1
|
He Q, Lin Y, Chen B, Chen C, Zeng J, Dou X, Cai D, Zhou C, Wang H. Vitamin K2 ameliorates osteoarthritis by suppressing ferroptosis and extracellular matrix degradation through activation GPX4's dual functions. Biomed Pharmacother 2024; 175:116697. [PMID: 38759289 DOI: 10.1016/j.biopha.2024.116697] [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: 02/15/2024] [Revised: 04/23/2024] [Accepted: 05/01/2024] [Indexed: 05/19/2024] Open
Abstract
Vitamin K2 (VK2) is an effective compound for anti-ferroptosis and anti-osteoporosis, and Semen sojae praeparatum (Dandouchi in Chinese) is the main source of VK2. Chondrocyte ferroptosis and extracellular matrix (ECM) degradation playing a role in the pathogenesis of osteoarthritis (OA). Glutathione peroxidase 4 (GPX4) is the intersection of two mechanisms in regulating OA progression. But no studies have elucidated the therapeutic effects and mechanisms of VK2 on OA. This study utilized an in vivo rat OA model created via anterior cruciate ligament transection (ACLT) and an in vitro chondrocyte oxidative damage model induced by TBHP to investigate the protective effects and mechanisms of action of VK2 in OA. Knee joint pain in mice was evaluated using the Von Frey test. Micro-CT and Safranin O-Fast Green staining were employed to observe the extent of damage to the tibial cartilage and subchondral bone, while immunohistochemistry and PCR were used to examine GPX4 levels in joint cartilage. The effects of VK2 on rat chondrocyte viability were assessed using CCK-8 and flow cytometry assays, and chondrocyte morphology was observed with toluidine blue and alcian blue staining. The impact of VK2 on intracellular ferroptosis-related markers was observed using fluorescent staining and flow cytometry. Protein expression changes were detected by immunofluorescence and Western blot analysis. Furthermore, specific protein inhibitors were applied to confirm the dual-regulatory effects of VK2 on GPX4. VK2 can increase bone mass and cartilage thickness in the subchondral bone of the tibia, and reduce pain and the OARSI score induced by OA. Immunohistochemistry results indicate that VK2 exerts its anti-OA effects by regulating GPX4 to delay ECM degradation. VK2 can inhibit the activation of the MAPK/NFκB signaling pathway caused by reduced expression of intracellular GPX4, thereby decreasing ECM degradation. Additionally, VK2 can reverse the inhibitory effect of RSL3 on GPX4, increase intracellular GSH content and the GSH/GSSG ratio, reduce MDA content, and rescue chondrocyte ferroptosis. The protective mechanism of VK2 may involve its dual-target regulation of GPX4, reducing chondrocyte ferroptosis and inhibiting the MAPK/NFκB signaling pathway to decelerate the degradation of the chondrocyte extracellular matrix.
Collapse
Affiliation(s)
- Qi He
- First School of Clinical Medicine, Guangzhou University of Chinese Medicine, 12 Jichang Road, Baiyun Area, Guangzhou 510405, PR China; The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, PR China; Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Yuewei Lin
- First School of Clinical Medicine, Guangzhou University of Chinese Medicine, 12 Jichang Road, Baiyun Area, Guangzhou 510405, PR China; The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, PR China
| | - Baihao Chen
- First School of Clinical Medicine, Guangzhou University of Chinese Medicine, 12 Jichang Road, Baiyun Area, Guangzhou 510405, PR China; The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, PR China
| | - Chuyi Chen
- First School of Clinical Medicine, Guangzhou University of Chinese Medicine, 12 Jichang Road, Baiyun Area, Guangzhou 510405, PR China; The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, PR China
| | - Jiaxu Zeng
- First School of Clinical Medicine, Guangzhou University of Chinese Medicine, 12 Jichang Road, Baiyun Area, Guangzhou 510405, PR China; The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, PR China
| | - Xiangyun Dou
- First School of Clinical Medicine, Guangzhou University of Chinese Medicine, 12 Jichang Road, Baiyun Area, Guangzhou 510405, PR China; The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, PR China
| | - Dongling Cai
- Department of Orthopedics, Panyu Hospital of Chinese Medicine, Guangzhou 510405, PR China.
| | - Chi Zhou
- Department of Orthopaedics, The First Affiliated Hospital, Guangzhou University of Chinese Medicine, 16 Jichang Road, Baiyun Area, Guangzhou 510405, PR China; Department of Orthopaedics, Maoming Hospital of Guangzhou University of Chinese Medicine, Maoming 525000, PR China.
| | - Haibin Wang
- Department of Orthopaedics, The First Affiliated Hospital, Guangzhou University of Chinese Medicine, 16 Jichang Road, Baiyun Area, Guangzhou 510405, PR China.
