1
|
Xu D, Gao K, Zhang W, Tao XR, Zhao MN, Mu F, Wang JW. Shenqi Funeng Xingnao prescription alleviates exercise-induced fatigue by regulating the HIF-1/FoxO signaling pathway. JOURNAL OF ETHNOPHARMACOLOGY 2025; 348:119881. [PMID: 40287116 DOI: 10.1016/j.jep.2025.119881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/16/2025] [Revised: 04/23/2025] [Accepted: 04/24/2025] [Indexed: 04/29/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Shenqi Funeng Xingnao Prescription (SQFNXNP) is a traditional Chinese medicine that has been used successfully to treat exercise-induced fatigue in clinical therapy. However, the pharmacological effects and underlying mechanisms of action of SQFNXNP on exercise-induced fatigue remain unexplored. AIM OF THE STUDY We performed a preliminary investigation of the therapeutic effects of SQFNXNP in alleviating exercise-induced fatigue. METHODS An exercise-induced fatigue model was used to evaluate the anti-fatigue effects of SQFNXNP. UHPLC-MS analysis identified the compounds with prototype structures in plasma after SQFNXNP administration. Network pharmacology predicted potential targets of SQFNXNP in exercise-induced fatigue. Genes obtained via weighted gene co-expression network analysis in transcriptomics were integrated with those identified via network pharmacology to reveal the targets and pathways of SQFNXNP activity against exercise-induced fatigue. Differential metabolites of SQFNXNP against exercise-induced fatigue were discovered via correlation analysis of genes in the Hypoxia-inducible factor 1 (HIF-1) and Forkhead box O (FoxO) signaling pathways and differential metabolites of SQFNXNP vs. Model. The CytoHubba plugin and virtual screening were used to further screen core genes and proteins. The potential targets of SQFNXNP in exercise-induced fatigue were verified using reverse transcription quantitative polymerase chain reaction (RT-qPCR) and Western blotting (WB). RESULTS SQFNXNP improved fatigue symptoms, enhanced athletic ability, delayed glycogen consumption, inhibited excessive metabolite accumulation, increased antioxidant enzyme levels, inhibited lipid peroxidation, relieved muscle fiber contraction caused by excessive fatigue or exercise, increased the number of mitochondria, and improved the pathological morphology of the gastrocnemius muscle. A total of 38 compounds with prototype structures in the plasma were identified in SQFNXNP. The potential HIF-1 and FoxO signaling pathways were identified via the integrated results of network pharmacology and transcriptomics. The top 15 hub genes in the HIF-1 and FoxO signaling pathways were screened using the CytoHubba plugin and verified by RT-qPCR. Key protein targets, including IFNG, IL-6, MAPK14, and CCND1, were obtained via virtual screening and verified by WB. CONCLUSION This study suggests that SQFNXNP, a potential anti-fatigue TCM prescription, alleviates exercise-induced fatigue, addressing the lack of anti-fatigue drugs in clinical therapy.
Collapse
Affiliation(s)
- Dong Xu
- Department of Pharmacy, Xijing Hospital, The Fourth Military Medical University, Xi'an, 710032, China
| | - Kai Gao
- Department of Pharmacy, Xijing Hospital, The Fourth Military Medical University, Xi'an, 710032, China
| | - Wei Zhang
- Department of Pharmacy, Xijing Hospital, The Fourth Military Medical University, Xi'an, 710032, China
| | - Xing-Ru Tao
- Department of Pharmacy, Xijing Hospital, The Fourth Military Medical University, Xi'an, 710032, China
| | - Mei-Na Zhao
- Department of Pharmacy, Xijing Hospital, The Fourth Military Medical University, Xi'an, 710032, China
| | - Fei Mu
- Department of Pharmacy, Xijing Hospital, The Fourth Military Medical University, Xi'an, 710032, China
| | - Jing-Wen Wang
- Department of Pharmacy, Xijing Hospital, The Fourth Military Medical University, Xi'an, 710032, China.
| |
Collapse
|
2
|
Xu S, Yu Y, Xie Q, Liu X, Zhang A, Tang H, Zhu Z, Bian X, Guo L. Revealing the molecular mechanism of Buzhong Yiqi Decoction for tendon bone healing on the basis of network pharmacology, molecular docking and experimental validation. JOURNAL OF ETHNOPHARMACOLOGY 2025; 347:119726. [PMID: 40185260 DOI: 10.1016/j.jep.2025.119726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 03/17/2025] [Accepted: 03/30/2025] [Indexed: 04/07/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Buzhong Yiqi Decoction (BD), a traditional formula in Chinese medicine, is clinically and historically recognized for its effectiveness in reducing physical fatigue and promoting strength, as well as enhancing bone remodeling. Nevertheless, its specific molecular mechanisms related to bone formation have yet to be thoroughly characterized. AIM OF THE STUDY This study aims to investigate the effects and mechanisms of BD on osteogenesis in bone marrow mesenchymal stem cells and in a model of tendon-bone junction injury in mice. MATERIALS AND METHODS By establishing a mouse model of tendon-bone junction injury, the pathological morphology of the tendon-bone junction in mice was observed. Determining the mechanism of action of BD in regulating osteogenic differentiation through network pharmacology and molecular docking. Flow analysis and osteogenic induction assay were utilized to verify the effect of BD in promoting BMSCs osteogenic differentiation in vitro. In vivo experiments were performed to validate the impact of BD in improving healing after tendon-bone junction injury in mice by promoting osteogenic differentiation. RESULTS Bone loss at the heel bone end is an essential pathophysiologic process in the natural healing process after injury to the tendon-bone junction. Using network pharmacology and molecular docking, we identified the PI3K-Akt signaling pathway as a critical mediator of BD-induced osteogenic differentiation. In vitro experiments demonstrated that BD promoted BMSC osteogenesis, while in vivo assays confirmed the enhancement of tendon-bone healing in mice models. CONCLUSION These results suggest that BD can effectively promote tendon-bone repair, with the PI3K-Akt pathway playing a crucial role in its therapeutic effects, positioning BD as a promising candidate for improving musculoskeletal injury recovery.
