1
|
Zhang X, Yang L, Feng K, Zhang H, Chen Y, Li W, Wang X, Zhang M, Wu Y, Wei S, Zheng Y, Meng G, Meng W, Chen X, Tang J. Shuxuening injection improves myocardial injury after myocardial infarction by regulating macrophage polarization via the TLR4/NF-κB and PI3K/Akt signaling pathways. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 138:156418. [PMID: 39879705 DOI: 10.1016/j.phymed.2025.156418] [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: 11/16/2024] [Revised: 01/05/2025] [Accepted: 01/21/2025] [Indexed: 01/31/2025]
Abstract
BACKGROUND Macrophage activation and polarization play pivotal roles in the inflammatory response and myocardial injury associated with myocardial infarction (MI). Modulating macrophage polarization from the pro-inflammatory M1 phenotype to the anti-inflammatory M2 phenotype is a promising therapeutic approach for MI. Shuxuening injection (SXNI) is extensively utilized in clinical settings for MI treatment and has demonstrated therapeutic efficacy. However, the effects of SXNI on macrophage polarization post-MI and its underlying mechanisms remain insufficiently understood. AIM OF THE STUDY This study is aimed to evaluate the effects of SXNI on macrophage polarization following MI and to elucidate its potential mechanisms of action. METHODS A rat model of MI was established by ligation of the left anterior descending coronary artery. The cardioprotective effects of SXNI were assessed through echocardiography, TTC staining, Masson's trichrome staining, HE staining, TUNEL staining, and western blotting (WB). Macrophage polarization was evaluated using ELISA, immunofluorescence staining, and WB. An in vitro model of oxygen-glucose deprivation (OGD) was utilized to simulate MI in macrophages, and qRT-PCR was employed to examine M1/M2 polarization markers. UPLC-Q-TOF/MS was used to identify active components in SXNI. Network pharmacology analysis and molecular docking were utilized to predict the key targets and pathways, which were subsequently validated through WB and immunohistochemistry. RESULTS SXNI improved cardiac function, reduced infarct size, and attenuated myocardial tissue damage and apoptosis in MI rats. Staining analyses indicated a reduction in M1 macrophages (CD86+/CD68+) and an increase in M2 macrophages (CD206+/CD68+) in SXNI-treated animals. In vivo and in vitro experiments demonstrated that SXNI decreased M1 markers and pro-inflammatory cytokines levels while increasing M2 markers and the production of anti-inflammatory and pro-angiogenic cytokines. UPLC-Q-TOF/MS analysis identified 18 active components in SXNI. Network pharmacology analysis and molecular docking implicated the TLR4/NF-κB and PI3K/Akt pathways as central mechanisms, which were further confirmed by WB and immunohistochemistry. SXNI inhibited the expression of TLR4 and phosphorylated NF-κB while enhancing phosphorylated PI3 K and Akt levels. CONCLUSIONS SXNI modulates the TLR4/NF-κB and PI3K/Akt signaling pathways to promote the polarization of macrophages from the M1 to the M2 phenotype, thereby alleviating myocardial inflammation and injury. These findings provide a scientific basis for the clinical application of SXNI in MI management, and establish a scientific foundation for exploring novel therapeutic strategies for cardiovascular diseases based on macrophage polarization.
Collapse
Affiliation(s)
- Xiaoshuai Zhang
- Department of Pharmacy, the First Affiliated Hospital of Henan University of Chinese Medicine, Henan Zhengzhou, 450003, China; School of Pharmacy, Henan University of Chinese Medicine, Henan Zhengzhou 450046, China
| | - Liuqing Yang
- Department of Pharmacy, the First Affiliated Hospital of Henan University of Chinese Medicine, Henan Zhengzhou, 450003, China; Henan Province Engineering Research Center for Clinical Application, Evaluation and Transformation of Traditional Chinese Medicine, Henan Provincial Key Laboratory for Clinical Pharmacy of Traditional Chinese Medicine, Henan Province Engineering Research Center of Safety Evaluation and Risk Management of Traditional Chinese Medicine, Zhengzhou 450003, Henan, China.
