1
|
Chen Y, Shu C, Yan Z, Zhang S, Zhang W, Zhao J, Wang A, Li J, Zeng Y, Zhu J, Huang JA, Liu Z. Liensinine overcomes EGFR-TKI resistance in lung adenocarcinoma through DRP1-mediated autophagy. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 140:156593. [PMID: 40054177 DOI: 10.1016/j.phymed.2025.156593] [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: 12/07/2024] [Revised: 02/17/2025] [Accepted: 02/28/2025] [Indexed: 03/25/2025]
Abstract
INTRODUCTION Persistent upregulation of autophagy contributes to tumour cells' resistance to EGFR-TKI therapy, and hence, inhibiting autophagy could be a valuable strategy for overcoming such resistance. OBJECTIVES This study investigated the effects of liensinine in EGFR-TKI resistant lung adenocarcinoma (LUAD) and to explore the underlying mechanism. METHODS CCK-8 assay, colony formation, EdU assay and apoptosis assays were conducted for investigating the effect of EGFR-TKI and liensinine combination treatment in LUAD. Furthermore, autophagic flux were detected by western blot, fluorescence assays and TEM. In addition, by employing a DARTS approach, a CETSA assay, and SPR analysis, we identified DRP1 as a target of liensinine. Finally, by establishing a xenograft model of the disease, the impact of combination treatment in vivo was assessed. RESULT In vitro and in vivo experiments revealed that the novel autophagy inhibitor liensinine enhanced the sensitivity of LUAD to EGFR-TKIs. This effect was achieved by inhibiting autophagic flux. We then examined whether liensinine inhibits autophagic flux through the impairment of autophagosome and autolysosome degradation. Furthermore, we identified DRP1 as a target of liensinine. The activation of DRP1 by liensinine through dephosphorylation at Ser637 promotes the accumulation of autophagosomes and autolysosomes while simultaneously blocking autophagic flux, thereby enhancing the cancer cell-killing effects of EGFR-TKIs. CONCLUSIONS Our study validated the efficacy of liensinine in overcoming EGFR-TKI resistance and elucidated the mechanism underlying liensinine's inhibition of autophagy.
Collapse
Affiliation(s)
- Yuling Chen
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China; Institute of Respiratory Diseases, Soochow University, Suzhou, 215006, China
| | - Chenying Shu
- Institute of Respiratory Diseases, Soochow University, Suzhou, 215006, China; Department of Pharmacy, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Zhaowei Yan
- Department of Pharmacy, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Saiqun Zhang
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China; Institute of Respiratory Diseases, Soochow University, Suzhou, 215006, China
| | - Weijie Zhang
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China; Institute of Respiratory Diseases, Soochow University, Suzhou, 215006, China
| | - Jian Zhao
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China; Institute of Respiratory Diseases, Soochow University, Suzhou, 215006, China
| | - Anqi Wang
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China; Institute of Respiratory Diseases, Soochow University, Suzhou, 215006, China
| | - Jianjun Li
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China; Institute of Respiratory Diseases, Soochow University, Suzhou, 215006, China
| | - Yuanyuan Zeng
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China; Institute of Respiratory Diseases, Soochow University, Suzhou, 215006, China; Suzhou Key Laboratory for Respiratory Diseases, Suzhou 215006, China
| | - Jianjie Zhu
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China; Institute of Respiratory Diseases, Soochow University, Suzhou, 215006, China; Suzhou Key Laboratory for Respiratory Diseases, Suzhou 215006, China
| | - Jian-An Huang
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China; Institute of Respiratory Diseases, Soochow University, Suzhou, 215006, China; Suzhou Key Laboratory for Respiratory Diseases, Suzhou 215006, China.
| | - Zeyi Liu
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China; Institute of Respiratory Diseases, Soochow University, Suzhou, 215006, China; Suzhou Key Laboratory for Respiratory Diseases, Suzhou 215006, China.
| |
Collapse
|
2
|
Tang X, Shi Y, Chen Y, Sun Z, Wang L, Tang P, Cui H, Zhao W, Xu W, Kopylov P, Shchekochikhin D, Afina B, Han W, Liu X, Zhang Y. Tetrahydroberberrubine exhibits preventive effect on obesity by activating PGC1α-mediated thermogenesis in white and brown adipose tissue. Biochem Pharmacol 2024; 226:116381. [PMID: 38909786 DOI: 10.1016/j.bcp.2024.116381] [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: 12/05/2023] [Revised: 06/03/2024] [Accepted: 06/20/2024] [Indexed: 06/25/2024]
Abstract
The escalating prevalence of obesity presents formidable challenges, necessitating the development of effective therapeutic strategies. In this study, we aimed to elucidate the preventive effects on obesity of tetrahydroberberrubine (THBru), a derivative of berberine (BBR) and to unravel its underlying mechanism. Using an obese mouse model induced by a high-fat diet (HFD), THBru was found to markedly ameliorate obesity, as evidenced by reduced body weight, decreased Lee's index, diminished fat mass in epididymal white adipose tissue (WAT) and brown adipose tissue (BAT), alongside improved dyslipidemia. Notably, at the same dose, THBru exhibited superior efficacy compared to BBR. RNA-sequencing and gene set enrichment analysis indicated THBru activated thermogenesis, which was further confirmed in WAT, BAT, and 3T3-L1 cells. Bioinformatics analysis of RNA-sequencing data revealed the candidate gene Pgc1α, a key regulator involved in thermogenesis. Moreover, THBru was demonstrated to elevate the expression of PGC1α by stabilizing its mRNA in WAT, BAT and 3T3-L1 cells. Furthermore, PGC1α knockdown blocked the pro-thermogenic and anti-obesity action of THBru both in vivo and in vitro. This study unravels the preventive effects of THBru on obesity through the activation of PGC1α-mediated thermogenesis, thereby delineating its potential therapeutic implications for obesity and associated disorders.
