1
|
An Y, Zhao H, He C, Shi L, Su X, Zhang H, Huang Y, Shan Z, Wang M, Du Y, Xie J, Zhao Y, Yang Y, Huang Z, Wan A, Zhao Y, Zhao B. Xiasangju alleviates hepatic insulin resistance in db/db mice via AMPK pathway: Mechanisms and active components study. Int Immunopharmacol 2025; 156:114675. [PMID: 40286785 DOI: 10.1016/j.intimp.2025.114675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Revised: 03/30/2025] [Accepted: 04/12/2025] [Indexed: 04/29/2025]
Abstract
Type 2 diabetes mellitus (T2DM), one of the prevalent chronic diseases, significantly impacts individuals and society. Xiasangju (XSJ), a herbal tea formulation, has been commonly used in traditional Chinese medicine. Accumulating evidence suggests that XSJ can alleviate metabolic syndrome by regulating glucose and lipid metabolism, lowering liver index and improving glucose tolerance. In the present study, db/db mice were used to examine the effect of XSJ on treating T2DM, and Western blotting was performed to explore the underlying anti-T2DM pharmacological mechanisms. With AMP-activated protein kinase (AMPK) chosen as the target protein, surface plasmon resonance (SPR)-LC-MS technology was used to identify potential active ingredients of XSJ. To further explore the role of potential active ingredients of XSJ, their effects were investigated in insulin resistance (IR)-HepG2 cells. Our results demonstrate that in diabetic db/db mice, XSJ activated the AMPK pathway, which regulated hepatic glucose metabolism and inhibited oxidative stress caused by hepatic NADPH oxidase 4 (NOX4), thereby ameliorating hepatic IR. By means of SPR-LC-MS experiments, 4-Methylesculetin was identified as an important active ingredient in XSJ. Subsequently, to further elucidate the effects of this ingredient, in IR-HepG2 cells, 4-Methylesculetin was found to mitigate oxidative stress, enhance glucose consumption, and promote glycogen synthesis. This study demonstrated that XSJ improved T2DM and mitigated oxidative stress by activating the AMPK pathway. Specifically, 4-Methylesculetin emerged as a promising therapeutic agent for T2DM.
Collapse
Affiliation(s)
- Yongcheng An
- Department of Pharmacology, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Hongbin Zhao
- Department of Oncology and Hematology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, China
| | - Changhao He
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Lu Shi
- Central Laboratories, Qingdao Municipal Hospital, University of Health and Rehabilitation Sciences, Qingdao 266001, China
| | - Xiaohua Su
- Guangzhou Baiyunshan Xingqun Pharmaceutical Co., Ltd., Guangzhou 510288, China
| | - Huilin Zhang
- Department of Pharmacology, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Yan Huang
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Ziyi Shan
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Menglu Wang
- Department of Pharmacology, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Yuhang Du
- Department of Pharmacology, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Jiamei Xie
- Department of Pharmacology, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Yige Zhao
- Department of Pharmacology, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Yang Yang
- Department of Pharmacology, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Zhiyun Huang
- Guangzhou Baiyunshan Xingqun Pharmaceutical Co., Ltd., Guangzhou 510288, China
| | - Anfeng Wan
- Guangzhou Baiyunshan Xingqun Pharmaceutical Co., Ltd., Guangzhou 510288, China
| | - Ying Zhao
- Third Affiliated Hospital, Beijing University of Chinese Medicine, Beijing 100029, China.
| | - Baosheng Zhao
- Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China.
| |
Collapse
|
2
|
Zhu YJ, Dai XY, Zhao YL, Ma YG, Zhao ZZ, Su CF, Hao ZY, Wang D, Chen H. Lyciumines A and B: Two Pyrrole-Fused Alkaloids from the Fruits of Lycium barbarum. JOURNAL OF NATURAL PRODUCTS 2025; 88:1237-1243. [PMID: 40320641 DOI: 10.1021/acs.jnatprod.5c00385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2025]
Abstract
Two novel pyrrole-fused alkaloids, lyciumines A (1) and B (2), were isolated from the fruits of Lycium barbarum. Their structures were elucidated by analysis of NMR spectroscopic and MS spectrometric data, along with computational studies. Compound 1 represents a novel pyrroloindoline alkaloid with a rare 6/5/5/6 tetracyclic system connected with a 2-formyl-5-methylpyrrole moiety via a C-C bond. Compound 2 features an unusual pyrrole alkaloid with a 5/5/5 tricyclic skeleton. Compound 1 significantly increases glucose consumption in insulin-resistant HepG2 cells in a dose-dependent manner. Further mechanism investigations demonstrated that compound 1 regulates glucose metabolism via activating the AMPK/PI3K/Akt signaling pathway.
Collapse
Affiliation(s)
- Yan-Jie Zhu
- School of Pharmacy, Henan University of Chinese Medicine, Zhengzhou 450046, People's Republic of China
| | - Xiang-Yang Dai
- School of Pharmacy, Henan University of Chinese Medicine, Zhengzhou 450046, People's Republic of China
| | - Yi-Lin Zhao
- School of Pharmacy, Henan University of Chinese Medicine, Zhengzhou 450046, People's Republic of China
| | - Yu-Guo Ma
- School of Pharmacy, Henan University of Chinese Medicine, Zhengzhou 450046, People's Republic of China
| | - Zhen-Zhu Zhao
- School of Pharmacy, Henan University of Chinese Medicine, Zhengzhou 450046, People's Republic of China
| | - Cheng-Fu Su
- School of Pharmacy, Henan University of Chinese Medicine, Zhengzhou 450046, People's Republic of China
| | - Zhi-You Hao
- School of Pharmacy, Henan University of Chinese Medicine, Zhengzhou 450046, People's Republic of China
| | - Dongdong Wang
- Molecular Targets Program, Center for Cancer Research, National Cancer Institute, Frederick, Maryland 21702-1201, United States
| | - Hui Chen
- School of Pharmacy, Henan University of Chinese Medicine, Zhengzhou 450046, People's Republic of China
- Co-Construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases by Henan & Education Ministry of People's Republic of China, Zhengzhou 450046, People's Republic of China
| |
Collapse
|
3
|
Li G, Yi S, Wang H, Qiu H, Wang W, Gao L, Xu Q, Han B, Yin X. Salidroside production through cascade biocatalysis with a thermostability-enhanced UDP-glycosyltransferase. Int J Biol Macromol 2025; 299:140261. [PMID: 39855494 DOI: 10.1016/j.ijbiomac.2025.140261] [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: 09/19/2024] [Revised: 12/24/2024] [Accepted: 01/21/2025] [Indexed: 01/27/2025]
Abstract
Salidroside is a phenylpropanoid glycoside with wide applications in the food, pharmaceutical, and cosmetic industries; however, the plant genus Rhodiola, the natural source of salidroside, has slow growth and limited distribution. In this study, we designed a novel six-enzyme biocatalytic cascade for the efficient production of salidroside, utilizing cost-effective bio-based L-Tyrosine as the starting material. A preliminary analysis revealed that the poor thermostability of the Bacillus licheniformis UDP-glycosyltransferase (EC 2.4.1.384) BlYjiC M6 is a bottleneck in the cascade. Therefore, a combined computational strategy was used to engineer it and finally obtained a mutant TSM6 (T304V/G307A/N309W/F123W/T344V/D271G) with a 134-fold longer half-life at 40 °C and a 13 °C higher Tmapp compared to M6. The integration of TSM6 into the cascade improved salidroside productivity significantly, while reducing residual intermediates. After further optimization, the whole-cell biocatalytic cascade achieved a high salidroside titer of 12.8 g·L-1 in a 5 L bioreactor, giving a productivity of 0.53 g·L-1·h-1. This study provides a green and efficient biosynthetic process for salidroside production and highlights the potential of enzyme engineering to enhance the biocatalytic cascade.