| | - Kairui Feng
- School of Pharmacy, Shandong Second Medical University, Weifang 261053, China
| | - Hui Zhang
- Department of Pharmacy, the First Affiliated Hospital of Henan University of Chinese Medicine, Henan Zhengzhou, 450003, China; Henan Province Engineering Research Center for Clinical Application, Evaluation and Transformation of Traditional Chinese Medicine, Henan Provincial Key Laboratory for Clinical Pharmacy of Traditional Chinese Medicine, Henan Province Engineering Research Center of Safety Evaluation and Risk Management of Traditional Chinese Medicine, Zhengzhou 450003, Henan, China
| | - Yulong Chen
- College of pharmaceutical engineering of traditional Chinese medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Weixia Li
- Department of Pharmacy, the First Affiliated Hospital of Henan University of Chinese Medicine, Henan Zhengzhou, 450003, China; Henan Province Engineering Research Center for Clinical Application, Evaluation and Transformation of Traditional Chinese Medicine, Henan Provincial Key Laboratory for Clinical Pharmacy of Traditional Chinese Medicine, Henan Province Engineering Research Center of Safety Evaluation and Risk Management of Traditional Chinese Medicine, Zhengzhou 450003, Henan, China; School of Pharmacy, Henan University of Chinese Medicine, Henan Zhengzhou 450046, China
| | - Xiaoyan Wang
- Department of Pharmacy, the First Affiliated Hospital of Henan University of Chinese Medicine, Henan Zhengzhou, 450003, China; Henan Province Engineering Research Center for Clinical Application, Evaluation and Transformation of Traditional Chinese Medicine, Henan Provincial Key Laboratory for Clinical Pharmacy of Traditional Chinese Medicine, Henan Province Engineering Research Center of Safety Evaluation and Risk Management of Traditional Chinese Medicine, Zhengzhou 450003, Henan, China; School of Pharmacy, Henan University of Chinese Medicine, Henan Zhengzhou 450046, China
| | - Mingliang Zhang
- Department of Pharmacy, the First Affiliated Hospital of Henan University of Chinese Medicine, Henan Zhengzhou, 450003, China; Henan Province Engineering Research Center for Clinical Application, Evaluation and Transformation of Traditional Chinese Medicine, Henan Provincial Key Laboratory for Clinical Pharmacy of Traditional Chinese Medicine, Henan Province Engineering Research Center of Safety Evaluation and Risk Management of Traditional Chinese Medicine, Zhengzhou 450003, Henan, China
| | - Yali Wu
- Department of Pharmacy, the First Affiliated Hospital of Henan University of Chinese Medicine, Henan Zhengzhou, 450003, China; Henan Province Engineering Research Center for Clinical Application, Evaluation and Transformation of Traditional Chinese Medicine, Henan Provincial Key Laboratory for Clinical Pharmacy of Traditional Chinese Medicine, Henan Province Engineering Research Center of Safety Evaluation and Risk Management of Traditional Chinese Medicine, Zhengzhou 450003, Henan, China
| | - Shiting Wei
- Department of Pharmacy, the First Affiliated Hospital of Henan University of Chinese Medicine, Henan Zhengzhou, 450003, China; School of Pharmacy, Henan University of Chinese Medicine, Henan Zhengzhou 450046, China
| | - Yajuan Zheng
- Department of Pharmacy, the First Affiliated Hospital of Henan University of Chinese Medicine, Henan Zhengzhou, 450003, China; School of Pharmacy, Henan University of Chinese Medicine, Henan Zhengzhou 450046, China
| | - Gaoquan Meng
- Department of Pharmacy, the First Affiliated Hospital of Henan University of Chinese Medicine, Henan Zhengzhou, 450003, China; School of Pharmacy, Henan University of Chinese Medicine, Henan Zhengzhou 450046, China
| | - Weiting Meng
- Department of Pharmacy, the First Affiliated Hospital of Henan University of Chinese Medicine, Henan Zhengzhou, 450003, China; School of Pharmacy, Henan University of Chinese Medicine, Henan Zhengzhou 450046, China
| | - Xiaofei Chen
- Department of Pharmacy, the First Affiliated Hospital of Henan University of Chinese Medicine, Henan Zhengzhou, 450003, China; Henan Province Engineering Research Center for Clinical Application, Evaluation and Transformation of Traditional Chinese Medicine, Henan Provincial Key Laboratory for Clinical Pharmacy of Traditional Chinese Medicine, Henan Province Engineering Research Center of Safety Evaluation and Risk Management of Traditional Chinese Medicine, Zhengzhou 450003, Henan, China.