Collapse
Affiliation(s)
- Xueqing Tang
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology, College of Pharmacy, and Department of Cardiology, the Second Affiliated Hospital, Harbin Medical University, Harbin, China; State Key Laboratory -Province Key Laboratories of Biomedicine-Pharmaceutics of China, and Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin, China; Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin, China
| | - Yang Shi
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology, College of Pharmacy, and Department of Cardiology, the Second Affiliated Hospital, Harbin Medical University, Harbin, China; State Key Laboratory -Province Key Laboratories of Biomedicine-Pharmaceutics of China, and Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin, China; Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin, China
| | - Yongchao Chen
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology, College of Pharmacy, and Department of Cardiology, the Second Affiliated Hospital, Harbin Medical University, Harbin, China; State Key Laboratory -Province Key Laboratories of Biomedicine-Pharmaceutics of China, and Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin, China; Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin, China
| | - Zeqi Sun
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology, College of Pharmacy, and Department of Cardiology, the Second Affiliated Hospital, Harbin Medical University, Harbin, China; State Key Laboratory -Province Key Laboratories of Biomedicine-Pharmaceutics of China, and Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin, China; Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin, China
| | - Lei Wang
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology, College of Pharmacy, and Department of Cardiology, the Second Affiliated Hospital, Harbin Medical University, Harbin, China; State Key Laboratory -Province Key Laboratories of Biomedicine-Pharmaceutics of China, and Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin, China; Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin, China
| | - Pingping Tang
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology, College of Pharmacy, and Department of Cardiology, the Second Affiliated Hospital, Harbin Medical University, Harbin, China; State Key Laboratory -Province Key Laboratories of Biomedicine-Pharmaceutics of China, and Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin, China; Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin, China
| | - Hao Cui
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology, College of Pharmacy, and Department of Cardiology, the Second Affiliated Hospital, Harbin Medical University, Harbin, China; State Key Laboratory -Province Key Laboratories of Biomedicine-Pharmaceutics of China, and Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin, China; Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin, China
| | - Wenjie Zhao
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology, College of Pharmacy, and Department of Cardiology, the Second Affiliated Hospital, Harbin Medical University, Harbin, China; State Key Laboratory -Province Key Laboratories of Biomedicine-Pharmaceutics of China, and Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin, China; Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin, China
| | - Wanqing Xu
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology, College of Pharmacy, and Department of Cardiology, the Second Affiliated Hospital, Harbin Medical University, Harbin, China; State Key Laboratory -Province Key Laboratories of Biomedicine-Pharmaceutics of China, and Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin, China; Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin, China
| | - Philipp Kopylov
- Department of Preventive and Emergency Cardiology, Sechenov First Moscow State Medical University, Moscow, Russian Federation
| | - Dmitry Shchekochikhin
- Department of Preventive and Emergency Cardiology, Sechenov First Moscow State Medical University, Moscow, Russian Federation
| | - Bestavashvili Afina
- Department of Cardiology, Functional and Ultrasound Diagnostics, N.V. Sklifosofsky, I. M. Sechenov First Moscow State Medical University, Moscow, Russian Federation
| | - Weina Han
- Department of Medicinal Chemistry and Natural Medicine Chemistry, College of Pharmacy, Harbin Medical University, Harbin, China
| | - Xin Liu
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology, College of Pharmacy, and Department of Cardiology, the Second Affiliated Hospital, Harbin Medical University, Harbin, China; State Key Laboratory -Province Key Laboratories of Biomedicine-Pharmaceutics of China, and Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin, China; Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin, China.
| | - Yong Zhang
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology, College of Pharmacy, and Department of Cardiology, the Second Affiliated Hospital, Harbin Medical University, Harbin, China; State Key Laboratory -Province Key Laboratories of Biomedicine-Pharmaceutics of China, and Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin, China; Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin, China.
| |
Collapse
|
3
|
Zhou X, Wang H, Huang M, Chen J, Chen J, Cheng H, Ye X, Wang W, Liu D. Role of bitter contributors and bitter taste receptors: a comprehensive review of their sources, functions and future development. FOOD SCIENCE AND HUMAN WELLNESS 2024; 13:1806-1824. [DOI: 10.26599/fshw.2022.9250151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
4
|
Zhang X, Yuan S, Fan H, Zhang W, Zhang H. Liensinine alleviates sepsis-induced acute liver injury by inhibiting the NF-κB and MAPK pathways in an Nrf2-dependent manner. Chem Biol Interact 2024; 396:111030. [PMID: 38692452 DOI: 10.1016/j.cbi.2024.111030] [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: 01/13/2024] [Revised: 04/24/2024] [Accepted: 04/29/2024] [Indexed: 05/03/2024]
Abstract
Sepsis remains a serious public health issue that needs to be addressed globally. Severe liver injury caused by sepsis increases the risk of death in patients with sepsis. Liensinine (Lie) is one of the primary active components in Plumula nelumbinis and has anti-inflammatory and antioxidant effects. Nevertheless, the effects of Lie on septic liver injury are unclear. This research investigated the protective effect of Lie (10, 20 and 40 mg/kg) on liver damage via intraperitoneal administration of LPS (10 mg/kg) to C57BL/6 mice. Lie was given through intraperitoneal injection once a day for five days. Mice were treated with LPS intraperitoneally for 6 h at 1 h after Lie administration on the last day. The results suggested that Lie could decrease AST and ALT levels in serum, ameliorate histopathological changes and inhibit cell apoptosis in mice with LPS-induced septic liver injury. In addition, Lie inhibited increases in the mRNA levels of TNF-α, IL-1β, iNOS and IL-6. Lie also increased the mRNA level of IL-10. Lie reduced the content of MDA, a marker of lipid peroxidation, and increased the activity of the antioxidant enzymes GSH-Px, CAT and SOD. Our results also showed that Lie could suppress the LPS-activated MAPK and NF-κB pathways and trigger the Nrf2 signaling pathway both in vitro and in vivo. Additionally, an Nrf2 inhibitor (ML385) weakened the suppressive effect of Lie on the MAPK and NF-κB pathways. Our results demonstrated that the suppressive effect of Lie on the MAPK and NF-κB pathways was partially reliant on activation of the Nrf2 pathway. In summary, these results indicate that Lie can improve inflammation and oxidative stress by activating Nrf2, which is a prospective therapeutic drug for alleviating septic liver injury.