Collapse
Affiliation(s)
- Guosi Li
- Traditional Chinese Medicine Institute of Anhui Dabie Mountain, Generic Technology Research Center for Anhui Traditional Chinese Medicine Industry, Anhui Engineering Research Center for Eco-agriculture of Traditional Chinese Medicine, West Anhui University, Lu'an 237012, Anhui, China
| | - Shanyong Yi
- Traditional Chinese Medicine Institute of Anhui Dabie Mountain, Generic Technology Research Center for Anhui Traditional Chinese Medicine Industry, Anhui Engineering Research Center for Eco-agriculture of Traditional Chinese Medicine, West Anhui University, Lu'an 237012, Anhui, China
| | - Haijiao Wang
- Key State Laboratory of Plant Diversity and Specialty Crops, Guangdong Provincial Key Laboratory of Applied Botany, South China Botanical Garden, Chinese Academy of Sciences, Guangzhou 510650, China
| | - Hulin Qiu
- School of Marine Sciences, Sun Yat-Sen University, Zhuhai 519080, China
| | - Wei Wang
- Traditional Chinese Medicine Institute of Anhui Dabie Mountain, Generic Technology Research Center for Anhui Traditional Chinese Medicine Industry, Anhui Engineering Research Center for Eco-agriculture of Traditional Chinese Medicine, West Anhui University, Lu'an 237012, Anhui, China
| | - Leilei Gao
- Traditional Chinese Medicine Institute of Anhui Dabie Mountain, Generic Technology Research Center for Anhui Traditional Chinese Medicine Industry, Anhui Engineering Research Center for Eco-agriculture of Traditional Chinese Medicine, West Anhui University, Lu'an 237012, Anhui, China
| | - Qilin Xu
- Traditional Chinese Medicine Institute of Anhui Dabie Mountain, Generic Technology Research Center for Anhui Traditional Chinese Medicine Industry, Anhui Engineering Research Center for Eco-agriculture of Traditional Chinese Medicine, West Anhui University, Lu'an 237012, Anhui, China
| | - Bangxing Han
- Traditional Chinese Medicine Institute of Anhui Dabie Mountain, Generic Technology Research Center for Anhui Traditional Chinese Medicine Industry, Anhui Engineering Research Center for Eco-agriculture of Traditional Chinese Medicine, West Anhui University, Lu'an 237012, Anhui, China.
| | - Xinjian Yin
- School of Marine Sciences, Sun Yat-Sen University, Zhuhai 519080, China.
| |
Collapse
|
4
|
Zhu D, Li Y, Liu M, Yang Y, Fu J, Su L, Wang F, Cen Y, Zhou Y, Li Y. Study on the role of mitophagy and pyroptosis induced by nano-silver in testicular injury. Food Chem Toxicol 2025; 197:115245. [PMID: 39788479 DOI: 10.1016/j.fct.2025.115245] [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: 09/20/2024] [Revised: 12/10/2024] [Accepted: 01/06/2025] [Indexed: 01/12/2025]
Abstract
Silver nanoparticles(AgNPs)have been widely used in biomedicine and industry. Growing studies have shown that AgNPs can induce sperm motility decrease and spermiogenesis disorders. In this study, animal experiments were used to investigate the role of mitophagy and pyroptosis caused by AgNPs (25.93 nm) in testicular injury. Results showed that AgNPs induced the production of NLRP3, IL-1β and IL-18, activated Caspase-1, increased the expression of GSDMD protein, and activated the PINK1/Parkin signaling pathway, which induced in mitophagy in mice testicle tissue. In summary, AgNPs induced mitophagy and pyroptosis in mice testis at the highest dose, which lead to damage of testis tissue.
Collapse
Affiliation(s)
- Deyu Zhu
- School of Public Health, Zunyi Medical University, Zunyi, Guizhou, 563000, PR China; Key Laboratory of Maternal & Child Health and Exposure Science of Guizhou Higher Education Institutes, Zunyi, Guizhou, 563000, PR China
| | - Yingyi Li
- School of Public Health, Zunyi Medical University, Zunyi, Guizhou, 563000, PR China
| | - Min Liu
- School of Public Health, Zunyi Medical University, Zunyi, Guizhou, 563000, PR China
| | - Yufen Yang
- School of Public Health, Zunyi Medical University, Zunyi, Guizhou, 563000, PR China
| | - Jiayu Fu
- School of Public Health, Zunyi Medical University, Zunyi, Guizhou, 563000, PR China
| | - Liyu Su
- School of Public Health, Zunyi Medical University, Zunyi, Guizhou, 563000, PR China
| | - Feng Wang
- School of Public Health, Zunyi Medical University, Zunyi, Guizhou, 563000, PR China
| | - Yuyan Cen
- School of Public Health, Zunyi Medical University, Zunyi, Guizhou, 563000, PR China
| | - Yanna Zhou
- School of Public Health, Zunyi Medical University, Zunyi, Guizhou, 563000, PR China
| | - Yan Li
- School of Public Health, Zunyi Medical University, Zunyi, Guizhou, 563000, PR China; Key Laboratory of Maternal & Child Health and Exposure Science of Guizhou Higher Education Institutes, Zunyi, Guizhou, 563000, PR China
| |
Collapse
|
5
|
Wang Y, Gao P, Wu Z, Jiang B, Wang Y, He Z, Zhao B, Tian X, Gao H, Cai L, Li W. Exploring the therapeutic potential of Chinese herbs on comorbid type 2 diabetes mellitus and Parkinson's disease: A mechanistic study. JOURNAL OF ETHNOPHARMACOLOGY 2025; 338:119095. [PMID: 39537117 DOI: 10.1016/j.jep.2024.119095] [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: 08/21/2024] [Revised: 10/12/2024] [Accepted: 11/08/2024] [Indexed: 11/16/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Type 2 diabetes mellitus (T2DM) and Parkinson's disease (PD) are chronic conditions that affect the aging population, with increasing prevalence globally. The rising prevalence of comorbidity between these conditions, driven by demographic shifts, severely impacts the quality of life of patients, posing a significant burden on healthcare resources. Chinese herbal medicine has been used to treat T2DM and PD for millennia. Pharmacological studies have demonstrated that medicinal herbs effectively lower blood glucose levels and exert neuroprotective effects, suggesting their potential as adjunctive therapy for concurrent management of T2DM and PD. AIM OF THE STUDY To elucidate the shared mechanisms underlying T2DM and PD, particularly focusing on the potential mechanisms by which medicinal herbs (including herbal formulas, single herbs, and active compounds) may treat these diseases, to provide valuable insights for developing therapeutics targeting comorbid T2DM and PD. MATERIALS AND METHODS Studies exploring the mechanisms underlying T2DM and PD, as well as the treatment of these conditions with medicinal herbs, were extracted from several electronic databases, including PubMed, Web of Science, Google Scholar, and China National Knowledge Infrastructure (CNKI). RESULTS Numerous studies have shown that inflammation, oxidative stress, insulin resistance, impaired autophagy, gut microbiota dysbiosis, and ferroptosis are shared mechanisms underlying T2DM and PD mediated through the NLRP3 inflammasome, NF-κB, MAPK, Keap1/Nrf2/ARE, PI3K/AKT, AMPK/SIRT1, and System XC--GSH-GPX4 signaling pathways. Thirty-four medicinal herbs, including 2 herbal formulas, 4 single herbs, and 28 active compounds, have been reported to potentially exert anti-T2DM and anti-PD effects by targeting these shared mechanisms. CONCLUSIONS Traditional Chinese medicine effectively combats T2DM and PD through shared pathological mechanisms, highlighting their potential for application in treating these comorbid conditions.
