1
|
Li J, Yu Y, Zhang Y, Zhou Y, Ding S, Dong S, Jin S, Li Q. Flavonoids Derived from Chinese Medicine: Potential Neuroprotective Agents. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2024; 52:1613-1640. [PMID: 39343989 DOI: 10.1142/s0192415x24500630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Due to their complex pathological mechanisms, neurodegenerative diseases have brought great challenges to drug development and clinical treatment. Studies have shown that many traditional Chinese medicines have neuroprotective pharmacological activities such as anti-inflammatory and anti-oxidation properties and have certain effects on improving the symptoms of neurodegenerative diseases and delaying disease progression. Flavonoids are the main active components of many traditional Chinese medicines for the treatment of neurodegenerative diseases. These compounds have a wide range of biological activities, including anti-inflammatory, anti-oxidative stress, regulation of autophagy balance, inhibition of apoptosis, and promotion of neuronal regeneration. This paper focuses on the neuroprotective effects of six common flavonoids: quercetin, rutin, luteolin, kaempferol, baicalein, and puerarin. It then systematically reviews their characteristics, mechanisms, and key signaling pathways, summarizes the common characteristics and laws of their neuroprotective effects, and discusses the significance of strengthening the research on the neuroprotective effects of these compounds, aiming to provide reference for more research and drug development of these substances as neuroprotective drugs.
Collapse
Affiliation(s)
- Jinhua Li
- Hangzhou Medical College, Xihu District, Hangzhou, Zhejiang, 310013, P. R. China
- Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, Hangzhou Medical College, Hangzhou, Zhejiang, 310013, P. R. China
| | - Ye Yu
- Hangzhou Medical College, Xihu District, Hangzhou, Zhejiang, 310013, P. R. China
- Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, Hangzhou Medical College, Hangzhou, Zhejiang, 310013, P. R. China
| | - Yanjie Zhang
- Hangzhou Medical College, Xihu District, Hangzhou, Zhejiang, 310013, P. R. China
- Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, Hangzhou Medical College, Hangzhou, Zhejiang, 310013, P. R. China
| | - Yilin Zhou
- Hangzhou Medical College, Xihu District, Hangzhou, Zhejiang, 310013, P. R. China
- Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, Hangzhou Medical College, Hangzhou, Zhejiang, 310013, P. R. China
| | - Shuxian Ding
- Hangzhou Medical College, Xihu District, Hangzhou, Zhejiang, 310013, P. R. China
- Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, Hangzhou Medical College, Hangzhou, Zhejiang, 310013, P. R. China
| | - Shuze Dong
- Hangzhou Medical College, Xihu District, Hangzhou, Zhejiang, 310013, P. R. China
- Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, Hangzhou Medical College, Hangzhou, Zhejiang, 310013, P. R. China
| | - Sainan Jin
- Hangzhou Medical College, Xihu District, Hangzhou, Zhejiang, 310013, P. R. China
- Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, Hangzhou Medical College, Hangzhou, Zhejiang, 310013, P. R. China
| | - Qin Li
- Hangzhou Medical College, Xihu District, Hangzhou, Zhejiang, 310013, P. R. China
- Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, Hangzhou Medical College, Hangzhou, Zhejiang, 310013, P. R. China
| |
Collapse
|
2
|
Yin J, Huang Y, Wang K, Zhong Q, Liu Y, Ji Z, Liao Y, Ma Z, Bei W, Wang W. Ginseng extract improves pancreatic islet injury and promotes β-cell regeneration in T2DM mice. Front Pharmacol 2024; 15:1407200. [PMID: 38989151 PMCID: PMC11234855 DOI: 10.3389/fphar.2024.1407200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 06/03/2024] [Indexed: 07/12/2024] Open
Abstract
