1
|
Wang L, Lian YJ, Dong JS, Liu MK, Liu HL, Cao ZM, Wang QN, Lyu WL, Bai YN. Traditional Chinese medicine for chronic atrophic gastritis: Efficacy, mechanisms and targets. World J Gastroenterol 2025; 31:102053. [DOI: 10.3748/wjg.v31.i9.102053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 12/06/2024] [Accepted: 01/21/2025] [Indexed: 02/18/2025] Open
Abstract
Chronic atrophic gastritis (CAG) is an important stage of precancerous lesions of gastric cancer. Effective treatment and regulation of CAG are essential to prevent its progression to malignancy. Traditional Chinese medicine (TCM) has shown multi-targeted efficacy in CAG treatment, with advantages in enhancing gastric mucosal barrier defense, improving microcirculation, modulating inflammatory and immune responses, and promoting lesion healing, etc. Clinical studies and meta-analyses indicate that TCM provides significant benefits, with specific Chinese herbal compounds and monomers demonstrating protective effects on the gastric mucosa through mechanisms including anti-inflammation, anti-oxidation, and regulation of cellular proliferation and apoptosis, etc. Finally, it is pointed out that the efficacy of TCM in the treatment of CAG requires standardized research and unified standards, and constantly clarifies and improves the evaluation criteria of each dimension of gastric mucosal barrier function.
Collapse
Affiliation(s)
- Li Wang
- Department of Gastroenterology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Yan-Jie Lian
- Division of Cardiovascular, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing 100010, China
| | - Jin-Sheng Dong
- Department of Gastroenterology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Ming-Kun Liu
- Department of Gastroenterology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Hong-Liang Liu
- Department of Gastroenterology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Zheng-Min Cao
- Department of Infectious Diseases, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Qing-Nan Wang
- Department of Dermatology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Wen-Liang Lyu
- Department of Infectious Diseases, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Yu-Ning Bai
- Department of Gastroenterology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| |
Collapse
|
2
|
Shi P, Zheng B, Zhang S, Guo Q. A review of the sources and pharmacological research of morroniside. Front Pharmacol 2024; 15:1423062. [PMID: 39301568 PMCID: PMC11411571 DOI: 10.3389/fphar.2024.1423062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 08/19/2024] [Indexed: 09/22/2024] Open
Abstract
Introduction Morroniside (Mor) is a bioactive compound found in Corni Fructus (CF) [Cornaceae; Cornus officinalis Siebold & Zucc.], which has been used as medicine and food in China, Korea, and Japan for over 2,000 years. This review summarizes recent progress on Mor, specifically focusing on its distribution, isolation, detection, and various pharmacological effects. Methods A literature survey on Mor was conducted using electronic databases such as PubMed, ScienceDirect, CNKI, and Google Scholar. After removing TCM prescription-related standards, medicinal herb processing-related research, and other irrelevant works of literature, we obtained relevant information on Mor's biological and pharmacological properties. Results The main conclusions are as follows: Mor is widely distributed in the plant kingdom; the methods for extracting and isolating Mor are well established; and the technology for detecting it is accurate. Mor exhibits numerous pharmacological effects. Along with CF, Mor has shown renoprotective effects against diabetes, hepatoprotective effects against diabetes, triptolide, and nonalcoholic steatohepatitis, and boneprotective effects against osteoporosis and osteoarthritis. In addition, researchers have also explored other pharmacological effects of Mor, including neuroprotective effects against focal cerebral ischemia, spinal cord injury, and Alzheimer's disease; cardioprotective effects against acute myocardial infarction; protection of the digestive system from gastritis, inflammatory bowel disease, and colitis; protection of the skin by promoting hair growth, wound healing, and flap survival; and protection of the lungs from acute lung injury and pulmonary fibrosis. Moreover, Mor has anti-obesity effects, anti-inflammatory effects in the eye, and improves follicular development. Discussion Overall, this review provides a comprehensive understanding of the pharmacological effects of Mor, from which the limitations of the current research can be understood, which will help facilitate future research.
