1
|
Wang X, Abu Bakar MH, Liqun S, Kassim MA, Shariff KA, Karunakaran T. Targeting metabolic diseases with celastrol: A comprehensive review of anti-inflammatory mechanisms and therapeutic potential. JOURNAL OF ETHNOPHARMACOLOGY 2025; 344:119560. [PMID: 40015541 DOI: 10.1016/j.jep.2025.119560] [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: 12/10/2024] [Revised: 02/15/2025] [Accepted: 02/23/2025] [Indexed: 03/01/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Tripterygium wilfordii is a traditional Chinese medicine used to treat rheumatic diseases, with properties such as clearing heat, detoxifying, dispelling wind, and relieving pain. In recent years, its active compound, celastrol, garnered significant attention for its potential therapeutic effects on metabolic diseases. Celastrol exhibits bioactivities such as regulating metabolic functions and anti-inflammatory effects, positioning it as a promising candidate for the treatment of obesity, diabetes, atherosclerosis (AS), and non-alcoholic fatty liver disease (NAFLD). AIM OF THE REVIEW This review aims to explore the pharmacological mechanisms of celastrol in metabolic diseases, focusing on its anti-inflammatory mechanisms and metabolic regulation effects, providing theoretical support for further investigation of its therapeutic potential in metabolic diseases. METHODS Literature was retrieved from PubMed, Web of Science, Scopus, Cochrane, and Google Scholar. This review primarily focuses on anti-inflammatory mechanisms of celastrol, its metabolic regulation, and toxicity studies, by systematically analyzing its effects in obesity, diabetes, AS, and NAFLD, providing scientific evidence for its potential clinical applications. RESULTS Celastrol regulates multiple signaling pathways, particularly inhibiting NF-κB and activating AMPK, reducing the production of pro-inflammatory cytokines and improving insulin sensitivity, enhancing its therapeutic potential in metabolic diseases. Additionally, celastrol regulates adipogenesis and energy metabolism by influencing key transcription factors such as PPARγ and SREBP-1c. Numerous studies highlight its role in alleviating oxidative stress and improving mitochondrial function, further enhancing its metabolic benefits. CONCLUSION In summary, celastrol holds great promise as a multi-target therapeutic agent for metabolic diseases, offering anti-inflammatory, metabolic regulatory, and antioxidative benefits. Despite these, challenges remain for the clinical application of celastrol due to its poor bioavailability and potential toxicity. Advanced formulation strategies and targeted delivery systems are urgently needed to overcome challenges related to bioavailability and clinical translation.
Collapse
Affiliation(s)
- Xiaojuan Wang
- Bioprocess Technology Division, School of Industrial Technology, Universiti Sains Malaysia, Gelugor, 11800, Penang, Malaysia; Department of Pharmacy, Taishan Vocational College of Nursing, 271099, Tai'an, Shandong, China
| | - Mohamad Hafizi Abu Bakar
- Bioprocess Technology Division, School of Industrial Technology, Universiti Sains Malaysia, Gelugor, 11800, Penang, Malaysia.
| | - Song Liqun
- Department of Pharmacy, Taishan Vocational College of Nursing, 271099, Tai'an, Shandong, China
| | - Mohd Asyraf Kassim
- Bioprocess Technology Division, School of Industrial Technology, Universiti Sains Malaysia, Gelugor, 11800, Penang, Malaysia
| | - Khairul Anuar Shariff
- School of Materials & Mineral Resources Engineering, Universiti Sains Malaysia, Nibong Tebal, 14300, Penang, Malaysia
| | | |
Collapse
|
2
|
Jack BU, Dias S, Pheiffer C. Comparative Effects of Tumor Necrosis Factor Alpha, Lipopolysaccharide, and Palmitate on Mitochondrial Dysfunction in Cultured 3T3-L1 Adipocytes. Cell Biochem Biophys 2025; 83:905-918. [PMID: 39269560 PMCID: PMC11870959 DOI: 10.1007/s12013-024-01522-3] [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] [Accepted: 08/29/2024] [Indexed: 09/15/2024]
Abstract
We have previously reported that dysregulated lipid metabolism and inflammation in 3T3-L1 adipocytes is attributed to tumor necrosis factor alpha (TNFα) rather than lipopolysaccharide (LPS) and palmitate (PA). In this study, we further compared the modulative effects of TNFα, LPS, and PA on mitochondrial function by treating 3T3-L1 adipocytes with TNFα (10 ng/mL), LPS (100 ng/mL), and PA (0.75 mM) individually or in combination for 24 h. Results showed a significant reduction in intracellular adenosine triphosphate (ATP) content, mitochondrial bioenergetics, total antioxidant capacity, and the mRNA expression of citrate synthase (Cs), sirtuin 3 (Sirt3), protein kinase AMP-activated catalytic subunit alpha 2 (Prkaa2), peroxisome proliferator-activated receptor gamma coactivator 1 alpha (Ppargc1α), nuclear respiratory factor 1 (Nrf1), and superoxide dismutase 1 (Sod1) in cells treated with TNFα individually or in combination with LPS and PA. Additionally, TNFα treatments decreased insulin receptor substrate 1 (Irs1), insulin receptor substrate 2 (Irs2), solute carrier family 2, facilitated glucose transporter member 4 (Slc2a4), and phosphoinositide 3 kinase regulatory subunit 1 (Pik3r1) mRNA expression. Treatment with LPS and PA alone, or in combination, did not affect the assessed metabolic parameters, while the combination of LPS and PA increased lipid peroxidation. These results show that TNFα but not LPS and PA dysregulate mitochondrial function, thus inducing oxidative stress and impaired insulin signaling in 3T3-L1 adipocytes. This suggests that TNFα treatment can be used as a basic in vitro model for studying the pathophysiology of mitochondrial dysfunction and related metabolic complications and screening potential anti-obesity therapeutics in 3T3-L1 adipocytes.
Collapse
Affiliation(s)
- Babalwa Unice Jack
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg, Cape Town, 7505, South Africa.
- Centre for Cardiometabolic Research in Africa, Division of Medical Physiology, Stellenbosch University, Tygerberg, Cape Town, 7505, South Africa.
| | - Stephanie Dias
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg, Cape Town, 7505, South Africa
| | - Carmen Pheiffer
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg, Cape Town, 7505, South Africa
- Department of Obstetrics and Gynaecology, Faculty of Health Sciences, University of Pretoria, Pretoria, 0001, South Africa
| |
Collapse
|
3
|
Gu J, Shi YN, Zhu N, Li HF, Zhang CJ, Qin L. Celastrol functions as an emerging manager of lipid metabolism: Mechanism and therapeutic potential. Biomed Pharmacother 2023; 164:114981. [PMID: 37285754 DOI: 10.1016/j.biopha.2023.114981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 05/27/2023] [Accepted: 06/01/2023] [Indexed: 06/09/2023] Open
Abstract
Lipid metabolism disorders are pivotal in the development of various lipid-related diseases, such as obesity, atherosclerosis, non-alcoholic fatty liver disease, type 2 diabetes, and cancer. Celastrol, a bioactive compound extracted from the Chinese herb Tripterygium wilfordii Hook F, has recently demonstrated potent lipid-regulating abilities and promising therapeutic effects for lipid-related diseases. There is substantial evidence indicating that celastrol can ameliorate lipid metabolism disorders by regulating lipid profiles and related metabolic processes, including lipid synthesis, catabolism, absorption, transport, and peroxidation. Even wild-type mice show augmented lipid metabolism after treatment with celastrol. This review aims to provide an overview of recent advancements in the lipid-regulating properties of celastrol, as well as to elucidate its underlying molecular mechanisms. Besides, potential strategies for targeted drug delivery and combination therapy are proposed to enhance the lipid-regulating effects of celastrol and avoid the limitations of its clinical application.