Introduction Panax ginseng C. A. Mey. (Araliaceae; Ginseng Radix et Rhizoma), a traditional plant commonly utilized in Eastern Asia, has demonstrated efficacy in treating neuro-damaging diseases and diabetes mellitus. However, its precise roles and mechanism in alleviating type 2 diabetes mellitus (T2DM) need further study. The objective of this study is to explore the pharmacological effects of ginseng extract and elucidate its potential mechanisms in protecting islets and promoting β-cell regeneration. Methods The T2DM mouse model was induced through streptozotocin combined with a high-fat diet. Two batches of mice were sacrificed on the 7th and 28th days following ginseng extract administration. Body weight, fasting blood glucose levels, and glucose tolerance were detected. Morphological changes in the pancreatic islets were examined via H & E staining. Levels of serum insulin, glucagon, GLP-1, and inflammatory factors were measured using ELISA. The ability of ginseng extract to promote pancreatic islet β-cell regeneration was evaluated through insulin & PCNA double immunofluorescence staining. Furthermore, the mechanism behind β-cells regeneration was explored through insulin & glucagon double immunofluorescence staining, accompanied by immunohistochemical staining and western blot analyses. Results and Discussion The present research revealed that ginseng extract alleviates symptoms of T2DM in mice, including decreased blood glucose levels and improved glucose tolerance. Serum levels of insulin, GLP-1, and IL-10 increased following the administration of ginseng extract, while levels of glucagon, TNF-α, and IL-1β decreased. Ginseng extract preserved normal islet morphology, increased nascent β-cell population, and inhibited inflammatory infiltration within the islets, moreover, it decreased α-cell proportion while increasing β-cell proportion. Mechanistically, ginseng extract might inhibit ARX and MAFB expressions, increase MAFA level to aid in α-cell to β-cell transformation, and activate AKT-FOXM1/cyclin D2 to enhance β-cell proliferation. Our study suggests that ginseng extract may be a promising therapy in treating T2DM, especially in those with islet injury.
Collapse
Affiliation(s)
- Jianying Yin
- Traditional Chinese Medicine Research Institute, Guangdong Pharmaceutical University, Guangzhou, Guangdong, China
- Guangdong Provincial Research Center of Integration of Traditional Chinese Medicine and Western Medicine in Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou, Guangdong, China
| | - Yuanfeng Huang
- Traditional Chinese Medicine Research Institute, Guangdong Pharmaceutical University, Guangzhou, Guangdong, China
- Guangdong Provincial Research Center of Integration of Traditional Chinese Medicine and Western Medicine in Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou, Guangdong, China
| | - Ke Wang
- Traditional Chinese Medicine Research Institute, Guangdong Pharmaceutical University, Guangzhou, Guangdong, China
- Guangdong Provincial Research Center of Integration of Traditional Chinese Medicine and Western Medicine in Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou, Guangdong, China
| | - Qin Zhong
- Traditional Chinese Medicine Research Institute, Guangdong Pharmaceutical University, Guangzhou, Guangdong, China
- Guangdong Provincial Research Center of Integration of Traditional Chinese Medicine and Western Medicine in Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou, Guangdong, China
| | - Yuan Liu
- Traditional Chinese Medicine Research Institute, Guangdong Pharmaceutical University, Guangzhou, Guangdong, China
- Guangdong Provincial Research Center of Integration of Traditional Chinese Medicine and Western Medicine in Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou, Guangdong, China
| | - Zirui Ji
- Traditional Chinese Medicine Research Institute, Guangdong Pharmaceutical University, Guangzhou, Guangdong, China
- Guangdong Provincial Research Center of Integration of Traditional Chinese Medicine and Western Medicine in Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou, Guangdong, China
| | - Yiwen Liao
- Traditional Chinese Medicine Research Institute, Guangdong Pharmaceutical University, Guangzhou, Guangdong, China
