1
|
Ye H, Wang Y, Zhang X, Yang L, Cai B, Zhang D, Peng B. Characterization of global research trends and prospects on celastrol, a principal bioactive ingredient of Tripterygium wilfordii Hook F: bibliometric analysis. PHARMACEUTICAL BIOLOGY 2025; 63:15-26. [PMID: 39745069 DOI: 10.1080/13880209.2024.2443424] [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: 04/12/2024] [Revised: 11/25/2024] [Accepted: 12/12/2024] [Indexed: 01/04/2025]
Abstract
CONTEXT Celastrol, acknowledged as a prominent exemplar of the potential for transforming traditional medicinal compounds into contemporary pharmaceuticals, has garnered considerable attention owing to its extensive pharmacological activities. The increasing volume of publications concerning celastrol highlights its importance in current scientific inquiry. Despite the growing interest in this compound, a bibliometric analysis focused on this subject remains to be undertaken. OBJECTIVE Our study explored a bibliometric approach to identify and characterize global research trends and frontiers related to celastrol, including mapping research outputs, influential contributors, and thematic areas, as well as highlighting gaps and opportunities for future investigations. MATERIALS AND METHODS In this study, we utilized the Web of Science Core Collection (WoSCC) to source and review articles related to celastrol published from 1997 to 2023. The bibliometric analysis was conducted using the R package 'Bibliometrix,' supplemented by visualization tools including CiteSpace, VOSviewer, and GraphPad Prism 10. RESULTS Celastrol related research papers have exhibited an upward trend annually and can be categorized into three distinct phases, each highlighting different areas of focus. China, the United States, and South Korea rank as the top three nations for publication volume, with varied research interests across these countries. Several prolific research teams have emerged, each with distinct areas of interest. Examining the primary research domains of celastrol (anti-inflammatory, anticancer, and toxicity) reveals a notable intersection between the first two domains. DISCUSSION AND CONCLUSIONS The scope and depth of celastrol research have been steadily expanding, with regional and team-specific variations. Key research areas include anti-inflammatory, anticancer, and toxicity studies. Future research is expected to focus on enhancing the effectiveness and reducing the toxicity of celastrol. Meanwhile, given the multi-target characteristics of celastrol's effects, integrating methods such as network biology and molecular simulation will provide a novel perspective for celastrol research.
Collapse
Affiliation(s)
- Huizi Ye
- Postgraduate training base at Shanghai Gongli Hospital, Ningxia Medical University, Shanghai, China
| | - Yufang Wang
- Postgraduate training base at Shanghai Gongli Hospital, Ningxia Medical University, Shanghai, China
| | - Xue Zhang
- Postgraduate training base at Shanghai Gongli Hospital, Ningxia Medical University, Shanghai, China
- Shanghai Health Commission Key Lab of Artificial Intelligence (AI)-Based Management of Inflammation and Chronic Diseases, Department of Central Laboratory, Gongli Hospital of Shanghai Pudong New Area, Shanghai, China
- School of Basic Medicine, Ningxia Medical University, Yinchuan, Ningxia, China
| | - Lin Yang
- Postgraduate training base at Shanghai Gongli Hospital, Ningxia Medical University, Shanghai, China
| | - Banglan Cai
- Postgraduate training base at Shanghai Gongli Hospital, Ningxia Medical University, Shanghai, China
- School of Basic Medicine, Ningxia Medical University, Yinchuan, Ningxia, China
| | - Denghai Zhang
- Shanghai Health Commission Key Lab of Artificial Intelligence (AI)-Based Management of Inflammation and Chronic Diseases, Department of Central Laboratory, Gongli Hospital of Shanghai Pudong New Area, Shanghai, China
| | - Bin Peng
- Shanghai Health Commission Key Lab of Artificial Intelligence (AI)-Based Management of Inflammation and Chronic Diseases, Department of Central Laboratory, Gongli Hospital of Shanghai Pudong New Area, Shanghai, China
| |
Collapse
|
2
|
Fan X, Ai Y, Mu G, Wang F, Feng J, Jia J, Chen Y, Zhou J. Celastrol attenuates ferroptosis-mediated intestinal ischemia/reperfusion-induced acute lung injury via Hippo-YAP signaling. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 143:156865. [PMID: 40414049 DOI: 10.1016/j.phymed.2025.156865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 04/17/2025] [Accepted: 05/14/2025] [Indexed: 05/27/2025]
Abstract
BACKGROUND Acute lung injury commonly arises as a secondary complication following intestinal ischemia/reperfusion (II/R) injury. Celastrol (CEL), recognized for its therapeutic effects on inflammation-related conditions such as acute lung injury. Its protective efficacy against II/R-induced acute lung injury remains insufficiently investigated. The Hippo-YAP signaling pathway regulates ferroptosis and plays a pivotal role in II/R injury. PURPOSE To evaluate whether CEL can activate the Hippo-YAP signaling pathway, suppress ferroptosis, and mitigate II/R-induced acute lung injury. METHODS Firstly, an II/R model in mice was established, Immunofluorescence staining and Western blot were used to evaluate the effects of CEL on the Hippo signaling pathway and ferroptosis regulation. Network pharmacology predicted the relevance of the Hippo-YAP