Collapse
Affiliation(s)
- Xiao Zhang
- Department of Vascular Surgery, The First People's Hospital of Lianyungang, Lianyungang, 222005, China; Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Silong Yuan
- Department of Vascular Surgery, The First People's Hospital of Lianyungang, Lianyungang, 222005, China
| | - Hui Fan
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Wei Zhang
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Honggang Zhang
- Department of Vascular Surgery, The First People's Hospital of Lianyungang, Lianyungang, 222005, China.
| |
Collapse
|
5
|
Wang Z, Yang T, Zeng M, Wang Z, Chen Q, Chen J, Christian M, He Z. Mitophagy suppression by miquelianin-rich lotus leaf extract induces 'beiging' of white fat via AMPK/DRP1-PINK1/PARKIN signaling axis. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2024; 104:2597-2609. [PMID: 37991930 DOI: 10.1002/jsfa.13143] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 11/07/2023] [Accepted: 11/23/2023] [Indexed: 11/24/2023]
Abstract
BACKGROUND Lotus (Nelumbo nucifera) leaf has been described to have anti-obesity activity, but the role of white fat 'browning' or 'beiging' in its beneficial metabolic actions remains unclear. Here, 3T3-L1 cells and high-fat-diet (HFD)-fed mice were used to evaluate the effects of miquelianin-rich lotus leaf extract (LLE) on white-to-beige fat conversion and its regulatory mechanisms. RESULTS Treatment with LLE increased mitochondrial abundance, mitochondrial membrane potential and NAD+ /NADH ratio in 3T3-L1 cells, suggesting its potential in promoting mitochondrial activity. qPCR and/or western blotting analysis confirmed that LLE induced the expression of beige fat-enriched gene signatures (e.g. Sirt1, Cidea, Dio2, Prdm16, Ucp1, Cd40, Cd137, Cited1) and mitochondrial biogenesis-related markers (e.g. Nrf1, Cox2, Cox7a, Tfam) in 3T3-L1 cells and inguinal white adipose tissue of HFD-fed mice. Furthermore, we found that LLE treatment inhibited mitochondrial fission protein DRP1 and blocked mitophagy markers such as PINK1, PARKIN, BECLIN1 and LC-3B. Chemical inhibition experiments revealed that AMPK/DRP1 signaling was required for LLE-induced beige fat formation via suppressing PINK1/PARKIN/mitophagy. CONCLUSION Our data reveal a novel mechanism underlying the anti-obesity effect of LLE, namely the induction of white fat beiging via AMPK/DRP1/mitophagy signaling. © 2023 Society of Chemical Industry.
Collapse
Affiliation(s)
- Zhenyu Wang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, China
- International Joint Laboratory on Food Safety, Jiangnan University, Wuxi, China
| | - Tian Yang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, China
- International Joint Laboratory on Food Safety, Jiangnan University, Wuxi, China
| | - Maomao Zeng
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, China
- International Joint Laboratory on Food Safety, Jiangnan University, Wuxi, China
| | - Zhaojun Wang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, China
- International Joint Laboratory on Food Safety, Jiangnan University, Wuxi, China
| | - Qiuming Chen
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, China
- International Joint Laboratory on Food Safety, Jiangnan University, Wuxi, China
| | - Jie Chen
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, China
- International Joint Laboratory on Food Safety, Jiangnan University, Wuxi, China
| | - Mark Christian
- School of Science and Technology, Nottingham Trent University, Nottingham, UK
| | - Zhiyong He
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, China
- International Joint Laboratory on Food Safety, Jiangnan University, Wuxi, China
| |
Collapse
|
6
|
Wang Z, Yang T, Zeng M, Wang Z, Chen Q, Chen J, Christian M, He Z. Miquelianin in Folium Nelumbinis extract promotes white-to-beige fat conversion via blocking AMPK/DRP1/mitophagy and modulating gut microbiota in HFD-fed mice. Food Chem Toxicol 2023; 181:114089. [PMID: 37804915 DOI: 10.1016/j.fct.2023.114089] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 08/16/2023] [Accepted: 10/04/2023] [Indexed: 10/09/2023]
Abstract
The main purpose of the present study was to investigate the effect of miquelianin (quercetin 3-O-glucuronide, Q3G), one of the main flavonoids in the Folium Nelumbinis extract (FNE), on beige adipocyte formation and its underlying mechanisms. In 3T3-L1 adipocytes Q3G (12.8%)-rich FNE treatment upregulated beige-related markers such as SIRT1, COX2, PGC-1α, TFAM, and UCP1. Furthermore, Q3G enhanced mitochondrial biosynthesis and inhibited mitophagy by downregulating the expression of PINK1, PARKIN, BECLIN1 and LC-3B in 3T3-L1 cells. Moreover, in high-fat-diet (HFD)-fed mice, Q3G markedly inhibited body weight gain, reduced blood glucose/lipid levels, reduced white adipose tissues (WAT) and mitigated hepatic steatosis. Meanwhile, the induced beiging accompanied by suppressed mitophagy was also demonstrated in inguinal WAT (iWAT). Chemical intervention of AMPK activity with Compound C (Com C) and Acadesine (AICAR) revealed that AMPK/DRP1 signaling was involved in Q3G-mediated mitophagy and the beiging process. Importantly, 16S rRNA sequencing analysis showed that Q3G beneficially reshaped gut microbiota structure, specifically inhibiting unclassified_Lachnospiraceae, Faecalibaculum, Roseburia and Colidextribacter while increasing Bacteroides, Akkermansia and Mucispirillum, which may potentially facilitate WAT beiging. Collectively, our findings provide a novel biological function for Folium Nelumbinis and Q3G in the fight against obesity through activating the energy-dissipating capacity of beige fat.