Collapse
Affiliation(s)
- Shibo Xu
- State Key Laboratory of Trauma, Burn and Combined Injury, Department of Orthopedics/Sports Medicine Center, First Affiliated Hospital of Army Medical University, Chongqing, 400038, China
| | - Yihang Yu
- State Key Laboratory of Trauma, Burn and Combined Injury, Department of Orthopedics/Sports Medicine Center, First Affiliated Hospital of Army Medical University, Chongqing, 400038, China
| | - Qizhong Xie
- State Key Laboratory of Trauma, Burn and Combined Injury, Department of Orthopedics/Sports Medicine Center, First Affiliated Hospital of Army Medical University, Chongqing, 400038, China
| | - Xiao Liu
- State Key Laboratory of Trauma, Burn and Combined Injury, Department of Orthopedics/Sports Medicine Center, First Affiliated Hospital of Army Medical University, Chongqing, 400038, China
| | - Anyang Zhang
- State Key Laboratory of Trauma, Burn and Combined Injury, Department of Orthopedics/Sports Medicine Center, First Affiliated Hospital of Army Medical University, Chongqing, 400038, China
| | - Hong Tang
- State Key Laboratory of Trauma, Burn and Combined Injury, Department of Orthopedics/Sports Medicine Center, First Affiliated Hospital of Army Medical University, Chongqing, 400038, China
| | - Zhiquan Zhu
- State Key Laboratory of Trauma, Burn and Combined Injury, Department of Orthopedics/Sports Medicine Center, First Affiliated Hospital of Army Medical University, Chongqing, 400038, China
| | - Xuting Bian
- State Key Laboratory of Trauma, Burn and Combined Injury, Department of Orthopedics/Sports Medicine Center, First Affiliated Hospital of Army Medical University, Chongqing, 400038, China; Shigatse Branch, Xinqiao Hospital, Third Military Medical University, Shigatse, 857000, China.
| | - Lin Guo
- State Key Laboratory of Trauma, Burn and Combined Injury, Department of Orthopedics/Sports Medicine Center, First Affiliated Hospital of Army Medical University, Chongqing, 400038, China.
| |
Collapse
|
3
|
Cai W, Jiang L, Zhao C, Zhou X. Advances in omics technologies for traditional Chinese medicine in the prevention and treatment of metabolic bone diseases. Front Pharmacol 2025; 16:1576286. [PMID: 40290428 PMCID: PMC12021879 DOI: 10.3389/fphar.2025.1576286] [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: 02/13/2025] [Accepted: 03/25/2025] [Indexed: 04/30/2025] Open
Abstract
Metabolic bone disease (MBD), as one of the most severe metabolic disorders, remains a focal point and challenge in medical research. Numerous studies have demonstrated the efficacy of Traditional Chinese Medicine (TCM) in preventing and treating MBD. However, the inherent complexity of TCM metabolites poses significant limitations in elucidating their mechanisms of action. The advancement of omics technologies, including metabolomics, proteomics, and transcriptomics, has greatly facilitated research on MBD. These approaches enable the identification of potential biomarkers and the exploration of metabolic pathways and mechanisms underlying TCM interventions for MBD. Evidence indicates that TCM monomers, single botanical drugs, and herbal formulations are effective, safe, and well-tolerated in MBD prevention and treatment. This review summarizes recent applications and key findings of transcriptomics, proteomics, and metabolomics in studying the mechanisms of TCM interventions for MBD. It highlights the role of omics technologies in uncovering relevant metabolites and pathways under TCM treatment, providing valuable insights and clinical references for TCM-based strategies in managing MBD.