Collapse
Affiliation(s)
- Yan Wang
- Clinical College of Chinese Medicine, Gansu University of Chinese Medicine, Lanzhou, 730000, China; Encephalopathy Department, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, China
| | - Pengpeng Gao
- Department of Preventive Treatment, Ningxia Integrated Chinese and Western Medicine Hospital, Yinchuan, 750004, China
| | - Zicong Wu
- Encephalopathy Department, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, China
| | - Bing Jiang
- Department of Integrated Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, 730000, China
| | - Yanru Wang
- Gansu University Key Laboratory for Molecular Medicine & Chinese Medicine Prevention and Treatment of Major Diseases, Gansu University of Chinese Medicine, Lanzhou, 730000, China
| | - Zhaxicao He
- Clinical College of Chinese Medicine, Gansu University of Chinese Medicine, Lanzhou, 730000, China
| | - Bing Zhao
- Clinical College of Chinese Medicine, Gansu University of Chinese Medicine, Lanzhou, 730000, China
| | - Xinyun Tian
- Encephalopathy Department, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, China
| | - Han Gao
- Encephalopathy Department, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, China
| | - Li Cai
- Encephalopathy Department, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, China.
| | - Wentao Li
- Encephalopathy Department, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, China.
| |
Collapse
|
6
|
Li M, Ding L, Cao L, Zhang Z, Li X, Li Z, Xia Q, Yin K, Song S, Wang Z, Du H, Zhao D, Li X, Wang Z. Natural products targeting AMPK signaling pathway therapy, diabetes mellitus and its complications. Front Pharmacol 2025; 16:1534634. [PMID: 39963239 PMCID: PMC11830733 DOI: 10.3389/fphar.2025.1534634] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Accepted: 01/15/2025] [Indexed: 02/20/2025] Open
Abstract
Diabetes mellitus (DM) ranks among the most prevalent chronic metabolic diseases, characterized primarily by a persistent elevation in blood glucose levels. This condition typically stems from either insufficient insulin secretion or a functional defect in the insulin itself. Clinically, diabetes is primarily classified into type 1 diabetes mellitus (T1DM) and type 2 diabetes mellitus (T2DM), with T2DM comprising nearly 90% of all diagnosed cases. Notably, the global incidence of T2DM has surged dramatically over recent decades. The adenylate-activated protein kinase (AMPK) signaling pathway is crucial in regulating cellular energy metabolism, marking it as a significant therapeutic target for diabetes and related complications. Natural products, characterized by their diverse origins, multifaceted bioactivities, and relative safety, hold considerable promise in modulating the AMPK pathway. This review article explores the advances in research on natural products that target the AMPK signaling pathway, aiming to inform the development of innovative antidiabetic therapies.
Collapse
Affiliation(s)
- Min Li
- Northeast Asia Research Institute of Traditional Chinese Medicine, Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efcacy, Ministry of Education, Jilin Provincial Key Laboratory of Bio-Macromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Jilin, China
| | - Lu Ding
- Northeast Asia Research Institute of Traditional Chinese Medicine, Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efcacy, Ministry of Education, Jilin Provincial Key Laboratory of Bio-Macromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Jilin, China
- Research Center of Traditional Chinese Medicine, The Affiliated Hospital to Changchun University of Chinese Medicine, Jilin, China
| | - Liyuan Cao
- Northeast Asia Research Institute of Traditional Chinese Medicine, Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efcacy, Ministry of Education, Jilin Provincial Key Laboratory of Bio-Macromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Jilin, China
| | - Zepeng Zhang
- Research Center of Traditional Chinese Medicine, The Affiliated Hospital to Changchun University of Chinese Medicine, Jilin, China
| | - Xueyan Li
- Northeast Asia Research Institute of Traditional Chinese Medicine, Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efcacy, Ministry of Education, Jilin Provincial Key Laboratory of Bio-Macromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Jilin, China
| | - Zirui Li
- Northeast Asia Research Institute of Traditional Chinese Medicine, Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efcacy, Ministry of Education, Jilin Provincial Key Laboratory of Bio-Macromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Jilin, China
| | - Qinjing Xia
- Northeast Asia Research Institute of Traditional Chinese Medicine, Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efcacy, Ministry of Education, Jilin Provincial Key Laboratory of Bio-Macromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Jilin, China
| | - Kai Yin
- Northeast Asia Research Institute of Traditional Chinese Medicine, Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efcacy, Ministry of Education, Jilin Provincial Key Laboratory of Bio-Macromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Jilin, China
| | - Siyu Song
- Northeast Asia Research Institute of Traditional Chinese Medicine, Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efcacy, Ministry of Education, Jilin Provincial Key Laboratory of Bio-Macromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Jilin, China
| | - Zihan Wang
- Northeast Asia Research Institute of Traditional Chinese Medicine, Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efcacy, Ministry of Education, Jilin Provincial Key Laboratory of Bio-Macromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Jilin, China
| | - Haijian Du
- Northeast Asia Research Institute of Traditional Chinese Medicine, Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efcacy, Ministry of Education, Jilin Provincial Key Laboratory of Bio-Macromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Jilin, China
| | - Daqing Zhao
- Northeast Asia Research Institute of Traditional Chinese Medicine, Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efcacy, Ministry of Education, Jilin Provincial Key Laboratory of Bio-Macromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Jilin, China
| | - Xiangyan Li
- Northeast Asia Research Institute of Traditional Chinese Medicine, Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efcacy, Ministry of Education, Jilin Provincial Key Laboratory of Bio-Macromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Jilin, China
| | - Zeyu Wang
- Northeast Asia Research Institute of Traditional Chinese Medicine, Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efcacy, Ministry of Education, Jilin Provincial Key Laboratory of Bio-Macromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Jilin, China
- Research Center of Traditional Chinese Medicine, The Affiliated Hospital to Changchun University of Chinese Medicine, Jilin, China
| |
Collapse
|
7
|
Zhou Y, Zhang Y, Li Y, Liu L, Zhuang M, Xiao Y. IL-27 attenuated macrophage injury and inflammation induced by Mycobacterium tuberculosis by activating autophagy. In Vitro Cell Dev Biol Anim 2025; 61:245-256. [PMID: 39455490 DOI: 10.1007/s11626-024-00989-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 10/14/2024] [Indexed: 10/28/2024]
Abstract
Interleukin-27 (IL-27) is a cytokine that is reported to be highly expressed in the peripheral blood of patients with pulmonary tuberculosis (PTB). IL-27-mediated signaling pathways, which exhibit anti- Mycobacterium tuberculosis (Mtb) properties, have also been demonstrated in macrophages infected with Mtb. However, the exact mechanism remains unclear. This study aimed to clarify the potential molecular mechanisms through which IL-27 enhances macrophage resistance to Mtb infection. Both normal and PTB patients provided bronchoalveolar lavage fluid (BALF). Peripheral blood mononuclear cells (PBMCs) were isolated from healthy individuals and stimulated with 50 ng/mL macrophage-colony stimulating factor (M-CSF) to obtain monocyte-derived macrophages (MDMs). Using 100 ng/mL phorbol 12-myristate 13-acetate (PMA), THP-1 cells were induced to differentiate into THP-1-derived macrophage-like cells (TDMs). Both MDMs and TDMs were subsequently infected with the Mtb strain H37Rv and treated with 50 ng/mL IL-27 prior to infection. The damage and inflammation of macrophages were examined using flow cytometry, enzyme-linked immunosorbent assay (ELISA), and Western blotting. Patients with PTB had elevated levels of IL-27 in their BALF. Preconditioning with IL-27 was shown to reduce H37Rv-induced MDMs and TDMs apoptosis while also decreasing the levels of Cleaved Caspase-3, Bax and the proinflammatory cytokines TNF-α, IL-1β, and IL-6, promoting the expression of Bcl-2 and the anti-inflammatory factors IL-10 and IL-4. Silencing of the IL-27 receptor IL-27Ra increased macrophage damage and inflammation triggered by H37Rv. Mechanistically, IL-27 activates autophagy by inhibiting TLR4/NF-κB signaling and activating the PI3K/AKT signaling pathway, thereby inhibiting H37Rv-induced macrophage apoptosis and the inflammatory response. Our study suggests that IL-27 alleviates H37Rv-induced macrophage injury and the inflammatory response by activating autophagy and that IL-27 may be a new target for the treatment of PTB.