Collapse
Affiliation(s)
- Zhenyu Wang
- State Key Laboratory of Food Science and Resource, Jiangnan University, Wuxi, Jiangsu, 214122, China; International Joint Laboratory on Food Safety, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Tian Yang
- State Key Laboratory of Food Science and Resource, Jiangnan University, Wuxi, Jiangsu, 214122, China; International Joint Laboratory on Food Safety, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Maomao Zeng
- State Key Laboratory of Food Science and Resource, Jiangnan University, Wuxi, Jiangsu, 214122, China; International Joint Laboratory on Food Safety, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Zhaojun Wang
- State Key Laboratory of Food Science and Resource, Jiangnan University, Wuxi, Jiangsu, 214122, China; International Joint Laboratory on Food Safety, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Qiuming Chen
- State Key Laboratory of Food Science and Resource, Jiangnan University, Wuxi, Jiangsu, 214122, China; International Joint Laboratory on Food Safety, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Jie Chen
- State Key Laboratory of Food Science and Resource, Jiangnan University, Wuxi, Jiangsu, 214122, China; International Joint Laboratory on Food Safety, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Mark Christian
- School of Science and Technology, Trent University, Clifton, Nottingham, NG11 8NS, United Kingdom.
| | - Zhiyong He
- State Key Laboratory of Food Science and Resource, Jiangnan University, Wuxi, Jiangsu, 214122, China; International Joint Laboratory on Food Safety, Jiangnan University, Wuxi, Jiangsu, 214122, China.
| |
Collapse
|
7
|
Shi L, Wang S, Zhang S, Wang J, Chen Y, Li Y, Liu Z, Zhao S, Wei B, Zhang L. Research progress on pharmacological effects and mechanisms of cepharanthine and its derivatives. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2023; 396:2843-2860. [PMID: 37338575 DOI: 10.1007/s00210-023-02537-y] [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: 04/08/2023] [Accepted: 05/16/2023] [Indexed: 06/21/2023]
Abstract
Cepharanthine (CEP) is a bisbenzylisoquinoline alkaloid compound found in plants of the Stephania genus, which has biological functions such as regulating autophagy, inhibiting inflammation, oxidative stress, and apoptosis. It is often used for the treatment of inflammatory diseases, viral infections, cancer, and immune disorders and has great clinical translational value. However, there is no detailed research on its specific mechanism and dosage and administration methods, especially clinical research is limited. In recent years, CEP has shown significant effects in the prevention and treatment of COVID-19, suggesting its potential medicinal value waiting to be discovered. In this article, we comprehensively introduce the molecular structure of CEP and its derivatives, describe in detail the pharmacological mechanisms of CEP in various diseases, and discuss how to chemically modify and design CEP to improve its bioavailability. In summary, this work will provide a reference for further research and clinical application of CEP.
Collapse
Affiliation(s)
- Liangliang Shi
- Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Shuaizhe Wang
- Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Shangzu Zhang
- Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Jiawei Wang
- Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Yaping Chen
- Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Yangyang Li
- Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Zhiwei Liu
- Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Sichen Zhao
- Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Benjun Wei
- Gansu University of Traditional Chinese Medicine, Lanzhou, China.
- Key Laboratory of Dunhuang Medicine and Transformation at Provincial and Ministerial Level, Lanzhou, China.
| | - Liying Zhang
- Gansu University of Traditional Chinese Medicine, Lanzhou, China.
- Key Laboratory of Traditional Chinese Medicine Exploration and Innovation Transformation in Gansu Province, Lanzhou, China.
| |
Collapse
|
8
|
Li J, Huang Q, Lv M, Ma W, Sun J, Zhong X, Hu R, Ma M, Han Z, Zhang W, Feng W, Sun X, Zhou X. Role of liensinine in sensitivity of activated macrophages to ferroptosis and in acute liver injury. Cell Death Discov 2023; 9:189. [PMID: 37353487 DOI: 10.1038/s41420-023-01481-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 05/14/2023] [Accepted: 06/12/2023] [Indexed: 06/25/2023] Open
Abstract
Acute liver injury (ALI) is an acute inflammatory liver disease with a high mortality rate. Alternatively, activated macrophages (AAMs) have been linked to the inflammation and recovery of ALI. However, the mechanism underlying AAM death in ALI has not been studied sufficiently. We used liensinine (Lie) as a drug of choice after screening a library of small-molecule monomers with 1488 compounds from traditional Chinese remedies. In ALI, we evaluated the potential therapeutic effects and underlying mechanisms of action of the drug in ALI and found that it effectively inhibited RSL3-induced ferroptosis in AAM. Lie significantly reduced lipid peroxidation in RSL3-generated AAM. It also improved the survival rate of LPS/D-GalN-treated mice, reduced serum transaminase activity, suppressed inflammatory factor production, and may have lowered AAM ferroptosis in ALI. Lie also inhibited ferritinophagy and blocked Fe2+ synthesis. Following combined treatment with RSL3 and Lie, super-resolution microscopy revealed a close correlation between ferritin and LC3-positive vesicles in the AAM. The co-localization of ferritin and LC3 with LAMP1 was significantly reduced. These findings suggest that Lie may ameliorate ALI by inhibiting ferritinophagy and enhancing AMM resistance to ferroptosis by inhibiting autophagosome-lysosome fusion. Therefore, Lie may be used as a potential therapeutic agent for patients with ALI.
