI was established, where anti-HMGB1 antibody and RAGE inhibitor (FPS-ZM1) were respectively injected after 10 min of reperfusion. The results showed that treatment with anti-HMGB1 significantly reduced the infarct size, serum LDH, and CK-MB level. Similar situations also occurred in mice administrated with FPS-ZM1, though the HMGB1 level was unchanged. Then, we found that treatment with anti-HMGB1 or FPS-ZM1 performed the same effects in suppressing the autophagy and apoptosis, as reflected by the results of lower LAMP2 and LC3B levels, increased Bcl-2 level, reduced BAX and caspase-3 levels. Moreover, the Pink1/Parkin levels were also inhibited at the same time. Collectively, this study indicates that the HMGB1-RAGE axis aggravated diabetic MI/RI via apoptosis and Pink1/Parkin mediated autophagy pathways, and inhibition of HMGB1 or RAGE contributes to alleviating those adverse situations.
Collapse
Affiliation(s)
- De-Wei He
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Guangxi Key Laboratory of Precision Medicine in Cardio-cerebrovascular Diseases Control and Prevention, Guangxi Clinical Research Center for Cardio-Cerebrovascular Diseases, No.6 Shuangyong Road, Nanning 530021, Guangxi, China
| | - De-Zhao Liu
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Guangxi Key Laboratory of Precision Medicine in Cardio-cerebrovascular Diseases Control and Prevention, Guangxi Clinical Research Center for Cardio-Cerebrovascular Diseases, No.6 Shuangyong Road, Nanning 530021, Guangxi, China
| | - Xiao-Zhi Luo
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Guangxi Key Laboratory of Precision Medicine in Cardio-cerebrovascular Diseases Control and Prevention, Guangxi Clinical Research Center for Cardio-Cerebrovascular Diseases, No.6 Shuangyong Road, Nanning 530021, Guangxi, China
| | - Chuan-Bin Chen
- Department of Cardiology, The Second Affiliated Hospital of Hainan Medical University, 368 Yihai Avenue, Haikou 570216, Hainan, China
| | - Chuang-Hong Lu
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Guangxi Key Laboratory of Precision Medicine in Cardio-cerebrovascular Diseases Control and Prevention, Guangxi Clinical Research Center for Cardio-Cerebrovascular Diseases, No.6 Shuangyong Road, Nanning 530021, Guangxi, China
| | - Na Na
- Department of Chemistry, Scripps Research Institute, No.10550 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Feng Huang
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Guangxi Key Laboratory of Precision Medicine in Cardio-cerebrovascular Diseases Control and Prevention, Guangxi Clinical Research Center for Cardio-Cerebrovascular Diseases, No.6 Shuangyong Road, Nanning 530021, Guangxi, China
| |
Collapse
|
5
|
Yang X, Dong X, Li J, Zheng A, Shi W, Shen C, Liu J. Nanocurcumin attenuates pyroptosis and inflammation through inhibiting NF-κB/GSDMD signal in high altitude-associated acute liver injury. J Biochem Mol Toxicol 2024; 38:e23606. [PMID: 38050447 DOI: 10.1002/jbt.23606] [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: 05/07/2022] [Revised: 07/12/2023] [Accepted: 11/21/2023] [Indexed: 12/06/2023]
Abstract
Exposure to a hypobaric hypoxic environment at high altitudes can lead to liver injury, and mounting evidence indicates that pyroptosis and inflammation play important roles in liver injury. Curcumin (Cur) can inhibit pyroptosis and inflammation. Therefore, our purpose here was to clarify the mechanism underlying the protective effect of nanocurcumin (Ncur) and Cur in a rat model of high altitude-associated acute liver injury. Eighty healthy rats were selected and exposed to different altitudes (6000 or 7000 m) for 0, 24, 48, or 72 h. Fifty normal healthy rats were divided into normal control, high-altitude control, salidroside (40 mg/kg [Sal-40]), Cur (200 mg/kg [Cur-200]), and Ncur (25 mg/kg [Ncur-25]) groups and exposed to a high-altitude hypobaric hypoxic environment (48 h, 7000 m). Serum-liver enzyme activities (alanine transaminase, aspartate transaminase, and lactate dehydrogenase were detected and histopathology of liver injury was evaluated by hematoxylin and eosin staining, and inflammatory factors were detected in liver tissues by enzyme-linked immunosorbent assays. Pyroptosis-associated proteins (gasdermin D, gasdermin D N-terminal [GSDMD-N], pro-Caspase-1, and cleaved-Caspase-1 [cleaved-Casp1]) and inflammation-associated proteins (nuclear factor-κB [NF-κB], phospho-NF-κB [P-NF-κB], and high-mobility group protein B1 [HMGB1]) levels were analyzed by immunoblotting. Ncur and Cur inhibited increased serum-liver enzyme activities, alleviated liver injury in rats caused by high-altitude hypobaric hypoxic exposure, and downregulated inflammatory factors, including tumor necrosis factor-α, interleukin (IL)-1β, IL-6, and IL-18, in rat liver tissues. The level of P-NF-κB, GSDMD-N, cleaved-Casp1, and HMGB1 in rat liver tissues increased significantly after high-altitude exposure. Ncur and Cur downregulated P-NF-κB, GSDMD-N, cleaved-Casp-1, and HMGB1. Ncur and Cur may inhibit inflammatory responses and pyroptosis in a rat model of high altitude-associated acute liver injury.