signaling pathway in CEL's improvement of acute lung injury. Molecular docking experiment indicated that CEL binds effectively to yes-associated protein (YAP), and overexpression of YAP significantly alleviated both lung injury and ferroptosis. Furthermore, the oxygen-glucose deprivation/recovery (OGD/R) model of MLE-12 cells was developed to further confirm CEL's inhibition of ferroptosis via the Hippo-YAP signaling pathway. RESULTS CEL ameliorated II/R-induced acute lung injury and inhibited inflammation. In vivo and in vitro studies further revealed that CEL significantly reduced ferroptosis and reactive oxygen species (ROS) accumulation in the lung epithelial cells. CONCLUSION CEL effectively mitigated ferroptosis and II/R-induced acute lung injury through elevating YAP protein level, reducing lipid peroxidation, and decreasing intracellular iron accumulation. This study highlights CEL's therapeutic potential for inhibiting ferroptosis, provides mechanistic insights to support CEL's broader therapeutic utility.
Collapse
Affiliation(s)
- Xin Fan
- Department of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, China; Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Yinxin Ai
- Department of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, China; Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Guo Mu
- Department of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, China; Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Feixiang Wang
- Department of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, China; Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Jianguo Feng
- Department of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, China; Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Jing Jia
- Department of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, China; Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Ye Chen
- Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, Southwest Medical University, Luzhou, Sichuan, 646000, China; Department of Traditional Chinese Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, China.
| | - Jun Zhou
- Department of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, China; Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, Southwest Medical University, Luzhou, Sichuan, 646000, China.
| |
Collapse
|
3
|
Li R, Feng K, Du Y, Fan X, Zhang Y, Wang C, Zhang J, Ahmad S, Amna R, Liu C, Niu R, Sun Z. Identification of potential natural compounds to relieve deoxynivalenol-induced intestinal damage based on bioinformatics and reverse network pharmacology. Food Chem Toxicol 2025; 202:115551. [PMID: 40379078 DOI: 10.1016/j.fct.2025.115551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Revised: 05/07/2025] [Accepted: 05/13/2025] [Indexed: 05/19/2025]
Abstract
Deoxynivalenol (DON) is one of the most prevalent mycotoxins globally, causing a variety of toxic effects in both humans and animals. Numerous studies have demonstrated the considerable efficacy of natural medicines in treating and preventing DON-induced damage. Therefore, it is crucial to predict and screen highly efficient natural medicines and further investigate their mechanisms. In this study, we employed bioinformatics approaches to explore DON's pathogenic mechanism and targets. Utilizing drug prediction and screening databases, we conducted reverse prediction and screening of differentially expressed genes (DEGs) and key targets to obtain optimal natural medicines, ultimately identifying quercetin as the most promising candidate. Subsequently, network pharmacology analyses revealed that quercetin alleviated DON-induced intestinal damage by modulating inflammatory targets and the TNF/NF-κB pathways. Our experiments demonstrated that quercetin treatment improved DON-induced growth inhibition and intestinal damage in mice, while successfully reversing the abnormal expression of key target genes. Furthermore, quercetin restored the intestinal microbial imbalance induced by DON. Overall, these findings suggest that quercetin is a promising natural medicine capable of alleviating DON-induced intestinal dysfunction by regulating inflammation-related factor levels and gut microbiota, thereby providing new insights for the future prevention and treatment of mycotoxins.
Collapse
Affiliation(s)
- Rui Li
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, 030801, Shanxi, China
| | - Kang Feng
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, 030801, Shanxi, China
| | - Yu Du
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, 030801, Shanxi, China
| | - Xuebin Fan
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, 030801, Shanxi, China
| | - Yaqin Zhang
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, 030801, Shanxi, China
| | - Chenli Wang
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, 030801, Shanxi, China
| | - Jingdi Zhang
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, 030801, Shanxi, China
| | - Sheraz Ahmad
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, 030801, Shanxi, China
| | - Rafique Amna
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, 030801, Shanxi, China
| | - Ci Liu
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, 030801, Shanxi, China
| | - Ruiyan Niu
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, 030801, Shanxi, China
| | - Zilong Sun
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, 030801, Shanxi, China.
