1
|
Xu SY, Cao HY, Yang RH, Xu RX, Zhu XY, Ma W, Liu XB, Yan XY, Fu P. Genus Paeonia monoterpene glycosides: A systematic review on their pharmacological activities and molecular mechanisms. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 127:155483. [PMID: 38432036 DOI: 10.1016/j.phymed.2024.155483] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 02/11/2024] [Accepted: 02/24/2024] [Indexed: 03/05/2024]
Abstract
BACKGROUND Genus Paeonia, which is the main source of Traditional Chinese Medicine (TCM) Paeoniae Radix Rubra (Chishao in Chinese), Paeoniae Radix Alba (Baishao in Chinese) and Moutan Cortex (Mudanpi in Chinese), is rich in active pharmaceutical ingredient such as monoterpenoid glycosides (MPGs). MPGs from Paeonia have extensive pharmacological effects, but the pharmacological effects and molecular mechanisms of MPGs has not been comprehensively reviewed. PURPOSE MPGs compounds are one of the main chemical components of the genus Paeonia, with a wide variety of compounds and strong pharmacological activities, and the structure of the mother nucleus-pinane skeleton is similar to that of a cage. The purpose of this review is to summarize the pharmacological activity and mechanism of action of MPGs from 2012 to 2023, providing reference direction for the development and utilization of Paeonia resources and preclinical research. METHODS Keywords and phrases are widely used in database searches, such as PubMed, Web of Science, Google Scholar and X-Mol to search for citations related to the new compounds, extensive pharmacological research and molecular mechanisms of MPGs compounds of genus Paeonia. RESULTS Modern research confirms that MPGs are the main compounds in Paeonia that exert pharmacological effects. MPGs with extensive pharmacological characteristics are mainly concentrated in two categories: paeoniflorin derivatives and albiflflorin derivatives among MPGs, which contains 32 compounds. Among them, 5 components including paeoniflorin, albiflorin, oxypaeoniflorin, 6'-O-galloylpaeoniflorin and paeoniflorigenone have been extensively studied, while the other 28 components have only been confirmed to have a certain degree of anti-inflammatory and anticomplementary effects. Studies of pharmacological effects are widely involved in nervous system, endocrine system, digestive system, immune system, etc., and some studies have identified clear mechanisms. MPGs exert pharmacological activity through multilateral mechanisms, including anti-inflammatory, antioxidant, inhibition of cell apoptosis, regulation of brain gut axis, regulation of gut microbiota and downregulation of mitochondrial apoptosis, etc. CONCLUSION: This systematic review delved into the pharmacological effects and related molecular mechanisms of MPGs. However, there are still some compounds in MPGs whose pharmacological effects and pharmacological mechanisms have not been clarified. In addition, extensive clinical randomized trials are needed to verify the efficacy and dosage of MPGs.
Collapse
Affiliation(s)
- Shi-Yi Xu
- School of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin 150040, China; Experimental Training Center, Heilongjiang University of Chinese Medicine, Harbin 150040, China
| | - Hui-Yan Cao
- School of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin 150040, China
| | - Rui-Hong Yang
- School of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin 150040, China
| | - Rong-Xue Xu
- The Health Center of Longjiang Airlines, Harbin 150000, China; Qiqihar Medical University, Qiqihar 161003, China
| | - Xing-Yu Zhu
- Experimental Training Center, Heilongjiang University of Chinese Medicine, Harbin 150040, China
| | - Wei Ma
- School of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin 150040, China; Experimental Training Center, Heilongjiang University of Chinese Medicine, Harbin 150040, China
| | - Xiu-Bo Liu
- Jiamusi College, Heilongjiang University of Chinese Medicine, Jiamusi 154007, China
| | - Xue-Ying Yan
- School of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin 150040, China.
| | - Peng Fu
- First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin 150040, China.