Collapse
Affiliation(s)
- Pengliang Shi
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Bingqing Zheng
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Shiyao Zhang
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Qingmei Guo
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
3
|
Műzes G, Sipos F. Inflammasomes Are Influenced by Epigenetic and Autophagy Mechanisms in Colorectal Cancer Signaling. Int J Mol Sci 2024; 25:6167. [PMID: 38892354 PMCID: PMC11173330 DOI: 10.3390/ijms25116167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Revised: 05/31/2024] [Accepted: 05/31/2024] [Indexed: 06/21/2024] Open
Abstract
Inflammasomes contribute to colorectal cancer signaling by primarily inducing inflammation in the surrounding tumor microenvironment. Its role in inflammation is receiving increasing attention, as inflammation has a protumor effect in addition to inducing tissue damage. The inflammasome's function is complex and controlled by several layers of regulation. Epigenetic processes impact the functioning or manifestation of genes that are involved in the control of inflammasomes or the subsequent signaling cascades. Researchers have intensively studied the significance of epigenetic mechanisms in regulation, as they encompass several potential therapeutic targets. The regulatory interactions between the inflammasome and autophagy are intricate, exhibiting both advantageous and harmful consequences. The regulatory aspects between the two entities also encompass several therapeutic targets. The relationship between the activation of the inflammasome, autophagy, and epigenetic alterations in CRC is complex and involves several interrelated pathways. This article provides a brief summary of the newest studies on how epigenetics and autophagy control the inflammasome, with a special focus on their role in colorectal cancer. Based on the latest findings, we also provide an overview of the latest therapeutic ideas for this complex network.
Collapse
Affiliation(s)
- Györgyi Műzes
- Immunology Division, Department of Internal Medicine and Hematology, Semmelweis University, 1088 Budapest, Hungary
| | - Ferenc Sipos
- Immunology Division, Department of Internal Medicine and Hematology, Semmelweis University, 1088 Budapest, Hungary
| |
Collapse
|
4
|
Bhuia MS, Chowdhury R, Ara I, Mamun M, Rouf R, Khan MA, Uddin SJ, Shakil MAK, Habtemariam S, Ferdous J, Calina D, Sharifi-Rad J, Islam MT. Bioactivities of morroniside: A comprehensive review of pharmacological properties and molecular mechanisms. Fitoterapia 2024; 175:105896. [PMID: 38471574 DOI: 10.1016/j.fitote.2024.105896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 03/07/2024] [Accepted: 03/08/2024] [Indexed: 03/14/2024]
Abstract
Morroniside (MOR) is an iridoid glycoside and the main active principle of the medicinal plant, Cornus officinalis Sieb. This phytochemical is associated with numerous health benefits due to its antioxidant properties. The primary objective of the present study was to assess the pharmacological effects and underlying mechanisms of MOR, utilizing published data obtained from literature databases. Data collection involved accessing various sources, including PubMed/Medline, Scopus, Science Direct, Google Scholar, Web of Science, and SpringerLink. Our findings demonstrate that MOR can be utilized for the treatment of several diseases and disorders, as numerous studies have revealed its significant therapeutic activities. These activities encompass anti-inflammatory, antidiabetic, lipid-lowering capability, anticancer, trichogenic, hepatoprotective, gastroprotective, osteoprotective, renoprotective, and cardioprotective effects. MOR has also shown promising benefits against various neurological ailments, including Alzheimer's disease, Parkinson's disease, spinal cord injury, cerebral ischemia, and neuropathic pain. Considering these therapeutic features, MOR holds promise as a lead compound for the treatment of various ailments and disorders. However, further comprehensive preclinical and clinical trials are required to establish MOR as an effective and reliable therapeutic agent.
Collapse
Affiliation(s)
- Md Shimul Bhuia
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj 8100, Bangladesh
| | - Raihan Chowdhury
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj 8100, Bangladesh
| | - Iffat Ara
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj 8100, Bangladesh
| | - Md Mamun
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj 8100, Bangladesh
| | - Razina Rouf
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj 8100, Bangladesh
| | - Muahmmad Ali Khan
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj 8100, Bangladesh
| | | | - Md Abdul Kader Shakil
- Research Center, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj 8100, Bangladesh
| | - Solomon Habtemariam
- Pharmacognosy Research & Herbal Analysis Services UK, Central Avenue, Chatham-Maritime, Kent ME4 4TB, UK
| | - Jannatul Ferdous
- Department of Biotechnology and Genetic Engineering, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj 8100, Bangladesh
| | - Daniela Calina
- Department of Clinical Pharmacy, University of Medicine and Pharmacy of Craiova, Craiova 200349, Romania.