| |
Collapse
|
4
|
Shi J, Cheng Y, Wang C, Liu M, Qu M, Zhou S, Chen L, Li X, Luo J, Luo Y, Luo C, An P. Effects of Celastrol-Enriched Peanuts on Metabolic Health and the Development of Atherosclerosis. Nutrients 2025; 17:1418. [PMID: 40362727 PMCID: PMC12073367 DOI: 10.3390/nu17091418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2025] [Revised: 04/16/2025] [Accepted: 04/17/2025] [Indexed: 05/15/2025] Open
Abstract
BACKGROUND Celastrol, a pentacyclic triterpenoid active component isolated from the root bark of the traditional medicinal plant Tripterygium wilfordii, displays significant anti-inflammatory, antioxidant, and immunomodulatory properties. However, its clinical application remains limited due to inadequate bioavailability. METHODS Regarding these issues, we innovatively developed a novel peanut cultivar (cel-peanut) enriched with celastrol through distant hybridization combined with metabolomics screening. Guided by the research concept of "natural anti-inflammatory diets for metabolic disease management", we established a high-fat diet-induced ApoE-/- atherosclerotic mouse model to systematically evaluate the anti-atherosclerosis effects and mechanisms of cel-peanut. RESULTS Our results revealed that cel-peanut significantly reduced serum levels of triglycerides (TGs) and increased high-density lipoprotein cholesterol (HDL-C). Concurrently, cel-peanut markedly decreased the atherosclerotic lesion area and enhanced collagen content within plaques. Mechanistic investigations demonstrated that cel-peanut reduced serum malondialdehyde (MDA) levels and suppressed the concentration of pro-inflammatory cytokine IL-6 in atherosclerotic lesions. Furthermore, cel-peanut promoted intestinal health by modulating the composition and functionality of gut microbiota, thereby attenuating atherosclerosis progression. CONCLUSIONS Overall, these findings indicate that cel-peanut exerts therapeutic effects against atherosclerosis through its anti-inflammatory, antioxidant, and gut microbiota-modulating properties. This study proposes a novel nutritional intervention strategy for atherosclerosis and provides a promising adjuvant strategy for clinical atherosclerosis treatment.
Collapse
Affiliation(s)
- Jiaxin Shi
- Department of Nutrition and Health, Key Laboratory of Precision Nutrition and Food Quality, China Agricultural University, Beijing 100193, China; (J.S.); (Y.C.); (C.W.); (J.L.)
| | - Yitong Cheng
- Department of Nutrition and Health, Key Laboratory of Precision Nutrition and Food Quality, China Agricultural University, Beijing 100193, China; (J.S.); (Y.C.); (C.W.); (J.L.)
| | - Chenxuan Wang
- Department of Nutrition and Health, Key Laboratory of Precision Nutrition and Food Quality, China Agricultural University, Beijing 100193, China; (J.S.); (Y.C.); (C.W.); (J.L.)
| | - Min Liu
- Guangdong Agricultural Antibiotic Reduction and Replacement Technology Promotion Association, Shenzhen 518114, China
| | - Mingxuan Qu
- Guangdong Agricultural Antibiotic Reduction and Replacement Technology Promotion Association, Shenzhen 518114, China
| | - Shuaishuai Zhou
- Department of Nutrition and Health, Key Laboratory of Precision Nutrition and Food Quality, China Agricultural University, Beijing 100193, China; (J.S.); (Y.C.); (C.W.); (J.L.)
| | - Leon Chen
- Guangdong Agricultural Antibiotic Reduction and Replacement Technology Promotion Association, Shenzhen 518114, China
| | - Xiaohao Li
- Guangdong Agricultural Antibiotic Reduction and Replacement Technology Promotion Association, Shenzhen 518114, China
| | - Junjie Luo
- Department of Nutrition and Health, Key Laboratory of Precision Nutrition and Food Quality, China Agricultural University, Beijing 100193, China; (J.S.); (Y.C.); (C.W.); (J.L.)
| | - Yongting Luo
- Department of Nutrition and Health, Key Laboratory of Precision Nutrition and Food Quality, China Agricultural University, Beijing 100193, China; (J.S.); (Y.C.); (C.W.); (J.L.)
| | - Chao Luo
- Guangdong Agricultural Antibiotic Reduction and Replacement Technology Promotion Association, Shenzhen 518114, China
| | - Peng An
- Department of Nutrition and Health, Key Laboratory of Precision Nutrition and Food Quality, China Agricultural University, Beijing 100193, China; (J.S.); (Y.C.); (C.W.); (J.L.)