Collapse
Affiliation(s)
- Yushan Zhou
- Respiratory and Critical Care Medicine Ward 1, Yan'an Hospital Affiliated to Kunming Medical University, Kunming, 650051, Yunnan, China
| | - Yuxuan Zhang
- Respiratory and Critical Care Medicine Ward 1, Yan'an Hospital Affiliated to Kunming Medical University, Kunming, 650051, Yunnan, China
| | - Yanli Li
- Respiratory and Critical Care Medicine Ward 1, Yan'an Hospital Affiliated to Kunming Medical University, Kunming, 650051, Yunnan, China
| | - Liqiong Liu
- Respiratory and Critical Care Medicine Ward 1, Yan'an Hospital Affiliated to Kunming Medical University, Kunming, 650051, Yunnan, China
| | - Min Zhuang
- Respiratory and Critical Care Medicine Ward 1, Yan'an Hospital Affiliated to Kunming Medical University, Kunming, 650051, Yunnan, China
| | - Yi Xiao
- Respiratory and Critical Care Medicine Ward 1, Yan'an Hospital Affiliated to Kunming Medical University, Kunming, 650051, Yunnan, China.
| |
Collapse
|
8
|
Alshawaf E, Marafie SK, Abu-Farha M, Albatineh AN, Alramah T, Albuhairi A, Al Qassar Y, Zinoun R, Shalabi R, Behbehani S, Mohammed D, Alajmi F, Abdalla MA, Al-Ozairi E, Shehab M, Abdul-Ghani M, Al-Mulla F, Abubaker J. Circulating Isthmin-1 Levels and Their Relationship with Diabetes and Metabolic Diseases in Kuwaiti Adults. Biomedicines 2025; 13:101. [PMID: 39857685 PMCID: PMC11762165 DOI: 10.3390/biomedicines13010101] [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: 12/16/2024] [Revised: 12/30/2024] [Accepted: 01/02/2025] [Indexed: 01/27/2025] Open
Abstract
BACKGROUND/OBJECTIVES Obesity and type 2 diabetes (T2D) are associated with significant alterations in various metabolic biomarkers. Isthmin-1 (Ism1) has recently emerged as a potential marker of metabolic health and was shown in animal studies to associate with metabolic-associated fatty liver disease (MAFLD). In this study, we aimed to investigate the circulatory levels of Ism1 in individuals with obesity compared to non-obese individuals and evaluate their association with insulin resistance, MAFLD, and T2D. The primary outcomes of this study are obesity, insulin resistance, MAFLD, and T2D, while the secondary outcome is hypertension; Methods: This is a cross-sectional study involving 450 participants, who were divided based on their obesity status into people with obesity (n = 182) and those without obesity (n = 265). Circulating Ism1 levels were measured by ELISA and were compared between the groups. Insulin resistance was assessed using the homeostasis model assessment of insulin resistance (HOMA-IR), and fatty liver was evaluated using Fibroscan; Results: Our results showed a significant reduction in circulating Ism1 levels in individuals with obesity (p-value = 0.002). Ism1 levels were negatively associated with the odds of T2D, possibly suggesting a protective role. Additionally, individuals with higher CAP scores demonstrated significantly lower Ism1 levels, and the Spearman's rank correlation revealed a negative association between Ism1 and both CAP scores (r = -0.109, p-value = 0.025) and insulin resistance (r = -0.141, p-value = 0.004). Logistic regression analysis further supported Ism1 as an independent significant protective factor against obesity-related metabolic dysfunction. This significance persisted after adjusting for several confounders. Furthermore, our ROC results indicate that circulatory Ism1 levels possess significant diagnostic capability for identifying individuals with obesity-related metabolic imbalances with an area under the curve of 0.764 (95% CI = 0.718, 0.811). Finally, the adjusted multinomial analysis suggested that higher levels of Ism1 may play a protective role against pre-diabetes (AOR = 0.88, 95% CI = 0.838, 0.925) and T2D (AOR = 0.87, 95% CI = 0.814, 0.934); Conclusions: This study suggests that reduced Ism1 levels are linked to increased insulin resistance, MAFLD, and T2D in obese individuals. Our findings further corroborate the protective role of Ism1 and highlight its potential utility as a biomarker for monitoring obesity-related metabolic diseases.
Collapse
Affiliation(s)
- Eman Alshawaf
- Department of Biochemistry and Molecular Biology, Dasman Diabetes Institute, Dasman 15462, Kuwait; (E.A.); (S.K.M.); (M.A.-F.); (T.A.); (A.A.); (Y.A.Q.); (R.Z.); (R.S.); (S.B.); (D.M.)
| | - Sulaiman K. Marafie
- Department of Biochemistry and Molecular Biology, Dasman Diabetes Institute, Dasman 15462, Kuwait; (E.A.); (S.K.M.); (M.A.-F.); (T.A.); (A.A.); (Y.A.Q.); (R.Z.); (R.S.); (S.B.); (D.M.)
| | - Mohamed Abu-Farha
- Department of Biochemistry and Molecular Biology, Dasman Diabetes Institute, Dasman 15462, Kuwait; (E.A.); (S.K.M.); (M.A.-F.); (T.A.); (A.A.); (Y.A.Q.); (R.Z.); (R.S.); (S.B.); (D.M.)
- Department of Translational Research, Dasman Diabetes Institute, Dasman 15462, Kuwait; (F.A.); (M.A.A.); (M.A.-G.)
| | - Ahmed N. Albatineh
- Department of Community and Population Health, College of Health, Lehigh University, Bethlehem, PA 18015, USA;
| | - Tahani Alramah
- Department of Biochemistry and Molecular Biology, Dasman Diabetes Institute, Dasman 15462, Kuwait; (E.A.); (S.K.M.); (M.A.-F.); (T.A.); (A.A.); (Y.A.Q.); (R.Z.); (R.S.); (S.B.); (D.M.)
| | - Aldana Albuhairi
- Department of Biochemistry and Molecular Biology, Dasman Diabetes Institute, Dasman 15462, Kuwait; (E.A.); (S.K.M.); (M.A.-F.); (T.A.); (A.A.); (Y.A.Q.); (R.Z.); (R.S.); (S.B.); (D.M.)
| | - Yafa Al Qassar
- Department of Biochemistry and Molecular Biology, Dasman Diabetes Institute, Dasman 15462, Kuwait; (E.A.); (S.K.M.); (M.A.-F.); (T.A.); (A.A.); (Y.A.Q.); (R.Z.); (R.S.); (S.B.); (D.M.)
| | - Reem Zinoun
- Department of Biochemistry and Molecular Biology, Dasman Diabetes Institute, Dasman 15462, Kuwait; (E.A.); (S.K.M.); (M.A.-F.); (T.A.); (A.A.); (Y.A.Q.); (R.Z.); (R.S.); (S.B.); (D.M.)
| | - Rawan Shalabi
- Department of Biochemistry and Molecular Biology, Dasman Diabetes Institute, Dasman 15462, Kuwait; (E.A.); (S.K.M.); (M.A.-F.); (T.A.); (A.A.); (Y.A.Q.); (R.Z.); (R.S.); (S.B.); (D.M.)
| | - Sarah Behbehani
- Department of Biochemistry and Molecular Biology, Dasman Diabetes Institute, Dasman 15462, Kuwait; (E.A.); (S.K.M.); (M.A.-F.); (T.A.); (A.A.); (Y.A.Q.); (R.Z.); (R.S.); (S.B.); (D.M.)
| | - Dalal Mohammed
- Department of Biochemistry and Molecular Biology, Dasman Diabetes Institute, Dasman 15462, Kuwait; (E.A.); (S.K.M.); (M.A.-F.); (T.A.); (A.A.); (Y.A.Q.); (R.Z.); (R.S.); (S.B.); (D.M.)
| | - Fahad Alajmi
- Department of Translational Research, Dasman Diabetes Institute, Dasman 15462, Kuwait; (F.A.); (M.A.A.); (M.A.-G.)
| | - Mohammed A. Abdalla
- Department of Translational Research, Dasman Diabetes Institute, Dasman 15462, Kuwait; (F.A.); (M.A.A.); (M.A.-G.)
| | - Ebaa Al-Ozairi
- Clinical Research Unit, Dasman Diabetes Institute, Dasman 15462, Kuwait;
| | - Mohammad Shehab
- Division of Gastroenterology, Department of Internal Medicine, Mubarak Alkabeer University Hospital, Kuwait University, Kuwait City 31470, Kuwait;
| | - Muhammad Abdul-Ghani
- Department of Translational Research, Dasman Diabetes Institute, Dasman 15462, Kuwait; (F.A.); (M.A.A.); (M.A.-G.)