Collapse
Affiliation(s)
- Jia Gu
- Laboratory of Stem Cell Regulation with Chinese Medicine and Its Application, School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, Hunan, China
| | - Ya-Ning Shi
- Laboratory of Stem Cell Regulation with Chinese Medicine and Its Application, School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, Hunan, China; Science and Technology Innovation Center, Hunan University of Chinese Medicine, Changsha 410208, Hunan, China
| | - Neng Zhu
- Department of Urology, The First Hospital of Hunan University of Chinese Medicine, Changsha 410021, Hunan, China
| | - Hong-Fang Li
- Laboratory of Stem Cell Regulation with Chinese Medicine and Its Application, School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, Hunan, China
| | - Chan-Juan Zhang
- Laboratory of Stem Cell Regulation with Chinese Medicine and Its Application, School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, Hunan, China
| | - Li Qin
- Laboratory of Stem Cell Regulation with Chinese Medicine and Its Application, School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, Hunan, China; Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Hunan University of Chinese Medicine, Changsha 410208, Hunan, China; Hunan Engineering Technology Research Center for Bioactive Substance Discovery of Chinese Medicine, Hunan University of Chinese Medicine, Changsha 410208, Hunan, China.
| |
Collapse
|
4
|
Wu Y, Zhao R, Li M, Li H, Chen Z, Zhao Y. Novel soybean peptide iglycin ameliorates insulin resistance of high-fat diet fed C57BL/6J mice and differentiated 3T3L1 adipocytes with improvement of insulin signaling and mitochondrial function. FOOD SCIENCE AND HUMAN WELLNESS 2022. [DOI: 10.1016/j.fshw.2022.06.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
5
|
Bakar MHA, Shahril NSN, Khalid MSFM, Mohammad S, Shariff KA, Karunakaran T, Salleh RM, Rosdi MN. Celastrol alleviates high-fat diet-induced obesity via enhanced muscle glucose utilization and mitochondrial oxidative metabolism-mediated upregulation of pyruvate dehydrogenase complex. Toxicol Appl Pharmacol 2022; 449:116099. [DOI: 10.1016/j.taap.2022.116099] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 05/23/2022] [Accepted: 06/01/2022] [Indexed: 12/25/2022]
|
6
|
Abu Bakar MH, Mohamad Khalid MSF, Nor Shahril NS, Shariff KA, Karunakaran T. Celastrol attenuates high-fructose diet-induced inflammation and insulin resistance via inhibition of 11β-hydroxysteroid dehydrogenase type 1 activity in rat adipose tissues. Biofactors 2022; 48:111-134. [PMID: 34676604 DOI: 10.1002/biof.1793] [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: 05/15/2021] [Accepted: 09/27/2021] [Indexed: 11/10/2022]
Abstract
High fructose consumption has been linked to low-grade inflammation and insulin resistance that results in increased intracellular 11ß-hydroxysteroid dehydrogenase type 1 (11β-HSD1) activity. Celastrol, a pentacyclic triterpene, has been demonstrated to exhibit multifaceted targets to attenuate various metabolic diseases associated with inflammation. However, the underlying mechanisms by which celastrol exerts its attributive properties on high fructose diet (HFrD)-induced metabolic syndrome remain elusive. Herein, the present study was aimed to elucidate the mechanistic targets of celastrol co-administrations upon HFrD in rats and evaluate its potential to modulate 11β-HSD1 activity. Celastrol remarkably improved glucose tolerance, lipid profiles, and insulin sensitivity along with suppression of hepatic glucose production. In rat adipose tissues, celastrol attenuated nuclear factor-kappa B (NF-κB)-driven inflammation, reduced c-Jun N-terminal kinases (JNK) phosphorylation, and mitigated oxidative stress via upregulated genes expression involved in mitochondrial biogenesis. Furthermore, insulin signaling pathways were significantly improved through the restoration of Akt phosphorylation levels at Ser473 and Thr308 residues. Celastrol exhibited a potent, selective and specific inhibitor of intracellular 11β-HSD1 towards oxidoreductase activity (IC50 value = 4.3 nM) in comparison to other HSD-related enzymes. Inhibition of 11β-HSD1 expression in rat adipose microsomes reduced the availability of its cofactor NADPH and substrate H6PDH in couple to upregulated mRNA and protein expressions of glucocorticoid receptor. In conclusion, our results underscore the most likely conceivable mechanisms exhibited by celastrol against HFrD-induced metabolic dysregulations mainly through attenuating inflammation and insulin resistance, at least via specific inhibitions on 11β-HSD1 activity in adipose tissues.
Collapse
Affiliation(s)
- Mohamad Hafizi Abu Bakar
- Bioprocess Technology Division, School of Industrial Technology, Universiti Sains Malaysia, 11800, Gelugor, Penang, Malaysia
| | | | - Nor Shafiqah Nor Shahril
- Bioprocess Technology Division, School of Industrial Technology, Universiti Sains Malaysia, 11800, Gelugor, Penang, Malaysia
| | - Khairul Anuar Shariff
- School of Materials and Mineral Resources Engineering, Universiti Sains Malaysia, 14300, Nibong Tebal, Penang, Malaysia
| | | |
Collapse
|
7
|
Zhao J, Zhang F, Xiao X, Wu Z, Hu Q, Jiang Y, Zhang W, Wei S, Ma X, Zhang X. Tripterygium hypoglaucum (Lévl.) Hutch and Its Main Bioactive Components: Recent Advances in Pharmacological Activity, Pharmacokinetics and Potential Toxicity. Front Pharmacol 2021; 12:715359. [PMID: 34887747 PMCID: PMC8650721 DOI: 10.3389/fphar.2021.715359] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 11/04/2021] [Indexed: 01/12/2023] Open
Abstract
Tripterygium hypoglaucum (Lévl.) Hutch (THH) is believed to play an important role in health care and disease treatment according to traditional Chinese medicine. Moreover, it is also the representative of medicine with both significant efficacy and potential toxicity. This characteristic causes THH hard for embracing and fearing. In order to verify its prospect for clinic, a wide variety of studies were carried out in the most recent years. However, there has not been any review about THH yet. Therefore, this review summarized its characteristic of components, pharmacological effect, pharmacokinetics and toxicity to comprehensively shed light on the potential clinical application. More than 120 secondary metabolites including terpenoids, alkaloids, glycosides, sugars, organic acids, oleanolic acid, polysaccharides and other components were found in THH based on phytochemical research. All these components might be the pharmacological bases for immunosuppression, anti-inflammatory and anti-tumour effect. In addition, recent studies found that THH and its bioactive compounds also demonstrated remarkable effect on obesity, insulin resistance, fertility and infection of virus. The main mechanism seemed to be closely related to regulation the balance of immune, inflammation, apoptosis and so on in various disease. Furthermore, the study of pharmacokinetics revealed quick elimination of the main component triptolide. The feature of celastrol was also investigated by several models. Finally, the side effect of THH was thought to be the key for its limitation in clinical application. A series of reports indicated that multiple organs or systems including liver, kidney and genital system were involved in the toxicity. Its potential serious problem in liver was paid specific attention in recent years. In summary, considering the significant effect and potential toxicity of THH as well as its components, the combined medication to inhibit the toxicity, maintain effect might be a promising method for clinical conversion. Modern advanced technology such as structure optimization might be another way to reach the efficacy and safety. Thus, THH is still a crucial plant which remains for further investigation.