| |
Collapse
|
5
|
Zhao H, Tao H, Gao J, Wang J, Hui G, Zhu Y, Wang J, Ding X, Dai Y. IL-6 Affects Liver Metabolic Abnormalities Caused by Silicon Exposure by Regulating the PKC/YY1 Signaling Pathway. Genes (Basel) 2025; 16:456. [PMID: 40282416 PMCID: PMC12026785 DOI: 10.3390/genes16040456] [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: 03/03/2025] [Revised: 03/31/2025] [Accepted: 04/06/2025] [Indexed: 04/29/2025] Open
Abstract
BACKGROUND This study aims to investigate the impact of coal dust (silicon dioxide) exposure on dyslipidemia and its underlying mechanisms, with a focus on the association between coal dust exposure and hepatic metabolic disorders. METHODS Clinical data were collected from 5433 coal mine workers to compare the incidence of dyslipidemia between the dust-exposed group and the non-exposed group. A mouse model of silicon dioxide exposure was established to observe hepatic fat accumulation and pathological changes. Liver tissue sequencing was performed to screen for key differential genes. In vitro cell experiments were utilized to identify the molecular mechanisms underlying hepatocyte metabolic abnormalities induced by silicon dioxide exposure. RESULTS Clinical data revealed that 69.2% of miners in the dust-exposed group developed dyslipidemia, which was higher than the 30.7% in the non-exposed group. Animal data showed that silicon dioxide exposure led to hepatic fat deposition and pathological damage, with the degree of injury positively correlated with exposure time. Liver sequencing identified a significant upregulation of the FMO3 (flavin monooxygenase 3) gene in mouse liver tissue following silicon dioxide exposure, accompanied by enhanced inflammatory responses. Mechanistic studies demonstrated that silicon dioxide activates Kupffer cells to secrete IL-6 (interleukin-6), which induces high expression of FMO3 in hepatocytes through the PKC/YY1 signaling pathway, thereby disrupting lipid metabolism. CONCLUSIONS Silicon dioxide exposure can promote the upregulation of FMO3 expression in hepatocytes by activating Kupffer cells to release IL-6 via the PKC/YY1 pathway, ultimately leading to lipid metabolic disorders and dyslipidemia.
Collapse
Affiliation(s)
- Hui Zhao
- Department of Pharmacology, School of Medicine, Anhui University of Science and Technology, Huainan 232001, China; (H.Z.); (H.T.); (J.G.); (J.W.); (G.H.); (Y.Z.); (J.W.)
- Key Laboratory of Industrial Dust Deep Reduction and Occupational Health and Safety of Anhui Higher Education Institutes, Huainan 232001, China
- Anhui Province Engineering Laboratory of Occupational Health and Safety, Huainan 232001, China
| | - Huihui Tao
- Department of Pharmacology, School of Medicine, Anhui University of Science and Technology, Huainan 232001, China; (H.Z.); (H.T.); (J.G.); (J.W.); (G.H.); (Y.Z.); (J.W.)
- Key Laboratory of Industrial Dust Deep Reduction and Occupational Health and Safety of Anhui Higher Education Institutes, Huainan 232001, China
| | - Jian Gao
- Department of Pharmacology, School of Medicine, Anhui University of Science and Technology, Huainan 232001, China; (H.Z.); (H.T.); (J.G.); (J.W.); (G.H.); (Y.Z.); (J.W.)
- Key Laboratory of Industrial Dust Deep Reduction and Occupational Health and Safety of Anhui Higher Education Institutes, Huainan 232001, China
- Anhui Province Engineering Laboratory of Occupational Health and Safety, Huainan 232001, China
| | - Jingjing Wang
- Department of Pharmacology, School of Medicine, Anhui University of Science and Technology, Huainan 232001, China; (H.Z.); (H.T.); (J.G.); (J.W.); (G.H.); (Y.Z.); (J.W.)
| | - Guangliang Hui
- Department of Pharmacology, School of Medicine, Anhui University of Science and Technology, Huainan 232001, China; (H.Z.); (H.T.); (J.G.); (J.W.); (G.H.); (Y.Z.); (J.W.)
| | - Ye Zhu
- Department of Pharmacology, School of Medicine, Anhui University of Science and Technology, Huainan 232001, China; (H.Z.); (H.T.); (J.G.); (J.W.); (G.H.); (Y.Z.); (J.W.)
| | - Jialin Wang
- Department of Pharmacology, School of Medicine, Anhui University of Science and Technology, Huainan 232001, China; (H.Z.); (H.T.); (J.G.); (J.W.); (G.H.); (Y.Z.); (J.W.)