| |
Collapse
|
2
|
He Q, Yang J, Chen W, Pan Z, Chen B, Zeng J, Zhang N, Lin Y, Chen C, Xiao J, Li M, Li S, Wang H, Chen P. Biochanin A abrogates osteoclastogenesis in type 2 diabetic osteoporosis via regulating ROS/MAPK signaling pathway based on integrating molecular docking and experimental validation. BMC Complement Med Ther 2024; 24:24. [PMID: 38191438 PMCID: PMC10773052 DOI: 10.1186/s12906-023-04332-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 12/27/2023] [Indexed: 01/10/2024] Open
Abstract
BACKGROUND There are accumulating type 2 diabetes patients who have osteoporosis simultaneously. More effective therapeutic strategies should be discovered. Biochanin A (BCA) has been indicated that can play a role in improving metabolic disorders of type 2 diabetes and preventing osteoporosis. But whether BCA can treat type 2 diabetic osteoporosis has not been studied. PURPOSE To investigate if the BCA can protect against type 2 diabetic osteoporosis and clarify the mechanism. METHODS Micro-CT and histology assays were performed to detect the trabecular bone and analyze the bone histomorphology effect of BCA. CCK-8 assay was performed to detect the toxicity of BCA. TRAcP staining, immunofluorescence and hydroxyapatite resorption assay were used to observe osteoclasts differentiation and resorptive activity. Molecular docking provided evidence about BCA regulating the MAPK axis via prediction by the algorithm. QRT-PCR and Western Blotting were utilized to detect the expression of osteoclastogenesis-related markers and MAPK signaling pathway. RESULTS Accumulation of bone volume after BCA treatment could be found based on the 3D reconstruction. Besides, there were fewer osteoclasts in db/db mice treated with BCA than db/db mice treated with saline. In vitro, we found that BCA hadn't toxicity in osteoclasts precursor, but also inhibited differentiation of osteoclasts. Further, we found that BCA suppresses osteoclastogenesis via ROS/MAPK signaling pathway. CONCLUSION BCA can prevent type 2 diabetic osteoporosis by restricting osteoclast differentiation via ROS/MAPK signaling pathway.
Collapse
Affiliation(s)
- Qi He
- First School of Medicine, Guangzhou University of Chinese Medicine, 12 Jichang Road, Baiyun Area, Guangzhou, 510405, P.R. China
- The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, P.R. China
| | - Junzheng Yang
- The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, P.R. China
- Fifth School of Medicine, Guangzhou University of Chinese Medicine, 12 Jichang Road, Baiyun Area, Guangzhou, 510405, P.R. China
| | - Weijian Chen
- Fifth School of Medicine, Guangzhou University of Chinese Medicine, 12 Jichang Road, Baiyun Area, Guangzhou, 510405, P.R. China
| | - Zhaofeng Pan
- First School of Medicine, Guangzhou University of Chinese Medicine, 12 Jichang Road, Baiyun Area, Guangzhou, 510405, P.R. China
- The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, P.R. China
| | - Baihao Chen
- First School of Medicine, Guangzhou University of Chinese Medicine, 12 Jichang Road, Baiyun Area, Guangzhou, 510405, P.R. China
- The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, P.R. China
| | - Jiaxu Zeng
- First School of Medicine, Guangzhou University of Chinese Medicine, 12 Jichang Road, Baiyun Area, Guangzhou, 510405, P.R. China
- The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, P.R. China
| | - Nenling Zhang
- The State Key Laboratory of Functions and Applications of Medicinal Plants, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, 550025, P.R. China
| | - Yuewei Lin
- First School of Medicine, Guangzhou University of Chinese Medicine, 12 Jichang Road, Baiyun Area, Guangzhou, 510405, P.R. China
- The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, P.R. China
| | - Chuyi Chen
- First School of Medicine, Guangzhou University of Chinese Medicine, 12 Jichang Road, Baiyun Area, Guangzhou, 510405, P.R. China
- The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, P.R. China
| | - Jiacong Xiao
- First School of Medicine, Guangzhou University of Chinese Medicine, 12 Jichang Road, Baiyun Area, Guangzhou, 510405, P.R. China
- The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, P.R. China
| | - Miao Li
- First School of Medicine, Guangzhou University of Chinese Medicine, 12 Jichang Road, Baiyun Area, Guangzhou, 510405, P.R. China
- The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, P.R. China
| | - Shaocong Li
- First School of Medicine, Guangzhou University of Chinese Medicine, 12 Jichang Road, Baiyun Area, Guangzhou, 510405, P.R. China
- The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, P.R. China
| | - Haibin Wang
- Department of Orthopaedics, First Affiliated Hospital, Guangzhou University of Chinese Medicine, 12 Jichang Road, Baiyun Area, Guangzhou, 510405, P.R. China.
| | - Peng Chen
- Department of Orthopaedics, First Affiliated Hospital, Guangzhou University of Chinese Medicine, 12 Jichang Road, Baiyun Area, Guangzhou, 510405, P.R. China.