- Guangdong Provincial Research Center of Integration of Traditional Chinese Medicine and Western Medicine in Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou, Guangdong, China
| | - Zhiyuan Ma
- Baishan Institute of Science and Technology, Baishan, Jilin, China
| | - Weijian Bei
- Traditional Chinese Medicine Research Institute, Guangdong Pharmaceutical University, Guangzhou, Guangdong, China
- Guangdong Provincial Research Center of Integration of Traditional Chinese Medicine and Western Medicine in Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou, Guangdong, China
| | - Weixuan Wang
- Traditional Chinese Medicine Research Institute, Guangdong Pharmaceutical University, Guangzhou, Guangdong, China
- Guangdong Provincial Research Center of Integration of Traditional Chinese Medicine and Western Medicine in Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou, Guangdong, China
| |
Collapse
|
3
|
Yu M, Kong XY, Chen TT, Zou ZM. In vivo metabolism combined network pharmacology to identify anti-constipation constituents in Aloe barbadensis Mill. JOURNAL OF ETHNOPHARMACOLOGY 2024; 319:117200. [PMID: 37726070 DOI: 10.1016/j.jep.2023.117200] [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: 07/30/2023] [Revised: 09/03/2023] [Accepted: 09/16/2023] [Indexed: 09/21/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Aloe barbadensis Mill. (Aloe vera), is a homology of medicine and food plant, widely applied in functional food, cosmetics, and medicine. Aloe vera whole leaf extract, one of the most popular traditional Chinese medicines (TCMs), is mainly used in China to treat functional constipation. AIM OF THE STUDY To elucidate the active constituents of Aloe vera for treating functional constipation. MATERIALS AND METHODS Prototype constituents and metabolites in rat plasma and excreta after oral administration of Aloe vera whole leaf extract were identified by UPLC-Q-TOF/MS, and the pharmacokinetics (PK) properties of its key anti-constipation constituents speculated by network pharmacology were investigated via the established UFLC-MS/MS method. RESULTS A total of 13 prototype constituents and 56 metabolites were identified in rat plasma, urine, and feces after oral administration of Aloe vera. Among them, aloesin, aloenin, aloin B, aloin A, and aloe-emodin were intimately connected to the core targets of constipation in network pharmacology analysis, and recognized as major anti-constipation constituents in Aloe vera. The validated quantitative method of the six active constituents in rat plasma exhibited good linearity, and lower limits of quantification (0.64-1.95 ng/mL). Aloin A, aloin B, aloeresin D and aloe-emodin exhibited better absorption and slower elimination rate, whereas the others, including aloesin and aloenin showed fast absorption and elimination in rat plasma after oral administration of Aloe vera. Aloin A and its isomer aloin B present similar Tmax and t1/2 but different AUC and Cmax values, indicating different relative bioavailability. The results suggested that aloin A, aloin B and aloe-emodin may be key constituents of Aloe vera for the treatment of constipation, and the other constituents including aloeresin D also contribute to its anti-constipation. CONCLUSIONS This study will benefit understanding the contributions of those constituents for the anti-constipation effect of Aloe vera and also provide valuable information for its application in functional food development and clinics.
Collapse
Affiliation(s)
- Meng Yu
- The Institute of Medicinal Plant Development, State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100193, China.
| | - Xin-Yu Kong
- Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China.
| | - Tong-Tong Chen
- Tianjin Institute for Drug Control, Tianjin, 300070, China.
| | - Zhong-Mei Zou
- The Institute of Medicinal Plant Development, State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100193, China.