- Division of Diabetes, University of Texas Health Science Center, San Antonio, TX 78229, USA
| | - Fahd Al-Mulla
- Department of Translational Research, Dasman Diabetes Institute, Dasman 15462, Kuwait; (F.A.); (M.A.A.); (M.A.-G.)
| | - Jehad Abubaker
- Department of Biochemistry and Molecular Biology, Dasman Diabetes Institute, Dasman 15462, Kuwait; (E.A.); (S.K.M.); (M.A.-F.); (T.A.); (A.A.); (Y.A.Q.); (R.Z.); (R.S.); (S.B.); (D.M.)
| |
Collapse
|
9
|
Hong S, Heo H, Kim HJ, Jeong HS, Lee H, Lee J. Avenanthramides Ameliorate Insulin Resistance by Modulating Gluconeogenesis and Glycogen Synthesis in HepG2 Cells. J Med Food 2025; 28:18-27. [PMID: 39527253 DOI: 10.1089/jmf.2024.k.0199] [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] [Indexed: 11/16/2024] Open
Abstract
Diabetes mellitus (DM) is a multifaceted metabolic condition, mainly defined by elevated blood glucose levels. A feature of type 2 DM includes insulin resistance (IR), which involves impairments within the insulin signaling pathways. Avenanthramides (AVNs) are phenolic alkaloids found in Avena sativa L. The major AVNs are AVN A, AVN B, and AVN C. They have been reported to offer benefits in preventing inflammation, cancer, and cardiovascular diseases. However, the effects of AVNs on the liver glucose metabolism pathways remain unknown. This study examined the effects and underlying mechanisms through which AVNs alleviate IR induced by free fatty acid (FFA) in HepG2 cells. The results indicated that FFA treatment significantly decreased glucose consumption by 34.54% compared to the control. However, treatments with AVN A, B, and C at 100 μM increased glucose uptake by 57.93%, 58.28%, and 53.10%, respectively, compared to FFA treatment alone. This effect occurs through the increased expression of glucose transporter 4. Furthermore, AVNs significantly enhanced the glycogen content. AVNs induced increased phosphorylation of insulin receptor substrate-1 (IRS-1), phosphatidylinositol-3-kinase (PI3K), and protein kinase B (Akt). AVNs treatment decreased the levels of phosphoenolpyruvate carboxykinase and glucose-6-phosphatase in HepG2 cells. This effect was attributed to AMP-activated protein kinase activation and inhibition of forkhead box protein O1. Collectively, these results suggest that AVNs regulate glucose metabolism by activating the IRS-1/PI3K/Akt pathway, which is related to glycogen synthesis, and by inhibiting key molecules that promote gluconeogenesis.
Collapse
Affiliation(s)
- Seonghwa Hong
- Department of Food Science and Biotechnology, Chungbuk National University, Cheongju, Korea
| | - Huijin Heo
- Department of Food Science and Biotechnology, Chungbuk National University, Cheongju, Korea
| | - Hyun-Joo Kim
- Department of Central Area Crop Science, National Institute of Crop Science, Rural Development Administration, Suwon, Korea
| | - Heon Sang Jeong
- Department of Food Science and Biotechnology, Chungbuk National University, Cheongju, Korea
| | - Hana Lee
- Department of Food Science and Biotechnology, Chungbuk National University, Cheongju, Korea
| | - Junsoo Lee
- Department of Food Science and Biotechnology, Chungbuk National University, Cheongju, Korea
| |
Collapse
|
10
|
Ren W, Huang Y, Meng S, Cao Z, Qin N, Zhao J, Huang T, Guo X, Chen X, Zhou Z, Zhu Y, Yu L, Wang H. Salidroside treatment decreases the susceptibility of atrial fibrillation in diabetic mice by reducing mTOR-STAT3-MCP-1 signaling and atrial inflammation. Int Immunopharmacol 2024; 142:113196. [PMID: 39306893 DOI: 10.1016/j.intimp.2024.113196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 07/19/2024] [Accepted: 09/14/2024] [Indexed: 10/12/2024]
Abstract
Atrial fibrillation (AF) is the most common sustained cardiac arrhythmia in clinic, and type 2 diabetes mellitus (T2DM) is an independent risk factor for AF. Salidroside (Sal), the active ingredient of the Rhodiola rosea, has hypoglycemic, anti-inflammatory, anti-fibrotic and anti-arrhythmic effects. The aim of this study is to investigate the effects and underlying molecular mechanisms of Sal on T2DM associated atrial inflammation and the pathogenesis of AF. In the in vivo study, T2DM mice model was established by high-fat diet and intraperitoneal injection of streptozotocin (STZ). Sal (25 mg/kg/d, 50 mg/kg/d, and 100 mg/kg/d) was administered orally for 4 weeks. T2DM caused atrial electrical and structural remodeling and significantly increased the susceptibility of AF. Meanwhile, mTOR-STAT3-MCP-1 signaling and inflammatory markers were also significantly enhanced in diabetic atria. However, Sal dose-dependently ameliorated cardiac dysfunction, mitigated atrial structural and electrical remodeling, and reduced atrial inflammation. Moreover, Sal-treated group exhibited remarkably down-regulated activity of mTOR-STAT3-MCP-1 pathway, and decreased atrial monocyte/macrophage infiltration. In palmitic acid (PA)-challenged HL-1 cells, Sal attenuated cytotoxicity, downregulated the expressions of TNF-α, IL-6, MCP-1, and inhibited the activation of mTOR-STAT3 signaling. However, co-treatment with MHY1485 (a mTOR agonist) reversed these effects. Taken together, the present study demonstrates that Sal treatment decreases the susceptibility of AF in diabetic mice by reducing mTOR-STAT3-MCP-1 signaling and atrial monocyte/macrophage infiltration. Sal treatment may represent a novel preventive therapy for cardiac arrhythmia and atrial fibrillation in diabetic patients.
Collapse
Affiliation(s)
- Wenpu Ren
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Department of Cardiovascular Surgery, General Hospital of Northern Theater Command, 83 Wenhua Road, Shenyang, Liaoning 110016, PR China; Liaoning University of Traditional Chinese Medicine, Shenyang, Liaoning 110847, PR China
| | - Yuting Huang
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Department of Cardiovascular Surgery, General Hospital of Northern Theater Command, 83 Wenhua Road, Shenyang, Liaoning 110016, PR China
| | - Shan Meng
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Department of Cardiovascular Surgery, General Hospital of Northern Theater Command, 83 Wenhua Road, Shenyang, Liaoning 110016, PR China; Jinzhou Medical University, Jinzhou, Liaoning 121001, PR China
| | - Zijun Cao
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Department of Cardiovascular Surgery, General Hospital of Northern Theater Command, 83 Wenhua Road, Shenyang, Liaoning 110016, PR China; Liaoning University of Traditional Chinese Medicine, Shenyang, Liaoning 110847, PR China
| | - Nana Qin
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Department of Cardiovascular Surgery, General Hospital of Northern Theater Command, 83 Wenhua Road, Shenyang, Liaoning 110016, PR China; Liaoning University of Traditional Chinese Medicine, Shenyang, Liaoning 110847, PR China
| | - Jikai Zhao
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Department of Cardiovascular Surgery, General Hospital of Northern Theater Command, 83 Wenhua Road, Shenyang, Liaoning 110016, PR China
| | - Tao Huang
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Department of Cardiovascular Surgery, General Hospital of Northern Theater Command, 83 Wenhua Road, Shenyang, Liaoning 110016, PR China
| | - Xiaodong Guo
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Department of Cardiovascular Surgery, General Hospital of Northern Theater Command, 83 Wenhua Road, Shenyang, Liaoning 110016, PR China
| | - Xin Chen
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Department of Cardiovascular Surgery, General Hospital of Northern Theater Command, 83 Wenhua Road, Shenyang, Liaoning 110016, PR China; Jinzhou Medical University, Jinzhou, Liaoning 121001, PR China
| | - Zijun Zhou
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Department of Cardiovascular Surgery, General Hospital of Northern Theater Command, 83 Wenhua Road, Shenyang, Liaoning 110016, PR China
| | - Yan Zhu
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Department of Cardiovascular Surgery, General Hospital of Northern Theater Command, 83 Wenhua Road, Shenyang, Liaoning 110016, PR China.