| |
Collapse
|
3
|
Zhang P, He J, Gan Y, Shang Q, Chen H, Zhao W, Shen G, Jiang X, Ren H. Plastrum testudinis Ameliorates Oxidative Stress in Nucleus Pulposus Cells via Downregulating the TNF-α Signaling Pathway. Pharmaceuticals (Basel) 2023; 16:1482. [PMID: 37895953 PMCID: PMC10610230 DOI: 10.3390/ph16101482] [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: 08/31/2023] [Revised: 10/05/2023] [Accepted: 10/13/2023] [Indexed: 10/29/2023] Open
Abstract
BackgroundPlastrum testudinis (PT), a widely used traditional Chinese medicine, exerts protective effects against bone diseases such as intervertebral disc degeneration (IDD). Despite its effectiveness, the molecular mechanisms underlying the effects of PT on IDD remain unclear. Methods In this study, we used a comprehensive strategy combining bioinformatic analysis with experimental verification to investigate the possible molecular mechanisms of PT against IDD. We retrieved targets for PT and IDD, and then used their overlapped targets for protein-protein interaction (PPI) analysis. In addition, we used Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses to investigate the anti-IDD mechanisms of PT. Moreover, in vivo and in vitro experiment validations including hematoxylin-eosin (HE) and safranine O-green staining, senescence-associated β-galactosidase (SA-β-gal) assay, cell immunofluorescence staining, intracellular ROS measurement and Western blot analysis were performed to verify bioinformatics findings. Results We identified 342 and 872 PT- and IDD-related targets (32 overlapping targets). GO enrichment analysis yielded 450 terms related to oxidative stress and inflammatory response regulation. KEGG analysis identified 48 signaling pathways, 10 of which were significant; the TNF-α signaling pathway had the highest p-value, and prostaglandin G/H synthase 2 (PTGS2), endothelin-1 (EDN1), TNF-α, JUN and FOS were enriched in this pathway. Histopathological results and safranin O/green staining demonstrated that PT attenuated IDD, and SA-β-gal assay showed that PT ameliorated nucleus pulposus cell (NPC) senescence. An ROS probe was adopted to confirm the protective effect of PT against oxidative stress. Western blot analyses confirmed that PT downregulated the protein expression of PTGS2, EDN1, TNF-α, JUN and FOS in the TNF-α signaling pathway as well as cellular senescence marker p16, proinflammatory cytokine interleukin-6 (IL6), while PT upregulated the expression of NPC-specific markers including COL2A1 and ACAN in a concentration-dependent manner. Conclusions To the best of our knowledge, this study is the first to report that PT alleviates IDD by downregulating the protein expression of PTGS2, EDN1, TNF-α, JUN and FOS in the TNF-α signaling pathway and upregulating that of COL2A1 and ACAN, thus suppressing inflammatory responses and oxidative stress in NPCs.
Collapse
Affiliation(s)
- Peng Zhang
- Guangzhou University of Chinese Medicine, Guangzhou 510405, China; (P.Z.); (Y.G.); (Q.S.); (H.C.)
| | - Jiahui He
- The Affiliated TCM Hospital of Guangzhou Medical University, Guangzhou 510130, China;
| | - Yanchi Gan
- Guangzhou University of Chinese Medicine, Guangzhou 510405, China; (P.Z.); (Y.G.); (Q.S.); (H.C.)
| | - Qi Shang
- Guangzhou University of Chinese Medicine, Guangzhou 510405, China; (P.Z.); (Y.G.); (Q.S.); (H.C.)
| | - Honglin Chen
- Guangzhou University of Chinese Medicine, Guangzhou 510405, China; (P.Z.); (Y.G.); (Q.S.); (H.C.)
| | - Wenhua Zhao
- Department of Spinal Surgery, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, China; (W.Z.); (G.S.)
| | - Gengyang Shen
- Department of Spinal Surgery, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, China; (W.Z.); (G.S.)
| | - Xiaobing Jiang
- Department of Spinal Surgery, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, China; (W.Z.); (G.S.)
| | - Hui Ren
- Department of Spinal Surgery, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, China; (W.Z.); (G.S.)
| |
Collapse
|
4
|
Zhao XL, Lin GY, Liu T. Anti-diabetic effect of Ornithogalum caudatum Jacq. polysaccharides via the PI3K/Akt/GSK-3β signaling pathway and regulation of gut microbiota. Heliyon 2023; 9:e20808. [PMID: 37860571 PMCID: PMC10582383 DOI: 10.1016/j.heliyon.2023.e20808] [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: 06/19/2023] [Revised: 09/12/2023] [Accepted: 10/06/2023] [Indexed: 10/21/2023] Open
Abstract
This study evaluated the anti-diabetic effect of polysaccharides isolated from Ornithogalum caudatum and their underlying mechanisms. To achieve this, a type 2 diabetes mellitus mouse model was established using a combination of a high-fat diet and low-dose streptozotocin injection. The mice were treated with Ornithogalumcaudatum polysaccharides (OCPs) for 4 weeks. OCPs treatment significantly decreased body weight loss, fasting blood glucose levels, and plasma insulin levels in diabetic mice. Additionally, compared with the untreated group, OCPs treatment significantly decreased total cholesterol, triacylglycerol, and low-density lipoprotein-cholesterol levels, but increased those of high-density lipoprotein-cholesterol in diabetic mice. Moreover, antioxidant enzyme activity and histopathology results revealed that OCPs effectively alleviated oxidative stress and streptozotocin-induced lesions by increasing antioxidant enzyme activity. Results from mechanistic studies showed that OCPs treatment significantly increased the expression of p-PI3K, p-Akt, and p-GSK-3β in the liver. Moreover, OCPs optimized the gut microbiota composition of diabetic mice by significantly decreasing the Firmicutes/Bacteroidetes ratio and increasing the levels of beneficial bacteria (Muribaculaceae_norank, Prevotellaceae_UCG-001 and Alloprevotella). Overall, these findings suggest that OCPs exert anti-diabetic effects by triggering the PI3K/Akt/GSK-3β signaling pathway and regulating the gut microbiota.