| |
Collapse
|
4
|
Mu N, Li J, Zeng L, You J, Li R, Qin A, Liu X, Yan F, Zhou Z. Plant-Derived Exosome-Like Nanovesicles: Current Progress and Prospects. Int J Nanomedicine 2023; 18:4987-5009. [PMID: 37693885 PMCID: PMC10492547 DOI: 10.2147/ijn.s420748] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 08/28/2023] [Indexed: 09/12/2023] Open
Abstract
Exosomes are small extracellular vesicles, ranging in size from 30-150nm, which can be derived from various types of cells. In recent years, mammalian-derived exosomes have been extensively studied and found to play a crucial role in regulating intercellular communication, thereby influencing the development and progression of numerous diseases. Traditional Chinese medicine has employed plant-based remedies for thousands of years, and an increasing body of evidence suggests that plant-derived exosome-like nanovesicles (PELNs) share similarities with mammalian-derived exosomes in terms of their structure and function. In this review, we provide an overview of recent advances in the study of PELNs and their potential implications for human health. Specifically, we summarize the roles of PELNs in respiratory, digestive, circulatory, and other diseases. Furthermore, we have extensively investigated the potential shortcomings and challenges in current research regarding the mechanism of action, safety, administration routes, isolation and extraction methods, characterization and identification techniques, as well as drug-loading capabilities. Based on these considerations, we propose recommendations for future research directions. Overall, our review highlights the potential of PELNs as a promising area of research, with broad implications for the treatment of human diseases. We anticipate continued interest in this area and hope that our summary of recent findings will stimulate further exploration into the implications of PELNs for human health.
Collapse
Affiliation(s)
- Nai Mu
- Department of Clinical Medicine, North Sichuan Medical College, Nanchong, Sichuan Province, People’s Republic of China
- Geriatric Diseases Institute of Chengdu, Department of Orthopedics, Chengdu Fifth People’s Hospital, Chengdu, Sichuan Province, People’s Republic of China
| | - Jie Li
- Center for Medicine Research and Translation, Chengdu Fifth People’s Hospital, Chengdu, Sichuan Province, People’s Republic of China
| | - Li Zeng
- Department of Pharmacy, Chengdu Fifth People’s Hospital, Chengdu, Sichuan Province, People’s Republic of China
| | - Juan You
- Department of Pharmacy, Chengdu Fifth People’s Hospital, Chengdu, Sichuan Province, People’s Republic of China
| | - Rong Li
- Department of Pharmacy, Chengdu Fifth People’s Hospital, Chengdu, Sichuan Province, People’s Republic of China
| | - Anquan Qin
- Department of Pharmacy, Chengdu Fifth People’s Hospital, Chengdu, Sichuan Province, People’s Republic of China
| | - Xueping Liu
- Department of Clinical Medicine, North Sichuan Medical College, Nanchong, Sichuan Province, People’s Republic of China
| | - Fang Yan
- Center for Medicine Research and Translation, Chengdu Fifth People’s Hospital, Chengdu, Sichuan Province, People’s Republic of China
- Geriatric Diseases Institute of Chengdu, Department of Geriatrics, Chengdu Fifth People’s Hospital, Chengdu, Sichuan Province, People’s Republic of China
| | - Zheng Zhou
- Department of Clinical Medicine, North Sichuan Medical College, Nanchong, Sichuan Province, People’s Republic of China
- Geriatric Diseases Institute of Chengdu, Department of Orthopedics, Chengdu Fifth People’s Hospital, Chengdu, Sichuan Province, People’s Republic of China
| |
Collapse
|
5
|
Li J, Liao R, Zhang S, Weng H, Liu Y, Tao T, Yu F, Li G, Wu J. Promising remedies for cardiovascular disease: Natural polyphenol ellagic acid and its metabolite urolithins. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 116:154867. [PMID: 37257327 DOI: 10.1016/j.phymed.2023.154867] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 04/17/2023] [Accepted: 05/08/2023] [Indexed: 06/02/2023]
Abstract
BACKGROUND Cardiovascular disease (CVD) is a significant worldwide factor contributing to human fatality and morbidity. With the increase of incidence rates, it is of concern that there is a lack of current therapeutic alternatives because of multiple side effects. Ellagic acid (EA), the natural polyphenol (C14H6O8), is abundant in pomegranates, berries, and nuts. EA and its intestinal microflora metabolite, urolithins, have recently attracted much attention as a potential novel "medicine" because of their wide pharmacological properties. PURPOSE This study aimed to critically analyze available literature to summarize the beneficial effects of EA and urolithins, and highlights their druggability and therapeutic potential in various CVDs. METHODS We systematically studied research and review articles between 1984 and 2022 available on various databases to obtain the data on EA and urolithins with no language restriction. Their cardiovascular protective activities, underlying