| |
Collapse
|
2
|
The Multiple Pharmacologic Functions and Mechanisms of Action of Guizhi Fuling Formulation. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:6813421. [PMID: 35529925 PMCID: PMC9076289 DOI: 10.1155/2022/6813421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 04/09/2022] [Indexed: 11/17/2022]
Abstract
Objectives Guizhi Fuling Formulation (GZFL), a traditional Chinese medical formulation, consists of Cinnamomi Ramulus, Paeoniae Radix Alba (or Paeoniae Radix Rubra), Moutan Cortex, Persicae Semen, and Poria, with multiple therapeutic functions such as sedation, antitumor activity, anti-inflammation, and neuroprotection. However, its clinical applications remain relatively fragmented, and the underlying mechanisms of GZFL in different diseases are still not very certain. Further research and summary in both application and mechanisms remain to be needed for human health and the best use of GZFL. Therefore, we summarized the multiple pharmacologic effects and possible mechanisms of action of GZFL according to recent 17 years of research. Methods We retrieved four English and two Chinese databases using these keywords (the formulation name or its synonyms) and searched articles written in English from January 2006 up to February 2022. Key Findings. GZFL exhibits multiple pharmacologic advantages in gynecologic diseases and other expanding diseases such as cancer, blood, and vascular disease, renal failure, inflammation, and brain injury. Possibly due to its diverse bioactive components and pharmacologic activities, GZFL could target the multiple signaling pathways involved in regulating blood circulation, inflammatory and immune factors, proliferation, apoptosis, and so on. Conclusion This review suggests that GZFL displays promising therapeutic effects for many kinds of diseases, which have been beyond the scope of the original prescription for gynecologic diseases. In this way, we wish to provide a reference and recommendation for further preclinic and clinic studies.
Collapse
|
3
|
Multiple Roles of Paeoniflorin in Alzheimer's Disease. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:2464163. [PMID: 35449815 PMCID: PMC9017479 DOI: 10.1155/2022/2464163] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 02/09/2022] [Indexed: 12/18/2022]
Abstract
Alzheimer's disease (AD) is a geriatric disease with the morbidity and mortality continuing to grow, partly due to the aging of the world population. As one of the most common types of primary neurodegenerative dementia, it is mainly due to environmental, epigenetic, immunological, and genetic factors. Paeoniflorin (PF), the main component of paeony extract, plays a more and more important role in the prevention and treatment of AD, including regulating protein, anti-inflammation, antioxidation, and antiapoptosis, protecting glial cells, regulating neurotransmitters and related enzymes and receptors, and inhibiting or activating related signal pathways. This article summarizes the latest researches on the multiple effects and the mechanisms of PF in the treatment to cure AD, providing new insights and research basis for further clinical application of traditional Chinese medicine (TCM) in the treatment of AD.
Collapse
|
4
|
Lv S, Wang X, Bai X, Ning H, Li Y, Wen H, Lu W, Wang J. Mesenchymal epithelial transition factor regulates tumor necrosis factor-related apoptotic induction ligand resistance in hepatocellular carcinoma cells through down-regulation of cyclin B1. Int J Biochem Cell Biol 2020; 128:105844. [PMID: 32882404 DOI: 10.1016/j.biocel.2020.105844] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 08/26/2020] [Accepted: 08/27/2020] [Indexed: 11/25/2022]
Abstract
Tumor necrosis factor-related apoptotic induction ligand can induce cell apoptosis in various tumor cells. However, many cancer cells are resistant to tumor necrosis factor-related apoptotic induction ligand. Therefore, overcoming the tumor necrosis factor-related apoptotic induction ligand resistance makes it possible for tumor necrosis factor-related apoptotic induction ligand-based anti-cancer therapies. In this study, we took mesenchymal epithelial transition factor as the research target to study its role in tumor necrosis factor-related apoptotic induction ligand-resistant hepatocellular carcinoma. Mesenchymal epithelial transition factor gene has been proved to be an effective predictor of recurrence after hepatocellular carcinoma resection. The expression of mesenchymal epithelial transition factor and cyclin B1 were measured in tumor necrosis factor-related apoptotic induction ligand-resistant and non-resistant hepatocellular carcinoma tissues. Cyclin B1-knockdown and cyclin B1-overexpression hepatocellular carcinoma cells were treated with tumor necrosis factor-related apoptotic induction ligand; mesenchymal epithelial transition factor knockout, mesenchymal epithelial transition factor re-introduction and cyclin B1 restored in hepatocellular carcinoma cells treated with tumor necrosis factor-related apoptotic induction ligand were established. And MTT, bromodeoxyuridine, flow cytometry and western blotting were performed to evaluate the effect of mesenchymal epithelial transition factor and cyclin B1 on hepatocellular carcinoma cells treated with tumor necrosis factor-related apoptotic induction ligand. In addition, subcutaneous tumor transplantation in nude mice was conducted to access the effect of mesenchymal epithelial transition factor and cyclin B1 on tumor formation in vivo. In conclusion, cyclin B1 enhanced the cell growth and inhibited apoptosis in tumor necrosis factor-related apoptotic induction ligand-resistant hepatocellular carcinoma cells. And mesenchymal epithelial transition factor promoted the cell growth and apoptosis in tumor necrosis factor-related apoptotic induction ligand-resistant hepatocellular carcinoma cells by regulating cyclin B1. Therefore, mesenchymal epithelial transition factor regulates the cyclin B1 to regulate tumor necrosis factor-related apoptotic induction ligand resistance in hepatocellular carcinoma cells. Our results suggest a novel molecular mechanism for regulating tumor necrosis factor-related apoptotic induction ligand resistance, which might be helpful to select drug targets in the treatment of liver cancer.