Collapse
Affiliation(s)
- Xue-liang Zhao
- Key Laboratory for Metabolic Regulation and Activity Research of Medicinal Plants, Baicheng Medical College, Baicheng, Jilin 137000, China
| | - Guang-yu Lin
- Jilin animal husbandry information center, Changchun, Jilin 130000, China
| | - Tong Liu
- Key Laboratory for Metabolic Regulation and Activity Research of Medicinal Plants, Baicheng Medical College, Baicheng, Jilin 137000, China
| |
Collapse
|
5
|
Liang F, Du L, Rao X, Li Y, Long W, Tian J, Zhu X, Zou A, Lu W, Wan B. Effect of electroacupuncture at ST36 on the cerebral metabolic kinetics of rheumatoid arthritis rats. Brain Res Bull 2023; 201:110700. [PMID: 37414302 DOI: 10.1016/j.brainresbull.2023.110700] [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: 02/25/2023] [Revised: 06/27/2023] [Accepted: 07/03/2023] [Indexed: 07/08/2023]
Abstract
Electroacupuncture (EA) has been shown to enhance the recovery of symptoms in rheumatoid arthritis (RA); however, the underlying mechanism remains unclear. Both the pathogenesis of RA and the therapeutic effects of EA are closely associated with the metabolic activity of the brain. In this study, we investigated the effect of EA at the "Zusanli" acupoint (ST36) on a rat model of collagen-induced rheumatoid arthritis (CIA). The results demonstrated that EA effectively alleviated joint swelling, synovial hyperplasia, cartilage erosion, and bone destruction in CIA rats. Additionally, the metabolic kinetics study revealed a significant increase in the 13C enrichment of GABA2 and Glu4 in the midbrain of CIA rats treated with EA. Correlation network analysis showed that changes in Gln4 levels in the hippocampus were strongly associated with the severity of rheumatoid arthritis. Immunofluorescence staining of c-Fos in the midbrain's periaqueductal gray matter (PAG) and hippocampus demonstrated increased c-Fos expression in these regions following EA treatment. These findings suggest that GABAergic and glutamatergic neurons in the midbrain, along with astrocytes in the hippocampus, may play vital roles in the beneficial effects of EA on RA. Furthermore, the PAG and hippocampus brain regions hold potential as critical targets for future RA treatments. Overall, this study provides valuable insights into the specific mechanism of EA in treating RA by elucidating the perspective of cerebral metabolism.
Collapse
Affiliation(s)
- Fangyuan Liang
- College of Acupuncture and Orthopedics, Hubei University of Chinese Medicine, Wuhan, China
| | - Lei Du
- Eye Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xiaoping Rao
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences-Wuhan National Laboratory for Optoelectronics, Wuhan, China
| | - Ying Li
- College of Acupuncture and Orthopedics, Hubei University of Chinese Medicine, Wuhan, China
| | - Wei Long
- College of Acupuncture and Orthopedics, Hubei University of Chinese Medicine, Wuhan, China
| | - Jiaxuan Tian
- College of Acupuncture and Orthopedics, Hubei University of Chinese Medicine, Wuhan, China
| | - Xuanai Zhu
- College of Acupuncture and Orthopedics, Hubei University of Chinese Medicine, Wuhan, China
| | - Aijia Zou
- College of Acupuncture and Orthopedics, Hubei University of Chinese Medicine, Wuhan, China
| | - Wei Lu
- Clinical College of Chinese Medicine, Hubei University of Chinese Medicine, Wuhan, China.
| | - Bijiang Wan
- College of Acupuncture and Orthopedics, Hubei University of Chinese Medicine, Wuhan, China; Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences-Wuhan National Laboratory for Optoelectronics, Wuhan, China.