mechanism, and druggability were highlighted and discussed comprehensively. RESULTS We found that EA and urolithins may exert preventive and curative effects on CVD with negligible side effects and possibly regulate lipid metabolism imbalance, pro-inflammatory factor production, vascular smooth muscle cell proliferation, cardiomyocyte apoptosis, endothelial cell dysfunction, and Ca2+ intake and release. Potentially, this may lead to the prevention and amelioration of atherosclerosis, hypertension, myocardial infarction, cardiac fibrosis, cardiomyopathy, cardiac arrhythmias, and cardiotoxicities in vivo. Several molecules and signaling pathways are associated with their therapeutic actions, including phosphatidylinositol 3-kinase/protein kinase B, mitogen-activated protein kinase, NF-κB, nuclear factor erythroid-2 related factor 2, sirtuin1, miRNA, and extracellular signal-regulated kinase 1/2. CONCLUSION In vitro and in vivo studies shows that EA and urolithins could be used as valid candidates for early prevention and effective therapeutic strategies for various CVDs.
Collapse
Affiliation(s)
- Jingyan Li
- Cardiovascular Surgery Department, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China; Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Drugability Evaluation for Chinese Materia Medica, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China; Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention of Cardiovascular Diseases, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan, China
| | - Ruixue Liao
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Drugability Evaluation for Chinese Materia Medica, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Shijia Zhang
- School of Pharmacy, Xuzhou Medical University, Xuzhou 221000, China
| | - Huimin Weng
- Cardiovascular Surgery Department, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Yuanzhi Liu
- Cardiovascular Surgery Department, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China; Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Drugability Evaluation for Chinese Materia Medica, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Tianyi Tao
- Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention of Cardiovascular Diseases, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan, China
| | - Fengxu Yu
- Cardiovascular Surgery Department, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China.
| | - Guang Li
- Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention of Cardiovascular Diseases, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan, China.
| | - Jianming Wu
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Drugability Evaluation for Chinese Materia Medica, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China; School of Basic Medical Sciences, Southwest Medical University, Luzhou, China.
| |
Collapse
|
6
|
Jin Z, Luo Y, Zhao H, Cui J, He W, Li J, Pi J, Qing L, Wu W. Qingre Huoxue Decoction regulates macrophage polarisation to attenuate atherosclerosis through the inhibition of NF-κB signalling-mediated inflammation. JOURNAL OF ETHNOPHARMACOLOGY 2023; 301:115787. [PMID: 36206868 DOI: 10.1016/j.jep.2022.115787] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 09/24/2022] [Accepted: 09/29/2022] [Indexed: 06/16/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Atherosclerosis (AS) is a common pathogenesis of cardiovascular diseases. Qingre Huoxue Decoction (QRHX) is an herbal formula used for the prevention and treatment of AS. However, the potential mechanism of QRHX is not clear. AIM OF THE STUDY In our study, RNA sequencing combined with preclinical models were used to analyse the effect and mechanism of QRHX for the treatment of AS. MATERIALS AND METHODS For in vivo studies, ApoE-/- mice were fed with a high-fat diet to induce AS. We measured weight, blood lipid, inflammatory cytokines, lipid deposition, plaque, and the M1/M2 macrophage. For in vitro studies, RAW264.7 were induced by lipopolysaccharides and treated with different concentrations of QRHX. We focusd on the relationship between QRHX, the NF-κB pathway, and macrophage polarisation, and performed simultaneous RNA sequencing both in vivo and in vitro. RESULTS In vivo, QRHX decreased weight, improved blood lipid, relieved the degree of lipid deposition, reduced plaque area, decreased the levels of inflammatory cytokines (MCP-1, NLRP3, and TNFα), down-regulated the expression of iNOS, and up-regulated the expression of Arg-1. In vitro, QRHX down-regulated M1 markers, iNOS and CCR7, with lower concentrations of IL-1β; furthermore, QRHX up-regulated M2 markers, Arg-1, CD163, Ym-1, and Fizz-1, with higher concentrations of IL-4 and IL-10. RNA sequencing of both samples in vivo and in vitro suggested that NF-κB was the target pathway of QRHX to regulate macrophage polarisation; this result was validated at the gene and protein levels. CONCLUSIONS QRHX induced M2 polarisation, reduced an inflammatory response, and played a role in stabilising plaque by mediating the NF-κB signalling pathway.
