1
|
Yu L, Wang X, Lei Q, Liu Y, Li Z, Dai X, Song Z, He Y, Gao S, Yu C, Li L. Tongmai Yangxin pill alleviates myocardial ischemia/reperfusion injury by regulating mitochondrial fusion and fission through the estrogen receptor alpha/peroxisome proliferator-activated receptor gamma coactivator-1 alpha signaling pathway. JOURNAL OF ETHNOPHARMACOLOGY 2025; 345:119639. [PMID: 40096900 DOI: 10.1016/j.jep.2025.119639] [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: 12/30/2024] [Revised: 03/11/2025] [Accepted: 03/13/2025] [Indexed: 03/19/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The Tongmai Yangxin Pill (TMYX) is derived from the Zhigancao decoction recorded in Treatise on Cold Damage Disorders (Shang Han Lun) by Zhang Zhongjing during the Han dynasty. The prescription of TMYX reflects a therapeutic rationale and efficacy unique to traditional Chinese medicine. TMYX is clinically effective in alleviating myocardial ischemia-reperfusion injury (MI/RI). However, the precise active ingredients and underlying mechanisms remain unclear. AIM OF THE STUDY The primary objective of this study was to investigate the potential of TMYX in addressing MI/RI by activating the estrogen receptor ERα. We hypothesized that this action upregulates PGC-1α activity, subsequently promoting a balanced regulation of mitochondrial fusion and fission. MATERIALS AND METHODS UPLC-Q-TOF-MS/MS was used to identify the active components of TMYX. Subsequently, a network pharmacology approach was used to uncover the therapeutic targets and underlying pharmacological mechanisms through which TMYX mitigates MI/RI. Lastly, the anticipated outcomes were confirmed through in vivo and in vitro experimental validations. RESULTS Using UPLC-Q-TOF-MS/MS, we successfully identified 53 distinct compounds in TMYX. Network pharmacology analysis revealed 20 key TMYX targets associated with MI/RI. Enrichment studies using GO and KEGG analyses revealed that these targets were mainly associated with mitochondrial processes and estrogen signaling pathways. Both in vivo and in vitro studies confirmed that TMYX markedly improved mitochondrial function through the ERα/PGC-1α signaling cascade, leading to a reduction in the size of myocardial infarctions and the incidence of apoptosis. Notably, combining TMYX with siERα abolished the protective effect of TMYX on the mitochondria. CONCLUSION TMYX therapy can improve cardiac function in MI/RI. This effect is likely mediated by the ERα/PGC-1α signaling pathway. However, given the complex multi-component composition of traditional Chinese medicine formulas, additional studies are necessary to confirm the findings of this research.
Collapse
Affiliation(s)
- Lu Yu
- Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin, 301617, China
| | - Xu Wang
- Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin, 301617, China
| | - Qina Lei
- Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin, 301617, China
| | - Yutong Liu
- Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin, 301617, China
| | - Zhu Li
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, No. 548 Binwen Road, Binjiang District, Hangzhou, 310053, China
| | - Xiangdong Dai
- Pharmaron Beijing Co., Ltd. (China), BDA, 6 Taihe Road, Beijing, 100176, China
| | - Zhihui Song
- Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin, 301617, China
| | - Yuanyuan He
- Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin, 301617, China
| | - Shan Gao
- Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin, 301617, China
| | - Chunquan Yu
- Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin, 301617, China
| | - Lin Li
- Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin, 301617, China.