| |
Collapse
|
6
|
Sun Z, Zhang L, Yin K, Zang G, Qian Y, Mao X, Li L, Jing Q, Wang Z. SIRT3-and FAK-mediated acetylation-phosphorylation crosstalk of NFATc1 regulates N ε-carboxymethyl-lysine-induced vascular calcification in diabetes mellitus. Atherosclerosis 2023; 377:43-59. [PMID: 37392543 DOI: 10.1016/j.atherosclerosis.2023.06.969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 06/21/2023] [Accepted: 06/23/2023] [Indexed: 07/03/2023]
Abstract
BACKGROUND AND AIMS Arterial calcification is the predictor of cardiovascular risk in diabetic patients. Nε-carboxymethyl-lysine (CML), a toxic metabolite, is associated with accelerated vascular calcification in diabetes mellitus (DM). However, the mechanism remains elusive. This study aims to explore the key regulators involved in CML-induced vascular calcification in DM. METHODS We used Western blot and immuno-staining to test the expression and localization of nuclear factor of activated T cells, cytoplasmic 1 (NFATc1) in human samples, a diabetic apolipoprotein E-deficient (ApoE-/-) mouse model, and a vascular smooth muscle cells (VSMC) model. Further, we confirmed the regulator of NFATc1 phosphorylation and acetylation induced by CML. The role of NFATc1 in VSMCs calcification and osteogenic differentiation was explored in vivo and in vitro. RESULTS In diabetic patients, CML and NFATc1 levels increased in the severe calcified anterior tibial arteries. CML significantly promoted NFATc1 expression and nuclear translocation in VSMCs and mouse aorta. Knockdown of NFATc1 significantly inhibited CML-induced calcification. CML promoted NFATc1 acetylation at K549 by downregulating sirtuin 3 (SIRT3), which antagonized the focal adhesion kinase (FAK) induced NFATc1 phosphorylation at the Y270 site. FAK and SIRT3 affected the nuclear translocation of NFATc1 by regulating the acetylation-phosphorylation crosstalk. NFATc1 dephosphorylation mutant Y270F and deacetylation mutant K549R had opposite effects on VSMC calcification. SIRT3 overexpression and FAK inhibitor could reverse CML-promoted VSMC calcification. CONCLUSIONS CML enhances vascular calcification in DM through NFATc1. In this process, CML increases NFATc1 acetylation by downregulating SIRT3 to antagonize FAK-induced NFATc1 phosphorylation.
Collapse
Affiliation(s)
- Zhen Sun
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Lili Zhang
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Kai Yin
- Department of General Practice, The Fifth Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China
| | - Guangyao Zang
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Yongjiang Qian
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Xiang Mao
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Lihua Li
- Department of Pathology, Affiliated Hospital of Jiangsu University, Zhenjiang, China.
| | - Qing Jing
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Innovation Center for Intervention of Chronic Disease and Promotion of Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, China.
| | - Zhongqun Wang
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, China.
| |
Collapse
|
7
|
Tawulie D, Jin L, Shang X, Li Y, Sun L, Xie H, Zhao J, Liao J, Zhu Z, Cui H, Wen W. Jiang-Tang-San-Huang pill alleviates type 2 diabetes mellitus through modulating the gut microbiota and bile acids metabolism. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 113:154733. [PMID: 36870307 DOI: 10.1016/j.phymed.2023.154733] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 02/18/2023] [Accepted: 02/22/2023] [Indexed: 06/18/2023]
Abstract
BACKGROUND Jiang-Tang-San-Huang (JTSH) pill, a traditional Chinese medicine (TCM) prescription, has long been applied to clinically treat type 2 diabetes mellitus (T2DM), while the underlying antidiabetic mechanism remains unclarified. Currently, it is believed that the interaction between intestinal microbiota and bile acids (BAs) metabolism mediates host metabolism and promotes T2DM. PURPOSE To elucidate the underlying mechanisms of JTSH for treating T2DM with animal models. METHODS In this study, male SD rats received high-fat diet (HFD) and streptozotocin (STZ) injection to induce T2DM and were treated with different dosages (0.27, 0.54 and 1.08 g/kg) of JTSH pill for 4 weeks; metformin was given as a positive control. Alterations of gut microbiota and BA profiles in the distal ileum were assessed by 16S ribosomal RNA gene sequencing and ultra-high performance liquid chromatography-tandem mass spectrometry (UPLC-MS/MS), respectively. Additionally, we conducted quantitative Real Time-PCR and western blotting to determine the mRNA and protein expression levels of intestinal farnesoid X receptor (FXR), fibroblast growth factor 15 (FGF15), Takeda G-protein-coupled receptor 5 (TGR5) and glucagon-like peptide 1 (GLP-1) as well as hepatic cytochrome P450, family 7, subfamily a, poly-peptide 1 (CYP7A1) and cytochrome P450, family 8, subfamily b, poly-peptide 1 (CYP8B1), which are involved in BAs metabolism and enterohepatic circulation. RESULTS Here, the results revealed that JTSH treatment significantly ameliorated hyperglycaemia, insulin resistance (IR), hyperlipidaemia, and pathological changes in the pancreas, liver, kidney and intestine and reduced the serum levels of pro-inflammatory cytokines in T2DM model rats. 16S rRNA sequencing and UPLC-MS/MS showed that JTSH treatment could modulate gut microbiota dysbiosis by preferentially increasing bacteria (e.g., Bacteroides, Lactobacillus, Bifidobacterium) with bile-salt hydrolase (BSH) activity, which might in turn lead to the accumulation of ileal unconjugated BAs (e.g., CDCA, DCA) and further upregulate the intestinal FXR/FGF15 and TGR5/GLP-1 signaling pathways. CONCLUSION The study demonstrated that JTSH treatment could alleviate T2DM by modulating the interaction between gut microbiota and BAs metabolism. These findings suggest that JTSH pill may serve as a promising oral therapeutic agent for T2DM.