Collapse
Affiliation(s)
- Xinyue Yang
- Key Laboratory of Special Environmental Medicine of Xinjiang, General Hospital of Xinjang Military Command, Urumqi, China
- Graduate School, Xinjiang Medical University, Urumqi, China
| | - Xiang Dong
- Graduate School, Xinjiang Medical University, Urumqi, China
| | - Jiajia Li
- Graduate School, Xinjiang Medical University, Urumqi, China
| | - Aiping Zheng
- Institute of Pharmacology and Toxicology, Academy of Military Medicine, Beijing, China
| | - Wenhui Shi
- Graduate School, Xinjiang Medical University, Urumqi, China
| | - Caifu Shen
- Graduate School, Xinjiang Medical University, Urumqi, China
| | - Jiangwei Liu
- Graduate School, Xinjiang Medical University, Urumqi, China
| |
Collapse
|
6
|
He W, Xu C, Mao D, Zheng Y, Wang N, Wang M, Mao N, Wang T, Li Y. Recent advances in pyroptosis, liver disease, and traditional Chinese medicine: A review. Phytother Res 2023; 37:5473-5494. [PMID: 37622684 DOI: 10.1002/ptr.7989] [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: 03/28/2023] [Revised: 07/29/2023] [Accepted: 08/09/2023] [Indexed: 08/26/2023]
Abstract
In recent years, the incidence of liver disease has increased, becoming a major cause of death. Various liver diseases are intricately linked to pyroptosis, which is one of the most common forms of programmed cell death. As a powerful weapon in the fight against liver diseases, traditional Chinese medicine (TCM) can affect pyroptosis via a number of routes, including the classical, nucleotide oligomerization domain-like receptors protein 3/caspase-1/gasdermin D (GSDMD) pathway, the nonclassical lipopolysaccharide/caspase-11/GSDMD pathway, the ROS/caspase-3/gasdermin E pathway, the caspase-9/caspase-3/GSDMD pathway, and the Apaf-1/caspase-11/caspase-3 pathway. In this review, we provide an overview of pyroptosis, the interplay between pyroptosis and liver diseases, and the mechanisms through which TCM regulates pyroptosis in liver diseases. The information used in the text was collected and compiled from the databases of PubMed, Web of Science, Scopus, CNKI, and Wanfang Data up to June 2023. The search was not limited with regard to the language and country of the articles. Research and review articles were included, and papers with duplicate results or unrelated content were excluded. We examined the current understanding of the relationship between pyroptosis and liver diseases as well as the advances in TCM interventions to provide a resource for the identification of potential targets for TCM in the treatment of liver diseases.