Collapse
Affiliation(s)
- Junqi Zhao
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Fangling Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiaolin Xiao
- Hospital of Chengdu University of Traditional Chinese Medicine, School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zhao Wu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qichao Hu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yinxiao Jiang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Wenwen Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Shizhang Wei
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiao Ma
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiaomei Zhang
- Institute of Medicinal Chemistry of Chinese Medicine, Chongqing Academy of Chinese Materia Medica, Chongqing, China
| |
Collapse
|
8
|
Zhang C, Zhao M, Wang B, Su Z, Guo B, Qin L, Zhang W, Zheng R. The Nrf2-NLRP3-caspase-1 axis mediates the neuroprotective effects of Celastrol in Parkinson's disease. Redox Biol 2021; 47:102134. [PMID: 34600334 PMCID: PMC8487081 DOI: 10.1016/j.redox.2021.102134] [Citation(s) in RCA: 106] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 09/10/2021] [Accepted: 09/12/2021] [Indexed: 12/14/2022] Open
Abstract
Parkinson's disease (PD) is a chronic neurodegenerative disorder that is characterized by motor symptoms as a result of a loss of dopaminergic neurons in the substantia nigra pars compacta (SNc), accompanied by chronic neuroinflammation, oxidative stress, formation of α-synuclein aggregates. Celastrol, a potent anti-inflammatory and anti-oxidative pentacyclic triterpene, has emerged as a neuroprotective agent. However, the mechanisms by which celastrol is neuroprotective in PD remain elusive. Here we show that celastrol protects against dopamine neuron loss, mitigates neuroinflammation, and relieves motor deficits in MPTP-induced PD mouse model and AAV-mediated human α-synuclein overexpression PD model. Whole-genome deep sequencing analysis revealed that Nrf2, NLRP3 and caspase-1 in SNc may be associated with the neuroprotective actions of celastrol in PD. By using multiple genetically modified mice (Nrf2-KO, NLRP3-KO and Caspase-1-KO), we identified that celastrol inhibits NLRP3 inflammasome activation, relieves motor deficits and nigrostriatal dopaminergic degeneration through Nrf2-NLRP3-caspase-1 pathway. Taken together, these findings suggest that Nrf2-NLRP3-caspase-1 axis may serve as a key target of celastrol in PD treatment, and highlight the favorable properties of celastrol for neuroprotection, making celastrol as a promising disease-modifying agent for PD.
Collapse
Affiliation(s)
- Chenyu Zhang
- Department of Anatomy, Histology and Embryology, Health Science Center, Peking University, Beijing, China
| | - Miao Zhao
- Department of Anatomy, Histology and Embryology, Health Science Center, Peking University, Beijing, China
| | - Bingwei Wang
- Department of Anatomy, Histology and Embryology, Health Science Center, Peking University, Beijing, China
| | - Zhijie Su
- Department of Anatomy, Histology and Embryology, Health Science Center, Peking University, Beijing, China
| | - Bingbing Guo
- Department of Anatomy, Histology and Embryology, Health Science Center, Peking University, Beijing, China
| | - Lihua Qin
- Department of Anatomy, Histology and Embryology, Health Science Center, Peking University, Beijing, China
| | - Weiguang Zhang
- Department of Anatomy, Histology and Embryology, Health Science Center, Peking University, Beijing, China
| | - Ruimao Zheng
- Department of Anatomy, Histology and Embryology, Health Science Center, Peking University, Beijing, China; Neuroscience Research Institute, Peking University, Beijing, China; Key Laboratory for Neuroscience of Ministry of Education, Peking University, Beijing, China; Key Laboratory for Neuroscience of National Health Commission, Peking University, Beijing, China.
| |
Collapse
|
9
|
Kim CW, Go RE, Lee HK, Kang BT, Cho WJ, Choi KC. Anti-obesity effects of Celastrus orbiculatus extract containing celastrol on canine adipocytes. CANADIAN JOURNAL OF VETERINARY RESEARCH = REVUE CANADIENNE DE RECHERCHE VETERINAIRE 2021; 85:177-185. [PMID: 34248261 PMCID: PMC8243799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 01/04/2021] [Indexed: 06/13/2023]
Abstract
From 50 to 60% of companion animals in the United States are overweight or obese and this obesity rate is rising. As obesity is associated with a number of health problems, an agent that can help weight loss in pets and assist in clinically managing obesity through veterinary prescription foods and medication would be beneficial. Many studies have shown that celastrol, a phytochemical compound found in Celastrus orbiculatus extract (COE), has anti-obesity and anti-inflammatory effects, although these effects have not yet been determined in canine or canine-derived cells. The objective of this study was to investigate the effects of celastrol on the adipogenic differentiation and lipolysis of canine adipocytes. Primary preadipocytes were isolated from the gluteal region of a beagle dog and the primary adipocytes were differentiated into mature adipocytes by adipocyte differentiation media containing isobutylmethylxanthine, dexamethasone, and insulin. In a water-soluble tetrazolium (WST) assay, the cell viability of mature adipocytes was decreased after treatment with COE (0, 0.93, 2.32, and 4.64 nM celastrol) in a concentration-dependent manner, although preadipocytes were not affected. Oil Red O (ORO) staining revealed that COE inhibited the differentiation into mature adipocytes and lipid accumulation in adipocytes. In addition, treatment with COE significantly reduced triglyceride content and increased lipolytic activities by 1.5-fold in canine adipocytes. Overall, it was concluded that COE may enhance anti-obesity activity in canine adipocytes by inhibiting lipid accumulation and increasing lipolytic activity.