| | | | - Muhammad Torequl Islam
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj 8100, Bangladesh.
| |
Collapse
|
5
|
Lee DY, Bahar ME, Kim CW, Seo MS, Song MG, Song SY, Kim SY, Kim DR, Kim DH. Autophagy in Osteoarthritis: A Double-Edged Sword in Cartilage Aging and Mechanical Stress Response: A Systematic Review. J Clin Med 2024; 13:3005. [PMID: 38792546 PMCID: PMC11122125 DOI: 10.3390/jcm13103005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 05/13/2024] [Accepted: 05/17/2024] [Indexed: 05/26/2024] Open
Abstract
Background: Although osteoarthritis (OA) development is epidemiologically multifactorial, a primary underlying mechanism is still under debate. Understanding the pathophysiology of OA remains challenging. Recently, experts have focused on autophagy as a contributor to OA development. Method: To better understand the pathogenesis of OA, we survey the literature on the role of autophagy and the molecular mechanisms of OA development. To identify relevant studies, we used controlled vocabulary and free text keywords to search the MEDLINE, EMBASE, the Cochrane Central Register of Controlled Trials, Web of Science, and SCOPUS database. Thirty-one studies were included for data extraction and systematic review. Among these studies, twenty-five studies investigated the effects of autophagy in aging and OA chondrocytes, six studies examined the effects of autophagy in normal human chondrocytes, and only one study investigated the effects of mechanical stress-induced autophagy on the development of OA in normal chondrocytes. Results: The studies suggest that autophagy activation prevents OA by exerting cell-protective effects in normal human chondrocytes. However, in aging and osteoarthritis (OA) chondrocytes, the role of autophagy is intricate, as certain studies indicate that stimulating autophagy in these cells can have a cytotoxic effect, while others propose that it may have a protective (cytoprotective) effect against damage or degeneration. Conclusions: Mechanical stress-induced autophagy is also thought to be involved in the development of OA, but further research is required to identify the precise mechanism. Thus, autophagy contributions should be interpreted with caution in aging and the types of OA cartilage.
Collapse
Affiliation(s)
- Dong-Yeong Lee
- Department of Orthopaedic Surgery, Barun Hospital, Jinju 52725, Republic of Korea;
| | - Md Entaz Bahar
- Department of Biochemistry and Convergence Medical Sciences, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Republic of Korea; (M.E.B.); (M.-S.S.)
| | - Chang-Won Kim
- Department of Orthopaedic Surgery, Institute of Medical Science, Gyeongsang National University Hospital, Jinju 52727, Republic of Korea; (C.-W.K.); (S.-Y.S.); (S.-Y.K.)
| | - Min-Seok Seo
- Department of Biochemistry and Convergence Medical Sciences, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Republic of Korea; (M.E.B.); (M.-S.S.)
| | - Myung-Geun Song
- Department of Orthopaedic Surgery, Inha University Hospital, Incheon 22212, Republic of Korea;
| | - Sang-Youn Song
- Department of Orthopaedic Surgery, Institute of Medical Science, Gyeongsang National University Hospital, Jinju 52727, Republic of Korea; (C.-W.K.); (S.-Y.S.); (S.-Y.K.)
| | - Soung-Yon Kim
- Department of Orthopaedic Surgery, Institute of Medical Science, Gyeongsang National University Hospital, Jinju 52727, Republic of Korea; (C.-W.K.); (S.-Y.S.); (S.-Y.K.)
| | - Deok-Ryong Kim
- Department of Biochemistry and Convergence Medical Sciences, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Republic of Korea; (M.E.B.); (M.-S.S.)
| | - Dong-Hee Kim
- Department of Orthopaedic Surgery, Institute of Medical Science, Gyeongsang National University Hospital, Jinju 52727, Republic of Korea; (C.-W.K.); (S.-Y.S.); (S.-Y.K.)