Collapse
Affiliation(s)
- Wenjun Cai
- Changchun University of Chinese Medicine, Changchun, China
- Department of Orthopedics, The Third Affiliated Clinical Hospital of Changchun University of Chinese Medicine, Changchun, China
| | - Lili Jiang
- Department of Endocrinology, Changchun Hospital of Chinese Medicine, Changchun, China
| | - Changwei Zhao
- Changchun University of Chinese Medicine, Changchun, China
- Department of Orthopedics, The Affiliated Hospital of Changchun University of Chinese Medicine. Changchun, China
| | - Xiaoling Zhou
- Changchun University of Chinese Medicine, Changchun, China
- Department of Geriatrics, The Affiliated Hospital of Changchun University of Chinese Medicine. Changchun, China
| |
Collapse
|
4
|
Wang W, Yang W, Wang F, Gao H, Liu K, Zhang J, Li Y, Zhang M, Zhou G, Hou Y, Bai G. Kunxinning granules alleviate perimenopausal syndrome by supplementing estrogen deficiency. Front Pharmacol 2025; 16:1554479. [PMID: 40206089 PMCID: PMC11979375 DOI: 10.3389/fphar.2025.1554479] [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: 01/02/2025] [Accepted: 03/10/2025] [Indexed: 04/11/2025] Open
Abstract
Introduction Ovarian function decline results in reduced estrogen levels, leading to endocrine disorders, oxidative stress damage, and excessive activation of inflammatory factors, all of which contribute to the development of premenstrual syndrome (PMS). Kunxinning Granules (KXN) has been clinically approved for PMS treatment, but its bioactive ingredients and mechanism of action remain unclear. This study aimed to investigate the active metabolites and molecular mechanism of KXN in treating PMS rats, laying a foundation for the clinical development of PMS treatment. Methods An ovariectomized (OVX) rat model was established to evaluate the efficacy of KXN in treating PMS. Molecular network (MN) analysis, combined with UPLC/Q-TOF-MS, identified prototype compounds in the samples and constructed a chemical classification map based on their structures. A network analysis and proteomics were conducted to predict potential pathways through which KXN regulates PMS. Quantitative metabolomics assays were used to confirm these potential pathways. Additionally, target prediction and binding enzyme activity detection elucidated the key active metabolites and mechanisms of action in KXN. Results KXN exhibited significant effectiveness in supplementing estrogen deficiency and uterine atrophy in the OVX model. We identified 16 absorbed metabolites as the potential pharmacological ingredients of KXN in vivo. The steroid hormone biosynthesis pathway, a crucial pathway of KXN in PMS, played a key role in KXN's effectiveness. KXN improved hormonal metabolic disorders by regulating this pathway. The main metabolites in KXN, including astragaloside IV, icariin and baohuoside I increased estradiol levels by enhancing the activity of CYP19A1, the representative enzyme in hormone biosynthesis pathway. Discussion This study shows that KXN could relieve anxiety, depression, and osteoporosis in PMS. This pharmacological effect is exerted through steroid hormone synthesis to address estrogen deficiency. The findings provide valuable insights into the underlying mechanisms and support its clinical application.
Collapse
Affiliation(s)
- Wenshuang Wang
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, China
| | - Wen Yang
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, China
| | - Fangwenting Wang
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, China
| | - He Gao
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, China
| | - Kaixin Liu
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, China
| | - Jinling Zhang
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, China
| | - Yunjuan Li
- State Key Laboratory of Chinese Medicine Modernization, Tasly Pharmaceutical Group Co., Ltd., Tianjin, China
- Tianjin Key Laboratory of Component-Based Chinese Medicine, Tianjin, China
| | - Man Zhang
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, China
| | - Guirong Zhou
- State Key Laboratory of Chinese Medicine Modernization, Tasly Pharmaceutical Group Co., Ltd., Tianjin, China
- Tianjin Key Laboratory of Component-Based Chinese Medicine, Tianjin, China
| | - Yuanyuan Hou
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, China
| | - Gang Bai
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, China
| |
Collapse
|
5
|
Zhang C, Fan Y, Qin Z, Su M, Yao F. Network pharmacology and experimental validation reveal dexmedetomidine's protective mechanisms against acute liver injury in mice. Sci Rep 2025; 15:9044. [PMID: 40090997 PMCID: PMC11911443 DOI: 10.1038/s41598-025-93998-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Accepted: 03/11/2025] [Indexed: 03/19/2025] Open
Abstract
This study explored the role and molecular mechanisms of dexmedetomidine (DEX), an α2-adrenergic receptor agonist, in the treatment of a mouse model of acute liver injury (ALI). DEX significantly mitigated hepatic tissue damage and reduced serum levels of liver function biomarkers and proinflammatory cytokines. Network pharmacology analysis revealed 81 common targets between DEX and ALI, identifying 10 crucial hub genes. Kyoto Encyclopedia of Genes and Genomes pathway analysis indicated that DEX's therapeutic effect on ALI is likely linked to the activation of the PI3K/AKT pathway. Immunohistochemical experiments verified DEX's activation of the PI3K/AKT pathway. Molecular docking and dynamic simulations confirmed the stable interaction between DEX and the epidermal growth factor receptor (EGFR). Immunohistochemistry and western blotting further validated that DEX pretreatment upregulated EGFR expression. Our findings indicate that DEX may mitigate ALI by interacting with EGFR and triggering the PI3K/AKT pathway. These findings provide a solid theoretical and experimental basis for using DEX as a potential therapeutic regimen for treating inflammatory liver diseases.
Collapse
Affiliation(s)
- Chong Zhang
- Department of Anesthesia, Sichuan Provincial Orthopedic Hospital (Chengdu Sports Hospital and Chengdu Research Institute for Sports Injury), Chengdu, China
| | - Yixin Fan
- Oncology Department, Chengdu BOE Hospital, Chengdu, China
| | - Zhijun Qin
- Department of Anesthesia, Sichuan Provincial Orthopedic Hospital (Chengdu Sports Hospital and Chengdu Research Institute for Sports Injury), Chengdu, China
| | - Mi Su
- Department of Anesthesia, Sichuan Provincial Orthopedic Hospital (Chengdu Sports Hospital and Chengdu Research Institute for Sports Injury), Chengdu, China
| | - Fu Yao
- Department of Anesthesia, Sichuan Provincial Orthopedic Hospital (Chengdu Sports Hospital and Chengdu Research Institute for Sports Injury), Chengdu, China.