Collapse
Affiliation(s)
- Zheng Jin
- Department of Cardiology and Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Yuan Luo
- Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510405, China
| | - Huanyi Zhao
- First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510405, China
| | - Jiayan Cui
- Department of Cardiology and Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Weifeng He
- Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510405, China
| | - Junlong Li
- First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510405, China
| | - Jianbin Pi
- Foshan Hospital of Traditional Chinese Medicine, Foshan, Guangdong, 528099, China
| | - Lijin Qing
- First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510405, China.
| | - Wei Wu
- First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510405, China.
| |
Collapse
|
7
|
|
8
|
Combined Therapy with Traditional Chinese Medicine and Antiplatelet Drugs for Ischemic Heart Disease: Mechanism, Efficacy, and Safety. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:9956248. [PMID: 34745309 PMCID: PMC8566037 DOI: 10.1155/2021/9956248] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 10/18/2021] [Indexed: 02/06/2023]
Abstract
Ischemic heart disease is a significant risk factor that threatens human health, and antiplatelet drugs are routinely used to treat cases in clinical settings. Chinese medicine for promoting blood circulation and removing blood stasis (PBCRBSCM) can often be combined with antiplatelet drugs to treat ischemic heart disease. PBCRBSCM can inhibit platelet adhesion, activation, and aggregation; moreover, PBCRBSCM in combination with antiplatelet drugs exerts antiplatelet effects. The mechanism is related to several factors, including the inhibition of platelet activation and aggregation, improvement of the hemodynamic status and coagulation function, and correction of metabolism and inflammation. PBCRBSCM can also regulate the absorption and metabolism of conventional antiplatelet drugs and protect the gastric mucosal epithelial cells against damage induced by conventional antiplatelet drugs. Randomized controlled trials have confirmed that PBCRBSCM preparations and the active ingredients in these preparations can reduce resistance to aspirin and clopidogrel so that the combination of these drugs can exert their antiplatelet effects. In the perioperative treatment of patients with stable angina pectoris, unstable angina pectoris, and acute coronary syndrome undergoing percutaneous coronary intervention therapy, preparations of the active ingredients of PBCRBSCM combined with antiplatelet drugs and other conventional Western medicine treatments have been proven effective. The efficacy and safety of such combinations have also been extensively verified. Considerable progress has been made to understand the antiplatelet mechanism of PBCRBSCM. However, most clinical studies had problems, such as limited sample size and inappropriate research design, which has limited the translational use of PBCRBSCM in antiplatelet therapy. A large-scale, multicenter, randomized controlled study with cardiovascular events as the endpoint is still to be conducted to provide evidence for the combined application of PBCRBSCM and antiplatelet drugs in the prevention and treatment of ischemic heart disease.