| |
Collapse
|
6
|
Yang HZ, Dong R, Jia Y, Li Y, Luo G, Li T, Long Y, Liang S, Li S, Jin X, Sun T. Morroniside ameliorates glucocorticoid-induced osteoporosis and promotes osteoblastogenesis by interacting with sodium-glucose cotransporter 2. PHARMACEUTICAL BIOLOGY 2023; 61:416-426. [PMID: 36786302 PMCID: PMC9930836 DOI: 10.1080/13880209.2023.2173787] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 12/10/2022] [Accepted: 01/23/2023] [Indexed: 06/18/2023]
Abstract
CONTEXT Morroniside (MOR) possesses antiosteoporosis (OP) effects, but its molecular target and relevant mechanisms remain unknown. OBJECTIVE We investigated the effects of MOR on glucocorticoid-induced OP and osteoblastogenesis and its underlying mechanisms. MATERIALS AND METHODS The effects of MOR (10-100 μM) on the proliferation and differentiation of MC3T3-E1 cells were studied in vitro. The glucocorticoid-induced zebrafish OP model was treated with 10, 20 and 40 μM MOR for five days to evaluate its effects on vertebral bone density and related osteogenic markers. In addition, molecular targets prediction and molecular docking analysis were carried out to explore the binding interactions of MOR with the target proteins. RESULTS In cultured MC3T3-E1 cells, 20 μM MOR significantly increased cell viability (1.64 ± 0.12 vs. 0.95 ± 0.16; p < 0.01) and cell differentiation (1.57 ± 0.01 vs. 1.00 ± 0.04; p < 0.01) compared to the control group. MOR treatment significantly ameliorated vertebral bone loss in the glucocorticoid-induced OP zebrafish model (0.86 ± 0.02 vs. 0.40 ± 0.03; p < 0.01) and restored the expression of osteoblast-specific markers, including ALP, Runx2 and Col-І. Ligand-based target prediction and molecular docking revealed the binding interaction between MOR and the glucose pockets in sodium-glucose cotransporter 2 (SGLT2). DISCUSSION AND CONCLUSIONS These findings demonstrated that MOR treatment promoted osteoblastogenesis and ameliorated glucocorticoid-induced OP by targeting SGLT2, which may provide therapeutic potential in managing glucocorticoid-induced OP.
Collapse
Affiliation(s)
| | | | - Yutao Jia
- Department of Spinal Surgery, Tianjin Union Medical Center, Tianjin, China
| | - Yuqiao Li
- Tianjin Medical University, Tianjin, China
| | - Gan Luo
- Tianjin Medical University, Tianjin, China
| | - Tianhao Li
- Tianjin Medical University, Tianjin, China
| | - Yao Long
- School of Medicine, Nankai University, Tianjin, China
| | - Shuang Liang
- School of Medicine, Nankai University, Tianjin, China
| | - Shanshan Li
- School of Medicine, Nankai University, Tianjin, China
| | - Xin Jin
- School of Medicine, Nankai University, Tianjin, China
| | - Tianwei Sun
- Tianjin Medical University, Tianjin, China
- Department of Spinal Surgery, Tianjin Union Medical Center, Tianjin, China
| |
Collapse
|
7
|
Wang L, Meng X, Zhou H, Liu Y, Zhang Y, Liang H, Hou G, Kang W, Liu Z. Iridoids and active ones in patrinia: A review. Heliyon 2023; 9:e16518. [PMID: 37292326 PMCID: PMC10245019 DOI: 10.1016/j.heliyon.2023.e16518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 04/27/2023] [Accepted: 05/19/2023] [Indexed: 06/10/2023] Open
Abstract
Iridoid is a special class of monoterpenoids, whose basic skeleton is the acetal derivative of antinodilaldehyde with a bicyclic H-5/H-9β, β-cisfused cyclopentan pyran ring. They were often existed in Valerianaceae, Rubiaceae, Scrophulariaceae and Labiaceae family, and has various biological activities, such as anti-inflammatory, hypoglycemic, neuroprotection, and soon. In this review, iridoids from Patrinia (Valerianaceae family), and the active ones as well as their mechanisms in recent 20 years were summarized. Up to now, a total of 115 iridoids had been identified in Patrinia, among which 48 had extensive biological activities mainly presented in anti-inflammatory, anti-tumor and neuroprotective. And the mechanisms involved in MAPK, NF-κB and JNK signal pathways. The summary for iridoids and their activities will provide the evidence to exploit the iridoids in Patrinia.