Collapse
Affiliation(s)
- Jing Li
- Department of Liver Disease, The Fourth Clinical Medical School, Guangzhou University of Chinese Medicine, Shenzhen, 518033, China
- Department of Liver Disease, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, 518033, China
- Macau University of Science and Technology, Faculty of Chinese Medicine, Taipa, Macao, 999078, China
| | - Qi Huang
- Department of Liver Disease, The Fourth Clinical Medical School, Guangzhou University of Chinese Medicine, Shenzhen, 518033, China
- Department of Liver Disease, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, 518033, China
| | - Minling Lv
- Department of Liver Disease, The Fourth Clinical Medical School, Guangzhou University of Chinese Medicine, Shenzhen, 518033, China
- Department of Liver Disease, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, 518033, China
| | - Wenfeng Ma
- Department of Liver Disease, The Fourth Clinical Medical School, Guangzhou University of Chinese Medicine, Shenzhen, 518033, China
- Department of Liver Disease, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, 518033, China
| | - Jialing Sun
- Department of Liver Disease, The Fourth Clinical Medical School, Guangzhou University of Chinese Medicine, Shenzhen, 518033, China
- Department of Liver Disease, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, 518033, China
| | - Xin Zhong
- Department of Liver Disease, The Fourth Clinical Medical School, Guangzhou University of Chinese Medicine, Shenzhen, 518033, China
- Department of Liver Disease, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, 518033, China
| | - Rui Hu
- Department of Liver Disease, The Fourth Clinical Medical School, Guangzhou University of Chinese Medicine, Shenzhen, 518033, China
- Department of Liver Disease, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, 518033, China
| | - MengQing Ma
- Department of Liver Disease, The Fourth Clinical Medical School, Guangzhou University of Chinese Medicine, Shenzhen, 518033, China
- Department of Liver Disease, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, 518033, China
| | - Zhiyi Han
- Department of Liver Disease, The Fourth Clinical Medical School, Guangzhou University of Chinese Medicine, Shenzhen, 518033, China
- Department of Liver Disease, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, 518033, China
| | - Wei Zhang
- Department of Liver Disease, The Fourth Clinical Medical School, Guangzhou University of Chinese Medicine, Shenzhen, 518033, China
- Department of Liver Disease, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, 518033, China
| | - Wenxing Feng
- Department of Liver Disease, The Fourth Clinical Medical School, Guangzhou University of Chinese Medicine, Shenzhen, 518033, China
- Department of Liver Disease, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, 518033, China
| | - Xinfeng Sun
- Department of Liver Disease, The Fourth Clinical Medical School, Guangzhou University of Chinese Medicine, Shenzhen, 518033, China
- Department of Liver Disease, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, 518033, China
| | - Xiaozhou Zhou
- Department of Liver Disease, The Fourth Clinical Medical School, Guangzhou University of Chinese Medicine, Shenzhen, 518033, China.
- Department of Liver Disease, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, 518033, China.
| |
Collapse
|
9
|
Wang Y, Chen W, Han Y, Xu X, Yang A, Wei J, Hong D, Fang X, Chen T. Neuroprotective effect of engineered Clostridiumbutyricum-pMTL007-GLP-1 on Parkinson's disease mice models via promoting mitophagy. Bioeng Transl Med 2023; 8:e10505. [PMID: 37206220 PMCID: PMC10189449 DOI: 10.1002/btm2.10505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 03/01/2023] [Accepted: 03/04/2023] [Indexed: 03/19/2023] Open
Abstract
Parkinson's disease (PD) is a common neurodegenerative disease with limited treatment and no cure, hence, broadening PD drug spectrum is of great significance. At present, engineered microorganisms are attracting increasing attention. In this study, we constructed an engineered strain of Clostridium butyricum-GLP-1, a C. butyricum (a probiotic) that consistently expresses glucagon-like peptide-1 (GLP-1, a peptide-based hormone with neurological advantage) in anticipation of its use in PD treatment. We further investigated the neuroprotective mechanism of C. butyricum-GLP-1 on PD mice models induced by 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine. The results indicated that C. butyricum-GLP-1 could improve motor dysfunction and ameliorate neuropathological changes by increasing TH expression and reducing the expression of α-syn. Moreover, we confirmed that C. butyricum-GLP-1 improved microbiome imbalance of PD mice by decreasing the relative abundance of Bifidobacterium at the genus level, improved gut integrity, and upregulated the levels of GPR41/43. Surprisingly, we found it could exert its neuroprotective effects via promoting PINK1/Parkin mediated mitophagy and attenuating oxidative stress. Together, our work showed that C. butyricum-GLP-1 improves PD by promoting mitophagy, which provides an alternative therapeutic modality for PD.