| | - Jinfa Tang
- Department of Pharmacy, the First Affiliated Hospital of Henan University of Chinese Medicine, Henan Zhengzhou, 450003, China; Henan Province Engineering Research Center for Clinical Application, Evaluation and Transformation of Traditional Chinese Medicine, Henan Provincial Key Laboratory for Clinical Pharmacy of Traditional Chinese Medicine, Henan Province Engineering Research Center of Safety Evaluation and Risk Management of Traditional Chinese Medicine, Zhengzhou 450003, Henan, China; School of Pharmacy, Henan University of Chinese Medicine, Henan Zhengzhou 450046, China.
| |
Collapse
|
2
|
Deng B, Zhang G, Zeng Y, Li N, Hu C, Pang M, Lu S, Gu Y, Chen G, Zhou Y, Liu Y, Hua Y. Gualou Xiebai Banxia Decoction suppresses cardiomyocyte apoptosis in mice after myocardial infarction through activation of acetaldehyde dehydrogenase 2. JOURNAL OF ETHNOPHARMACOLOGY 2025; 339:119143. [PMID: 39577675 DOI: 10.1016/j.jep.2024.119143] [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: 07/06/2024] [Revised: 11/11/2024] [Accepted: 11/18/2024] [Indexed: 11/24/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Cardiac apoptosis has been reported to be involved in the development of Heart failure (HF) after Myocardial infarction (MI). As a traditional Chinese medicine with cardioprotective properties, Gualou Xiebai Banxia Decoction (GXBD) is therapeutically effective in treating MI. However, whether GXBD regulates cardiac apoptosis in HF after MI remains unknown, and the underlying mechanisms still unclear. AIM OF THE STUDY This study aimed to explore the effects and potential mechanisms of GXBD on cardiac apoptosis after MI. MATERIALS AND METHODS The MI model was constructed by ligating the left anterior descending coronary artery (LAD) in mice. The cardioprotective effects of GXBD were determined by echocardiography, masson staining, and haematoxylin and eosin (HE) staining. Bioinformatics analysis and network Pharmacology were used to explore the underlying molecular mechanisms of GXBD in MI. The effects of GXBD on cardiomyocyte apoptosis as well as the ALDH2 were examined by TUNEL staining, Immunohistochemistry (IHC), and Western blot (WB). Additionally, the effects of GXBD on oxidative stress, apoptosis and the ALDH2 in H9c2 cells were investigated using reactive oxygen species (ROS) detection, Hoechst33342/PI stainingand and WB. Moreover, the effects of suppressing and overexpressing ALDH2 in H9c2 cells were further examined. RESULTS Target prediction analysis showed that ALDH2 was a key target of GXBD which could ameliorate myocardial infarction. GXBD dose-dependently reduced cardiomyocyte apoptosis and ventricular dysfunction. In vivo experiments, GXBD activated ALDH2 enzymatic activity and inhibited the expression levels of Bax, Bcl-2, Cleaved Caspase 3, and Caspase 9. In vitro experiments, GXBD inhibited apoptosis in H9c2 cells. The inhibitory effects of GXBD on these were at least partially attributed to ALDH2 activation while silencing of ALDH2 significantly reversed these inhibitory effects of GXBD. CONCLUSION GXBD exerts inhibitory effects on cardiomyocyte apoptosis in mice after MI and suppresses H9c2 cells oxidative stress and apoptosis through activation of the enzyme activity of ALDH2.