| |
Collapse
|
2
|
Wen C, Liao X, Ye X, Lai W. Pharmacokinetics and Biological Activities of Notoginsenoside R1: A Systematical Review. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2025; 53:205-249. [PMID: 39880667 DOI: 10.1142/s0192415x25500090] [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: 01/31/2025]
Abstract
Panax notoginseng (PN) root is a renowned nutritional supplement, health food additive, and traditional medicine that maintains homeostasis within the human microcirculatory system. Notoginsenoside R1 (NG-R1), an active compound derived from PN root, has been reported to possess various pharmacological activities, including anti-inflammatory, antioxidant, anticancer, antimicrobial, and angiogenic effects. However, NG-R1's pharmacokinetic properties and pharmacological activities have not been systematically elucidated. In this paper, the pharmacokinetic properties of NG-R1, its pharmacological effects, mechanisms of actions, and structure-activity relationship have been reviewed. Notably, NG-R1 inhibits tumor necrosis factor α (TNF-α) expression, enhances the expression of nuclear factor erythroid 2-related factor 2 (NRF2), and enhances the expression of vascular endothelial growth factor receptor (VEGFR). The pharmacological effects of NG-R1 are associated with the modulation of several signaling pathways, such as mitogen-activated protein kinase (MAPK)/nuclear factor κ-B (NF-κB), NRF2/antioxidant response element (ARE), Wnt/β-catenin, and phosphoinositide-3 kinase (PI3K)/protein kinase B (AKT). NG-R1 offers potentially protective effects against numerous diseases, including cardiovascular, neurological, renal, pulmonary, bone, and diabetes-related conditions. Although the pharmacological activities and diverse effects of NG-R1 have been demonstrated in various diseases, its clinical applications are limited by poor bioavailability. Several strategies have been explored to improve the pharmacokinetic profile of NG-R1, making it a promising candidate for drug development.
Collapse
Affiliation(s)
- Chao Wen
- School of Nursing, Gannan Medical University, Ganzhou 341000, P. R. China
| | - Xiaofei Liao
- Department of Pharmacy, Ganzhou People's Hospital, Ganzhou 341000, P. R. China
| | - Xinyun Ye
- Department of Neurosurgey, Ganzhou People's Hospital, Ganzhou 341000, P. R. China
| | - Wentao Lai
- Department of Neurosurgey, Ganzhou People's Hospital, Ganzhou 341000, P. R. China
| |
Collapse
|
3
|
Abstract
The PI3K/AKT signaling has crucial role in the regulation of numerous physiological functions through activation of downstream effectors and modulation of cell cycle transition, growth and proliferation. This pathway participates in the pathogenesis of several human disorders such as heart diseases through regulation of size and survival of cardiomyocytes, angiogenic processes as well as inflammatory responses. Moreover, PI3K/AKT pathway participates in the process of myocardial injury induced by a number of substances such as H2O2, Mercury, lipopolysaccharides, adriamycin, doxorubicin and epirubicin. In this review, we describe the contribution of this pathway in the pathoetiology of myocardial ischemia/reperfusion injury and myocardial infarction, heart failure, cardiac hypertrophy, cardiomyopathy and toxins-induced cardiac injury.
Collapse
|
4
|
Wang N, Song J, Zhou G, Li W, Ma H. Mechanism of salidroside relieving the acute hypoxia-induced myocardial injury through the PI3K/Akt pathway. Saudi J Biol Sci 2020; 27:1533-1537. [PMID: 32489291 PMCID: PMC7254047 DOI: 10.1016/j.sjbs.2020.04.035] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 04/14/2020] [Accepted: 04/17/2020] [Indexed: 02/01/2023] Open
Abstract
OBJECTIVE The objective was to investigate the anti-inflammatory effects of salidroside through the PI3K/Akt signaling pathway and its protective effects on acute hypoxia-induced myocardial injury in rats. METHODS A total of 24 healthy Sprague-Dawley male rats were selected as the experimental subjects. All rats were divided into 4 groups by using the random number table method, with 6 rats in each group. The groups included the normal control group, the salidroside group, the hypobaric hypoxia group, and the hypobaric hypoxia + salidroside group. Rats in the salidroside group were fed in the original animal laboratory and were intragastrically administered with salidroside every morning at a dosage of 35 mg/kg. Rats in the normal control group were intragastrically administered with an equal dosage of saline. Rats in the hypobaric hypoxia + salidroside group were intragastrically administered with salidroside every morning at a dosage of 35 mg/kg, who were fed in the hypoxic experiment module for animals. The altitude was increased to 4000 m, and the rats were kept in the module for 24 h. Rats in the hypobaric hypoxia group were intragastrically administered with an equal dosage of saline in the same environment, and the altitude was increased to 4000 m after administration. Parameters of blood gas analysis, histopathological changes in cardiac tissues, cardiac indexes, and inflammatory factors IL-6 and TNF-α levels of rats in groups were compared. RESULTS 1. The cardiac indexes of rats in groups were compared. The differences between the hypobaric hypoxia group and the hypobaric hypoxia + salidroside group were statistically significant (P < 0.05). 2. The results of blood gas analysis of rats in groups were compared. The differences between the hypobaric hypoxia group and the hypobaric hypoxia + salidroside group were significantly different (P < 0.05). 3. In the hypobaric hypoxia group, the myocardial cells of rats were arranged disorderly and shaped differently, with cases such as edema, degeneration, necrosis, nucleus pyknosis, and massive infiltration of inflammatory cells. In the hypobaric hypoxia + salidroside group, the above-mentioned pathological changes in myocardial cells were relieved. 4. Compared with the hypobaric hypoxia group, in the hypobaric hypoxia + salidroside group, the concentrations of IL-6 and TNF-α in rats decreased apparently, and the differences were statistically significant (P < 0.05). CONCLUSION Salidroside had the repairing and protective effects on the hypobaric hypoxia-induced myocardial injuries in rats. The application of salidroside could reduce the inflammatory responses of rats with hypobaric hypoxia-induced myocardial injuries through PI3K/Akt signaling pathway, thereby protecting the myocardial cells.