Collapse
Affiliation(s)
- Yun Wang
- Department of NeurologyThe First Affiliated Hospital of Nanchang UniversityNanchangJiangxi ProvinceP. R. China330006
| | - Wen‐jie Chen
- Institute of Translational MedicineNanchang UniversityNanchangJiangxi ProvinceP. R. China330031
| | - Yi‐yang Han
- Institute of Translational MedicineNanchang UniversityNanchangJiangxi ProvinceP. R. China330031
| | - Xuan Xu
- Institute of Translational MedicineNanchang UniversityNanchangJiangxi ProvinceP. R. China330031
| | - Ai‐xia Yang
- Department of NeurologyThe First Affiliated Hospital of Nanchang UniversityNanchangJiangxi ProvinceP. R. China330006
| | - Jing Wei
- Institute of Translational MedicineNanchang UniversityNanchangJiangxi ProvinceP. R. China330031
| | - Dao‐jun Hong
- Department of NeurologyThe First Affiliated Hospital of Nanchang UniversityNanchangJiangxi ProvinceP. R. China330006
| | - Xin Fang
- Department of NeurologyThe First Affiliated Hospital of Nanchang UniversityNanchangJiangxi ProvinceP. R. China330006
| | - Ting‐tao Chen
- Institute of Translational MedicineNanchang UniversityNanchangJiangxi ProvinceP. R. China330031
| |
Collapse
|
10
|
Li T, Bai H, Fang H, Yang L, Yan P. Growth hormone inhibits adipogenic differentiation and induces browning in bovine subcutaneous adipocytes. Growth Horm IGF Res 2022; 66:101498. [PMID: 36007464 DOI: 10.1016/j.ghir.2022.101498] [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: 05/15/2022] [Revised: 08/12/2022] [Accepted: 08/15/2022] [Indexed: 11/17/2022]
Abstract
OBJECTIVE It is well established that growth hormone (GH) has the ability to stimulate lipolysis. The effects of GH on adipocyte differentiation and browning have not been clearly described. Therefore, the present study aimed to elucidate the role of GH in the differentiation and browning of bovine subcutaneous adipocytes as well as its underlying molecular mechanisms. METHODS We first treated bovine subcutaneous preadipocytes with different concentrations (0, 10, 100, and 500 ng/mL) of GH for 8 days and measured lipid accumulation and gene expression. Afterward, we treated preadipocytes and mature adipocytes with 500 ng/mL GH and determined differentiation and browning-related indicators. Finally, we investigated the expression of STAT5B in both preadipocytes and mature adipocytes after GH treatment. RESULTS We demonstrated that GH inhibited lipid accumulation and decreased the expression levels of adipogenic key genes (SCD1, SREBP1, PPARγ, and CEBPα) during adipocyte differentiation. Moreover, we observed that the inhibitory effect of GH on the early stage of adipocyte differentiation (0-2 days) was stronger than that on the later stage of adipocyte differentiation (2-8 days). We also found that GH promoted the expression levels of browning-related genes such as uncoupling protein 1 (UCP1) in mature adipocytes. Concurrently, GH promoted mitochondrial biogenesis and increased the expression levels of mitochondrial biogenesis-related genes. In addition, GH promoted phosphorylation of signal transducers and activator of transcription 5 b (STAT5B) and contributed to translocation of STAT5B to nucleus. After blocking the expression of STAT5B protein, GH weakened the inhibition of adipogenic key genes and reduced the promotion of browning-related genes in bovine subcutaneous adipocytes. CONCLUSIONS GH can inhibit adipocyte differentiation and promote adipocyte browning by regulating STAT5B in bovine subcutaneous adipocytes.
Collapse
Affiliation(s)
- Tingting Li
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Hui Bai
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Haoyuan Fang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Liang Yang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Peishi Yan
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China.
| |
Collapse
|
11
|
Liensinine Inhibits Cell Growth and Blocks Autophagic Flux in Nonsmall-Cell Lung Cancer. JOURNAL OF ONCOLOGY 2022; 2022:1533779. [PMID: 35813859 PMCID: PMC9270144 DOI: 10.1155/2022/1533779] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 05/28/2022] [Indexed: 12/24/2022]
Abstract
Liensinine is a bioactive component of Plumula Nelumbinis extracted from the green embryo of the mature seeds of Nelumbonaceae and exhibits therapeutic functions and noteworthy anti-tumor effects in recent studies. However, the potential anti-tumor property and the underlying mechanisms of liensinine in nonsmall-cell lung cancer (NSCLC) have not been illustrated. In this study, we demonstrated that liensinine has the potential anti-tumor property, and it could inhibit growth of NSCLC in vitro and in vivo. In addition, we found that although it induced significant accumulation of autophagosomes, liensinine could quench them for degradation and blocked autophagic flux. Importantly, we observed that liensinine inhibited the normal function of mitochondrial energy supply and impaired the lysosomal function. This research firstly provides a possibility insight that liensinine could be a novel therapeutic strategy for NSCLC.
Collapse
|
12
|
Huo Y, Zhao G, Li J, Wang R, Ren F, Li Y, Wang X. Bifidobacterium animalis subsp. lactis A6 Enhances Fatty Acid β-Oxidation of Adipose Tissue to Ameliorate the Development of Obesity in Mice. Nutrients 2022; 14:598. [PMID: 35276956 PMCID: PMC8839083 DOI: 10.3390/nu14030598] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 01/20/2022] [Accepted: 01/25/2022] [Indexed: 12/17/2022] Open
Abstract
Fatty acid β-oxidation (FAO) is confirmed to be impaired in obesity, especially in adipose tissues. We previously proved that Bifidobacterium animalis subsp. lactis A6 (BAA6) had protective effects against diet-induced obesity. However, whether BAA6 enhances FAO to ameliorate the development of obesity has not been explored. After being fed with high-fat diet (HFD) for 9 weeks, male C57BL/6J mice were fed HFD or BAA6 for 8 weeks. In vitro study was carried out using 3T3-L1 adipocytes to determine the effect of BAA6 culture supernatant (BAA6-CM). Here, we showed that administration of BAA6 to mice fed with HFD decreased body weight gain (by 5.03 g) and significantly up-regulated FAO in epididymal adipose tissues. In parallel, FAO in 3T3-L1 cells was increased after BAA6-CM treatment. Acetate was identified as a constituent of BAA6-CM that showed a similar effect to BAA6-CM. Furthermore, acetate treatment activated the GPR43-PPARα signaling, thereby promoting FAO in 3T3-L1 cells. The levels of acetate were also elevated in serum and feces (by 1.92- and 2.27-fold) of HFD-fed mice following BAA6 administration. The expression levels of GPR43 and PPARα were increased by 55.45% and 69.84% after BAA6 supplement in the epididymal fat of mice. Together, these data reveal that BAA6 promotes FAO of adipose tissues through the GPR43-PPARα signaling, mainly by increasing acetate levels, leading to alleviating the development of obesity.