Collapse
|
9
|
CYP1B1 as a therapeutic target in cardio-oncology. Clin Sci (Lond) 2021; 134:2897-2927. [PMID: 33185690 PMCID: PMC7672255 DOI: 10.1042/cs20200310] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 10/12/2020] [Accepted: 10/28/2020] [Indexed: 02/06/2023]
Abstract
Cardiovascular complications have been frequently reported in cancer patients and survivors, mainly because of various cardiotoxic cancer treatments. Despite the known cardiovascular toxic effects of these treatments, they are still clinically used because of their effectiveness as anti-cancer agents. In this review, we discuss the growing body of evidence suggesting that inhibition of the cytochrome P450 1B1 enzyme (CYP1B1) can be a promising therapeutic strategy that has the potential to prevent cancer treatment-induced cardiovascular complications without reducing their anti-cancer effects. CYP1B1 is an extrahepatic enzyme that is expressed in cardiovascular tissues and overexpressed in different types of cancers. A growing body of evidence is demonstrating a detrimental role of CYP1B1 in both cardiovascular diseases and cancer, via perturbed metabolism of endogenous compounds, production of carcinogenic metabolites, DNA adduct formation, and generation of reactive oxygen species (ROS). Several chemotherapeutic agents have been shown to induce CYP1B1 in cardiovascular and cancer cells, possibly via activating the Aryl hydrocarbon Receptor (AhR), ROS generation, and inflammatory cytokines. Induction of CYP1B1 is detrimental in many ways. First, it can induce or exacerbate cancer treatment-induced cardiovascular complications. Second, it may lead to significant chemo/radio-resistance, undermining both the safety and effectiveness of cancer treatments. Therefore, numerous preclinical studies demonstrate that inhibition of CYP1B1 protects against chemotherapy-induced cardiotoxicity and prevents chemo- and radio-resistance. Most of these studies have utilized phytochemicals to inhibit CYP1B1. Since phytochemicals have multiple targets, future studies are needed to discern the specific contribution of CYP1B1 to the cardioprotective and chemo/radio-sensitizing effects of these phytochemicals.
Collapse
|
10
|
Magnetic Nanomaterials in Chinese Medicine Chemical Composition Analysis and Drug Metabolism and Its Industry Prospect and Development Path Research. J CHEM-NY 2020. [DOI: 10.1155/2020/1234269] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
The paramagnetism and superparamagnetism of magnetic nanomaterials are very important for in vivo applications. The magnetic particles with paramagnetism or superparamagnetism can redistribute the magnetic particles after the magnetic field is withdrawn, which is widely used for the separation and purification of biomolecules. At the same time, superparamagnetic particles can also be used as MRI imaging contrast agent. Compared with Western medicine, traditional Chinese medicine is different from Western medicine in that it is originated from nature and has thousands of years of clinical efficacy. Therefore, we hope to elaborate the complex mechanism of traditional Chinese medicine through some modern technical means: through the establishment of relevant quality control system, Chinese medicine will be recognized and popularized in the international field. Therefore, this paper discusses the application of magnetic nanomaterials in the chemical composition analysis and drug metabolism of traditional Chinese medicine and its industrial prospect and development path. Firstly, the advantages of magnetic nanomaterials and the shortcomings of chemical composition analysis technology of traditional Chinese medicine are analyzed theoretically. Then, through the experimental simulation, the results show that, under the optimal conditions, the magnetic nanomaterials can be used to analyze the chemical composition of traditional Chinese medicine. The peak current and concentration of THP showed a good linear relationship in the range of 5.2 × 10−8 ∼ 2.1 × 10−5 mol/L, and the detection limit was 1.9 × 10−7 mol/L. Moreover, it showed effective results in repeatability, stability, and interference tests. Therefore, magnetic nanomaterials play an important role in the chemical composition analysis and drug metabolism of traditional Chinese medicine as well as its industrial prospect and development path.