Collapse
Affiliation(s)
- Wenxing He
- Faculty of Chinese Medicine Science, Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Canli Xu
- Faculty of Chinese Medicine Science, Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Dewen Mao
- Department of Hepatology, The First Affiliated Hospital of Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Yang Zheng
- Faculty of Chinese Medicine Science, Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Na Wang
- Department of Hepatology, The First Affiliated Hospital of Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Minggang Wang
- Department of Hepatology, The First Affiliated Hospital of Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Nan Mao
- Department of Acupuncture-Moxibustion and Tuina, Jiangbin Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China
| | - Ting Wang
- The First Clinical Medical College, Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Yanjie Li
- Department of Hepatology, The First Affiliated Hospital of Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| |
Collapse
|
7
|
Meng XM, Yuan JH, Zhou ZF, Feng QP, Zhu BM. Evaluation of time-dependent phenotypes of myocardial ischemia-reperfusion in mice. Aging (Albany NY) 2023; 15:10627-10639. [PMID: 37819785 PMCID: PMC10599719 DOI: 10.18632/aging.205103] [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: 06/23/2023] [Accepted: 09/09/2023] [Indexed: 10/13/2023]
Abstract
BACKGROUND A mouse model of myocardial ischemia-reperfusion (I/R) is widely used to study myocardial ischemia-reperfusion injury (I/RI). However, few studies focus on the direct comparison of the extent of pathological events resulting from variant durations of ischemia and reperfusion process. METHODS A mouse model of I/RI was established by ligation and perfusion of the left anterior descending coronary artery (LAD), and the dynamic changes were recorded by electrocardiogram at different stages of I/R. Subsequently, reperfusion duration was used as a variable to directly compare the phenotypes of different myocardial injury degrees induced by 3 h, 6 h and 24 h reperfusion from myocardial infarct size, myocardial apoptosis, myocardial enzyme, and inflammatory cytokine levels. RESULTS All mice subjected to myocardial I/R surgery showed obvious myocardial infarction, extensive myocardial apoptosis, dynamic changes in serum myocardial enzyme and inflammatory cytokines, at least for the first 24 h of reperfusion. The infarct size and apoptosis rates gradually increased with the extension of reperfusion time. The peaks of serum myocardial enzyme and inflammatory cytokines occurred at 6 h and 3 h of reperfusion, respectively. We also established I/R mice models with 30 and 60 mins of ischemia. After 21 days of remodeling, longer periods of ischemia increased the degree of fibrosis and reduced cardiac function. CONCLUSIONS In summary, we conclude that reperfusion durations of 3 h, 6 h, and 24 h induces different injury phenotypes in ischemia-reperfusion mouse model. At the same time, the ischemia duration before reperfusion also affects the degree of cardiac remodeling.
Collapse
Affiliation(s)
- Xiang-Min Meng
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jing-Han Yuan
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zhen-Fang Zhou
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Qi-Pu Feng
- Animal Experiment Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Bing-Mei Zhu
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
8
|
Wu Z, Li M. High-Mobility Group Box 1 in Spinal Cord Injury and Its Potential Role in Brain Functional Remodeling After Spinal Cord Injury. Cell Mol Neurobiol 2023; 43:1005-1017. [PMID: 35715656 DOI: 10.1007/s10571-022-01240-5] [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: 03/20/2022] [Accepted: 06/04/2022] [Indexed: 11/30/2022]
Abstract
High-mobility group box 1 (HMGB1) is a nonhistone nuclear protein, the functions of which depend on its subcellular location. It is actively or passively secreted into the blood and/or cerebrospinal fluid (CSF) and can be used as a prognostic indicator of disease. HMGB1 released into the bloodstream can cause pathological reactions in distant organs, and entry into the CSF can destroy the blood-brain barrier and aggravate brain injuries. HMGB1 expression has been reported to be increased in the tissues of spinal cord injury (SCI) patients and involved in the regulation of neuroinflammation, neuronal apoptosis, and ferroptosis. SCI can lead to brain changes, resulting in neuropathic pain, depression, and cognitive dysfunction, but the specific mechanism is unknown. It remains unclear whether HMGB1 plays an important role in brain functional remodeling after SCI. Damaged cells at the site of SCI passively release HMGB1, which travels to the brain via the blood, CSF, and/or axonal transport, destroys the blood-brain barrier, and causes pathological changes in the brain. This may explain the remodeling of brain function that occurs after SCI. In this minireview, we introduce the structure and function of HMGB1 and its mechanism of action in SCI. Clarifying the functions of HMGB1 may provide insight into the links between SCI and various brain regions.