Collapse
Affiliation(s)
- Yanxiong Huo
- Key Laboratory of Precision Nutrition and Food Quality, College of Food Science & Nutritional Engineering, China Agricultural University, Beijing 100083, China; (Y.H.); (F.R.); (Y.L.)
| | - Guoping Zhao
- School of Food and Health, Beijing Technology and Business University, Beijing 100048, China; (G.Z.); (J.L.)
| | - Jinwang Li
- School of Food and Health, Beijing Technology and Business University, Beijing 100048, China; (G.Z.); (J.L.)
| | - Ran Wang
- Key Laboratory of Functional Dairy, Co-Constructed by Ministry of Education and Beijing Municipality, Department of Nutrition and Health, China Agricultural University, Beijing 100083, China;
| | - Fazheng Ren
- Key Laboratory of Precision Nutrition and Food Quality, College of Food Science & Nutritional Engineering, China Agricultural University, Beijing 100083, China; (Y.H.); (F.R.); (Y.L.)
- Key Laboratory of Functional Dairy, Co-Constructed by Ministry of Education and Beijing Municipality, Department of Nutrition and Health, China Agricultural University, Beijing 100083, China;
| | - Yixuan Li
- Key Laboratory of Precision Nutrition and Food Quality, College of Food Science & Nutritional Engineering, China Agricultural University, Beijing 100083, China; (Y.H.); (F.R.); (Y.L.)
- Key Laboratory of Functional Dairy, Co-Constructed by Ministry of Education and Beijing Municipality, Department of Nutrition and Health, China Agricultural University, Beijing 100083, China;
| | - Xiaoyu Wang
- Key Laboratory of Precision Nutrition and Food Quality, College of Food Science & Nutritional Engineering, China Agricultural University, Beijing 100083, China; (Y.H.); (F.R.); (Y.L.)
| |
Collapse
|
13
|
D'Urso O, Drago F. Pharmacological significance of extra-oral taste receptors. Eur J Pharmacol 2021; 910:174480. [PMID: 34496302 DOI: 10.1016/j.ejphar.2021.174480] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 08/23/2021] [Accepted: 09/01/2021] [Indexed: 01/17/2023]
Abstract
It has recently been shown that taste receptors, in addition to being present in the oral cavity, exist in various extra-oral organs and tissues such as the thyroid, lungs, skin, stomach, intestines, and pancreas. Although their physiological function is not yet fully understood, it appears that they can help regulate the body's homeostasis and provide an additional defense function against pathogens. Since the vast majority of drugs are bitter, the greatest pharmacological interest is in the bitter taste receptors. In this review, we describe how bitter taste 2 receptors (TAS2Rs) induce bronchodilation and mucociliary clearance in the airways, muscle relaxation in various tissues, inhibition of thyroid stimulating hormone (TSH) in thyrocytes, and release of glucagon-like peptide-1 (GLP-1) and ghrelin in the digestive system. In fact, substances such as dextromethorphan, chloroquine, methimazole and probably glimepiride, being agonists of TAS2Rs, lead to these effects. TAS2Rs and taste 1 receptors (TAS1R2/3) are G protein-coupled receptors (GPCR). TAS1R2/3 are responsible for sweet taste perception and may induce GLP-1 release and insulin secretion. Umami taste receptors, belonging to the same superfamily of receptors, perform a similar function with regard to insulin. The sour and salty taste receptors work in a similar way, both being channel receptors sensitive to amiloride. Finally, gene-protein coupled receptor 40 (GPR40) and GPR120 for fatty taste perception are also protein-coupled receptors and may induce GLP-1 secretion and insulin release, similar to those of other receptors belonging to the same superfamily.
Collapse
Affiliation(s)
- Ottavio D'Urso
- Department of Biomedical and Biotechnological Sciences, University of Catania, Via S. Sofia, 97, 95125 Catania, Italy
| | - Filippo Drago
- Department of Biomedical and Biotechnological Sciences, University of Catania, Via S. Sofia, 97, 95125 Catania, Italy.
| |
Collapse
|
14
|
Prediction of the Secretome and the Surfaceome: A Strategy to Decipher the Crosstalk between Adipose Tissue and Muscle during Fetal Growth. Int J Mol Sci 2020; 21:ijms21124375. [PMID: 32575512 PMCID: PMC7353064 DOI: 10.3390/ijms21124375] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 06/08/2020] [Accepted: 06/16/2020] [Indexed: 12/16/2022] Open
Abstract
Crosstalk between adipose and muscular tissues is hypothesized to regulate the number of muscular and adipose cells during fetal growth, with post-natal consequences on lean and fat masses. Such crosstalk largely remains, however, to be described. We hypothesized that a characterization of the proteomes of adipose and muscular tissues from bovine fetuses may enhance the understanding of the crosstalk between these tissues through the prediction of their secretomes and surfaceomes. Proteomic experiments have identified 751 and 514 proteins in fetal adipose tissue and muscle. These are mainly involved in the regulation of cell proliferation or differentiation, but also in pathways such as apoptosis, Wnt signalling, or cytokine-mediated signalling. Of the identified proteins, 51 adipokines, 11 myokines, and 37 adipomyokines were predicted, together with 26 adipose and 13 muscular cell surface proteins. Analysis of protein–protein interactions suggested 13 links between secreted and cell surface proteins that may contribute to the adipose–muscular crosstalk. Of these, an interaction between the adipokine plasminogen and the muscular cell surface alpha-enolase may regulate the fetal myogenesis. The in silico secretome and surfaceome analyzed herein exemplify a powerful strategy to enhance the elucidation of the crosstalk between cell types or tissues.