Collapse
Affiliation(s)
- Dina Tawulie
- The First School of Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Lulu Jin
- Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Xin Shang
- The First School of Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Yimei Li
- The First School of Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Le Sun
- The First School of Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Haixue Xie
- Yunnan Provincial Hospital of Chinese Medicine, Kunming 650021, China
| | - Jie Zhao
- Yunnan Provincial Hospital of Chinese Medicine, Kunming 650021, China
| | - Jiabao Liao
- Department of Emergency, Jiaxing Hospital of Traditional Chinese Medicine, Hangzhou 310003, China; Jiaxing Key Laboratory of Diabetic Angiopathy Research, Jiaxing Hospital of Traditional Chinese Medicine, Jiaxing 314033, China
| | - Zhangzhi Zhu
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510405, China.
| | - Huantian Cui
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology, School of Life Sciences, Shandong University, Qingdao 266237, China.
| | - Weibo Wen
- Yunnan Provincial Hospital of Chinese Medicine, Kunming 650021, China.
| |
Collapse
|
8
|
Chen J, Liu Y, Zhou K, Zhang W, Wen B, Xu K, Liu Y, Chen L, Huang Y, He B, Hang W, Chen J. DISC1 inhibits GSK3β activity to prevent tau hyperphosphorylation under diabetic encephalopathy. Biofactors 2023; 49:173-184. [PMID: 36070513 DOI: 10.1002/biof.1884] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Accepted: 07/01/2022] [Indexed: 11/10/2022]
Abstract
Diabetic encephalopathy (DE) is a common complication of type 2 diabetes (T2D), especially in those patients with long T2D history. Persistent high glucose (HG) stimulation leads to neuron damage and manifests like Alzheimer's disease's pathological features such as neurofilament tangle. However, the precise mechanism of high-glucose-induced tau hyperphosphorylation is not fully revealed. We here gave evidence that Disrupted in schizophrenia 1 protein (DISC1) could interact with glycogen synthase kinase 3β (GSK3β) and inhibit its activity to prevent tau hyperphosphorylation. By using DB/DB mice as animal model and HG-treated N2a cell as cell model, we found that DISC1 was downregulated both in vivo and in vitro, complicated with Tau hyperphosphorylation and GSK3β activation. Further, we identified DISC1 interacted with GSK3β by its 198th-237th amino acid residues. Overexpression of full length DISC1 but not mutated DISC1 lacking this domain could prevent HG induced tau hyperphosphorylation. Taken together, our work revealed DISC1 could be an important negative modulators of tau phosphorylation, and suggested that preservation of DISC1 could prevent HG induced neuron damage.
Collapse
Affiliation(s)
- Jiehui Chen
- Department of Biochemistry and Molecular Biology, School of Basic Medicine and the Collaborative Innovation Center for Brain Science, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yong Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medicine and the Collaborative Innovation Center for Brain Science, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Keru Zhou
- Neonatal Intensive Care Unit, Department of Pediatric, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wei Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medicine and the Collaborative Innovation Center for Brain Science, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- School of Basic Medicine, Anhui Medical University, Hefei, Anhui, China
| | - Bin Wen
- Department of Biochemistry and Molecular Biology, School of Basic Medicine and the Collaborative Innovation Center for Brain Science, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Cell Architecture Research Center, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Kai Xu
- Department of Biochemistry and Molecular Biology, School of Basic Medicine and the Collaborative Innovation Center for Brain Science, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Cell Architecture Research Center, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yazhou Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medicine and the Collaborative Innovation Center for Brain Science, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Cell Architecture Research Center, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Ling Chen
- Neonatal Intensive Care Unit, Department of Pediatric, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yue Huang
- Department of Cardiovascular Medicine, Lichuan People's Hospital, Lichuan, China
| | - Benhong He
- Department of Cardiovascular Medicine, Lichuan People's Hospital, Lichuan, China
| | - Weijian Hang
- Department of Biochemistry and Molecular Biology, School of Basic Medicine and the Collaborative Innovation Center for Brain Science, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Juan Chen
- Department of Biochemistry and Molecular Biology, School of Basic Medicine and the Collaborative Innovation Center for Brain Science, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Neonatal Intensive Care Unit, Department of Pediatric, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Cell Architecture Research Center, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
9
|
Chen B, He Q, Yang J, Pan Z, Xiao J, Chen W, Chi W, Li M, Li S, Zeng J, Chen C, Wang F, Pang X, Yi Y, Tu H, Wang H, Chen P. Metformin suppresses Oxidative Stress induced by High Glucose via Activation of the Nrf2/HO-1 Signaling Pathway in Type 2 Diabetic Osteoporosis. Life Sci 2022; 312:121092. [PMID: 36279968 DOI: 10.1016/j.lfs.2022.121092] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 10/10/2022] [Accepted: 10/13/2022] [Indexed: 11/07/2022]
Abstract
BACKGROUND Metformin (MET) is widely used as a first-line hypoglycemic agent for the treatment of type 2 diabetes mellitus (T2DM) and was also confirmed to have a therapeutic effect on type 2 diabetic osteoporosis (T2DOP). However, the potential mechanisms of MET in the treatment of T2DOP are unclear. OBJECTIVE To clarify the effect of MET in T2DOP and to explore the potential mechanism of MET in the treatment of T2DOP. METHODS In vitro, we used MC3T3-E1 cells to study the effects of MET on osteogenic differentiation and anti-oxidative stress injury in a high glucose (Glucose 25 mM) environment. In vivo, we directly used db/db mice as a T2DOP model and assessed the osteoprotective effects of MET by Micro CT and histological analysis. RESULTS In vitro, we found that MET increased ALP activity in MC3T3-E1 cells in a high-glucose environment, promoted the formation of bone mineralized nodules, and upregulated the expression of the osteogenesis-related transcription factors RUNX2, Osterix, and COL1A1-related genes. In addition, MET was able to reduce high glucose-induced reactive oxygen species (ROS) production. In studies on the underlying mechanisms, we found that MET activated the Nrf2/HO-1 signaling pathway and alleviated high-glucose-induced oxidative stress injury. In vivo results showed that MET reduced bone loss and bone microarchitecture destruction in db/db mice. CONCLUSION Our results suggest that MET can activate the Nrf2/HO-1 signaling pathway to regulate the inhibition of osteogenic differentiation induced by high glucose thereby protecting T2DOP.