Collapse
Affiliation(s)
- Cho-Won Kim
- Laboratory of Biochemistry and Immunology (Kim, Go, Lee, Choi) and Laboratory of Veterinary Dermatology and Neurology (Kang), College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk, 28644, Republic of Korea; Veterinary Nutrition Laboratory, JEIL Feed Co. Ltd., Daejeon, Chungnam, Republic of Korea (Cho)
| | - Ryeo-Eun Go
- Laboratory of Biochemistry and Immunology (Kim, Go, Lee, Choi) and Laboratory of Veterinary Dermatology and Neurology (Kang), College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk, 28644, Republic of Korea; Veterinary Nutrition Laboratory, JEIL Feed Co. Ltd., Daejeon, Chungnam, Republic of Korea (Cho)
| | - Hong Kyu Lee
- Laboratory of Biochemistry and Immunology (Kim, Go, Lee, Choi) and Laboratory of Veterinary Dermatology and Neurology (Kang), College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk, 28644, Republic of Korea; Veterinary Nutrition Laboratory, JEIL Feed Co. Ltd., Daejeon, Chungnam, Republic of Korea (Cho)
| | - Byeong-Teck Kang
- Laboratory of Biochemistry and Immunology (Kim, Go, Lee, Choi) and Laboratory of Veterinary Dermatology and Neurology (Kang), College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk, 28644, Republic of Korea; Veterinary Nutrition Laboratory, JEIL Feed Co. Ltd., Daejeon, Chungnam, Republic of Korea (Cho)
| | - Woo Jae Cho
- Laboratory of Biochemistry and Immunology (Kim, Go, Lee, Choi) and Laboratory of Veterinary Dermatology and Neurology (Kang), College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk, 28644, Republic of Korea; Veterinary Nutrition Laboratory, JEIL Feed Co. Ltd., Daejeon, Chungnam, Republic of Korea (Cho)
| | - Kyung-Chul Choi
- Laboratory of Biochemistry and Immunology (Kim, Go, Lee, Choi) and Laboratory of Veterinary Dermatology and Neurology (Kang), College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk, 28644, Republic of Korea; Veterinary Nutrition Laboratory, JEIL Feed Co. Ltd., Daejeon, Chungnam, Republic of Korea (Cho)
| |
Collapse
|
10
|
Kurosawa R, Satoh K, Nakata T, Shindo T, Kikuchi N, Satoh T, Siddique MAH, Omura J, Sunamura S, Nogi M, Takeuchi Y, Miyata S, Shimokawa H. Identification of Celastrol as a Novel Therapeutic Agent for Pulmonary Arterial Hypertension and Right Ventricular Failure Through Suppression of Bsg (Basigin)/CyPA (Cyclophilin A). Arterioscler Thromb Vasc Biol 2021; 41:1205-1217. [PMID: 33472404 DOI: 10.1161/atvbaha.120.315731] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Pulmonary arterial hypertension is characterized by abnormal proliferation of pulmonary artery smooth muscle cells and vascular remodeling, which leads to right ventricular (RV) failure. Bsg (Basigin) is a transmembrane glycoprotein that promotes myofibroblast differentiation, cell proliferation, and matrix metalloproteinase activation. CyPA (cyclophilin A) binds to its receptor Bsg and promotes pulmonary artery smooth muscle cell proliferation and inflammatory cell recruitment. We previously reported that Bsg promotes cardiac fibrosis and failure in the left ventricle in response to pressure-overload in mice. However, the roles of Bsg and CyPA in RV failure remain to be elucidated. Approach and Results: First, we found that protein levels of Bsg and CyPA were upregulated in the heart of hypoxia-induced pulmonary hypertension (PH) in mice and monocrotaline-induced PH in rats. Furthermore, cardiomyocyte-specific Bsg-overexpressing mice showed exacerbated RV hypertrophy, fibrosis, and dysfunction compared with their littermates under chronic hypoxia and pulmonary artery banding. Treatment with celastrol, which we identified as a suppressor of Bsg and CyPA by drug screening, decreased proliferation, reactive oxygen species, and inflammatory cytokines in pulmonary artery smooth muscle cells. Furthermore, celastrol treatment ameliorated RV systolic pressure, hypertrophy, fibrosis, and dysfunction in hypoxia-induced PH in mice and SU5416/hypoxia-induced PH in rats with reduced Bsg, CyPA, and inflammatory cytokines in the hearts and lungs. CONCLUSIONS These results indicate that elevated Bsg in pressure-overloaded RV exacerbates RV dysfunction and that celastrol ameliorates RV dysfunction in PH model animals by suppressing Bsg and its ligand CyPA. Thus, celastrol can be a novel drug for PH and RV failure that targets Bsg and CyPA. Graphic Abstract: A graphic abstract is available for this article.
Collapse
Affiliation(s)
- Ryo Kurosawa
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Kimio Satoh
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Takashi Nakata
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Tomohiko Shindo
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Nobuhiro Kikuchi
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Taijyu Satoh
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Mohammad A H Siddique
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Junichi Omura
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Shinichiro Sunamura
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Masamichi Nogi
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yutaro Takeuchi
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Satoshi Miyata
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Hiroaki Shimokawa
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| |
Collapse
|
11
|
Abu Bakar MH, Shariff KA, Tan JS, Lee LK. Celastrol attenuates inflammatory responses in adipose tissues and improves skeletal muscle mitochondrial functions in high fat diet-induced obese rats via upregulation of AMPK/SIRT1 signaling pathways. Eur J Pharmacol 2020; 883:173371. [DOI: 10.1016/j.ejphar.2020.173371] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Revised: 07/09/2020] [Accepted: 07/13/2020] [Indexed: 12/20/2022]
|
12
|
Hou W, Liu B, Xu H. Celastrol: Progresses in structure-modifications, structure-activity relationships, pharmacology and toxicology. Eur J Med Chem 2020; 189:112081. [DOI: 10.1016/j.ejmech.2020.112081] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2019] [Revised: 01/17/2020] [Accepted: 01/17/2020] [Indexed: 12/13/2022]
|
13
|
Hamon MP, Gergondey R, L'honoré A, Friguet B. Mitochondrial Lon protease - depleted HeLa cells exhibit proteome modifications related to protein quality control, stress response and energy metabolism. Free Radic Biol Med 2020; 148:83-95. [PMID: 31904544 DOI: 10.1016/j.freeradbiomed.2019.12.039] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 12/23/2019] [Accepted: 12/24/2019] [Indexed: 12/20/2022]
Abstract
The ATP-dependent Lon protease is located in the mitochondrial matrix and oxidized proteins are among its primary targets for their degradation. Impairment of mitochondrial morphology and function together with apoptosis were observed in lung fibroblasts depleted for Lon expression while accumulation of carbonylated mitochondrial proteins has been reported for yeast and HeLa Lon deficient cells. In addition, age-related mitochondrial dysfunction has been associated with an impairment of Lon expression. Using a HeLa cell line stably transfected with an inducible shRNA directed against Lon, we have previously observed that Lon depletion results in a mild phenotype characterized by an increase of both production of reactive oxygen species and level of oxidized proteins (Bayot et al., 2014, Biochimie, 100: 38-47). In this study using the same cell line, we now show that Lon knockdown leads to modifications of the expression of a number of specific proteins involved in protein quality control, stress response and energy metabolism, as evidenced using a 2D gel-based proteomic approach, and to alteration of the mitochondrial network morphology. We also show that these effects are associated with decreased proliferation and can be modulated by culture conditions in galactose versus glucose containing medium.
Collapse
Affiliation(s)
- Marie-Paule Hamon
- Sorbonne Université, CNRS, INSERM, Institut de Biologie Paris-Seine, Biological Adaptation and Aging, B2A-IBPS, F-75005, Paris, France
| | - Rachel Gergondey
- Sorbonne Université, CNRS, INSERM, Institut de Biologie Paris-Seine, Biological Adaptation and Aging, B2A-IBPS, F-75005, Paris, France
| | - Aurore L'honoré
- Sorbonne Université, CNRS, INSERM, Institut de Biologie Paris-Seine, Biological Adaptation and Aging, B2A-IBPS, F-75005, Paris, France
| | - Bertrand Friguet
- Sorbonne Université, CNRS, INSERM, Institut de Biologie Paris-Seine, Biological Adaptation and Aging, B2A-IBPS, F-75005, Paris, France.
| |
Collapse
|
14
|
Higashida K, Takeuchi N, Inoue S, Hashimoto T, Nakai N. Iron deficiency attenuates catecholamine‑stimulated lipolysis via downregulation of lipolysis‑related proteins and glucose utilization in 3T3‑L1 adipocytes. Mol Med Rep 2020; 21:1383-1389. [PMID: 32016466 DOI: 10.3892/mmr.2020.10929] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Accepted: 10/29/2019] [Indexed: 11/06/2022] Open
Abstract
Iron deficiency has been associated with obesity and related metabolic disorders. The aim of the present study was to evaluate the effect of iron deficiency on fat metabolism, particularly regarding the lipolytic activity, lipolysis‑related protein expression, and glucose utilization of adipocytes. Differentiated 3T3‑L1 adipocytes were incubated with an iron chelator, deferoxamine mesylate (DFO), for 48 h. Subsequently, basal and isoproterenol‑stimulated lipolytic activities, the proteins involved in lipolysis and glucose utilization were compared with a control (CON). The results revealed that treatment with DFO significantly decreased the free iron content but did not affect total protein and lipid contents in adipocytes. Iron deprivation caused a significant reduction in isoproterenol‑stimulated lipolysis, but not basal lipolysis. Lipolysis‑related proteins, including perilipin A, adipose triglyceride lipase, hormone sensitive lipase and comparative gene identification‑58, were decreased in the DFO compared with the CON group. Furthermore, glucose utilization, a major precursor of 3‑glycerol phosphate for micro‑lipid droplet synthesis during lipolysis and the expression of glucose transporter (GLUT) 4 were significantly lower in the DFO group when compared with the CON group. However, hypoxia‑inducible factor‑1α and GLUT1 expressions were upregulated in DFO‑treated adipocytes. In conclusion, the results indicated that low iron availability attenuated catecholamine‑stimulated lipolysis by downregulating lipolytic enzymes and glucose utilization in 3T3‑L1 adipocytes.