Collapse
|
11
|
Natural Drugs as a Treatment Strategy for Cardiovascular Disease through the Regulation of Oxidative Stress. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:5430407. [PMID: 33062142 PMCID: PMC7537704 DOI: 10.1155/2020/5430407] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 09/02/2020] [Accepted: 09/09/2020] [Indexed: 02/07/2023]
Abstract
Oxidative stress (OS) refers to the physiological imbalance between oxidative and antioxidative processes leading to increased oxidation, which then results in the inflammatory infiltration of neutrophils, increased protease secretion, and the production of a large number of oxidative intermediates. Oxidative stress is considered an important factor in the pathogenesis of cardiovascular disease (CVD). At present, active components of Chinese herbal medicines (CHMs) have been widely used for the treatment of CVD, including coronary heart disease and hypertension. Since the discovery of artemisinin for the treatment of malaria by Nobel laureate Youyou Tu, the therapeutic effects of active components of CHM on various diseases have been widely investigated by the medical community. It has been found that various active CHM components can regulate oxidative stress and the circulatory system, including ginsenoside, astragaloside, and resveratrol. This paper reviews advances in the use of active CHM components that modulate oxidative stress, suggesting potential drugs for the treatment of various CVDs.
Collapse
|
12
|
Guo S, Wu J, Zhou W, Liu X, Zhang J, Jia S, Meng Z, Liu S, Ni M, Liu Y. Investigating the multi-target pharmacological mechanism of danhong injection acting on unstable angina by combined network pharmacology and molecular docking. BMC Complement Med Ther 2020; 20:66. [PMID: 32122353 PMCID: PMC7076845 DOI: 10.1186/s12906-020-2853-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Accepted: 02/11/2020] [Indexed: 02/06/2023] Open
Abstract
Background Danhong injection (DHI), which is one of the most well-known Traditional Chinese Medicine (TCM) injections, widely used to treat unstable angina (UA). However, its underlying pharmacological mechanisms need to be further clarified. Methods In the present study, network pharmacology was adopted. Firstly, the relative compounds were obtained by a wide-scaled literatures-mining and potential targets of these compounds by target fishing were collected. Then, we built the UA target database by DisGeNET, DigSee, TTD, OMIM. Based on data, protein-protein interaction (PPI) analysis, GO and KEGG pathway enrichment analysis were performed and screen the hub targets by topology. Furthermore, evaluation of the binding potential of key targets and compounds through molecular docking. Results The results showed that 12 ingredients of DHI and 27 putative known therapeutic targets were picked out. By systematic analysis, identified 4 hub targets (TNF, TLR4, NFKB1 and SERPINE1) mainly involved in the complex treating effects associated with coagulation and hemostasis, cell membrane region, platelet alpha granule, NF-kappa B signaling pathway and TNF signaling pathway. Conclusion The results of this study preliminarily explained the potential targets and signaling pathways of DHI in the treatment of UA, which may help to laid a good foundation for experimental research and further clinical application.
Collapse
Affiliation(s)
- Siyu Guo
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, No. 11 of North Three-ring East Road, Chao Yang District, Beijing, China
| | - Jiarui Wu
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, No. 11 of North Three-ring East Road, Chao Yang District, Beijing, China.
| | - Wei Zhou
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, No. 11 of North Three-ring East Road, Chao Yang District, Beijing, China
| | - Xinkui Liu
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, No. 11 of North Three-ring East Road, Chao Yang District, Beijing, China
| | - Jingyuan Zhang
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, No. 11 of North Three-ring East Road, Chao Yang District, Beijing, China
| | - Shanshan Jia
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, No. 11 of North Three-ring East Road, Chao Yang District, Beijing, China
| | - Ziqi Meng
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, No. 11 of North Three-ring East Road, Chao Yang District, Beijing, China
| | - Shuyu Liu
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, No. 11 of North Three-ring East Road, Chao Yang District, Beijing, China
| | - Mengwei Ni
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, No. 11 of North Three-ring East Road, Chao Yang District, Beijing, China
| | - Yingying Liu
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, No. 11 of North Three-ring East Road, Chao Yang District, Beijing, China
| |
Collapse
|