Collapse
Affiliation(s)
- Bohao Chen
- 1st School of Medicine, Guangzhou University of Chinese Medicine, 12 Jichang Road, Baiyun Area, Guangzhou 510405, PR China; The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, PR China
| | - Qi He
- 1st School of Medicine, Guangzhou University of Chinese Medicine, 12 Jichang Road, Baiyun Area, Guangzhou 510405, PR China; The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, PR China
| | - Junzheng Yang
- 1st School of Medicine, Guangzhou University of Chinese Medicine, 12 Jichang Road, Baiyun Area, Guangzhou 510405, PR China; The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, PR China
| | - Zhaofeng Pan
- 1st School of Medicine, Guangzhou University of Chinese Medicine, 12 Jichang Road, Baiyun Area, Guangzhou 510405, PR China; The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, PR China
| | - Jiacong Xiao
- 1st School of Medicine, Guangzhou University of Chinese Medicine, 12 Jichang Road, Baiyun Area, Guangzhou 510405, PR China; The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, PR China
| | - Weijian Chen
- Guangdong Second Traditional Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Guangzhou 510405, PR China
| | - Weijin Chi
- 1st School of Medicine, Guangzhou University of Chinese Medicine, 12 Jichang Road, Baiyun Area, Guangzhou 510405, PR China; The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, PR China
| | - Miao Li
- 1st School of Medicine, Guangzhou University of Chinese Medicine, 12 Jichang Road, Baiyun Area, Guangzhou 510405, PR China; The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, PR China
| | - Shaocong Li
- 1st School of Medicine, Guangzhou University of Chinese Medicine, 12 Jichang Road, Baiyun Area, Guangzhou 510405, PR China; The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, PR China
| | - Jiaxu Zeng
- 1st School of Medicine, Guangzhou University of Chinese Medicine, 12 Jichang Road, Baiyun Area, Guangzhou 510405, PR China; The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, PR China
| | - Chuyi Chen
- 1st School of Medicine, Guangzhou University of Chinese Medicine, 12 Jichang Road, Baiyun Area, Guangzhou 510405, PR China; The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, PR China
| | - FanChen Wang
- 1st School of Medicine, Guangzhou University of Chinese Medicine, 12 Jichang Road, Baiyun Area, Guangzhou 510405, PR China; The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, PR China
| | - Xinyuan Pang
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Yanzi Yi
- The Third Affiliated Medical College of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510405, PR China
| | - Haitao Tu
- Department of Orthopaedics, First Affiliated Hospital, Guangzhou University of Chinese Medicine, 12 Jichang Road, Baiyun Area, Guangzhou 510405, China
| | - Haibin Wang
- Department of Orthopaedics, First Affiliated Hospital, Guangzhou University of Chinese Medicine, 12 Jichang Road, Baiyun Area, Guangzhou 510405, China.
| | - Peng Chen
- Department of Orthopaedics, First Affiliated Hospital, Guangzhou University of Chinese Medicine, 12 Jichang Road, Baiyun Area, Guangzhou 510405, China.
| |
Collapse
|
10
|
He Q, Yang J, Chen D, Li Y, Gong D, Ge H, Wang Z, Wang H, Chen P. 12-Deoxyphorbol-13-Hexadecanoate Abrogates OVX-Induced Bone Loss in Mice and Osteoclastogenesis via Inhibiting ROS Level and Regulating RANKL-Mediated NFATc1 Activation. Front Pharmacol 2022; 13:899776. [PMID: 35721216 PMCID: PMC9204068 DOI: 10.3389/fphar.2022.899776] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Accepted: 04/25/2022] [Indexed: 01/07/2023] Open
Abstract
Osteoporosis is a major health problem in the elderly. Almost every bone can fracture due to the increased bone fragility in osteoporosis, posing a major challenge to public health. 12-Deoxyphorbol-13-hexadecanoate (DHD), one of the main bioactive components of Stellera chamaejasme L. (Lang Du), is considered to have antitumor, antibacterial, and antifungal properties. However, the role of DHD in osteoporosis is still elusive. In this study, we demonstrated for the first time that DHD inhibits the receptor activator of nuclear factor-κB ligand (RANKL)-induced osteoclastogenesis and bone resorption in a dose- and time-dependent manner without exhibiting cytotoxicity in vitro. Mechanistically, we found that DHD not only represses the expression of osteoclasts marker genes by suppressing RANKL-induced mitogen-activated protein kinase (MAPK) and calcium signaling pathways but also scavenges reactive oxygen species (ROS) through enhancing cytoprotective enzymes expression. Furthermore, DHD inhibits the activation of nuclear factor of activated T cells 1 (NFATc1) during RANKL-induced osteoclasts formation. Preclinical studies revealed that DHD protects against bone loss in ovariectomy (OVX) mice. In sum, our data confirmed that DHD could potentially inhibit osteoclastogenesis by abrogating RANKL-induced MAPK, calcium, and NFATc1 signaling pathways and promoting the expression of ROS scavenging enzymes, thereby preventing OVX-induced bone loss. Thus, DHD may act as a novel therapeutic agent to manage osteoporosis.