| |
Collapse
|
6
|
Fan K, Hua X, Wang S, Efferth T, Tan S, Wang Z. A promising fusion: Traditional Chinese medicine and probiotics in the quest to overcome osteoporosis. FASEB J 2025; 39:e70428. [PMID: 40047492 DOI: 10.1096/fj.202403209r] [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: 12/12/2024] [Revised: 01/23/2025] [Accepted: 02/19/2025] [Indexed: 05/13/2025]
Abstract
Botanical drugs and probiotic supplements present safer alternative options for the prevention and treatment of osteoporosis (OP). However, pathological disorders of the gut microbiota and the specific properties of probiotics and traditional Chinese medicine (TCM) significantly limit their therapeutic efficacy. Given the favorable synergistic and complementary effects between probiotics and herbal medicines, a creative combination of these approaches may address the issue of their current limited efficacy. A comprehensive analysis is necessary to provide a detailed review of their potential for combination, the mechanisms behind their synergy, scientific applications, and future developments. There exists a complex relationship between gut microbiota and OP, and the underlying regulatory mechanisms are multidimensional, involving the production of pro-inflammatory metabolites, immune system disruption, and the impairment of the intestinal mucosal barrier. Furthermore, we analyzed the complex mechanisms and potential connections between probiotics, TCM, and their combined applications. We highlighted the principle of complementary gain and the substantial therapeutic potential of their organic combination, which facilitates the release of active substances in TCM, increases the bioavailability of TCM, enhances probiotic delivery efficiency, and exerts synergistic effects. The combined use of probiotics and TCM offers a safe and effective strategy for managing OP and presents an innovative and promising direction for the future development of modern phytomedicine.
Collapse
Affiliation(s)
- Kangcheng Fan
- College of Exercise and Health, Shenyang Sport University, Shenyang, China
| | - Xin Hua
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Hainan University, Haikou, China
| | - Shuwan Wang
- College of Exercise and Health, Shenyang Sport University, Shenyang, China
| | - Thomas Efferth
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Mainz, Germany
| | - Shengnan Tan
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Hainan University, Haikou, China
| | - Zhuo Wang
- College of Exercise and Health, Shenyang Sport University, Shenyang, China
| |
Collapse
|
7
|
Li CL, Zhang YX, Zheng XJ, Li S, Feng J. The traditional Chinese medicine formula Zhihan Anshen Tang (ZHAST) against obstructive sleep apnea hypopnea syndrome: network pharmacology and molecular docking approach. Front Chem 2025; 13:1524087. [PMID: 40129771 PMCID: PMC11931058 DOI: 10.3389/fchem.2025.1524087] [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: 11/07/2024] [Accepted: 01/28/2025] [Indexed: 03/26/2025] Open
Abstract
Introduction The current treaments for Obstructive Sleep Apnea Hypopnea (OSAHS) are Continuous Positive Airway Pressure (CPAP) and lifestyle modifications, which is not suitable for all patients. Traditional Chinese medicine (TCM) has increasingly demonstrated its efficacy and benefits in treating OSAHS. Zhihan Anshen Tang (ZHAST), has been demonstrated its efficacy and clinical metrics for treating OSAHS patients. However, its key ingredients and mechanisms of action are still unknown. Methods Using network pharmacology, we investigated the potential mechanisms of ZHAST through which OSAHS. Results In addition, the key targets, including TNF, IL6, GAPDH, STAT3, HIF1A, and JUN, are revealed by the topological analysis. According to the findings of the GO enrichment analysis, genes were enriched in inflammatory responses, hypoxia responses, positive regulation of angiogenesis, protein phosphorylation, and regulation of cell proliferation. KEGG pathway enrichment analysis suggests that the signaling pathway of ZHAST in OSAHS are MAPK and AGE-RAGE signaling pathway, especially in diabetic complications. In addition, it is demonstrated that the enoxolone in ZHASTs have high affinity with the relevant targets by molecular docking and molecular dynamics simulations. Disscussion To my knowledge, this is the first network pharmacological molecular docking study about a Chinese medicine effective against OSA. This investigation integrates molecular docking and network pharmacology to identify the effective compounds, related targets, and potential mechanism of ZHASTs in the treatment of OSAHS, providing the prospect of traditional Chinese medicines with modern medical research.