Collapse
Affiliation(s)
- Nan Wang
- Department of Cardiology, Gansu Provincial Hospital, Lanzhou City 730000, Gansu Province, China
| | - Jiyang Song
- Department of Cardiology, Gansu Provincial Hospital, Lanzhou City 730000, Gansu Province, China
| | - Gang Zhou
- Department of Cardiology, Gansu Provincial Hospital, Lanzhou City 730000, Gansu Province, China
| | - Wenli Li
- Department of Cardiology, Gansu Provincial Hospital, Lanzhou City 730000, Gansu Province, China
| | - Huiyuan Ma
- Department of Cardiology, Gansu Provincial Hospital, Lanzhou City 730000, Gansu Province, China
| |
Collapse
|
5
|
Dai YL, Jiang YF, Lee HG, Jeon YJ, Kang MC. Characterization and screening of anti-tumor activity of fucoidan from acid-processed hijiki (Hizikia fusiforme). Int J Biol Macromol 2019; 139:170-180. [PMID: 31336117 DOI: 10.1016/j.ijbiomac.2019.07.119] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 06/14/2019] [Accepted: 07/19/2019] [Indexed: 01/12/2023]
Abstract
The aim of this study was to investigate the antiproliferative effects of fucoidan from three regional hijiki (Hizikia fusiforme) samples (Zhejiang-China, Jeju-Korea [JH], and Wando-Korea) in East Asia. Hijiki was processed using 1% citric acid to decrease heavy metal content. The JH sample was separated using diethylaminoethyl-cellulose-ion exchange chromatography to obtain four active fractions (JHCF1-JHCF4) and their monosaccharide composition was detected using high-performance liquid chromatography. The structure of the crude polysaccharides and four fucoidan fractions was analyzed using Fourier-transform infrared spectroscopy. JHCF4 showed the highest fucose and sulfate content and decreased Hep3B cell growth in 48 h with a half-maximal inhibitory concentration of 33.53 ± 2.50 μg/ml, which represented the strongest anticancer activity. Further, nuclear staining with Hoechst 33342 and acridine orange-ethidium bromide staining demonstrated that the anticancer activity of JHCF4 was mediated by apoptosis. Moreover, JHCF4 down-regulated B-cell lymphoma extra-large, while up-regulating Bcl-2-associated X protein, caspase-3, and apoptotic bodies to different degrees in Hep3B cells. JHCF4 induced apoptosis via the generation of reactive oxygen species along with the concurrent loss of mitochondrial membrane potential, indicating the potential role of the mitochondria-mediated pathway. Therefore, these results indicate that JHCF4 exhibited antiproliferative effects on the investigated cancer cell lines.
Collapse
Affiliation(s)
- Yu-Lin Dai
- Department of Marine Life Science, Jeju National University, Jeju 63243, Republic of Korea; Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun 130117, China
| | - Yun-Fei Jiang
- Department of Marine Life Science, Jeju National University, Jeju 63243, Republic of Korea
| | - Hyo Geun Lee
- Department of Marine Life Science, Jeju National University, Jeju 63243, Republic of Korea
| | - You-Jin Jeon
- Department of Marine Life Science, Jeju National University, Jeju 63243, Republic of Korea; Marine Science Institute, Jeju National University, Jeju Self-Governing Province 63333, Republic of Korea.
| | - Min-Cheol Kang
- Research group of Food Processing, Research Division of Strategic Food Technology, Korea Food Research Institute (KFRI), Wanju 55365, Republic of Korea.