Collapse
Affiliation(s)
- Shuai Lv
- Department of Gastroenterology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450018, Henan Province, China.
| | - Xijuan Wang
- Department of Pediatrics, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, 450003, Henan Province, China
| | - Xia Bai
- Department of Gastroenterology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450018, Henan Province, China
| | - Hanbing Ning
- Department of Gastroenterology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450018, Henan Province, China
| | - Yingxia Li
- Department of Gastroenterology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450018, Henan Province, China
| | - Hongtao Wen
- Department of Gastroenterology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450018, Henan Province, China
| | - Wenquan Lu
- Department of Gastroenterology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450018, Henan Province, China
| | - Jingyun Wang
- Department of Gastroenterology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450018, Henan Province, China
| |
Collapse
|
5
|
Bupivacaine-Induced Neurotoxicity Is Modulated by Epigenetic Axis of Long Noncoding RNA SNHG16 and Hsa-miR-132-3p. Neurotox Res 2020; 38:175-183. [PMID: 32335807 DOI: 10.1007/s12640-020-00202-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 03/24/2020] [Accepted: 04/03/2020] [Indexed: 12/16/2022]
Abstract
Clinical application of local anesthetic reagent, liposomal bupivacaine (BUP), may cause irreversible damage to human nerve system. In this study, we explored the functional role of long non-coding RNA (lncRNA) small nucleolar RNA host gene 16 (SNHG16) in BUP-induced neurotoxicity in SH-SY5Y cells. SH-SY5Y cells were treated with BUP in vitro, whose dose-dependent effects on cell viability and SNHG16 expression were explored. SNHG16 was upregulated in SH-SY5Y cells. The protection of SNHG16 upregulation on BUP-induced neurotoxicity was examined by viability assay, apoptosis assay, and caspase activity assay, respectively. The endogenously competing target of SNHG16, human mature microRNA-132-3p (hsa-miR-132-3p), was explored by dual-luciferase assay and quantitative real-time PCR (qRT-PCR). Hsa-miR-132-3p was then further overexpressed in SNHG16-upregulated SH-SY5Y cells to explore its functional role in BUP-induced neurotoxicity. BUP induced dose-dependent cell death and SNHG16 downregulation in SH-SY5Y cells. Inversely, lentivirus-mediated SNHG16 upregulation mitigated cell death. In addition, SNHG16 upregulation rescued BUP-induced apoptosis and caspase 3/7 augmentation. Hsa-miR-132-3p was found to be reversely expressed with SNHG16 in BUP-treated SH-SY5Y cells. Overexpressing hsa-miR-132-3p reduced the protection of SNHG16 on BUP-induced neurotoxicity. We demonstrated that epigenetic axis of SNHG16/hsa-miR-132-3p had a functional role in regulating anesthesia-induced neurotoxicity in human lineage neural cells.
Collapse
|
6
|
Zhang H, Wang W, Du Q. Andrographolide attenuates bupivacaine-induced cytotoxicity in SH-SY5Y cells through preserving Akt/mTOR activity. DRUG DESIGN DEVELOPMENT AND THERAPY 2019; 13:1659-1666. [PMID: 31190744 PMCID: PMC6529178 DOI: 10.2147/dddt.s201122] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 04/03/2019] [Indexed: 12/11/2022]
Abstract
Background: Bupivacaine (Bup) is the most commonly used local anesthetic. However, Bup induces cytotoxicity, especially in older patients. Recent reports have indicated that andrographolide (Andro) exhibits protective effects on human neurons. Nevertheless, whether Andro can inhibit Bup-induced cytotoxicity remains unclear. As such, we investigated the effect of Andro on Bup-induced cytotoxicity of SH-SY5Y cells in the present study. Methods: Western blotting was used to examine expression of Bax, Bcl2, active caspase 3, p-Akt, and p-mTOR in SH-SY5Y cells. In addition, ELISA was used to detect levels of total glutathione and reactive oxygen species in cells. Results: We found that Andro attenuated Bup-induced cytotoxicity of SH-SY5Y cells. In addition, Andro inhibited Bup-induced apoptosis via downregulating the expression of Bax and active caspase 3 and upregulating the proteins Bcl2, p-Akt, and p-mTOR in SH-SY5Y cells. Moreover, Andro alleviated Bup-induced oxidative damage in SH-SY5Y cells via downregulating the level of reactive oxygen species and upregulating of the level of total glutathione. More significantly, inhibition of Akt abolished the protective effect of Andro in Bup-treated SH-SY5Y cells. Conclusion: Our findings indicated that Andro played a neuroprotective role via preserving Akt/mTOR activity and increasing antioxidative status in Bup-treated SH-SY5Y cells. Therefore, Andro may be a potential agent for the treatment of human cytotoxicity induced by Bup.