| | - Liming Yu
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Department of Cardiovascular Surgery, General Hospital of Northern Theater Command, 83 Wenhua Road, Shenyang, Liaoning 110016, PR China.
| | - Huishan Wang
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Department of Cardiovascular Surgery, General Hospital of Northern Theater Command, 83 Wenhua Road, Shenyang, Liaoning 110016, PR China.
| |
Collapse
|
11
|
Cheng S, Meng X, Wang Z, Lan T, Zhou Z, Wang Z, Abliz Z. Mass Spectrometry Imaging Reveals Spatial Metabolic Alterations and Salidroside's Effects in Diabetic Encephalopathy. Metabolites 2024; 14:670. [PMID: 39728451 DOI: 10.3390/metabo14120670] [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: 11/03/2024] [Revised: 11/23/2024] [Accepted: 11/25/2024] [Indexed: 12/28/2024] Open
Abstract
Background: Diabetic encephalopathy (DE) is a neurological complication of diabetes marked by cognitive decline and complex metabolic disturbances. Salidroside (SAL), a natural compound with antioxidant and neuroprotective properties, has shown promise in alleviating diabetic complications. Exploring the spatial metabolic reprogramming in DE and elucidating SAL's metabolic effects are critical for deepening our understanding of its pathogenesis and developing effective therapeutic strategies. Methods: Air-flow-assisted desorption electrospray ionization-mass spectrometry imaging (AFADESI-MSI) was employed to investigate spatial metabolic alterations in the brains of db/db mice, a spontaneous DE model. The mice were treated with SAL (30 and 150 mg/kg, orally) for 12 weeks. Differential metabolites were identified and characterized using high-resolution mass spectrometry and validated against public databases. Results: Our AFADESI-MSI analysis revealed significant changes in 26 metabolites in the brains of DE mice compared to the controls. These metabolic changes indicated disruptions in glucose, glutamate-glutamine, nucleotide, lipid, choline, aspartate, and L-carnitine metabolism. Notably, glucose 6-phosphate (G6P), glutamine, adenosine, L-carnitine, and choline exhibited similar trends in both db/db mice and STZ-induced rat models of DE, suggesting their potential as reliable biomarkers. Twelve weeks of SAL treatment demonstrated a positive regulatory effect on glucose metabolism, the glutamate-glutamine cycle, and lipid metabolism. Conclusions: This study identifies key metabolic alterations in DE and demonstrates the therapeutic potential of SAL in modulating these disturbances, offering valuable insights for targeted interventions in diabetic complications.
Collapse
Affiliation(s)
- Shuohan Cheng
- Key Laboratory of Mass Spectrometry Imaging and Metabolomics, Minzu University of China, National Ethnic Affairs Commission, Beijing 100081, China
- Key Laboratory of Ethnomedicine of Ministry of Education, School of Pharmacy, Minzu University of China, Beijing 100081, China
| | - Xianyue Meng
- Key Laboratory of Mass Spectrometry Imaging and Metabolomics, Minzu University of China, National Ethnic Affairs Commission, Beijing 100081, China
- Center for Imaging and Systems Biology, College of Life and Environmental Sciences, Minzu University of China, 27 Zhongguancun South Avenue, Beijing 100081, China
| | - Zixuan Wang
- Key Laboratory of Mass Spectrometry Imaging and Metabolomics, Minzu University of China, National Ethnic Affairs Commission, Beijing 100081, China
- Center for Imaging and Systems Biology, College of Life and Environmental Sciences, Minzu University of China, 27 Zhongguancun South Avenue, Beijing 100081, China
| | - Tianfang Lan
- Key Laboratory of Mass Spectrometry Imaging and Metabolomics, Minzu University of China, National Ethnic Affairs Commission, Beijing 100081, China
- Center for Imaging and Systems Biology, College of Life and Environmental Sciences, Minzu University of China, 27 Zhongguancun South Avenue, Beijing 100081, China
| | - Zhi Zhou
- Key Laboratory of Mass Spectrometry Imaging and Metabolomics, Minzu University of China, National Ethnic Affairs Commission, Beijing 100081, China
- Center for Imaging and Systems Biology, College of Life and Environmental Sciences, Minzu University of China, 27 Zhongguancun South Avenue, Beijing 100081, China
| | - Zhonghua Wang
- Key Laboratory of Mass Spectrometry Imaging and Metabolomics, Minzu University of China, National Ethnic Affairs Commission, Beijing 100081, China
- Center for Imaging and Systems Biology, College of Life and Environmental Sciences, Minzu University of China, 27 Zhongguancun South Avenue, Beijing 100081, China
| | - Zeper Abliz
- Key Laboratory of Mass Spectrometry Imaging and Metabolomics, Minzu University of China, National Ethnic Affairs Commission, Beijing 100081, China
- Key Laboratory of Ethnomedicine of Ministry of Education, School of Pharmacy, Minzu University of China, Beijing 100081, China
- Center for Imaging and Systems Biology, College of Life and Environmental Sciences, Minzu University of China, 27 Zhongguancun South Avenue, Beijing 100081, China
| |
Collapse
|
12
|
Zhong RF, Liu CJ, Hao KX, Fan XD, Jiang JG. Polysaccharides from Flos Sophorae Immaturus ameliorates insulin resistance in IR-HepG2 cells by co-regulating signaling pathways of AMPK and IRS-1/PI3K/AKT. Int J Biol Macromol 2024; 280:136088. [PMID: 39366625 DOI: 10.1016/j.ijbiomac.2024.136088] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Revised: 09/18/2024] [Accepted: 09/25/2024] [Indexed: 10/06/2024]
Abstract
Four polysaccharides, named FSIP, FSIP-I, FSIP-II and FSIP-III, were isolated from Flos Sophorae Immaturus. Structure characterization revealed that FSIP-I and FSIP-II were types of AG-II-like polysaccharides while FSIP-III featured a RG-II-like structure with high content of GalpA. In vitro experiments showed that FSIPs upregulated HK and PK activities in glycolysis while downregulated G-6-Pase activities in gluconeogenesis. This increased glucose utilization while decreased the glucose synthesis in IR-HepG2 cells, potentially reducing elevated blood sugar levels induced by excess insulin. In terms of antioxidant system, FSIPs decreased the levels of ROS and MDA, and increased the activities of SOD and CAT, enhancing antioxidant capacity to counteract damage caused by insulin resistance in IR-HepG2 cells. To further explore the mechanism, related genes expressions were analyzed. The results found that FSIPs ameliorated insulin resistance via regulating AMPK and IRS-1/PI3K/AKT signal pathways. In the case of AMPK, glucose can be channeled into oxidative (catabolic) pathway, whereas, in the case of IRS-1/PI3K/AKT, glucose can be stored as glycogen (anabolic). This co-modulation could ameliorate insulin resistance by upregulating the glycolysis and repressing the gluconeogenesis in catabolism, and upregulating the glycogen synthesis in anabolism. Additionally, FSIP-III exhibited better anti-insulin resistance activity, attributed to its high content of GalpA.