Collapse
Affiliation(s)
- Kazuhiko Higashida
- Department of Nutrition, Laboratory of Exercise Nutrition, University of Shiga Prefecture, Hikone, Shiga 522‑8533, Japan
| | - Nodoka Takeuchi
- Department of Nutrition, Laboratory of Exercise Nutrition, University of Shiga Prefecture, Hikone, Shiga 522‑8533, Japan
| | - Sachika Inoue
- Department of Nutrition, Laboratory of Exercise Nutrition, University of Shiga Prefecture, Hikone, Shiga 522‑8533, Japan
| | - Takeshi Hashimoto
- Faculty of Sport and Health Science, Ritsumeikan University, Kusatsu, Shiga 525‑8577, Japan
| | - Naoya Nakai
- Department of Nutrition, Laboratory of Exercise Nutrition, University of Shiga Prefecture, Hikone, Shiga 522‑8533, Japan
| |
Collapse
|
15
|
Boran T, Gunaydin A, Jannuzzi AT, Ozcagli E, Alpertunga B. Celastrol pretreatment as a therapeutic option against cisplatin-induced nephrotoxicity. Toxicol Res (Camb) 2019; 8:723-730. [PMID: 31588349 PMCID: PMC6762010 DOI: 10.1039/c9tx00141g] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Accepted: 07/29/2019] [Indexed: 12/31/2022] Open
Abstract
Celastrol is a natural bioactive compound extracted from the medicinal plant Tripterygium wilfordii Hook F. It exhibits immunosuppressive, anti-inflammatory, and antioxidant activities. Cisplatin is a commonly used chemotherapeutic drug in the treatment of a wide range of tumors. Although very effective therapeutically, it can cause nephrotoxicity leading to dose reduction or discontinuation of treatment. This study aims to clarify the therapeutic potential of celastrol in cisplatin-induced nephrotoxicity. The possible protective effects of celastrol pretreatment against cisplatin-induced oxidative stress and genotoxicity were investigated. A rat kidney epithelial cell line NRK-52E was pretreated with the desired concentrations of celastrol (200 nM, 100 nM, and 50 nM) for 24 h. The cells were treated with 50 μM cisplatin for a further 24 h to see whether cisplatin caused the same or less toxicity compared to the vehicle control group. Alkaline comet assay was performed for genotoxicity assessment. Genotoxicity evaluation revealed that celastrol caused a statistically significant reduction in DNA damage. Oxidative stress parameters were evaluated by measuring the glutathione (GSH) and protein carbonyl (PC) levels and also by measuring the enzyme activities of glutathione peroxidase (GPx), glutathione reductase (GR), catalase (CAT) and superoxide dismutase (SOD) enzymes. Celastrol pretreatment increased the GSH content of the cells and ameliorated the protein carbonylation level. Likewise, celastrol pretreatment improved the GR and CAT activities. However, no significant difference was observed in GPx and SOD activities. In the light of these findings, celastrol treatment could be a therapeutic option to reduce cisplatin-induced nephrotoxicity. Further studies are needed for the clarification of its therapeutic potential.
Collapse
Affiliation(s)
- Tugce Boran
- Istanbul University , Faculty of Pharmacy , Department of Pharmaceutical Toxicology , 34116 , Beyazıt , Istanbul , Turkey . ; ; Tel: +902124400000
| | - Aysenur Gunaydin
- Istanbul University , Faculty of Pharmacy , Department of Pharmaceutical Toxicology , 34116 , Beyazıt , Istanbul , Turkey . ; ; Tel: +902124400000
- Bezmialem Vakif University , Faculty of Pharmacy , Department of Pharmaceutical Toxicology , Vatan Street , 34093 , Fatih , Istanbul , Turkey
| | - Ayse Tarbin Jannuzzi
- Istanbul University , Faculty of Pharmacy , Department of Pharmaceutical Toxicology , 34116 , Beyazıt , Istanbul , Turkey . ; ; Tel: +902124400000
| | - Eren Ozcagli
- Istanbul University , Faculty of Pharmacy , Department of Pharmaceutical Toxicology , 34116 , Beyazıt , Istanbul , Turkey . ; ; Tel: +902124400000
| | - Buket Alpertunga
- Istanbul University , Faculty of Pharmacy , Department of Pharmaceutical Toxicology , 34116 , Beyazıt , Istanbul , Turkey . ; ; Tel: +902124400000
| |
Collapse
|
16
|
Lv H, Jiang L, Zhu M, Li Y, Luo M, Jiang P, Tong S, Zhang H, Yan J. The genus Tripterygium: A phytochemistry and pharmacological review. Fitoterapia 2019; 137:104190. [DOI: 10.1016/j.fitote.2019.104190] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 05/29/2019] [Accepted: 05/30/2019] [Indexed: 12/15/2022]
|
17
|
Ng SW, Chan Y, Chellappan DK, Madheswaran T, Zeeshan F, Chan YL, Collet T, Gupta G, Oliver BG, Wark P, Hansbro N, Hsu A, Hansbro PM, Dua K, Panneerselvam J. Molecular modulators of celastrol as the keystones for its diverse pharmacological activities. Biomed Pharmacother 2019; 109:1785-1792. [DOI: 10.1016/j.biopha.2018.11.051] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2018] [Revised: 11/10/2018] [Accepted: 11/10/2018] [Indexed: 12/30/2022] Open
|
18
|
Abu Bakar MH, Azmi MN, Shariff KA, Tan JS. Withaferin A Protects Against High-Fat Diet-Induced Obesity Via Attenuation of Oxidative Stress, Inflammation, and Insulin Resistance. Appl Biochem Biotechnol 2018; 188:241-259. [PMID: 30417321 DOI: 10.1007/s12010-018-2920-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Accepted: 11/05/2018] [Indexed: 01/07/2023]
Abstract
Withaferin A (WA), a bioactive constituent derived from Withania somnifera plant, has been shown to exhibit many qualifying properties in attenuating several metabolic diseases. The current investigation sought to elucidate the protective mechanisms of WA (1.25 mg/kg/day) on pre-existing obese mice mediated by high-fat diet (HFD) for 12 weeks. Following dietary administration of WA, significant metabolic improvements in hepatic insulin sensitivity, adipocytokines with enhanced glucose tolerance were observed. The hepatic oxidative functions of obese mice treated with WA were improved via augmented antioxidant enzyme activities. The levels of serum pro-inflammatory cytokines and hepatic mRNA expressions of toll-like receptor (TLR4), nuclear factor κB (NF-κB), tumor necrosis factor-α (TNF-α), chemokine (C-C motif) ligand-receptor, and cyclooxygenase 2 (COX2) in HFD-induced obese mice were reduced. Mechanistically, WA increased hepatic mRNA expression of peroxisome proliferator-activated receptors (PPARs), cluster of differentiation 36 (CD36), fatty acid synthase (FAS), carnitine palmitoyltransferase 1 (CPT1), glucokinase (GCK), phosphofructokinase (PFK), and phosphoenolpyruvate carboxykinase (PCK1) that were associated with enhanced lipid and glucose metabolism. Taken together, these results indicate that WA exhibits protective effects against HFD-induced obesity through attenuation of hepatic inflammation, oxidative stress, and insulin resistance in mice.