| | - Xuansheng Ding
- Department of Pharmacology, China Pharmaceutical University, Nanjing 211198, China
| | - Yong Dai
- Key Laboratory of Industrial Dust Deep Reduction and Occupational Health and Safety of Anhui Higher Education Institutes, Huainan 232001, China
- Anhui Province Engineering Laboratory of Occupational Health and Safety, Huainan 232001, China
- Joint Research Center for Occupational Medicine and Health of IHM, Anhui University of Science and Technology, Huainan 232001, China
| |
Collapse
|
6
|
Vithalkar MP, Sandra KS, Bharath HB, Krishnaprasad B, Fayaz SM, Sathyanarayana B, Nayak Y. Network Pharmacology-driven therapeutic interventions for Interstitial Lung Diseases using Traditional medicines: A Narrative Review. Int Immunopharmacol 2025; 147:113979. [PMID: 39746273 DOI: 10.1016/j.intimp.2024.113979] [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: 08/01/2024] [Revised: 12/06/2024] [Accepted: 12/28/2024] [Indexed: 01/04/2025]
Abstract
This review explores the progressive domain of network pharmacology and its potential to revolutionize therapeutic approaches for Interstitial Lung Diseases (ILDs), a collective term encompassing Interstitial Pneumonia, Pneumoconiosis, Connective Tissue Disease-related ILDs, and Sarcoidosis. The exploration focuses on the profound legacy of traditional medicines, particularly Ayurveda and Traditional Chinese Medicines (TCM), and their largely unexplored capacity in ILD treatment. These ancient healing systems, characterized by their holistic methodologies and multifaceted treatment modalities, offer a promising foundation for discovering innovative therapeutic strategies. Moreover, the review underscores the amalgamation of artificial intelligence (AI) and machine learning (ML) methodologies with bioinformatics, creating a computational synergy capable of deciphering the intricate biological networks associated with ILDs. Network pharmacology has tailored the hypothesis from the conventional "one target, one drug" towards a "network target, multi-component therapeutics" approach. The fusion of traditional literature and computational technology can unveil novel drugs, targets, and pathways, augmenting effective therapies and diminishing adverse effects related to current medications. In conclusion, this review provides a comprehensive exposition of how Network Pharmacology tools can leverage the insights of Ayurveda and TCM to craft efficacious therapeutic solutions for ILDs. It sets the stage for future investigations in this captivating interdisciplinary domain, validating the use of traditional medicines worldwide.
Collapse
Affiliation(s)
- Megh Pravin Vithalkar
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
| | - K S Sandra
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
| | - H B Bharath
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
| | - B Krishnaprasad
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
| | - S M Fayaz
- Department of Biotechnology, Manipal Institute of Technology, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - B Sathyanarayana
- Muniyal Institute of Ayurveda Medical Sciences, Manipal, Karnataka 576104, India
| | - Yogendra Nayak
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India.
| |
Collapse
|
7
|
Wang S, Yang X, Liu K, Xiong D, Yalikun A, Hamiti Y, Yusufu A. Therapeutic potential of omaveloxolone in counteracting muscle atrophy post-denervation: a multi-omics approach. J Transl Med 2024; 22:991. [PMID: 39487481 PMCID: PMC11531194 DOI: 10.1186/s12967-024-05810-7] [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: 09/04/2024] [Accepted: 10/23/2024] [Indexed: 11/04/2024] Open
Abstract
BACKGROUND Muscle atrophy caused by denervation is common in neuromuscular diseases, leading to loss of muscle mass and function. However, a comprehensive understanding of the overall molecular network changes during muscle denervation atrophy is still deficient, hindering the development of effective treatments. METHOD In this study, a sciatic nerve transection model was employed in male C57BL/6 J mice to induce muscle denervation atrophy. Gastrocnemius muscles were harvested at 3 days, 2 weeks, and 4 weeks post-denervation for transcriptomic and proteomic analysis. An integrative multi-omics approach was utilized to identify key genes essential for disease progression. Targeted proteomics using PRM was then employed to validate the differential expression of central genes. Combine single-nucleus sequencing results to observe the expression levels of PRM-validated genes in different cell types within muscle tissue.Through upstream regulatory analysis, NRF2 was identified as a potential therapeutic target. The therapeutic potential of the NRF2-targeting drug Omaveloxolone was evaluated in the mouse model. RESULT This research examined the temporal alterations in transcripts and proteins during muscle atrophy subsequent to denervation. A comprehensive analysis identified 54,534 transcripts and 3,218 proteins, of which 23,282 transcripts and 1,852 proteins exhibited statistically significant changes at 3 days, 2 weeks, and 4 weeks post-denervation. Utilizing multi-omics approaches, 30 hubgenes were selected, and PRM validation confirmed significant expression variances in 23 genes. The findings highlighted the involvement of mitochondrial dysfunction, oxidative stress, and metabolic disturbances in the pathogenesis of muscle atrophy, with a pronounced impact on type II muscle fibers, particularly type IIb fibers. The potential therapeutic benefits of Omaveloxolone in mitigating oxidative stress and preserving mitochondrial morphology were confirmed, thereby presenting novel strategies for addressing muscle atrophy induced by denervation. GSEA analysis results show that Autophagy, glutathione metabolism, and PPAR signaling pathways are significantly upregulated, while inflammation-related and neurodegenerative disease-related pathways are significantly inhibited in the Omaveloxolone group.GSR expression and the GSH/GSSG ratio were significantly higher in the Omaveloxolone group compared to the control group, while MuSK expression was significantly lower than in the control group. CONCLUSION In our study, we revealed the crucial role of oxidative stress, glucose metabolism, and mitochondrial dysfunction in denervation-induced muscle atrophy, identifying NRF2 as a potential therapeutic target. Omaveloxolone was shown to stabilize mitochondrial function, enhance antioxidant capacity, and protect neuromuscular junctions, thereby offering promising therapeutic potential for treating denervation-induced muscle atrophy.