Collapse
Affiliation(s)
- Bingying Deng
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Guoyong Zhang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Yixuan Zeng
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Nireng Li
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Changlei Hu
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Mingjie Pang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Sifan Lu
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Yufeng Gu
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Guanghong Chen
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Yingchun Zhou
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Yi Liu
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Yue Hua
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China; Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Southern Medical University, Guangzhou, 510515, China; Guangdong Basic Research Center of Excellence for Integrated Traditional and Western Medicine for Qingzhi Diseases, Guangzhou, 510515, China.
| |
Collapse
|
3
|
Wu YT, Zhang GY, Li L, Liu B, Wang RY, Song RQ, Hua Y, Bi YM, Han X, Zhang F, Wang D, Xie LP, Zhou YC. Salvia miltiorrhiza suppresses cardiomyocyte ferroptosis after myocardial infarction by activating Nrf2 signaling. JOURNAL OF ETHNOPHARMACOLOGY 2024; 330:118214. [PMID: 38641076 DOI: 10.1016/j.jep.2024.118214] [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: 01/18/2024] [Revised: 04/08/2024] [Accepted: 04/16/2024] [Indexed: 04/21/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Ferroptosis, a recently identified non-apoptotic form of cell death reliant on iron, is distinguished by an escalation in lipid reactive oxygen species (ROS) that are iron-dependent. This phenomenon has a strong correlation with irregularities in iron metabolism and lipid peroxidation. Salvia miltiorrhiza Bunge (DS), a medicinal herb frequently utilized in China, is highly esteemed for its therapeutic effectiveness in enhancing blood circulation and ameliorating blood stasis, particularly during the treatment of cardiovascular diseases (CVDs). Numerous pharmacological studies have identified that DS manifests antioxidative stress effects as well as inhibits lipid peroxidation. However, ambiguity persists regarding the potential of DS to impede ferroptosis in cardiomyocytes and subsequently improve myocardial damage post-myocardial infarction (MI). AIM OF THE STUDY The present work focused on investigating whether DS could be used to prevent the ferroptosis of cardiomyocytes and improve post-MI myocardial damage. MATERIALS AND METHODS In vivo experiments: Through ligation of the left anterior descending coronary artery, we constructed both a wild-type (WT) and NF-E2 p45-related factor 2 knockout (Nrf2-/-) mouse model of MI. Effects of DS and ferrostatin-1 (Fer-1) on post-MI cardiomyocyte ferroptosis were examined through detecting ferroptosis and myocardial damage-related indicators as well as Nrf2 signaling-associated protein levels. In vitro experiments: Erastin was used for stimulating H9C2 cardiomyocytes to construct an in vitro ferroptosis cardiomyocyte model. Effects of DS and Fer-1 on cardiomyocyte ferroptosis were determined based on ferroptosis-related indicators and Nrf2 signaling-associated protein levels. Additionally, inhibitor and activator of Nrf2 were used for confirming the impact of Nrf2 signaling on DS's effect on cardiomyocyte ferroptosis. RESULTS In vivo: In comparison to the model group, DS suppressed ferroptosis in cardiomyocytes post-MI and ameliorated myocardial damage by inducing Nrf2 signaling-related proteins (Nrf2, xCT, GPX4), diminishing tissue ferrous iron and malondialdehyde (MDA) content. Additionally, it enhanced glutathione (GSH) levels and total superoxide dismutase (SOD) activity, effects that are aligned with those of Fer-1. Moreover, the effect of DS on alleviating cardiomyocyte ferroptosis after MI could be partly inhibited through Nrf2 knockdown. In vitro: Compared with the erastin group, DS inhibited cardiomyocyte ferroptosis by promoting the expression of Nrf2 signaling-related proteins, reducing ferrous iron, ROS, and MDA levels, but increasing GSH content and SOD activity, consistent with the effect of Fer-1. Additionally, Nrf2 inhibition increased erastin-mediated ferroptosis of cardiomyocytes through decreasing Nrf2 signaling-related protein expressions. Co-treatment with DS and Nrf2 activator failed to further enhance the anti-ferroptosis effect of DS. CONCLUSION MI is accompanied by cardiomyocyte ferroptosis, whose underlying mechanism is probably associated with Nrf2 signaling inhibition. DS possibly suppresses ferroptosis of cardiomyocytes and improves myocardial damage after MI through activating Nrf2 signaling.