Collapse
Affiliation(s)
- Mohamad Hafizi Abu Bakar
- Bioprocess Technology Division, School of Industrial Technology, Universiti Sains Malaysia, 11800, Gelugor, Penang, Malaysia.
| | - Mohamad Nurul Azmi
- School of Chemical Sciences, Universiti Sains Malaysia, 11800, Gelugor, Penang, Malaysia
| | - Khairul Anuar Shariff
- School of Materials & Mineral Resources Engineering, Universiti Sains Malaysia, 14300, Nibong Tebal, Penang, Malaysia
| | - Joo Shun Tan
- Bioprocess Technology Division, School of Industrial Technology, Universiti Sains Malaysia, 11800, Gelugor, Penang, Malaysia
| |
Collapse
|
19
|
Reduced expression of Twist 1 is protective against insulin resistance of adipocytes and involves mitochondrial dysfunction. Sci Rep 2018; 8:12590. [PMID: 30135600 PMCID: PMC6105588 DOI: 10.1038/s41598-018-30820-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Accepted: 08/07/2018] [Indexed: 01/06/2023] Open
Abstract
Insulin resistance (IR) has become a global epidemic that represents a serious hazard to public health. However, the precise mechanisms modulating IR have not been fully elucidated. The present study aimed to investigate the role of transcriptional factor Twist 1 in adipocyte IR and to further explore the molecular mechanism. An in vitro IR model based on cultured 3T3-L1 adipocytes was established under high glucose/insulin stimulation and an in vivo IR model in C57/BL6J mice induced by a high fat diet (HFD) was also developed. Lentivirus targeting Twist 1 silencing was introduced. The relationships between Twist 1 expression and IR state, mitochondrial dysfunction and the downstream insulin signaling pathway were assayed. Our results firstly showed the elevation of Twist 1 in IR adipocytes, and Twist 1 silencing attenuated IR. Then mitochondrial ultra-structural damage, elevated ROS, decreased MMP and ATP, and changes in mitochondrial biosynthesis-related genes in IR group indicated mitochondrial dysfunction. Further, the downstream IRS/PI3K/AKT/GluT4 pathway was showed involved in Twist 1-mediated IR. In total, we provide evidence of a protective role of Twist 1 silencing in relieving the IR state of adipocytes. Mitochondrial dysfunction and the downstream IRS/PI3K/AKT/GluT4 pathway were involved in this Twist 1-mediated IR.
Collapse
|
20
|
Abu Bakar MH, Sarmidi MR. Association of cultured myotubes and fasting plasma metabolite profiles with mitochondrial dysfunction in type 2 diabetes subjects. MOLECULAR BIOSYSTEMS 2018; 13:1838-1853. [PMID: 28726959 DOI: 10.1039/c7mb00333a] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Accumulating evidence implicates mitochondrial dysfunction-induced insulin resistance in skeletal muscle as the root cause for the greatest hallmarks of type 2 diabetes (T2D). However, the identification of specific metabolite-based markers linked to mitochondrial dysfunction in T2D has not been adequately addressed. Therefore, we sought to identify the markers-based metabolomics for mitochondrial dysfunction associated with T2D. First, a cellular disease model was established using human myotubes treated with antimycin A, an oxidative phosphorylation inhibitor. Non-targeted metabolomic profiling of intracellular-defined metabolites on the cultured myotubes with mitochondrial dysfunction was then determined. Further, a targeted MS-based metabolic profiling of fasting blood plasma from normal (n = 32) and T2D (n = 37) subjects in a cross-sectional study was verified. Multinomial logical regression analyses for defining the top 5% of the metabolites within a 95% group were employed to determine the differentiating metabolites. The myotubes with mitochondrial dysfunction exhibited insulin resistance, oxidative stress and inflammation with impaired insulin signalling activities. Four metabolic pathways were found to be strongly associated with mitochondrial dysfunction in the cultured myotubes. Metabolites derived from these pathways were validated in an independent pilot investigation of the fasting blood plasma of healthy and diseased subjects. Targeted metabolic analysis of the fasting blood plasma with specific baseline adjustment revealed 245 significant features based on orthogonal partial least square discriminant analysis (PLS-DA) with a p-value < 0.05. Among these features, 20 significant metabolites comprised primarily of branched chain and aromatic amino acids, glutamine, aminobutyric acid, hydroxyisobutyric acid, pyroglutamic acid, acylcarnitine species (acetylcarnitine, propionylcarnitine, dodecenoylcarnitine, tetradecenoylcarnitine hexadecadienoylcarnitine and oleylcarnitine), free fatty acids (palmitate, arachidonate, stearate and linoleate) and sphingomyelin (d18:2/16:0) were identified as predictive markers for mitochondrial dysfunction in T2D subjects. The current study illustrates how cellular metabolites provide potential signatures associated with the biochemical changes in the dysregulated body metabolism of diseased subjects. Our finding yields additional insights into the identification of robust biomarkers for T2D associated with mitochondrial dysfunction in cultured myotubes.
Collapse
Affiliation(s)
- Mohamad Hafizi Abu Bakar
- Bioprocess Technology Division, School of Industrial Technology, Universiti Sains Malaysia, 11800 Gelugor, Penang, Malaysia.
| | | |
Collapse
|
21
|
Jannuzzi AT, Kara M, Alpertunga B. Celastrol ameliorates acetaminophen-induced oxidative stress and cytotoxicity in HepG2 cells. Hum Exp Toxicol 2017; 37:742-751. [DOI: 10.1177/0960327117734622] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Acetaminophen (APAP) is the most commonly used analgesic and antipyretic drug in the world. However, hepatotoxicity caused by APAP overdose is the most frequent cause of acute liver failure worldwide and oxidative stress involved in the pathogenesis of APAP hepatotoxicity. Celastrol is a natural triterpenoid derived from Tripterygium wilfordii Hook F. that exhibits antioxidant, anti-inflammatory, and antitumor activities. In this study, we aimed to investigate the potential ameliorative effects of celastrol against APAP-induced cytotoxicity and oxidative stress. Human hepatocellular carcinoma cells (HepG2) were incubated with 20 mM of APAP for 24 h and posttreated with 50 nM, 100 nM, or 200 nM of celastrol for a further 24 h. The methylthiazolyldiphenyl-tetrazolium bromide, lactate dehydrogenase, and neutral red uptake assays showed celastrol posttreatments recovered cell viability and cell membrane integrity in a concentration-dependent manner. Celastrol posttreatments exerted a significant increase in the glutathione content and a decrease in the malondialdehyde and protein carbonylation levels. Also, celastrol posttreatments attenuated the APAP-induced oxidative stress by raising glutathione peroxidase, glutathione reductase, and catalase activities. However, superoxide dismutase activity did not change. In conclusion, celastrol treatment may improve cell viability and increase cellular antioxidant defense in HepG2 cells. These results suggest that celastrol may have the potential to ameliorate the APAP-induced oxidative stress and cytotoxicity.