Collapse
Affiliation(s)
- Cai-Li Li
- Respiratory Department, Tianjin Medical University General Hospital, Tianjin, China
| | - Yu-Xiang Zhang
- Respiratory Department, Tianjin Medical University General Hospital, Tianjin, China
| | - Xing-Jie Zheng
- Infectious Disease Department, Tianjin Haihe Hospital, Tianjin, China
| | - Shuo Li
- Respiratory Department, Tianjin Medical University General Hospital, Tianjin, China
| | - Jing Feng
- Respiratory Department, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
8
|
Xie Y, Fan Y, Liu X, Li Z, Liu S. 4D-DIA-based proteomics analysis reveals the protective effects of Pidanjiangtang granules in IGT rat model. JOURNAL OF ETHNOPHARMACOLOGY 2025; 338:119012. [PMID: 39481621 DOI: 10.1016/j.jep.2024.119012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 10/25/2024] [Accepted: 10/28/2024] [Indexed: 11/02/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The Pidanjiangtang (PDJT) formula was founded on the "Pidan" theory from the "Nei Jing." PDJT is considered to eliminate the accumulation of pathological products, remove heat sources, and prevent damage to organs such as the liver and islets. It is widely used in clinical practice to treat impaired glucose tolerance (IGT). However, the bioactive ingredients and underlying mechanisms are still unclear and need further investigation. OBJECTIVE This study aimed to determine the therapeutic effect of PDJT on IGT rats and explore the mechanism of PDJT intervention on IGT by four-dimensional independent data acquisition (4D-DIA) proteomics analysis. MATERIALS AND METHODS The IGT model was established by a high-fat diet combined with Streptozotocin (STZ) injection. The IGT rats were treated with low, medium, and high doses of PDJT orally for 42 days and compared with the Metformin positive control group. The therapeutic effects of PDJT on IGT rats were evaluated using the oral glucose tolerance test (OGTT), serum lipoprotein detection, insulin detection, liver histopathology, and hepatic steatosis assessment. 4D-DIA proteomics analysis was used to explore the differential proteins (DEPs) and potential pathways of PDJT. Finally, Western blotting and ELISA techniques were used to verify DEPs and major targets. RESULTS PDJT can enhance glucose metabolism, restore islet β cell function, regulate lipoprotein metabolism, reduce hepatic steatosis, and consequently slow down the progression of IGT. In the proteomic analysis, a total of 355 DEPs were identified, and critical proteins were validated. The results indicated that the JAK2/STAT1 signaling pathway plays a pivotal role in the effects of PDJT. IκB-ζ may be a potential target for PDJT in regulating the inflammatory response of IGT. CONCLUSION PDJT is an effective formula for improving IGT, with its potential mechanism linked to the JAK2/STAT1/IκB-ζ signaling pathway. This study offers a novel approach to investigating the mechanisms of TCM formula through proteomics and offers new insight into exploring TCM treatment for IGT.
Collapse
Affiliation(s)
- Yu Xie
- Beijing University of Chinese Medicine, Beijing, China; Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Yue Fan
- Beijing University of Chinese Medicine, Beijing, China; Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Xinyi Liu
- Beijing University of Chinese Medicine, Beijing, China; Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Zirong Li
- Beijing University of Chinese Medicine, Beijing, China
| | - Shangjian Liu
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China.
| |
Collapse
|
9
|
Lin YK, Hong YL, Liu CY, Lin WQ, Liang K, Deng SQ, Zhang XJ, Zeng JX, Wang S. Jiawei Bai-Hu-decoction ameliorated heat stroke-induced brain injury by inhibiting TLR4/NF-κB signal and mitophagy of glial cell. JOURNAL OF ETHNOPHARMACOLOGY 2024; 334:118571. [PMID: 38996953 DOI: 10.1016/j.jep.2024.118571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 06/17/2024] [Accepted: 07/09/2024] [Indexed: 07/14/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Jiawei Bai-Hu-Decoction (JWBHD), a prescription formulated with seven traditional Chinese medicinal material has demonstrated clinical efficacy in mitigating brain injury among heat stroke (HS) patients. AIM OF THE STUDY This study aimed to evaluate the therapeutic efficacy of JWBHD on rat model of HS and to explore its therapeutic mechanisms by integrating network pharmacology and pharmacodynamic methodologies, which major components were analyzed by using UPLC-MS/MS. MATERIALS AND METHODS The network pharmacology analysis was firstly conducted to predict the potential active ingredients and therapeutic targets of JWBHD. The anti-HS effectiveness of JWBHD was then evaluated on rats experienced HS. Rat brain tissues were harvested for a comprehensive array of experiments, including Western blot, PCR, H&E staining, Nissl staining, ELISA, transmission electron microscope, flow cytometry and immunofluorescence to validate the protective effects of JWBHD against HS-induced brain damage. Furthermore, the inhibitory effects of JWBHD on TLR4/NF-κB signal and mitophagy of glial were further verified on HS-challenged F98 cell line. Finally, the chemical compositions of the water extract of JWBHD were analyzed by using UPLC-MS/MS. RESULTS Network pharmacology has identified fifty core targets and numerous HS-related signaling pathways as potential therapeutic targets of JWBHD. Analysis of protein-protein interaction (PPI) and GO suggests that JWBHD may suppress HS-induced inflammatory signals. In experiments conducted on HS-rats, JWBHD significantly reduced the core temperature, restored blood pressure and alleviated neurological defect. Furthermore, JWBHD downregulated the counts of white blood cells and monocytes, decreased the levels of inflammatory cytokines such as IL-1β, IL-6 and TNF-α in peripheral blood, and suppressed the expression of TLR4 and NF-κB in the cerebral cortex of HS-rats. Besides, JWBHD inhibited the apoptosis of cortical cells and mitigated the damage to the cerebral cortex in HS group. Conversely, overactive mitophagy was observed in the cerebral cortex of HS-rats. However, JWBHD restored the mitochondrial membrane potential and downregulated expressions of mitophagic proteins including Pink1, Parkin, LC3B and Tom20. JWBHD reduced the co-localization of Pink1 and GFAP, a specific marker of astrocytes in the cerebral cortex of HS-rats. In addition, the inhibitory effect of JWBHD on TLR4/NF-κB signaling and overactive mitophagy were further confirmed in F98 cells. Finally, UPLC-MS/MS analysis showed that the main components of JWBHD include isoliquiritigenin, liquiritin, dipotassium glycyrrhizinate, ginsenoside Rb1, ginsenoside Re, etc. CONCLUSIONS: JWBHD protected rats from HS and prevented HS-induced damage in the cerebral cortex by suppressing TLR4/NF-κB signaling and mitophagy of glial.