| |
Collapse
|
6
|
Qian D, Shao X, Li Y, Sun X. Retracted
: Notoginsenoside R1 protects WI‐38 cells against lipopolysaccharide‐triggered injury via adjusting the miR‐181a/TLR4 axis. J Cell Biochem 2019; 120:19764-19774. [DOI: 10.1002/jcb.29282] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 06/20/2019] [Indexed: 12/20/2022]
Affiliation(s)
- Daolin Qian
- Department of Pediatric Internal Medicine Juancheng People's Hospital Heze Shandong China
| | - Xiankun Shao
- Department of Pediatric Heze Municipal Hospital Heze Shandong China
| | - Yingchun Li
- Department of Neurology Heze No. 3 People's Hospital Heze Shandong China
| | - Xinyan Sun
- Department of Pediatric Internal Medicine Heze Municipal Hospital Heze Shandong China
| |
Collapse
|
7
|
Li Y, Ma G, Lv Y, Su J, Li G, Chen X. Efficacy of Obcordata A from Aspidopterys obcordata on Kidney Stones by Inhibiting NOX4 Expression. Molecules 2019; 24:E1957. [PMID: 31117291 PMCID: PMC6572403 DOI: 10.3390/molecules24101957] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Revised: 05/16/2019] [Accepted: 05/16/2019] [Indexed: 11/17/2022] Open
Abstract
Obcordata A (OA) is a polyoxypregnane glycoside derived from the Dai medicine Aspidopterys obcordata vines. This study aims to investigate the efficacy of OA on renal tubular epithelial cells exposed to calcium oxalate crystals. We incubated renal tubular cells with 28 μg·cm2 calcium oxalate crystals for 24 h with and without OA, GKT137831, phorbol-12-myristate-13-acetate (PMA), and tocopherol. The MTT [3-(4, 5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide] assay, microscopic examination, flow cytometry, and immunofluorescence staining revealed that calcium oxalate crystals decreased cell viability and elevated reactive oxygen species (ROS) levels. OA, GKT137831, and tocopherol protected cells and decreased ROS levels. However, OA did not exhibit direct DPPH scavenging ability. In addition, immunoblotting illustrated that OA inhibited the NOX4 (nicotinamide adenine dinucleotide phosphate oxidases 4) expression and downregulated the protein expression in the NOX4/ROS/p38 MAPK (p38 mitogen-activated protein kinase) pathway. The findings suggest that the cytoprotective and antioxidant effects of OA can be blocked by the NOX4 agonist PMA. In conclusion, OA could be used as a NOX4 inhibitor to prevent kidney stones.
Collapse
Affiliation(s)
- Yihang Li
- Yunnan Branch, Institute of Medicinal Plant, Chinese Academy of Medical Sciences, Peking Union Medical College, Jinghong 666100, China.
- Key Laboratory of Dai and Southern Medicine of Xishuangbanna Dai Autonomous Prefecture, Jinghong 666100, China.
| | - Guoxu Ma
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 151, Malianwa North Road, Haidian District, Beijing 100193, China.
| | - Yana Lv
- Yunnan Branch, Institute of Medicinal Plant, Chinese Academy of Medical Sciences, Peking Union Medical College, Jinghong 666100, China.
- Key Laboratory of Dai and Southern Medicine of Xishuangbanna Dai Autonomous Prefecture, Jinghong 666100, China.
| | - Jing Su
- Yunnan Branch, Institute of Medicinal Plant, Chinese Academy of Medical Sciences, Peking Union Medical College, Jinghong 666100, China.
- Key Laboratory of Dai and Southern Medicine of Xishuangbanna Dai Autonomous Prefecture, Jinghong 666100, China.
| | - Guang Li
- Yunnan Branch, Institute of Medicinal Plant, Chinese Academy of Medical Sciences, Peking Union Medical College, Jinghong 666100, China.
- Key Laboratory of Dai and Southern Medicine of Xishuangbanna Dai Autonomous Prefecture, Jinghong 666100, China.
| | - Xi Chen
- Yunnan Branch, Institute of Medicinal Plant, Chinese Academy of Medical Sciences, Peking Union Medical College, Jinghong 666100, China.
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 151, Malianwa North Road, Haidian District, Beijing 100193, China.
| |
Collapse
|