Collapse
Affiliation(s)
- AT Jannuzzi
- Department of Pharmaceutical Toxicology, Faculty of Pharmacy, Istanbul University, Beyazit, Istanbul, Turkey
| | - M Kara
- Department of Pharmaceutical Toxicology, Faculty of Pharmacy, Istanbul University, Beyazit, Istanbul, Turkey
| | - B Alpertunga
- Department of Pharmaceutical Toxicology, Faculty of Pharmacy, Istanbul University, Beyazit, Istanbul, Turkey
| |
Collapse
|
22
|
Improvement of mitochondrial function by celastrol in palmitate-treated C2C12 myotubes via activation of PI3K-Akt signaling pathway. Biomed Pharmacother 2017; 93:903-912. [PMID: 28715871 DOI: 10.1016/j.biopha.2017.07.021] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Revised: 07/04/2017] [Accepted: 07/05/2017] [Indexed: 12/16/2022] Open
Abstract
Compelling evidences posited that high level of saturated fatty acid gives rise to mitochondrial dysfunction and inflammation in the development of insulin resistance in skeletal muscle. Celastrol is a pentacyclic triterpenoid derived from the root extracts of Tripterygium wilfordii that possesses potent anti-inflammatory properties in a number of animal models with metabolic diseases. However, the cellular mechanistic action of celastrol in alleviating obesity-induced insulin resistance in skeletal muscle remains largely unknown. Therefore, the present investigation evaluated the attributive properties of celastrol at different concentrations (10, 20, 30 and 40nM) on insulin resistance in C2C12 myotubes evoked by palmitate. We demonstrated that celastrol improved mitochondrial functions through significant enhancement of intracellular ATP content, mitochondrial membrane potential, citrate synthase activity and decrease of mitochondrial superoxide productions. Meanwhile, augmented mitochondrial DNA (mtDNA) content with suppressed DNA oxidative damage were observed following celastrol treatment. Celastrol significantly enhanced fatty acid oxidation rate and increased the level of tricarboxylic acid (TCA) cycle intermediates in palmitate-treated cells. Further analysis revealed that the improvement of glucose uptake activity in palmitate-loaded myotubes was partly mediated by celastrol via activation of PI3K-Akt insulin signaling pathway. Collectively, these findings provided evidence for the first time that the protection from palmitate-mediated insulin resistance in C2C12 myotubes by celastrol is likely associated with the improvement of mitochondrial functions-related metabolic activities.
Collapse
|
23
|
Cascão R, Fonseca JE, Moita LF. Celastrol: A Spectrum of Treatment Opportunities in Chronic Diseases. Front Med (Lausanne) 2017; 4:69. [PMID: 28664158 PMCID: PMC5471334 DOI: 10.3389/fmed.2017.00069] [Citation(s) in RCA: 155] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Accepted: 05/19/2017] [Indexed: 01/02/2023] Open
Abstract
The identification of new bioactive compounds derived from medicinal plants with significant therapeutic properties has attracted considerable interest in recent years. Such is the case of the Tripterygium wilfordii (TW), an herb used in Chinese medicine. Clinical trials performed so far using its root extracts have shown impressive therapeutic properties but also revealed substantial gastrointestinal side effects. The most promising bioactive compound obtained from TW is celastrol. During the last decade, an increasing number of studies were published highlighting the medicinal usefulness of celastrol in diverse clinical areas. Here we systematically review the mechanism of action and the therapeutic properties of celastrol in inflammatory diseases, namely, rheumatoid arthritis, systemic lupus erythematosus, inflammatory bowel diseases, osteoarthritis and allergy, as well as in cancer, neurodegenerative disorders and other diseases, such as diabetes, obesity, atherosclerosis, and hearing loss. We will also focus in the toxicological profile and limitations of celastrol formulation, namely, solubility, bioavailability, and dosage issues that still limit its further clinical application and usefulness.
Collapse
Affiliation(s)
- Rita Cascão
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - João E Fonseca
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal.,Rheumatology Department, Centro Hospitalar de Lisboa Norte, EPE, Hospital de Santa Maria, Lisbon Academic Medical Centre, Lisbon, Portugal
| | - Luis F Moita
- Instituto Gulbenkian de Ciência, Oeiras, Portugal
| |
Collapse
|
24
|
Liu Z, Qiao Q, Sun Y, Chen Y, Ren B, Liu X. Sesamol ameliorates diet-induced obesity in C57BL/6J mice and suppresses adipogenesis in 3T3-L1 cells via regulating mitochondria-lipid metabolism. Mol Nutr Food Res 2017; 61. [DOI: 10.1002/mnfr.201600717] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Revised: 11/14/2016] [Accepted: 12/19/2016] [Indexed: 12/25/2022]
Affiliation(s)
- Zhigang Liu
- Laboratory of Functional Chemistry and Nutrition of Food; College of Food Science and Engineering; Northwest A&F University; Yangling China
| | - Qinglian Qiao
- Laboratory of Functional Chemistry and Nutrition of Food; College of Food Science and Engineering; Northwest A&F University; Yangling China
| | - Yali Sun
- Laboratory of Functional Chemistry and Nutrition of Food; College of Food Science and Engineering; Northwest A&F University; Yangling China
| | - Yuwei Chen
- Laboratory of Functional Chemistry and Nutrition of Food; College of Food Science and Engineering; Northwest A&F University; Yangling China
| | - Bo Ren
- Laboratory of Functional Chemistry and Nutrition of Food; College of Food Science and Engineering; Northwest A&F University; Yangling China
| | - Xuebo Liu
- Laboratory of Functional Chemistry and Nutrition of Food; College of Food Science and Engineering; Northwest A&F University; Yangling China
| |
Collapse
|
25
|
Abu Bakar MH, Sarmidi MR, Tan JS, Mohamad Rosdi MN. Celastrol attenuates mitochondrial dysfunction and inflammation in palmitate-mediated insulin resistance in C3A hepatocytes. Eur J Pharmacol 2017; 799:73-83. [PMID: 28161417 DOI: 10.1016/j.ejphar.2017.01.043] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Revised: 01/25/2017] [Accepted: 01/30/2017] [Indexed: 12/27/2022]
Abstract
Accumulating evidence indicates that mitochondrial dysfunction-induced inflammation is among the convergence points for the greatest hallmarks of hepatic insulin resistance. Celastrol, an anti-inflammatory compound from the root of Tripterygium Wilfordii has been reported to mitigate insulin resistance and inflammation in animal disease models. Nevertheless, the specific mechanistic actions of celastrol in modulating such improvements at the cellular level remain obscure. The present study sought to explore the mechanistic roles of celastrol upon insulin resistance induced by palmitate in C3A human hepatocytes. The hepatocytes exposed to palmitate (0.75mM) for 48h exhibited reduced both basal and insulin-stimulated glucose uptake, mitochondrial dysfunction, leading to increased mitochondrial oxidative stress with diminished fatty acid oxidation. Elevated expressions of nuclear factor-kappa B p65 (NF-κB p65), c-Jun NH(2)-terminal kinase (JNK) signaling pathways and the amplified release of pro-inflammatory cytokines including IL-8, IL-6, TNF-α and CRP were observed following palmitate treatment. Consistently, palmitate reduced and augmented phosphorylated Tyrosine-612 and Serine-307 of insulin receptor substrate-1 (IRS-1) proteins, respectively in hepatocytes. However, celastrol at the optimum concentration of 30nM was able to reverse these deleterious occasions and protected the cells from mitochondrial dysfunction and insulin resistance. Importantly, we presented evidence for the first time that celastrol efficiently prevented palmitate-induced insulin resistance in hepatocytes at least, via improved mitochondrial functions and insulin signaling pathways. In summary, the present investigation underlines a conceivable mechanism to elucidate the cytoprotective potential of celastrol in attenuating mitochondrial dysfunction and inflammation against the development of hepatic insulin resistance.