Collapse
|
15
|
Rahman MS, Kim YS. PINK1-PRKN mitophagy suppression by mangiferin promotes a brown-fat-phenotype via PKA-p38 MAPK signalling in murine C3H10T1/2 mesenchymal stem cells. Metabolism 2020; 107:154228. [PMID: 32289346 DOI: 10.1016/j.metabol.2020.154228] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 03/20/2020] [Accepted: 04/07/2020] [Indexed: 02/06/2023]
Abstract
OBJECTIVE Mangiferin (MF), a xanthonoid derived from Mangifera indica, has shown therapeutic effects on various human diseases including cancer, diabetes, and obesity. Nonetheless, the influence of MF on non-shivering thermogenesis and its underlying mechanism in browning remains unclear. Here, our aim was to investigate the effects of MF on browning and its molecular mechanisms in murine C3H10T1/2 mesenchymal stem cells (MSCs). MATERIALS/METHODS To determine the function of MF on browning, murine C3H10T1/2 MSCs were treated with MF in an adipogenic differentiation cocktail and the thermogenic and correlated metabolic responses were assessed using MF-mediated signalling. Human adipose-derived MSCs were differentiated and treated with MF to confirm its role in thermogenic induction. RESULTS MF treatment induced the expression of a brown-fat signature, UCP1, and reduced triglyceride (TG) in C3H10T1/2 MSCs. MF also induced the expression of major thermogenesis regulators: PGC1α, PRDM16, and PPARγ and up-regulated the expression of beiging markers CD137, HSPB7, TBX1, and COX2 in both murine C3H10T1/2 MSCs and human adipose-derived mesenchymal stem cells (hADMSC). We also observed that MF treatment increased the mitochondrial DNA and improved mitochondrial homeostasis by regulating mitofission-fusion plasticity via suppressing PINK1-PRKN-mediated mitophagy. Furthermore, MF treatment improved mitochondrial respiratory function by increasing mitochondrial oxygen consumption and expression of oxidative-phosphorylation (OXPHOS)-related proteins. Chemical-inhibition and gene knockdown experiments revealed that β3-AR-dependent PKA-p38 MAPK-CREB signalling is crucial for MF-mediated brown-fat formation via suppression of mitophagy in C3H10T1/2 MSCs. CONCLUSIONS MF promotes the brown adipocyte phenotype by suppressing mitophagy, which is regulated by PKA-p38MAPK-CREB signalling in C3H10T1/2 MSCs. Thus, we propose that MF may be a good browning inducer that can ameliorate obesity.
Collapse
Affiliation(s)
- Md Shamim Rahman
- Institute of Tissue Regeneration, College of Medicine, Soonchunhyang University, Cheonan, Chung-nam 31151, South Korea; Department of Microbiology, College of Medicine, Soonchunhyang University, Cheonan, Chung-nam 31151, South Korea
| | - Yong-Sik Kim
- Institute of Tissue Regeneration, College of Medicine, Soonchunhyang University, Cheonan, Chung-nam 31151, South Korea; Department of Microbiology, College of Medicine, Soonchunhyang University, Cheonan, Chung-nam 31151, South Korea.
| |
Collapse
|
16
|
Liang X, Wang S, Wang L, Ceylan AF, Ren J, Zhang Y. Mitophagy inhibitor liensinine suppresses doxorubicin-induced cardiotoxicity through inhibition of Drp1-mediated maladaptive mitochondrial fission. Pharmacol Res 2020; 157:104846. [PMID: 32339784 DOI: 10.1016/j.phrs.2020.104846] [Citation(s) in RCA: 98] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Revised: 03/17/2020] [Accepted: 04/16/2020] [Indexed: 01/04/2023]
Abstract
Doxorubicin (DOX) is one of the most effective antineoplastic drugs. However, its clinical application has been greatly limited due to the development of cardiotoxicity with DOX utilization. A number of theories have been postulated for DOX-induced cardiotoxicity with a pivotal contribution from unchecked (excess) mitophagy and mitochondrial fission. Liensinine (LIEN), a newly identified mitophagy inhibitor, strengthens the antineoplastic efficacy of DOX although its action on hearts remains elusive. This study was designed to examine the effect of LIEN on DOX-induced cardiotoxicity and the underlying mechanisms involved with a focus on mitochondrial dynamics. Our data revealed that LIEN alleviated DOX-induced cardiac dysfunction and apoptosis through inhibition of dynamin-related protein 1 (Drp1)-mediated excess (unchecked) mitochondrial fission. LIEN treatment decreased Drp1 phosphorylation at Ser616 site, inhibited mitochondrial fragmentation, mitophagy (assessed by TOM20 and TIM23), oxidative stress, cytochrome C leakage, cardiomyocyte apoptosis, as well as improved mitochondrial function and cardiomyocyte contractile function in DOX-induced cardiac injury. In DOX-challenged neonatal mouse ventricular myocytes (NMVMs), LIEN-suppressed Drp1 phosphorylation, mitochondrial fragmentation, and apoptosis were blunted by Rab7 overexpression, the effect of which was reversed by the ERK inhibitor U0126. Moreover, activation of ERK or Drp1 abolished the protective effects of LIEN on cardiomyocyte mechanical anomalies. These data shed some lights towards understanding the role of LIEN as a new protective agent against DOX-associated cardiotoxicity without compromising its anti-tumor effects.
Collapse
Affiliation(s)
- Xinyue Liang
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China; Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China
| | - Shuyi Wang
- Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China; Center for Cardiovascular Research and Alternative Medicine, Laramie, WY 82071, USA
| | - Lifeng Wang
- Center for Cardiovascular Research and Alternative Medicine, Laramie, WY 82071, USA; Department of Physiology, Basic Medicine College, Xinjiang Medical University, Urumqi, Xinjiang 830011, China
| | - Asli F Ceylan
- Center for Cardiovascular Research and Alternative Medicine, Laramie, WY 82071, USA
| | - Jun Ren
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China; Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China; Center for Cardiovascular Research and Alternative Medicine, Laramie, WY 82071, USA.
| | - Yingmei Zhang
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China; Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China.
| |
Collapse
|