Collapse
Affiliation(s)
- Sulong Wang
- Microsurgery Department of Orthopaedic Center, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, Xinjiang, China
| | - Xin Yang
- Microsurgery Department of Orthopaedic Center, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, Xinjiang, China
| | - Kai Liu
- Microsurgery Department of Orthopaedic Center, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, Xinjiang, China
| | - Debin Xiong
- Microsurgery Department of Orthopaedic Center, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, Xinjiang, China
| | - Ainizier Yalikun
- Microsurgery Department of Orthopaedic Center, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, Xinjiang, China
| | - Yimurang Hamiti
- Microsurgery Department of Orthopaedic Center, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, Xinjiang, China
| | - Aihemaitijiang Yusufu
- Microsurgery Department of Orthopaedic Center, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, Xinjiang, China.
- Key Laboratory of Trauma Repair and Reconstruction of Xinjiang Province, Xinjiang Medical University, Urumqi, 830054, Xinjiang, China.
| |
Collapse
|
8
|
Wang Y, Geng X, Sun X, Cui H, Guo Z, Chu D, Li J, Li Z. Celastrol alleviates subconjunctival fibrosis induced by silicone implants mimicking glaucoma surgery. Eur J Pharm Biopharm 2024; 201:114352. [PMID: 38851459 DOI: 10.1016/j.ejpb.2024.114352] [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: 02/05/2024] [Revised: 05/29/2024] [Accepted: 06/06/2024] [Indexed: 06/10/2024]
Abstract
Subconjunctival fibrosis is critical to the outcomes of several ophthalmic conditions or procedures, such as glaucoma filtering surgery. This study aimed to investigate the anti-fibrotic effect of celastrol on subconjunctival fibrosis and to further reveal the underlying mechanisms. We used celastrol-loaded nanomicelles hydrogel hybrid as a sustained-release drug. A rabbit model of subconjunctival fibrosis following silicone implantation was used for in vivo study, and TGF-β1-induced human pterygium fibroblast (HPF) activation as an in vitro model. The effects of celastrol on inhibiting TGF-β1-induced migration and proliferation of HPFs were evaluated by scratch wound assay and CCK-8, respectively. Immunofluorescence and western blotting were used to examine the effect of celastrol on the expression of α-SMA, collagen I, fibronectin, and the targets of the Hippo signaling pathway. We found that in vivo celastrol treatment reduced the expression of YAP and TAZ in subconjunctival tissue. Moreover, celastrol alleviated collagen deposition and subconjunctival fibrosis at 8 weeks. No obvious tissue toxicity was observed in the rabbit models. Mechanistically, celastrol significantly inhibited TGF-β1-induced proliferation and migration of HPFs. Pretreatment of HPFs with celastrol also suppressed the TGF-β1-induced protein expression of α-SMA, collagen I, fibronectin, TGF-βRII, phosphorylated Smad2/3, YAP, TAZ, and TEAD1. In conclusion, celastrol effectively prevented subconjunctival fibrosis through inhibiting TGF-β1/Smad2/3-YAP/TAZ pathway. Celastrol could serve as a promising therapy for subconjunctival fibrosis.