Collapse
Affiliation(s)
- Rui-Fang Zhong
- College of Food and Bioengineering, South China University of Technology, Guangzhou 510640, China
| | - Chang-Jun Liu
- College of Food and Bioengineering, South China University of Technology, Guangzhou 510640, China
| | - Ke-Xin Hao
- College of Food and Bioengineering, South China University of Technology, Guangzhou 510640, China
| | - Xiao-Dan Fan
- College of Food and Bioengineering, South China University of Technology, Guangzhou 510640, China.
| | - Jian-Guo Jiang
- College of Food and Bioengineering, South China University of Technology, Guangzhou 510640, China.
| |
Collapse
|
13
|
Seok JK, Yang G, Jee JI, Kang HC, Cho YY, Lee HS, Lee JY. Hepatocyte-specific RIG-I loss attenuates metabolic dysfunction-associated steatotic liver disease in mice via changes in mitochondrial respiration and metabolite profiles. Toxicol Res 2024; 40:683-695. [PMID: 39345739 PMCID: PMC11436585 DOI: 10.1007/s43188-024-00264-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 09/09/2024] [Accepted: 09/10/2024] [Indexed: 10/01/2024] Open
Abstract
Pattern recognition receptor (PRR)-mediated inflammation is an important determinant of the initiation and progression of metabolic diseases such as metabolic dysfunction-associated steatotic liver disease (MASLD). In this study, we investigated whether RIG-I is involved in hepatic metabolic reprogramming in a high-fat diet (HFD)-induced MASLD model in hepatocyte-specific RIG-I-KO (RIG-I∆hep) mice. Our study revealed that hepatic deficiency of RIG-I improved HFD-induced metabolic imbalances, including glucose impairment and insulin resistance. Hepatic steatosis and liver triglyceride levels were reduced in RIG-I-deficient hepatocytes in HFD-induced MASLD mice, and this was accompanied by the reduced expression of lipogenesis genes, such as PPARγ, Dga2, and Pck1. Hepatic RIG-I deficiency alters whole-body metabolic rates in the HFD-induced MASLD model; there is higher energy consumption in RIG-I∆hep mice. Deletion of RIG-I activated glycolysis and tricarboxylic acid (TCA) cycle-related metabolites in hepatocytes from both HFD-induced MASLD mice and methionine-choline-deficient diet (MCD)-fed mice. RIG-I deficiency enhanced AMPK activation and mitochondrial function in hepatocytes from HFD-induced MASLD mice. These findings indicate that deletion of RIG-I can activate cellular metabolism in hepatocytes by switching on both glycolysis and mitochondrial respiration, resulting in metabolic changes induced by a HFD and stimulation of mitochondrial activity. In summary, RIG-I may be a key regulator of cellular metabolism that influences the development of metabolic diseases such as MASLD. Supplementary Information The online version contains supplementary material available at 10.1007/s43188-024-00264-x.
Collapse
Affiliation(s)
- Jin Kyung Seok
- College of Pharmacy, The Catholic University of Korea, Bucheon, 14662 Republic of Korea
| | - Gabsik Yang
- Department of Pharmacology, College of Korean Medicine, Woosuk University, Jeonbuk, 55338 Republic of Korea
| | - Jung In Jee
- College of Pharmacy, The Catholic University of Korea, Bucheon, 14662 Republic of Korea
| | - Han Chang Kang
- College of Pharmacy, The Catholic University of Korea, Bucheon, 14662 Republic of Korea
| | - Yong-Yeon Cho
- College of Pharmacy, The Catholic University of Korea, Bucheon, 14662 Republic of Korea
| | - Hye Suk Lee
- College of Pharmacy, The Catholic University of Korea, Bucheon, 14662 Republic of Korea
| | - Joo Young Lee
- College of Pharmacy, The Catholic University of Korea, Bucheon, 14662 Republic of Korea
| |
Collapse
|
14
|
Wang L, Chi EZ, Zhao XH. Valorization of cell wall polysaccharides extracted from Liubao brick tea residues: chemical, structural, and hypoglycemic properties. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2024; 104:6933-6946. [PMID: 38597456 DOI: 10.1002/jsfa.13526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 01/29/2024] [Accepted: 04/10/2024] [Indexed: 04/11/2024]
Abstract
BACKGROUND Tea dregs, typically generated during the production of instant tea or tea beverages, have conventionally been regarded as waste material and routinely discarded. Nevertheless, contemporary research endeavors are concentrating on discovering efficient methods for utilizing the potential of this discarded resource. RESULTS In this study, we employed a sequential extraction method using chemical chelating agents to extract and isolate four distinct cell wall polysaccharides, designated as CWTPS-1 through CWTPS-4, from the tea dregs of Liubao brick tea. A comprehensive investigation into their physicochemical, structural, and hypoglycemic properties was conducted. The analysis of chemical composition and physicochemical characteristics revealed that all four CWTPSs were characterized as acidic polysaccharides, albeit with varying chemical compositions and physicochemical attributes. Specifically, the xyloglucan fractions, CWTPS-3 and CWTPS-4, were found to be rich in glucose and xylose, displaying a more uniform molecular weight distribution, greater structural stability, and a more irregular surface compared to the others. Moreover, they exhibited a higher diversity of monosaccharide residues. Importantly, our research unveiled that all four CWTPSs exhibited the capacity to modulate key glucose-regulated and antioxidant enzyme activities within HepG2 cells via the IRS-1-PI3K/AKT signaling pathway, thereby ameliorating cellular insulin resistance. Furthermore, our correlation analysis highlighted significant associations between monosaccharide composition and neutral sugar content with the observed hypoglycemic activity of CWTPSs. CONCLUSION This study highlights the potential of utilizing tea dregs as a valuable resource, making a significant contribution to the advancement of the tea industry. Furthermore, CWTPS-4 exhibits promising prospects for further development as a functional food ingredient or additive. © 2024 Society of Chemical Industry.
Collapse
Affiliation(s)
- Li Wang
- School of Biology and Food Engineering, Guangdong University of Petrochemical Technology, Maoming, China
| | - En-Zhong Chi
- School of Biology and Food Engineering, Guangdong University of Petrochemical Technology, Maoming, China
| | - Xin-Huai Zhao
- School of Biology and Food Engineering, Guangdong University of Petrochemical Technology, Maoming, China
| |
Collapse
|
15
|
Taheri R, Mokhtari Y, Yousefi AM, Bashash D. The PI3K/Akt signaling axis and type 2 diabetes mellitus (T2DM): From mechanistic insights into possible therapeutic targets. Cell Biol Int 2024; 48:1049-1068. [PMID: 38812089 DOI: 10.1002/cbin.12189] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 02/03/2024] [Accepted: 05/12/2024] [Indexed: 05/31/2024]
Abstract
Type 2 diabetes mellitus (T2DM) is an immensely debilitating chronic disease that progressively undermines the well-being of various bodily organs and, indeed, most patients succumb to the disease due to post-T2DM complications. Although there is evidence supporting the activation of the phosphoinositide 3-kinase (PI3K)/Akt signaling pathway by insulin, which is essential in regulating glucose metabolism and insulin resistance, the significance of this pathway in T2DM has only been explored in a few studies. The current review aims to unravel the mechanisms by which different classes of PI3Ks control the metabolism of glucose; and also to discuss the original data obtained from international research laboratories on this topic. We also summarized the role of the PI3K/Akt signaling axis in target tissues spanning from the skeletal muscle to the adipose tissue and liver. Furthermore, inquiries regarding the impact of disrupting this axis on insulin function and the development of insulin resistance have been addressed. We also provide a general overview of the association of impaired PI3K/Akt signaling pathways in the pathogenesis of the most prevalent diabetes-related complications. The last section provides a special focus on the therapeutic potential of this axis by outlining the latest advances in active compounds that alleviate diabetes via modulation of the PI3K/Akt pathway. Finally, we comment on the future research aspects in which the field of T2DM therapies using PI3K modulators might be developed.