Collapse
Affiliation(s)
- Zhiwu Wu
- Department of Neurosurgery & Jiangxi Key Laboratory of Neurosurgery, The First Affiliated Hospital of Nanchang University, 17th Yongwaizheng Street, Nanchang, 330006, China
| | - Meihua Li
- Department of Neurosurgery & Jiangxi Key Laboratory of Neurosurgery, The First Affiliated Hospital of Nanchang University, 17th Yongwaizheng Street, Nanchang, 330006, China.
| |
Collapse
|
9
|
He Y, Wang Y, Yang K, Jiao J, Zhan H, Yang Y, Lv D, Li W, Ding W. Maslinic Acid: A New Compound for the Treatment of Multiple Organ Diseases. Molecules 2022; 27:8732. [PMID: 36557864 PMCID: PMC9786823 DOI: 10.3390/molecules27248732] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 12/03/2022] [Accepted: 12/07/2022] [Indexed: 12/13/2022] Open
Abstract
Maslinic acid (MA) is a pentacyclic triterpene acid, which exists in many plants, including olive, and is highly safe for human beings. In recent years, it has been reported that MA has anti-inflammatory, antioxidant, anti-tumor, hypoglycemic, neuroprotective and other biological activities. More and more experimental data has shown that MA has a good therapeutic effect on multiple organ diseases, indicating that it has great clinical application potential. In this paper, the extraction, purification, identification and analysis, biological activity, pharmacokinetics in vivo and molecular mechanism of MA in treating various organ diseases are reviewed. It is hoped to provide a new idea for MA to treat various organ diseases.
Collapse
Affiliation(s)
- Yan He
- Department of Fundamental Medicine, Chengdu University of Traditional Chinese Medicine, 1166 Liutai Avenue, Chengdu 611137, China
| | - Yi Wang
- Department of Fundamental Medicine, Chengdu University of Traditional Chinese Medicine, 1166 Liutai Avenue, Chengdu 611137, China
| | - Kun Yang
- Department of Fundamental Medicine, Chengdu University of Traditional Chinese Medicine, 1166 Liutai Avenue, Chengdu 611137, China
| | - Jia Jiao
- Department of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, 1166 Liutai Avenue, Chengdu 611137, China
| | - Hong Zhan
- Department of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, 1166 Liutai Avenue, Chengdu 611137, China
| | - Youjun Yang
- Department of Fundamental Medicine, Chengdu University of Traditional Chinese Medicine, 1166 Liutai Avenue, Chengdu 611137, China
| | - De Lv
- Affiliated Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Weihong Li
- Department of Fundamental Medicine, Chengdu University of Traditional Chinese Medicine, 1166 Liutai Avenue, Chengdu 611137, China
| | - Weijun Ding
- Department of Fundamental Medicine, Chengdu University of Traditional Chinese Medicine, 1166 Liutai Avenue, Chengdu 611137, China
| |
Collapse
|
10
|
7-Hydroxyflavone Alleviates Myocardial Ischemia/Reperfusion Injury in Rats by Regulating Inflammation. Molecules 2022; 27:molecules27175371. [PMID: 36080137 PMCID: PMC9458087 DOI: 10.3390/molecules27175371] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 08/17/2022] [Accepted: 08/22/2022] [Indexed: 11/18/2022] Open
Abstract
Inflammation is the primary pathological process of myocardial ischemia/reperfusion injury (MI/RI). 7-Hydroxyflavone (HF), a natural flavonoid with a variety of bioactivities, plays a crucial role in various biological processes. However, its cardioprotective effects and the underlying mechanisms of MI/RI have not been investigated. This study aimed to explore whether pretreatment with HF could attenuate MI/RI-induced inflammation in rats and investigate its potential mechanisms. The results showed that pretreatment with HF could significantly improve the anatomic data and electrocardiograph parameters, reduce the myocardial infarct size, decrease markers of myocardial injury (aspartate transaminase, creatine kinase, lactate dehydrogenase, and cardiac troponin I), inhibit inflammatory cytokines (IL-1β, IL-6, and TNF-α), suppress oxidative stress, and recover the architecture of the cardiomyocytes. The cardioprotective effect of HF was connected with the regulation of the MAPK/NF-κB signaling pathway. What is more, molecular docking was carried out to prove that HF could be stably combined with p38, ERK1/2, JNK, and NF-κB. In summary, this is a novel study demonstrating the cardioprotective effects of HF against MI/RI in vivo. Consequently, these results demonstrate that HF can be considered a promising potential therapy for MI/RI.