Collapse
Affiliation(s)
- Yiwei Wang
- Henan Eye Hospital, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou 450003, China
| | - Xingchen Geng
- Henan Eye Hospital, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou 450003, China
| | - Xue Sun
- Henan Eye Hospital, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou 450003, China
| | - Haohao Cui
- Henan Eye Hospital, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou 450003, China
| | - Zhihua Guo
- Henan Eye Hospital, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou 450003, China
| | - Dandan Chu
- Henan Eye Hospital, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou 450003, China
| | - Jingguo Li
- Henan Eye Hospital, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou 450003, China.
| | - Zhanrong Li
- Henan Eye Hospital, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou 450003, China.
| |
Collapse
|
9
|
Qing TL, Jiang XY, Li JF, Shen Q, Zhao XY, Ren LJ, Dai XY, Zhang JQZ, Shi WJ, Zhang XF, Zhang B, Yan L, Chen JK, Zhu JB. Celastrol reduces lung inflammation induced by multiwalled carbon nanotubes in mice via NF-κb-signaling pathway. Inhal Toxicol 2024; 36:275-281. [PMID: 38836332 DOI: 10.1080/08958378.2024.2351098] [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: 10/27/2023] [Accepted: 04/29/2024] [Indexed: 06/06/2024]
Abstract
Multiwalled carbon nanotubes (MWCNTs) have numerous applications in the field of carbon nanomaterials. However, the associated toxicity concerns have increased significantly because of their widespread use. The inhalation of MWCNTs can lead to nanoparticle deposition in the lung tissue, causing inflammation and health risks. In this study, celastrol, a natural plant medicine with potent anti-inflammatory properties, effectively reduced the number of inflammatory cells, including white blood cells, neutrophils, and lymphocytes, and levels of inflammatory cytokines, such as IL-1β, IL-6, and TNF-α, in mice lungs exposed to MWCNTs. Moreover, celastrol inhibited the activation of the NF-κB-signaling pathway. This study confirmed these findings by demonstrating comparable reductions in inflammation upon exposure to MWCNTs in mice with the deletion of NF-κB (P50-/-). These results indicate the utility of celastrol as a promising pharmacological agent for preventing MWCNT-induced lung tissue inflammation.
Collapse
Affiliation(s)
- Tao-Lin Qing
- Department of Health Toxicology, Faculty of Naval Medicine, Naval Medical University, Shanghai, China
| | - Xuan-Yao Jiang
- Department of Health Toxicology, Faculty of Naval Medicine, Naval Medical University, Shanghai, China
| | - Jin-Feng Li
- Department of Health Toxicology, Faculty of Naval Medicine, Naval Medical University, Shanghai, China
| | - Qi Shen
- Department of Health Toxicology, Faculty of Naval Medicine, Naval Medical University, Shanghai, China
| | - Xin-Yi Zhao
- Department of Health Toxicology, Faculty of Naval Medicine, Naval Medical University, Shanghai, China
| | - Li-Jun Ren
- Department of Health Toxicology, Faculty of Naval Medicine, Naval Medical University, Shanghai, China
| | - Xiao-Yu Dai
- Department of Health Toxicology, Faculty of Naval Medicine, Naval Medical University, Shanghai, China
| | - Ji-Qian-Zhu Zhang
- Department of Health Toxicology, Faculty of Naval Medicine, Naval Medical University, Shanghai, China
| | - Wen-Jing Shi
- Department of Health Toxicology, Faculty of Naval Medicine, Naval Medical University, Shanghai, China
| | - Xiao-Fang Zhang
- Department of Health Toxicology, Faculty of Naval Medicine, Naval Medical University, Shanghai, China
| | - Bin Zhang
- Department of Health Toxicology, Faculty of Naval Medicine, Naval Medical University, Shanghai, China
| | - Lang Yan
- Department of Health Toxicology, Faculty of Naval Medicine, Naval Medical University, Shanghai, China
| | - Ji-Kuai Chen
- Department of Health Toxicology, Faculty of Naval Medicine, Naval Medical University, Shanghai, China
| | - Jiang-Bo Zhu
- Department of Health Toxicology, Faculty of Naval Medicine, Naval Medical University, Shanghai, China
| |
Collapse
|
10
|
Bai Y, Liang C, Gao L, Han T, Wang F, Liu Y, Zhou J, Guo J, Wu J, Hu D. Celastrol Pyrazine Derivative Alleviates Silicosis Progression via Inducing ROS-Mediated Apoptosis in Activated Fibroblasts. Molecules 2024; 29:538. [PMID: 38276616 PMCID: PMC10820882 DOI: 10.3390/molecules29020538] [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: 11/21/2023] [Revised: 12/27/2023] [Accepted: 01/19/2024] [Indexed: 01/27/2024] Open
Abstract
Silicosis is a complex occupational disease without recognized effective treatment. Celastrol, a natural product, has shown antioxidant, anti-inflammatory, and anti-fibrotic activities, but the narrow therapeutic window and high toxicity severely limit its clinical application. Through structural optimization, we have identified a highly efficient and low-toxicity celastrol derivative, CEL-07. In this study, we systematically investigated the therapeutic potential and underlying mechanisms of CEL-07 in silicosis fibrosis. By constructing a silicosis mouse model and analyzing with HE, Masson, Sirius Red, and immunohistochemical staining, CEL-07 significantly prevented the progress of inflammation and fibrosis, and it effectively improved the lung respiratory function of silicosis mice. Additionally, CEL-07 markedly suppressed the expression of inflammatory factors (IL-6, IL-1α, TNF-α, and TNF-β) and fibrotic factors (α-SMA, collagen I, and collagen III), and promoted apoptosis of fibroblasts by increasing ROS accumulation. Moreover, bioinformatics analysis combined with experimental validation revealed that CEL-07 inhibited the pathways associated with inflammation (PI3K-AKT and JAK2-STAT3) and the expression of apoptosis-related proteins. Overall, these results suggest that CEL-07 may serve as a potential candidate for the treatment of silicosis.