Collapse
Affiliation(s)
- Mohamad Hafizi Abu Bakar
- Bioprocess Technology Division, School of Industrial Technology, Universiti Sains Malaysia, 11800 Gelugor, Penang, Malaysia.
| | - Mohamad Roji Sarmidi
- Institute of Bioproduct Development, Universiti Teknologi Malaysia, 81310 Johor Bahru, Johor, Malaysia; Innovation Centre in Agritechnology for Advanced Bioprocessing (ICA), Universiti Teknologi Malaysia, 81310, Johor Bahru, Johor, Malaysia
| | - Joo Shun Tan
- Bioprocess Technology Division, School of Industrial Technology, Universiti Sains Malaysia, 11800 Gelugor, Penang, Malaysia
| | - Mohamad Norisham Mohamad Rosdi
- Department of Bioprocess and Polymer Engineering, Faculty of Chemical and Energy Engineering, Universiti Teknologi Malaysia, 81310 Johor Bahru, Johor, Malaysia
| |
Collapse
|
26
|
Sun R, Wu Y, Hou W, Sun Z, Wang Y, Wei H, Mo W, Yu M. Bromodomain-containing protein 2 induces insulin resistance via the mTOR/Akt signaling pathway and an inflammatory response in adipose tissue. Cell Signal 2016; 30:92-103. [PMID: 27865874 DOI: 10.1016/j.cellsig.2016.11.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2016] [Revised: 11/10/2016] [Accepted: 11/15/2016] [Indexed: 01/02/2023]
Abstract
Insulin resistance is a major metabolic abnormality in a large majority of patients with type II diabetes. Bromodomain-containing protein 2 (Brd2), a transcriptional co-activator/co-repressor with switch mating type/sucrose non-fermenting (SWI/SNF)-like functions that regulates chromatin, suppresses adipocyte differentiation and regulates pancreatic β-cell biology. However, the effects of Brd2 on insulin resistance remain unknown. Here, overexpression of Brd2 in white adipose tissue of wild-type (WT) mice led to insulin resistance. Brd2 overexpression induced the expression of nuclear Factor-κΒ (NF-κΒ) target genes, mainly involving proinflammatory and chemotactic factors, in adipocytes. Furthermore, it decreased the expression of DEP domain containing mTOR-interacting protein (Deptor) to enhance mechanistic target of rapamycin (mTOR) signaling, thus blocking insulin signaling. Collectively, these results provided evidence for a novel role of Brd2 in chronic inflammation and insulin resistance, suggesting its potential in improving insulin resistance and treating metabolic disorders.
Collapse
Affiliation(s)
- Ruixin Sun
- The Key Laboratory of Metabolism and Molecular Medicine, The Ministry of Education, Department of Biochemistry and Molecular Biology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Yi Wu
- The Key Laboratory of Metabolism and Molecular Medicine, The Ministry of Education, Department of Biochemistry and Molecular Biology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Weihua Hou
- The Key Laboratory of Metabolism and Molecular Medicine, The Ministry of Education, Department of Biochemistry and Molecular Biology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Zujun Sun
- The Key Laboratory of Metabolism and Molecular Medicine, The Ministry of Education, Department of Biochemistry and Molecular Biology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Yuxiong Wang
- The Key Laboratory of Metabolism and Molecular Medicine, The Ministry of Education, Department of Biochemistry and Molecular Biology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Huanhuan Wei
- The Key Laboratory of Metabolism and Molecular Medicine, The Ministry of Education, Department of Biochemistry and Molecular Biology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Wei Mo
- The Key Laboratory of Metabolism and Molecular Medicine, The Ministry of Education, Department of Biochemistry and Molecular Biology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Min Yu
- The Key Laboratory of Metabolism and Molecular Medicine, The Ministry of Education, Department of Biochemistry and Molecular Biology, Shanghai Medical College, Fudan University, Shanghai 200032, China.
| |
Collapse
|
27
|
Celastrol Protects against Antimycin A-Induced Insulin Resistance in Human Skeletal Muscle Cells. Molecules 2015; 20:8242-69. [PMID: 25961164 PMCID: PMC6272652 DOI: 10.3390/molecules20058242] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Revised: 04/29/2015] [Accepted: 05/04/2015] [Indexed: 01/04/2023] Open
Abstract
Mitochondrial dysfunction and inflammation are widely accepted as key hallmarks of obesity-induced skeletal muscle insulin resistance. The aim of the present study was to evaluate the functional roles of an anti-inflammatory compound, celastrol, in mitochondrial dysfunction and insulin resistance induced by antimycin A (AMA) in human skeletal muscle cells. We found that celastrol treatment improved insulin-stimulated glucose uptake activity of AMA-treated cells, apparently via PI3K/Akt pathways, with significant enhancement of mitochondrial activities. Furthermore, celastrol prevented increased levels of cellular oxidative damage where the production of several pro-inflammatory cytokines in cultures cells was greatly reduced. Celastrol significantly increased protein phosphorylation of insulin signaling cascades with amplified expression of AMPK protein and attenuated NF-κB and PKC θ activation in human skeletal muscle treated with AMA. The improvement of insulin signaling pathways by celastrol was also accompanied by augmented GLUT4 protein expression. Taken together, these results suggest that celastrol may be advocated for use as a potential therapeutic molecule to protect against mitochondrial dysfunction-induced insulin resistance in human skeletal muscle cells.
Collapse
|
28
|
Abu Bakar MH, Sarmidi MR, Cheng KK, Ali Khan A, Suan CL, Zaman Huri H, Yaakob H. Metabolomics – the complementary field in systems biology: a review on obesity and type 2 diabetes. MOLECULAR BIOSYSTEMS 2015; 11:1742-74. [DOI: 10.1039/c5mb00158g] [Citation(s) in RCA: 82] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
This paper highlights the metabolomic roles in systems biology towards the elucidation of metabolic mechanisms in obesity and type 2 diabetes.
Collapse
Affiliation(s)
- Mohamad Hafizi Abu Bakar
- Department of Bioprocess Engineering
- Faculty of Chemical Engineering
- Universiti Teknologi Malaysia
- 81310 Johor Bahru
- Malaysia
| | - Mohamad Roji Sarmidi
- Institute of Bioproduct Development
- Universiti Teknologi Malaysia
- 81310 Johor Bahru
- Malaysia
- Innovation Centre in Agritechnology for Advanced Bioprocessing (ICA)
| | - Kian-Kai Cheng
- Department of Bioprocess Engineering
- Faculty of Chemical Engineering
- Universiti Teknologi Malaysia
- 81310 Johor Bahru
- Malaysia
| | - Abid Ali Khan
- Institute of Bioproduct Development
- Universiti Teknologi Malaysia
- 81310 Johor Bahru
- Malaysia
- Department of Biosciences
| | - Chua Lee Suan
- Institute of Bioproduct Development
- Universiti Teknologi Malaysia
- 81310 Johor Bahru
- Malaysia
| | - Hasniza Zaman Huri
- Department of Pharmacy
- Faculty of Medicine
- University of Malaya
- 50603 Kuala Lumpur
- Malaysia
| | - Harisun Yaakob
- Institute of Bioproduct Development
- Universiti Teknologi Malaysia
- 81310 Johor Bahru
- Malaysia
| |
Collapse
|