Collapse
Affiliation(s)
- Li Wang
- National R & D Center for Edible Fungus Processing Technology, Henan University, Kaifeng 475004, China
- Functional Food Engineering Technology Research Center, Henan, Kaifeng, 475004, China
| | - Xinjing Meng
- National R & D Center for Edible Fungus Processing Technology, Henan University, Kaifeng 475004, China
- Functional Food Engineering Technology Research Center, Henan, Kaifeng, 475004, China
| | - Huihui Zhou
- National R & D Center for Edible Fungus Processing Technology, Henan University, Kaifeng 475004, China
- Joint International Research Laboratory of Food & Medicine Resource Function, Henan Province, Kaifeng 475004, China
| | - Yuhang Liu
- National R & D Center for Edible Fungus Processing Technology, Henan University, Kaifeng 475004, China
- Joint International Research Laboratory of Food & Medicine Resource Function, Henan Province, Kaifeng 475004, China
| | - Yadan Zhang
- National R & D Center for Edible Fungus Processing Technology, Henan University, Kaifeng 475004, China
- Joint International Research Laboratory of Food & Medicine Resource Function, Henan Province, Kaifeng 475004, China
| | - Haiyang Liang
- National R & D Center for Edible Fungus Processing Technology, Henan University, Kaifeng 475004, China
- Joint International Research Laboratory of Food & Medicine Resource Function, Henan Province, Kaifeng 475004, China
| | - Gaixia Hou
- National R & D Center for Edible Fungus Processing Technology, Henan University, Kaifeng 475004, China
- College of Physical Education, Henan University, Henan, Kaifeng, 475004, China
| | - Wenyi Kang
- National R & D Center for Edible Fungus Processing Technology, Henan University, Kaifeng 475004, China
- Joint International Research Laboratory of Food & Medicine Resource Function, Henan Province, Kaifeng 475004, China
- Shenzhen Research Institute of Henan University, Shenzhen, 518000, China
| | - Zhenhua Liu
- National R & D Center for Edible Fungus Processing Technology, Henan University, Kaifeng 475004, China
- Joint International Research Laboratory of Food & Medicine Resource Function, Henan Province, Kaifeng 475004, China
- Shenzhen Research Institute of Henan University, Shenzhen, 518000, China
| |
Collapse
|
8
|
Ke X, Yu S, Situ S, Lin Z, Yuan Y. Morroniside inhibits Beclin1-dependent autophagic death and Bax-dependent apoptosis in cardiomyocytes through repressing BCL2 phosphorylation. In Vitro Cell Dev Biol Anim 2023:10.1007/s11626-023-00768-0. [PMID: 37155079 DOI: 10.1007/s11626-023-00768-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 04/13/2023] [Indexed: 05/10/2023]
Abstract
Morroniside can prevent myocardial injury caused by ischemia and hypoxia, which can be used to treat acute myocardial infarction (AMI). Hypoxia can cause apoptosis and autophagic death of cardiomyocytes. Morroniside has the ability to inhibit apoptosis and autophagy. However, the relationship between Morroniside-protected cardiomyocytes and two forms of death is unclear. The effects of Morroniside on the proliferation, apoptosis level, and autophagic activity of rat cardiomyocyte line H9c2 under hypoxia were first observed. Next, the roles of Morroniside in the phosphorylation of JNK and BCL2, BCL2-Beclin1, and BCL2-Bax complexes as well as mitochondrial membrane potential in H9c2 cells were evaluated upon hypoxia. Finally, the significance of BCL2 or JNK in Morroniside-regulated autophagy, apoptosis, and proliferation in H9c2 cells was assessed by combining Morroniside and BCL2 competitive inhibitor (ABT-737) or JNK activator (Anisomycin). Our results showed that hypoxia promoted autophagy and apoptosis of H9c2 cells, and inhibited their proliferation. However, Morroniside could block the effect of hypoxia on H9c2 cells. In addition, Morroniside could inhibit JNK phosphorylation, BCL2 phosphorylation at the Ser70 and Ser87 sites, and the dissociation of BCL2-Beclin1 and BCL2-Bax complexes in H9c2 cells upon hypoxia. Moreover, the reduction of mitochondrial membrane potential in H9c2 cells caused by hypoxia was improved by Morroniside administration. Importantly, the inhibited autophagy, apoptosis, and promoted proliferation in H9c2 cells by Morroniside were reversed by the application of ABT-737 or Anisomycin. Overall, Morroniside inhibits Beclin1-dependent autophagic death and Bax-dependent apoptosis via JNK-mediated BCL2 phosphorylation, thereby improving the survival of cardiomyocytes under hypoxia.