Collapse
Affiliation(s)
- Ying Bai
- School of Medicine, Anhui University of Science and Technology, Huainan 232001, China; (Y.B.); (C.L.); (L.G.); (T.H.); (F.W.); (Y.L.); (J.Z.); (J.G.)
- Anhui Occupational Health and Safety Engineering Laboratory, Huainan 232001, China
| | - Chao Liang
- School of Medicine, Anhui University of Science and Technology, Huainan 232001, China; (Y.B.); (C.L.); (L.G.); (T.H.); (F.W.); (Y.L.); (J.Z.); (J.G.)
- Anhui Occupational Health and Safety Engineering Laboratory, Huainan 232001, China
| | - Lu Gao
- School of Medicine, Anhui University of Science and Technology, Huainan 232001, China; (Y.B.); (C.L.); (L.G.); (T.H.); (F.W.); (Y.L.); (J.Z.); (J.G.)
- Anhui Occupational Health and Safety Engineering Laboratory, Huainan 232001, China
| | - Tao Han
- School of Medicine, Anhui University of Science and Technology, Huainan 232001, China; (Y.B.); (C.L.); (L.G.); (T.H.); (F.W.); (Y.L.); (J.Z.); (J.G.)
- Anhui Occupational Health and Safety Engineering Laboratory, Huainan 232001, China
| | - Fengxuan Wang
- School of Medicine, Anhui University of Science and Technology, Huainan 232001, China; (Y.B.); (C.L.); (L.G.); (T.H.); (F.W.); (Y.L.); (J.Z.); (J.G.)
- Anhui Occupational Health and Safety Engineering Laboratory, Huainan 232001, China
| | - Yafeng Liu
- School of Medicine, Anhui University of Science and Technology, Huainan 232001, China; (Y.B.); (C.L.); (L.G.); (T.H.); (F.W.); (Y.L.); (J.Z.); (J.G.)
- Anhui Occupational Health and Safety Engineering Laboratory, Huainan 232001, China
| | - Jiawei Zhou
- School of Medicine, Anhui University of Science and Technology, Huainan 232001, China; (Y.B.); (C.L.); (L.G.); (T.H.); (F.W.); (Y.L.); (J.Z.); (J.G.)
- Anhui Occupational Health and Safety Engineering Laboratory, Huainan 232001, China
| | - Jianqiang Guo
- School of Medicine, Anhui University of Science and Technology, Huainan 232001, China; (Y.B.); (C.L.); (L.G.); (T.H.); (F.W.); (Y.L.); (J.Z.); (J.G.)
- Anhui Occupational Health and Safety Engineering Laboratory, Huainan 232001, China
| | - Jing Wu
- School of Medicine, Anhui University of Science and Technology, Huainan 232001, China; (Y.B.); (C.L.); (L.G.); (T.H.); (F.W.); (Y.L.); (J.Z.); (J.G.)
- Anhui Occupational Health and Safety Engineering Laboratory, Huainan 232001, China
- Key Laboratory of Industrial Dust Deep Reduction and Occupational Health and Safety of Anhui Higher Education Institute, Huainan 232001, China
- Key Laboratory of Industrial Dust Prevention and Control and Occupational Safety and Health Ministry of Education, Anhui University of Science and Technology, Huainan 232001, China
| | - Dong Hu
- School of Medicine, Anhui University of Science and Technology, Huainan 232001, China; (Y.B.); (C.L.); (L.G.); (T.H.); (F.W.); (Y.L.); (J.Z.); (J.G.)
- Anhui Occupational Health and Safety Engineering Laboratory, Huainan 232001, China
- Key Laboratory of Industrial Dust Deep Reduction and Occupational Health and Safety of Anhui Higher Education Institute, Huainan 232001, China
- Key Laboratory of Industrial Dust Prevention and Control and Occupational Safety and Health Ministry of Education, Anhui University of Science and Technology, Huainan 232001, China
| |
Collapse
|