Collapse
Affiliation(s)
- Yi-Ke Lin
- Guangzhou Hospital of Integrated Traditional and Western Medicine Affiliated to Guangzhou University of Chinese Medicine, No.87 Yingbin Avenue, Huadu District, Guangzhou, 510801, PR China; School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, No. 232, Waihuandong Road, Guangzhou, 510006, PR China
| | - Yu-Lin Hong
- School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, No. 232, Waihuandong Road, Guangzhou, 510006, PR China
| | - Chun-Yan Liu
- Guangzhou Hospital of Integrated Traditional and Western Medicine Affiliated to Guangzhou University of Chinese Medicine, No.87 Yingbin Avenue, Huadu District, Guangzhou, 510801, PR China; School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, No. 232, Waihuandong Road, Guangzhou, 510006, PR China
| | - Wan-Qiu Lin
- School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, No. 232, Waihuandong Road, Guangzhou, 510006, PR China
| | - Kang Liang
- School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, No. 232, Waihuandong Road, Guangzhou, 510006, PR China
| | - Si-Qi Deng
- School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, No. 232, Waihuandong Road, Guangzhou, 510006, PR China
| | - Xiao-Jun Zhang
- School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, No. 232, Waihuandong Road, Guangzhou, 510006, PR China.
| | - Jia-Xin Zeng
- School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, No. 232, Waihuandong Road, Guangzhou, 510006, PR China; The Sixth Clinical Medical College of Guangzhou University of Chinese Medicine, No. 6001, Beihuan Avenue, Futian District, Shenzhen, 518034, PR China.
| | - Shuai Wang
- Guangzhou Hospital of Integrated Traditional and Western Medicine Affiliated to Guangzhou University of Chinese Medicine, No.87 Yingbin Avenue, Huadu District, Guangzhou, 510801, PR China; School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, No. 232, Waihuandong Road, Guangzhou, 510006, PR China; Guangzhou Huadu District Women and Children's Health Hospital, No.51, Jianshe Road, Huadu District, Guangzhou, 510800, PR China.
| |
Collapse
|
10
|
Gong W, Zhu Y, Wang L. Qiang Jin Mixture Promotes Osteogenic Differentiation of MC3T3-E1 Cells via BMP2/Smads Pathway and its Network Pharmacology Study. Mol Biotechnol 2024:10.1007/s12033-024-01313-4. [PMID: 39527391 DOI: 10.1007/s12033-024-01313-4] [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: 07/24/2024] [Accepted: 10/18/2024] [Indexed: 11/16/2024]
Abstract
The study aimed to explore the potential of QiangJin mixture (QJM), a Chinese herbal compound prescription, in regulating MC3T3-E1 cell differentiation and to analyze the ingredients and therapeutic targets of QJM against osteoporosis based on network pharmacology. MC3T3-E1 cells were incubated with different concentrations of QJM-contained rat serum (5, 10, or 20%). After 14 days of cell culture, Alizarin Red staining was performed to assess the mineralization ability of osteoblasts. RT-qPCR was used to measure mRNA levels of osteogenesis-related genes. Western blot was conducted to measure protein levels of factors related to the BMP2/Smads pathway. Functional and pathway enrichment of overlapping targets for QJM and osteoporosis were analyzed using gene ontology and KEGG analyses. As shown by experimental results, QJM-contained serum led to calcium deposition, increased expression levels of osteogenesis-related genes, and activated BMP2/Smad/Runx2 signaling in MC3T3-E1 cells. A total of 125 active compounds and 162 disease-related targets were identified. The core targets were MAPK8, TP53, ESR1, STAT3, MAPK3, IL6, NFKB1, JUN, MAPK1 and AKT1. In conclusion, QJM promotes the osteogenic differentiation of MC3T3-E1 cells by activating the BMP2/Smads signaling. Additionally, QJM is an anti-osteoporotic mixture by regulating diverse therapeutic targets and signaling pathways.
Collapse
Affiliation(s)
- Weiyue Gong
- Department of Pharmacy, Huzhou Traditional Chinese Medicine Hospital Affiliated to Zhejiang Chinese Medical University, No. 315 South Street, Wuxing District, Huzhou, 313000, China
| | - Yao Zhu
- Department of Pharmacy, Huzhou Traditional Chinese Medicine Hospital Affiliated to Zhejiang Chinese Medical University, No. 315 South Street, Wuxing District, Huzhou, 313000, China
| | - Limin Wang
- Department of Pharmacy, Huzhou Traditional Chinese Medicine Hospital Affiliated to Zhejiang Chinese Medical University, No. 315 South Street, Wuxing District, Huzhou, 313000, China.