Collapse
Affiliation(s)
- Xueping Ke
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China
- The Sixth Affiliated Hospital, School of Medicine, South China University of Technology, Foshan, 528200, China
| | - Shicheng Yu
- Department of Medicine, Liwan Central Hospital of Guangzhou, Guangzhou, 510145, China
| | - Shubiao Situ
- Department of Medicine, Liwan Central Hospital of Guangzhou, Guangzhou, 510145, China
| | - Zhenqian Lin
- Department of Cardiology, Henan Provincial Chest Hospital, Zhengzhou, 450008, Henan, China
| | - Yiqiang Yuan
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China.
- Department of Cardiology, Zheng Zhou NO.7 People's Hospital, No.17, Jingnan 5th Road, Zhengzhou Economic and Technological Development Zone, Zhengzhou, 450016, China.
| |
Collapse
|
9
|
Santos TMR, Tavares CA, Pereira AF, da Cunha EFF, Ramalho TC. Evaluation of autophagy inhibition to combat cancer: (vanadium complex)-protein interactions, parameterization, and validation of a new force field. J Mol Model 2023; 29:123. [PMID: 36995564 PMCID: PMC10061415 DOI: 10.1007/s00894-023-05530-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 03/23/2023] [Indexed: 03/31/2023]
Abstract
Autophagy has drawn attention from the scientific community, mainly because of its significant advantages over chemotherapeutic processes. One of these advantages is its direct action on cancer cells, avoiding possible side effects, unlike chemotherapy, which reaches tumor cells and affects healthy cells in the body, leading to a great loss in the quality of life of patients. In this way, it is known that vanadium complex (VC) [VO(oda)(phen)] has proven inhibition effect on autophagy process in pancreatic cancer cells. Keeping that in mind, molecular dynamics (MD) simulations can be considered excellent strategies to investigate the interaction of metal complexes and their biological targets. However, simulations of this type are strongly dependent on the appropriate choice of force field (FF). Therefore, this work proposes the development of AMBER FF parameters for VC, having a minimum energy structure as a starting point, obtained through DFT calculations with B3LYP/def2-TZVP level of theory plus ECP for the vanadium atom. An MD simulation in vacuum was performed to validate the developed FF. From the structural analyses, satisfying values of VC bond lengths and angles were obtained, where a good agreement with the experimental data and the quantum reference was found. The RMSD analysis showed an average of only 0.3%. Finally, we performed docking and MD (120 ns) simulations with explicit solvent between VC and PI3K. Overall, our findings encourage new parameterizations of metal complexes with significant biological applications, as well as allow to contribute to the elucidation of the complex process of autophagy.
Collapse
Affiliation(s)
- Taináh M R Santos
- Laboratory of Molecular Modelling, Department of Chemistry, Federal University of Lavras, /MG, Lavras, 37200-000, Brazil.
| | - Camila A Tavares
- Laboratory of Molecular Modelling, Department of Chemistry, Federal University of Lavras, /MG, Lavras, 37200-000, Brazil
| | - Ander F Pereira
- Institute of Chemistry, University of Campinas, /SP, Campinas, 13083-970, Brazil
| | - Elaine F F da Cunha
- Laboratory of Molecular Modelling, Department of Chemistry, Federal University of Lavras, /MG, Lavras, 37200-000, Brazil
| | - Teodorico C Ramalho
- Laboratory of Molecular Modelling, Department of Chemistry, Federal University of Lavras, /MG, Lavras, 37200-000, Brazil.