Collapse
Affiliation(s)
- Huiyuan Zhang
- Department of Neurology, Liaocheng People's Hospital, Liaocheng, Shandong, 252000, People's Republic of China
| | - Weiwei Wang
- Department of Neurology, Liaocheng People's Hospital, Liaocheng, Shandong, 252000, People's Republic of China
| | - Qian Du
- EEG Room, Liaocheng People's Hospital, Liaocheng, Shandong 252000, People's Republic of China
| |
Collapse
|
7
|
Wang S, Xia B, Qiao Z, Duan L, Wang G, Meng W, Liu Z, Wang Y, Zhang M. Tetramethylpyrazine attenuated bupivacaine-induced neurotoxicity in SH-SY5Y cells through regulating apoptosis, autophagy and oxidative damage. DRUG DESIGN DEVELOPMENT AND THERAPY 2019; 13:1187-1196. [PMID: 31114159 PMCID: PMC6489565 DOI: 10.2147/dddt.s196172] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Accepted: 02/26/2019] [Indexed: 12/11/2022]
Abstract
Background: Bupivacaine (BUP) acts as a local anesthetic, which is extensively used for clinical patients but could generate neurotoxicity in neurons. Tetramethylpyrazine (TET) exhibits strong neuron protective effects against neurotoxicity. Hence, we investigate the effect of TET on BUP-induced neurotoxicity in SH-SY5Y cells. Methods: CCK-8 assay was used to detect cell proliferation in SH-SY5Y cells. In addition, Western blotting was used to examine Bax, Bcl-2, active caspase 3, LC3II, Beclin 1 and p-62 protein levels in cells. Moreover, ELISA assay was used to detect the levels of total glutathione (GS), superoxide dismutase (SOD) and malondialdehyde (MDA) in cells. Results: In this study, we found that TET attenuated the neurotoxicity of BUP on SH-SY5Y cells. Meanwhile, TET alleviated BUP-induced apoptosis in SH-SY5Y cell via decreasing the expressions of active caspase-3 and Bax and increasing the expression of Bcl-2. In addition, monodansylcadaverine staining assay and Western blotting results confirmed that TET induced autophagy in SH-SY5Y cells via increasing the LC3II/I and Beclin 1 levels. Furthermore, TET attenuated BUP-induced oxidative damage in SH-SY5Y cells via upregulation of the levels of total GS and SOD and downregulation of the level of MDA. Interesting, the protective effects of TET against BUP-induced neurotoxicity in SH-SY5Y cells were reversed by autophagy inhibitor 3-methyladenine (3MA). Conclusion: These data indicated that TET may play a neuroprotective role via inhibiting apoptosis and inducing autophagy in SH-SY5Y cells. Therefore, TET may be a potential agent for the treatment of human neurotoxicity induced by BUP.
Collapse
Affiliation(s)
- Shouliang Wang
- Department of Anesthesiology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, People's Republic of China
| | - Bin Xia
- Department of Anesthesiology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, People's Republic of China
| | - Zonglei Qiao
- Department of Anesthesiology, Qingyun County People's Hospital, Dezhou 253700, Shandong Province, People's Republic of China
| | - Lian Duan
- Department of Ophthalmology, Qianfoshan Hospital Affiliated to Medical School of Shandong University, Jinan 250014, Shandong Province, People's Republic of China
| | - Gongming Wang
- Department of Anesthesiology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, People's Republic of China
| | - Wenjun Meng
- Department of Anesthesiology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, People's Republic of China
| | - Zhifei Liu
- Department of Anesthesiology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, People's Republic of China
| | - Yu Wang
- Department of Anesthesiology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, People's Republic of China
| | - Mengyuan Zhang
- Department of Anesthesiology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, People's Republic of China
| |
Collapse
|