Collapse
|
11
|
Recent Advances Regarding the Molecular Mechanisms of Triterpenic Acids: A Review (Part II). Int J Mol Sci 2022; 23:ijms23168896. [PMID: 36012159 PMCID: PMC9408012 DOI: 10.3390/ijms23168896] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 08/04/2022] [Accepted: 08/08/2022] [Indexed: 12/18/2022] Open
Abstract
Triterpenic acids are a widespread class of phytocompounds which have been found to possess valuable therapeutic properties such as anticancer, anti-inflammatory, hepatoprotective, cardioprotective, antidiabetic, neuroprotective, lipolytic, antiviral, and antiparasitic effects. They are a subclass of triterpenes bearing a characteristic lipophilic structure that imprints unfavorable in vivo properties which subsequently limit their applications. The early investigation of the mechanism of action (MOA) of a drug candidate can provide valuable information regarding the possible side effects and drug interactions that may occur after administration. The current paper aimed to summarize the most recent (last 5 years) studies regarding the MOA of betulinic acid, boswellic acid, glycyrrhetinic acid, madecassic acid, moronic acid, and pomolic acid in order to provide scientists with updated and accessible material on the topic that could contribute to the development of future studies; the paper stands as the sequel of our previously published paper regarding the MOA of triterpenic acids with therapeutic value. The recent literature published on the topic has highlighted the role of triterpenic acids in several signaling pathways including PI3/AKT/mTOR, TNF-alpha/NF-kappa B, JNK-p38, HIF-α/AMPK, and Grb2/Sos/Ras/MAPK, which trigger their various biological activities.
Collapse
|
12
|
Jin L, Yin Q, Mao Y, Gao Y, Han Q, Mei R, Xue L, Tan H, Li H. Putative Prevention of XML Injection Against Myocardial Ischemia Is Mediated by PKC and PLA2 Proteins. Front Cell Dev Biol 2022; 10:827691. [PMID: 35141226 PMCID: PMC8819063 DOI: 10.3389/fcell.2022.827691] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 01/03/2022] [Indexed: 12/15/2022] Open
Abstract
Background: Xinmailong (XML) injection is a CFDA-approved traditional Chinese medicine with clinical value for heart failure treatment. The present investigation was aimed to evaluate the potential protective roles of this injection on myocardial ischemia and the underlying molecular mechanism. Methods: In our study, we selected two models of myocardial ischemia rats. Rats were randomly divided into six groups, with saline or XML administrated 4 days before ischemia model establishment. ECG of different time intervals and biochemical parameters of end point were measured. The potential mechanisms of the protective role of XML were explored using system pharmacology and molecular biology approaches. Results: Myocardial ischemia rats demonstrated abnormal ECG and serum levels of cTnT. Pretreatment with XML significantly attenuated these damages, especially the medium doses. GO and KEGG analysis revealed that the 90 putative target genes were associated with pathways of fatty acid absorption/metabolism, inflammation, RAAS, and vascular smooth muscle. Further network pharmacology method identified five main chemical ingredients and potential targets of XML injection for myocardial ischemia. Mechanically, the beneficial effect of XML injection was mediated by the reactive oxygen species (ROS) inhibition and inflammation attenuation via regulating the expression levels of targets of PKC and PLA2. Conclusion: These findings indicate that XML exerts protective effects against myocardial injury, with attenuated ROS production, apoptosis, and inflammation. Therefore, we speculate that XML may be an alternative supplementary therapeutic agent for myocardial ischemia prevention.
Collapse
Affiliation(s)
- Ling Jin
- Center of Basic Medical Research, Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, China
| | - Qianqian Yin
- Center of Basic Medical Research, Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, China
| | - Yiqing Mao
- Department of Pharmacology, Peking University, Health Science Center, Beijing, China
| | - Yuanxu Gao
- State Key Laboratory of Lunar and Planetary Sciences, Macau University of Science and Technology, Macau, China
| | - Qing Han
- Department of Pharmacology, Peking University, Health Science Center, Beijing, China
| | - Ruisi Mei
- Department of Pharmacology, Peking University, Health Science Center, Beijing, China
| | - Lixiang Xue
- Center of Basic Medical Research, Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, China
| | - Huanran Tan
- Department of Pharmacology, Peking University, Health Science Center, Beijing, China
- *Correspondence: Huanran Tan, ; Hui Li,
| | - Hui Li
- Department of Pharmacology, Peking University, Health Science Center, Beijing, China
- *Correspondence: Huanran Tan, ; Hui Li,
| |
Collapse
|