Collapse
Affiliation(s)
- Yu-Ting Wu
- Binzhou Medical University Hospital, Binzhou, 256603, China; School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China; Department of Traditional Chinese Medicine, Nanfang Hospital (ZengCheng Branch), Southern Medical University, Guangzhou, 510515, China
| | - Guo-Yong Zhang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China; Department of Traditional Chinese Medicine, Nanfang Hospital (ZengCheng Branch), Southern Medical University, Guangzhou, 510515, China
| | - Lei Li
- Binzhou Medical University Hospital, Binzhou, 256603, China
| | - Bin Liu
- Guangzhou Institute of Cardiovascular Disease, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, China
| | - Ru-Yu Wang
- School of Clinical Medicine, Jiangxi University of Traditional Chinese Medicine, Nanchang, 330004, China
| | | | - Yue Hua
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Yi-Ming Bi
- The Affiliated Traditional Chinese Medicine Hospital of Guangzhou Medical University, Guangzhou, 510000, China
| | - Xin Han
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China; Department of Traditional Chinese Medicine, Nanfang Hospital (ZengCheng Branch), Southern Medical University, Guangzhou, 510515, China
| | - Feng Zhang
- Binzhou Medical University Hospital, Binzhou, 256603, China
| | - Dong Wang
- Binzhou Medical University Hospital, Binzhou, 256603, China.
| | - Ling-Peng Xie
- Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510000, China.
| | - Ying-Chun Zhou
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China; Department of Traditional Chinese Medicine, Nanfang Hospital (ZengCheng Branch), Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
4
|
Chen H, Zhang G, Peng Y, Wu Y, Han X, Xie L, Xu H, Chen G, Liu B, Xu T, Pang M, Hu C, Fan H, Bi Y, Hua Y, Zhou Y, Luo S. Danggui Shaoyao San protects cyclophosphamide-induced premature ovarian failure by inhibiting apoptosis and oxidative stress through the regulation of the SIRT1/p53 signaling pathway. JOURNAL OF ETHNOPHARMACOLOGY 2024; 323:117718. [PMID: 38181933 DOI: 10.1016/j.jep.2024.117718] [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: 11/06/2023] [Revised: 12/28/2023] [Accepted: 01/03/2024] [Indexed: 01/07/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE It has been reported that apoptosis and oxidative stress are related to cyclophosphamide (CYC)-induced premature ovarian failure (POF). Therefore, anti-apoptotic and anti-oxidative stress treatments exhibit therapeutic efficacy in CYC-induced POF. Danggui Shaoyao San (DSS), which has been extensively used to treat gynecologic diseases, is found to inhibit apoptosis and reduce oxidative stress. However, the roles of DSS in regulating apoptosis and oxidative stress during CYC-induced POF, and its associated mechanisms are still unknown. AIM OF THE STUDY This work aimed to investigate the roles and mechanisms of DSS in inhibiting apoptosis and oxidative stress in CYC-induced POF. MATERIALS AND METHODS CYC (75 mg/kg) was intraperitoneally injected in mice to construct the POF mouse model for in vivo study. Thereafter, alterations of body weight, ovary morphology and estrous cycle were monitored to assess the ovarian protective properties of DSS. Serum LH and E2 levels were analyzed by enzyme-linked immunosorbent assay (ELISA). Hematoxylin-eosin (HE) staining was employed for examining ovarian pathological morphology and quantifying follicles in various stages. Meanwhile, TUNEL staining and apoptosis-related proteins were adopted for evaluating apoptosis. Oxidative stress was measured by the levels of ROS, MDA, and 4-HNE. Western blot (WB) assay was performed to detect proteins related to the SIRT1/p53 pathway. KGN cells were used for in vitro experiment. TBHP stimulation was carried out for establishing the oxidative stress-induced apoptosis cell model. Furthermore, MTT assay was employed for evaluating the protection of DSS from TBHP-induced oxidative stress. The anti-apoptotic ability of DSS was evaluated by hoechst/PI staining, JC-1 staining, and