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, Hradec Kralove, Czech Republic.
| |
Collapse
|
10
|
董 润, 贾 宇, 杨 厚, 罗 干, 李 玉, 孙 天. [Effects and mechanism of morroniside on osteogenic differentiation and proliferation of mouse MC3T3-E1 cells]. ZHONGGUO XIU FU CHONG JIAN WAI KE ZA ZHI = ZHONGGUO XIUFU CHONGJIAN WAIKE ZAZHI = CHINESE JOURNAL OF REPARATIVE AND RECONSTRUCTIVE SURGERY 2022; 36:889-895. [PMID: 35848187 PMCID: PMC9288899 DOI: 10.7507/1002-1892.202202088] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 05/25/2022] [Indexed: 01/24/2023]
Abstract
Objective To study the effects of morroniside (MOR) on the proliferation and osteogenic differentiation of mouse MC3T3-E1 cells. Methods The 4th generation MC3T3-E1 cells were randomly divided into 6 groups: control group (group A), MOR low dose group (10 μmol/L, group B), MOR medium-low dose group (20 μmol/L, group C), MOR medium dose group (40 μmol/L, group D), MOR medium-high dose group (80 μmol/L, group E), and MOR high dose group (100 μmol/L, group F). The proliferation activity of each group was detected by cell counting kit 8 (CCK-8) assay; the bone differentiation and mineralized nodule formation of each group were detected by alizarin red staining; real-time fluorescence quantitative PCR (RT-qPCR) was performed to detect cyclin-dependent kinase inhibitor 1A (P21), recombinant Cyclin D1 (CCND1), proliferating cell nuclear antigen (PCNA), alkaline phosphatase (ALP), collagen type Ⅰ (COL-1), bone morphogenetic protein 2 (BMP-2), and adenosine A2A receptor (A2AR) mRNA expressions; Western blot was used to detecte the expressions of osteopontin (OPN), Runt-related transcription factor 2 (RUNX2), and adenosine A2AR protein. Results The CCK-8 assay showed that the absorbance ( A) values of groups B to F were significantly higher than that of group A at 24 hours of culture, with group C significantly higher than the rest of the groups ( P<0.05). The MOR concentration (20 μmol/L) of group C was selected for the subsequent CCK-8 assay; the results showed that the A values of group C were significantly higher than those of group A at 24, 48, and 72 hours of culture ( P<0.05). Alizarin red staining showed that orange-red mineralized nodules were visible in all groups and the number of mineralized nodules was significantly higher in groups B and C than in group A ( P<0.05). RT-qPCR showed that the relative expressions of P21, CCND1, and PCNA mRNAs were significantly higher in group C than in group A ( P<0.05). The expressions of ALP, BMP-2, COL-1, and adenosine A2AR mRNAs in groups B to E were significantly higher than those in group A, with the expressions of ALP, BMP-2, COL-1 mRNAs in group C significantly higher than the rest of the groups ( P<0.05). Compared with group A, the expressions of OPN and RUNX2 proteins in groups B and C were significantly increased, while those in group D and E were significantly inhibited ( P<0.05). There was no significant difference between groups B and C and between groups D and E ( P>0.05). The relative expression of adenosine A2AR protein in groups B to E was significantly higher than that in group A, with group C significantly higher than the rest of the groups ( P<0.05). Conclusion MOR can promote the proliferation and osteogenic differentiation of MC3T3-E1 cells; the mechanism of MOR may be achieved by interacting with adenosine A2AR.