Collapse
Affiliation(s)
- Qi He
- First School of Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
- The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Junzheng Yang
- First School of Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
- The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Delong Chen
- Department of Orthopaedic Surgery, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Yejia Li
- Department of Orthopedics, Shunde Hospital, Guangzhou University of Chinese Medicine, Foshan, China
| | - Dawei Gong
- First School of Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
- Department of Orthopedics, Wendeng Orthopaedic and Traumatologic Hospital of Shandong Province, Weihai, China
| | - Hui Ge
- Department of Orthopedics, Guangzhou Hospital of Integrated Chinese and Western Medicine, Guangzhou, China
| | - Zihao Wang
- Queen’s University Belfast, Belfast, United Kingdom
| | - Haibin Wang
- Department of Orthopaedics, The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
- *Correspondence: Haibin Wang, ; Peng Chen,
| | - Peng Chen
- Department of Orthopaedics, The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
- *Correspondence: Haibin Wang, ; Peng Chen,
| |
Collapse
|
11
|
Zhang P, Chen H, Shen G, Zhang Z, Yu X, Shang Q, Zhao W, Li D, Li P, Chen G, Liang D, Jiang X, Ren H. Network pharmacology integrated with experimental validation reveals the regulatory mechanism of plastrum testudinis in treating senile osteoporosis. JOURNAL OF ETHNOPHARMACOLOGY 2021; 276:114198. [PMID: 33984459 DOI: 10.1016/j.jep.2021.114198] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Revised: 05/05/2021] [Accepted: 05/06/2021] [Indexed: 06/12/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Plastrum testudinis (PT) has been used in traditional Chinese medicine to treat bone diseases such as senile osteoporosis (SOP) for thousands of years. However, the underlying mechanisms remain largely unknown. AIM OF THE STUDY This study aims to investigate the possible molecular mechanism of PT in the treatment of SOP using an integrated strategy of network pharmacology and experimental validation. MATERIALS AND METHODS The compounds of PT and its targets were identified through the BATMAN-TCM database. The SOP-related targets were retrieved from the GeneCards database. Protein-protein interaction information was obtained by inputting the intersection targets into the STRING database. Cytoscape software was used to construct a protein-protein interaction network and a PT-compound-target-SOP network. Using Cytoscape and R software, we conducted GO function and KEGG pathway enrichment analyses. We also conducted in vivo and in vitro experiments to verify the network pharmacology findings. RESULTS In total, 6 active compounds and 342 targets of PT were screened, of which 57 common targets were related to SOP. The GO biological process enrichment analysis identified 880 entries, mainly relating to the regulation of hormone response, the cell apoptotic process, the apoptotic signaling pathway, NF-kappaB transcription factor activity, fatty acid transportation, osteoclast differentiation, macrophage activation, and inflammatory response. The KEGG pathway enrichment analysis identified 52 entries, including 14 related signaling pathways, which mainly involved the TNF, MAPK, IL-17, AGE-RAGE, estrogen, relaxin, and other signaling pathways. Our in vivo experiments confirmed that PT alleviates SOP, while the in vitro experiments demonstrated that PT exerts a suppressive effect on osteoclast differentiation and bone resorption in a concentration-dependent manner. Furthermore, we observed that PT downregulates the expression of osteoclast-specific genes, including C-FOS, TNF, and BDNF, in the MAPK signaling pathway. CONCLUSION Through network pharmacology and experimental validation, this study is the first to report that PT downregulates the expression of osteoclast-specific genes, including C-FOS, TNF, and BDNF, in the MAPK signaling pathway, thus exerting a suppressive effect on osteoclast differentiation and bone resorption, which may be the molecular mechanism for PT treatment of SOP.
Collapse
Affiliation(s)
- Peng Zhang
- The First Clinical School, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China; Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Honglin Chen
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China; Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Gengyang Shen
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China; Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Zhida Zhang
- The First Clinical School, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China; Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Xiang Yu
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China; Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Qi Shang
- The First Clinical School, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China; Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Wenhua Zhao
- The First Clinical School, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China; Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Danyun Li
- The First Clinical School, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Peixin Li
- The Second Clinical School, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Guifeng Chen
- The First Clinical School, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China; Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - De Liang
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Xiaobing Jiang
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China; Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China.
| | - Hui Ren
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China; Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China.
| |
Collapse
|