apoptosis-related proteins. Additionally, the anti-oxidative stress ability of DSS was measured by detecting the levels of ROS, MDA, and 4-HNE. Proteins related to SIRT1/p53 signaling pathway were also measured using WB and immunofluorescence (IF) staining. Besides, SIRT1 expression was suppressed by EX527 to further investigate the role of SIRT1 in the effects of DSS against apoptosis and oxidative stress. RESULTS In the in vivo experiment, DSS dose-dependently exerted its anti-apoptotic, anti-oxidative stress, and ovarian protective effects. In addition, apoptosis, apoptosis-related protein and oxidative stress levels were inhibited by DSS treatment. DSS treatment up-regulated SIRT1 and down-regulated p53 expression. From in vitro experiment, it was found that DSS treatment protected KGN cells from TBHP-induced oxidative stress injury. Besides, DSS administration suppressed the apoptosis ratio, apoptosis-related protein levels, mitochondrial membrane potential damage, and oxidative stress. SIRT1 suppression by EX527 abolished the anti-apoptotic, anti-oxidative stress, and ovarian protective effects, as discovered from in vivo and in vitro experiments. CONCLUSIONS DSS exerts the anti-apoptotic, anti-oxidative stress, and ovarian protective effects in POF mice, and suppresses the apoptosis and oxidative stress of KGN cells through activating SIRT1 and suppressing p53 pathway.
Collapse
Affiliation(s)
- Hongmei Chen
- The First School of Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Guoyong Zhang
- Department of Traditional Chinese Medicine, Nanfang Hospital (ZengCheng Branch), Southern Medical University, Guangzhou, 510515, China; School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Yan Peng
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Yuting Wu
- Binzhou Medical University Hospital, Binzhou, 256603, China
| | - Xin Han
- Department of Traditional Chinese Medicine, Nanfang Hospital (ZengCheng Branch), Southern Medical University, Guangzhou, 510515, China; School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Lingpeng Xie
- Department of Hepatology, Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510315, China
| | - Honglin Xu
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China; The Tenth Affiliated Hospital of Southern Medical University (Dongguan People's Hospital), Dongguan, 523058, China
| | - Guanghong Chen
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China; The First Affiliated Hospital of Guangzhou University of Chinese Medicine/Post- Doctoral Research Station, Guangzhou, 510405, China; Guangdong Clinical Research Academy of Chinese Medicine, Guangzhou, 510405, China
| | - Bin Liu
- Department of Traditional Chinese Medicine, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, 510260, China
| | - Tong Xu
- Department of Traditional Chinese Medicine, Nanfang Hospital (ZengCheng Branch), Southern Medical University, Guangzhou, 510515, China; School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Mingjie Pang
- Department of Traditional Chinese Medicine, Nanfang Hospital (ZengCheng Branch), Southern Medical University, Guangzhou, 510515, China; School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Changlei Hu
- Department of Traditional Chinese Medicine, Nanfang Hospital (ZengCheng Branch), Southern Medical University, Guangzhou, 510515, China; School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Huijie Fan
- Department of Traditional Chinese Medicine, Yangjiang People's Hospital, Yangjiang, 529599, China
| | - Yiming Bi
- Department of Acupuncture and Moxibustion, The Affliated TCM Hospital of Guangzhou Medical University, Guangzhou, 510130, China
| | - Yue Hua
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China.
| | - Yingchun Zhou
- Department of Traditional Chinese Medicine, Nanfang Hospital (ZengCheng Branch), Southern Medical University, Guangzhou, 510515, China; School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China.
| | - Songping Luo
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China.
| |
Collapse
|