| |
Collapse
|
11
|
Wang H, Zhao Y, Liu H, Zhang X, Lv S, Zhou T, Cui H, Zhao J, Li X. Untargeted metabolomics revealed the mechanism of aucubin on glucocorticoid-induced osteoporosis in mice through modulating arachidonic acid metabolism. J Pharm Biomed Anal 2024; 248:116273. [PMID: 38878451 DOI: 10.1016/j.jpba.2024.116273] [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: 05/09/2024] [Revised: 05/28/2024] [Accepted: 06/02/2024] [Indexed: 07/20/2024]
Abstract
Glucocorticoid-induced osteoporosis (GIOP) represents the most prevalent form of secondary osteoporosis. Aucubin (AU), a principal active component found in traditional herbal medicines such as Eucommia ulmoides, has been demonstrated to enhance osteoblast differentiation. Nonetheless, the precise therapeutic effects of AU on GIOP and the complex underlying regulatory mechanisms warrant further investigation. We first established a GIOP model in female mice and then assessed the therapeutic effects of AU using micro-CT analysis, biomechanical testing, measurements of serum calcium (Ca) and phosphorus (P) levels, and histological analyses using Hematoxylin and Eosin (HE) and Masson staining. Subsequently, non-targeted metabolomics was employed in order to study the effects of AU on serum metabolites in GIOP mice. The levels of the factors related to these metabolites were quantified using real-time quantitative reverse transcription polymerase chain reaction (RT-qPCR) and western blot analyses. Finally, the effects of AU on osteoblastic and osteoclastic differentiation were examined. We found that AU significantly ameliorated bone microarchitecture and strength in GIOP mice. It mitigated pathological damages such as impairment of trabecular bone structure and reduction in collagen fibers, while concurrently elevating serum levels of Ca and P. Non-targeted metabolomics revealed that Arachidonic acid (AA) metabolism serves as a common pathway between the control and GIOP groups, as well as between the high-dose AU (AUH) and GIOP groups. AU notably upregulates prostaglandin-endoperoxide synthase 2 (PTGS2) and microsomal prostaglandin-E synthase 1 (PTGES) expression and downregulates prostaglandin-H2 D-isomerase (PTGDS) expression. Furthermore, AU treatment increased the expression of runt-related transcription factor 2 (Runx2) and transcription factor Sp7 (Osterix), enhanced serum alkaline phosphatase (ALP) activity, and reduced osteoclast expression. These results indicate that AU is a potential drug for treating GIOP, and its mechanism is related to regulating AA metabolism and promoting osteoblast differentiation. However, the key targets of AU in treating GIOP still need further exploration.
Collapse
Affiliation(s)
- Hengjun Wang
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang, Hebei Province 050091, China; Department of Orthopedics, Cangzhou Hospital of Integrated Traditional Chinese Medicine and Western Medicine of Hebei Province Affiliated to Hebei University of Chinese Medicine, Cangzhou, Hebei Province 061013, China
| | - Yunchao Zhao
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang, Hebei Province 050091, China; Department of Orthopedics, Cangzhou Hospital of Integrated Traditional Chinese Medicine and Western Medicine of Hebei Province Affiliated to Hebei University of Chinese Medicine, Cangzhou, Hebei Province 061013, China
| | - Huan Liu
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang, Hebei Province 050091, China; Department of Orthopedics, Cangzhou Hospital of Integrated Traditional Chinese Medicine and Western Medicine of Hebei Province Affiliated to Hebei University of Chinese Medicine, Cangzhou, Hebei Province 061013, China
| | - Xuelei Zhang
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang, Hebei Province 050091, China; Department of Orthopedics, Cangzhou Hospital of Integrated Traditional Chinese Medicine and Western Medicine of Hebei Province Affiliated to Hebei University of Chinese Medicine, Cangzhou, Hebei Province 061013, China
| | - Shuquan Lv
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang, Hebei Province 050091, China; Department of Hebei Key Laboratory of Integrated Traditional Chinese and Western Medicine in Osteoarthrosis Research, Cangzhou Hospital of Integrated Traditional Chinese Medicine and Western Medicine of Hebei Province Affiliated to Hebei University of Chinese Medicine, Cangzhou, Hebei Province 061013, China; Department of Diabetes, Cangzhou Hospital of Integrated Traditional Chinese Medicine and Western Medicine of Hebei Province Affiliated to Hebei University of Chinese Medicine, Cangzhou, Hebei Province 061013, China
| | - Tingting Zhou
- Department of Orthopedics, Cangzhou Hospital of Integrated Traditional Chinese Medicine and Western Medicine of Hebei Province Affiliated to Hebei University of Chinese Medicine, Cangzhou, Hebei Province 061013, China; Department of Hebei Key Laboratory of Integrated Traditional Chinese and Western Medicine in Osteoarthrosis Research, Cangzhou Hospital of Integrated Traditional Chinese Medicine and Western Medicine of Hebei Province Affiliated to Hebei University of Chinese Medicine, Cangzhou, Hebei Province 061013, China; Department of Hebei Province Integrated Traditional Chinese and Western Medicine 3D Printing Technology Innovation Center, Cangzhou Hospital of Integrated Traditional Chinese Medicine and Western Medicine of Hebei Province Affiliated to Hebei University of Chinese Medicine, Cangzhou, Hebei Province 061013, China
| | - Huantian Cui
- The First School of Clinical Medicine, Yunnan University of Chinese Medicine, Kunming, Yunnan Province 650500, China
| | - Jianyong Zhao
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang, Hebei Province 050091, China; Department of Orthopedics, Cangzhou Hospital of Integrated Traditional Chinese Medicine and Western Medicine of Hebei Province Affiliated to Hebei University of Chinese Medicine, Cangzhou, Hebei Province 061013, China.