Collapse
Affiliation(s)
- 润北 董
- 天津医科大学研究生院(天津 300070)Graduate School of Tianjin Medical University, Tianjin, 300070, P. R. China
| | - 宇涛 贾
- 天津医科大学研究生院(天津 300070)Graduate School of Tianjin Medical University, Tianjin, 300070, P. R. China
| | - 厚志 杨
- 天津医科大学研究生院(天津 300070)Graduate School of Tianjin Medical University, Tianjin, 300070, P. R. China
| | - 干 罗
- 天津医科大学研究生院(天津 300070)Graduate School of Tianjin Medical University, Tianjin, 300070, P. R. China
| | - 玉乔 李
- 天津医科大学研究生院(天津 300070)Graduate School of Tianjin Medical University, Tianjin, 300070, P. R. China
| | - 天威 孙
- 天津医科大学研究生院(天津 300070)Graduate School of Tianjin Medical University, Tianjin, 300070, P. R. China
| |
Collapse
|
11
|
Tian Z, Zhang X, Sun M. Phytochemicals Mediate Autophagy Against Osteoarthritis by Maintaining Cartilage Homeostasis. Front Pharmacol 2022; 12:795058. [PMID: 34987406 PMCID: PMC8722717 DOI: 10.3389/fphar.2021.795058] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 12/01/2021] [Indexed: 12/11/2022] Open
Abstract
Osteoarthritis (OA) is a common degenerative joint disease and is a leading cause of disability and reduced quality of life worldwide. There are currently no clinical treatments that can stop or slow down OA. Drugs have pain-relieving effects, but they do not slow down the course of OA and their long-term use can lead to serious side effects. Therefore, safe and clinically appropriate long-term treatments for OA are urgently needed. Autophagy is an intracellular protective mechanism, and targeting autophagy-related pathways has been found to prevent and treat various diseases. Attenuation of the autophagic pathway has now been found to disrupt cartilage homeostasis and plays an important role in the development of OA. Therefore, modulation of autophagic signaling pathways mediating cartilage homeostasis has been considered as a potential therapeutic option for OA. Phytochemicals are active ingredients from plants that have recently been found to reduce inflammatory factor levels in cartilage as well as attenuate chondrocyte apoptosis by modulating autophagy-related signaling pathways, which are not only widely available but also have the potential to alleviate the symptoms of OA. We reviewed preclinical studies and clinical studies of phytochemicals mediating autophagy to regulate cartilage homeostasis for the treatment of OA. The results suggest that phytochemicals derived from plant extracts can target relevant autophagic pathways as complementary and alternative agents for the treatment of OA if subjected to rigorous clinical trials and pharmacological tests.
Collapse
Affiliation(s)
- Zheng Tian
- School of Kinesiology, Shenyang Sport University, Shenyang, China
| | - Xinan Zhang
- School of Kinesiology, Shenyang Sport University, Shenyang, China
| | - Mingli Sun
- School of Kinesiology, Shenyang Sport University, Shenyang, China
| |
Collapse
|
12
|
Liu H, Li X, Lin J, Lin M. Morroniside promotes the osteogenesis by activating PI3K/Akt/mTOR signaling. Biosci Biotechnol Biochem 2021; 85:332-339. [PMID: 33604633 DOI: 10.1093/bbb/zbaa010] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 09/11/2020] [Indexed: 12/16/2022]
Abstract
Morroniside exerts a proosteogenic effect, which can prevent bone loss. However, the detailed mechanism underlying Morroniside-regulated bone formation is unclear. Morroniside can maintain cell homeostasis by promoting PI3K/Akt/mTOR signaling. The purpose of this study is to explore the significance of PI3K/Akt/mTOR signaling in Morroniside-regulated osteogenesis. The results showed that Morroniside promoted the activities of PI3K, Akt, and mTOR in osteoblast precursor MC3T3-E1. The differentiation of MC3T3-E1 to mature osteoblasts promoted by Morroniside can be reversed by the pharmacological inhibition of PI3K or mTOR. Importantly, in the presence of Morroniside, the osteoblast differentiation suppressed by PI3K inhibitor was reversed by mTOR overexpression. In vivo assays showed that in bone tissue of ovariectomized mice, Morroniside-enhanced osteoblast formation was reversed by the pharmacological inhibition of PI3K or mTOR. In conclusion, Morroniside can promote the osteogenesis through PI3K/Akt/mTOR signaling, which provides a novel clue for the strategy of Morroniside in treating osteoporosis.
Collapse
Affiliation(s)
- Hui Liu
- Department of Orthopedics, Fuzhou Second Hospital Affiliated to Xiamen University, Fuzhou, Fujian, China
| | - Xi Li
- Department of Orthopedics, Fuzhou Second Hospital Affiliated to Xiamen University, Fuzhou, Fujian, China
| | - Jingui Lin
- Department of Orthopedics, Fuzhou Second Hospital Affiliated to Xiamen University, Fuzhou, Fujian, China
| | - Miaokuo Lin
- Department of Orthopedics, Fuzhou Second Hospital Affiliated to Xiamen University, Fuzhou, Fujian, China
| |
Collapse
|