| | - Xiaoming Li
- Department of Orthopedics, Cangzhou Hospital of Integrated Traditional Chinese Medicine and Western Medicine of Hebei Province Affiliated to Hebei University of Chinese Medicine, Cangzhou, Hebei Province 061013, China; Department of Hebei Key Laboratory of Integrated Traditional Chinese and Western Medicine in Osteoarthrosis Research, Cangzhou Hospital of Integrated Traditional Chinese Medicine and Western Medicine of Hebei Province Affiliated to Hebei University of Chinese Medicine, Cangzhou, Hebei Province 061013, China; Department of Hebei Province Integrated Traditional Chinese and Western Medicine 3D Printing Technology Innovation Center, Cangzhou Hospital of Integrated Traditional Chinese Medicine and Western Medicine of Hebei Province Affiliated to Hebei University of Chinese Medicine, Cangzhou, Hebei Province 061013, China.
| |
Collapse
|
12
|
Li G, Stampas A, Komatsu Y, Gao X, Huard J, Pan S. Proteomics in orthopedic research: Recent studies and their translational implications. J Orthop Res 2024; 42:1631-1640. [PMID: 38897819 DOI: 10.1002/jor.25917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 05/10/2024] [Accepted: 06/01/2024] [Indexed: 06/21/2024]
Abstract
Proteomics is a growing field that offers insights into various aspects of disease processes and therapy responses. Within the field of orthopedics, there are a variety of diseases that have a poor prognosis due to a lack of targeted curative therapy or disease modifying therapy. Other diseases have been difficult to manage in part due to lack of clinical biomarkers that offer meaningful insight into disease progression or severity. As an emerging technology, proteomics has been increasingly applied in studying bone biology and an assortment of orthopedics related diseases, such as osteoarthritis, osteosarcoma and bone tumors, osteoporosis, traumatic bone injury, spinal cord injury, hip and knee arthroplasty, and fragile healing. These efforts range from mechanistic studies for elucidating novel insights in tissue activity and metabolism to identification of candidate biomarkers for diagnosis, prognosis, and targeted treatment. The knowledge gained from these proteomic and functional studies has provided unique perspectives in studying orthopedic diseases. In this review, we seek to report on the current state of the proteomic study in the field of orthopedics, overview the advances in clinically applicable discoveries, and discuss the opportunities that may guide us for future research.
Collapse
Affiliation(s)
- George Li
- School of Medicine, Texas A&M University, Bryan, Texas, USA
| | - Argyrios Stampas
- Department of Physical Medicine and Rehabilitation, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, USA
- Department of Physical Medicine and Rehabilitation, TIRR Memorial Hermann Hospital, Houston, Texas, USA
| | - Yoshihiro Komatsu
- Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, USA
- Graduate Program in Genetics & Epigenetics, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, Texas, USA
| | - Xueqin Gao
- Linda and Mitch Hart Center for Regenerative and Personalized Medicine, Steadman Philippon Research Institute, Vail, Colorado, USA
| | - Johnny Huard
- Linda and Mitch Hart Center for Regenerative and Personalized Medicine, Steadman Philippon Research Institute, Vail, Colorado, USA
| | - Sheng Pan
- Graduate Program in Genetics & Epigenetics, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, Texas, USA
- The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, Texas, USA
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center at Houston, Houston, Texas, USA
| |
Collapse
|
13
|
Wang J, Xue M, Hu Y, Li J, Li Z, Wang Y. Proteomic Insights into Osteoporosis: Unraveling Diagnostic Markers of and Therapeutic Targets for the Metabolic Bone Disease. Biomolecules 2024; 14:554. [PMID: 38785961 PMCID: PMC11118602 DOI: 10.3390/biom14050554] [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: 03/29/2024] [Revised: 04/25/2024] [Accepted: 05/01/2024] [Indexed: 05/25/2024] Open
Abstract
Osteoporosis (OP), a prevalent skeletal disorder characterized by compromised bone strength and increased susceptibility to fractures, poses a significant public health concern. This review aims to provide a comprehensive analysis of the current state of research in the field, focusing on the application of proteomic techniques to elucidate diagnostic markers and therapeutic targets for OP. The integration of cutting-edge proteomic technologies has enabled the identification and quantification of proteins associated with bone metabolism, leading to a deeper understanding of the molecular mechanisms underlying OP. In this review, we systematically examine recent advancements in proteomic studies related to OP, emphasizing the identification of potential biomarkers for OP diagnosis and the discovery of novel therapeutic targets. Additionally, we discuss the challenges and future directions in the field, highlighting the potential impact of proteomic research in transforming the landscape of OP diagnosis and treatment.
Collapse
Affiliation(s)
- Jihan Wang
- Xi’an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, Xi’an 710072, China; (J.W.)
| | - Mengju Xue
- School of Medicine, Xi’an International University, Xi’an 710077, China
| | - Ya Hu
- Department of Medical College, Hunan Polytechnic of Environment and Biology, Hengyang 421000, China
| | - Jingwen Li
- Xi’an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, Xi’an 710072, China; (J.W.)
- Research and Development Institute of Northwestern Polytechnical University in Shenzhen, Shenzhen 518057, China
| | - Zhenzhen Li
- Xi’an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, Xi’an 710072, China; (J.W.)
- Research and Development Institute of Northwestern Polytechnical University in Shenzhen, Shenzhen 518057, China
| | - Yangyang Wang
- School of Electronics and Information, Northwestern Polytechnical University, Xi’an 710129, China
| |
Collapse
|