Collapse
Affiliation(s)
- Rana Taheri
- Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Yazdan Mokhtari
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Amir-Mohammad Yousefi
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Davood Bashash
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
16
|
Al Mamun A, Shao C, Geng P, Wang S, Xiao J. Pyroptosis in Diabetic Peripheral Neuropathy and its Therapeutic Regulation. J Inflamm Res 2024; 17:3839-3864. [PMID: 38895141 PMCID: PMC11185259 DOI: 10.2147/jir.s465203] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 06/05/2024] [Indexed: 06/21/2024] Open
Abstract
Pyroptosis is a pro-inflammatory form of cell death resulting from the activation of gasdermins (GSDMs) pore-forming proteins and the release of several pro-inflammatory factors. However, inflammasomes are the intracellular protein complexes that cleave gasdermin D (GSDMD), leading to the formation of robust cell membrane pores and the initiation of pyroptosis. Inflammasome activation and gasdermin-mediated membrane pore formation are the important intrinsic processes in the classical pyroptotic signaling pathway. Overactivation of the NOD-like receptor thermal protein domain associated protein 3 (NLRP3) inflammasome triggers pyroptosis and amplifies inflammation. Current evidence suggests that the overactivation of inflammasomes and pyroptosis may further induce the progression of cancers, nerve injury, inflammatory disorders and metabolic dysfunctions. Current evidence also indicates that pyroptosis-dependent cell death accelerates the progression of diabetes and its frequent consequences including diabetic peripheral neuropathy (DPN). Pyroptosis-mediated inflammatory reaction further exacerbates DPN-mediated CNS injury. Accumulating evidence shows that several molecular signaling mechanisms trigger pyroptosis in insulin-producing cells, further leading to the development of DPN. Numerous studies have suggested that certain natural compounds or drugs may possess promising pharmacological properties by modulating inflammasomes and pyroptosis, thereby offering potential preventive and practical therapeutic approaches for the treatment and management of DPN. This review elaborates on the underlying molecular mechanisms of pyroptosis and explores possible therapeutic strategies for regulating pyroptosis-regulated cell death in the pharmacological treatment of DPN.
Collapse
Affiliation(s)
- Abdullah Al Mamun
- Central Laboratory of The Lishui Hospital of Wenzhou Medical University, Lishui People’s Hospital, Lishui, Zhejiang, 323000, People’s Republic of China
- Molecular Pharmacology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325000, People’s Republic of China
| | - Chuxiao Shao
- Central Laboratory of The Lishui Hospital of Wenzhou Medical University, Lishui People’s Hospital, Lishui, Zhejiang, 323000, People’s Republic of China
| | - Peiwu Geng
- Central Laboratory of The Lishui Hospital of Wenzhou Medical University, Lishui People’s Hospital, Lishui, Zhejiang, 323000, People’s Republic of China
| | - Shuanghu Wang
- Central Laboratory of The Lishui Hospital of Wenzhou Medical University, Lishui People’s Hospital, Lishui, Zhejiang, 323000, People’s Republic of China
| | - Jian Xiao
- Central Laboratory of The Lishui Hospital of Wenzhou Medical University, Lishui People’s Hospital, Lishui, Zhejiang, 323000, People’s Republic of China
- Molecular Pharmacology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325000, People’s Republic of China
- Department of Wound Healing, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, People’s Republic of China
| |
Collapse
|
17
|
Song L, Li Y, Xu M. Exogenous Nucleotides Ameliorate Insulin Resistance Induced by Palmitic Acid in HepG2 Cells through the IRS-1/AKT/FOXO1 Pathways. Nutrients 2024; 16:1801. [PMID: 38931156 PMCID: PMC11206901 DOI: 10.3390/nu16121801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 06/01/2024] [Accepted: 06/04/2024] [Indexed: 06/28/2024] Open
Abstract
Nucleotides (NTs) act as pivotal regulatory factors in numerous biological processes, playing indispensable roles in growth, development, and metabolism across organisms. This study delves into the effects of exogenous NTs on hepatic insulin resistance using palmitic-acid-induced HepG2 cells, administering interventions at three distinct dosage levels of exogenous NTs. The findings underscore that exogenous NT intervention augments glucose consumption in HepG2 cells, modulates the expression of glycogen-synthesis-related enzymes (glycogen synthase kinase 3β and glycogen synthase), and influences glycogen content. Additionally, it governs the expression levels of hepatic enzymes (hexokinase, phosphoenolpyruvate carboxykinase, and glucose-6-phosphatase). Moreover, exogenous NT intervention orchestrates insulin signaling pathway (insulin receptor substrate-1, protein kinase B, and forkhead box protein O1) and AMP-activated protein kinase (AMPK) activity in HepG2 cells. Furthermore, exogenous NT intervention fine-tunes the expression levels of oxidative stress-related markers (malondialdehyde, glutathione peroxidase, and NADPH oxidase 4) and the expression of inflammation-related nuclear transcription factor (NF-κB). Lastly, exogenous NT intervention regulates the expression levels of glucose transporter proteins (GLUTs). Consequently, exogenous NTs ameliorate insulin resistance in HepG2 cells by modulating the IRS-1/AKT/FOXO1 pathways and regulate glucose consumption, glycogen content, insulin signaling pathways, AMPK activity, oxidative stress, and inflammatory status.
Collapse
Affiliation(s)
- Lixia Song
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Beijing 100191, China; (L.S.); (Y.L.)
- Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Peking University, Beijing 100191, China
| | - Yong Li
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Beijing 100191, China; (L.S.); (Y.L.)
- Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Peking University, Beijing 100191, China
| | - Meihong Xu
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Beijing 100191, China; (L.S.); (Y.L.)
- Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Peking University, Beijing 100191, China
- Institute of Medical Technology, Peking University Health Science Center, Beijing 100019, China
| |
Collapse
|
18
|
Zhou P, Dang J, Jiang Z, Dai S, Qu C, Wu Q. Transcriptome and metabolome analysis revealed the dynamic change of bioactive compounds of Fructus Ligustri Lucidi. BMC PLANT BIOLOGY 2024; 24:489. [PMID: 38825671 PMCID: PMC11145772 DOI: 10.1186/s12870-024-05096-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 05/02/2024] [Indexed: 06/04/2024]
Abstract
BACKGROUND The Fructus Ligustri Lucidi, the fruit of Ligustrum lucidum, contains a variety of bioactive compounds, such as flavonoids, triterpenoids, and secoiridoids. The proportions of these compounds vary greatly during the different fruit development periods of Fructus Ligustri Lucidi. However, a clear understanding of how the proportions of the compounds and their regulatory biosynthetic mechanisms change across the different fruit development periods of Fructus Ligustri Lucidi is still lacking. RESULTS In this study, metabolite profiling and transcriptome analysis of six fruit development periods (45 DAF, 75 DAF, 112 DAF, 135 DAF, 170 DAF, and 195 DAF) were performed. Seventy compounds were tentatively identified, of which secoiridoids were the most abundant. Eleven identified compounds were quantified by high performance liquid chromatography. A total of 103,058 unigenes were obtained from six periods of Fructus Ligustri Lucidi. Furthermore, candidate genes involved in triterpenoids, phenylethanols, and oleoside-type secoiridoid biosynthesis were identified and analyzed. The in vitro enzyme activities of nine glycosyltransferases involved in salidroside biosynthesis revealed that they can catalyze trysol and hydroxytyrosol to salidroside and hydroxylsalidroside. CONCLUSIONS These results provide valuable information to clarify the profile and molecular regulatory mechanisms of metabolite biosynthesis, and also in optimizing the harvest time of this fruit.
Collapse
Affiliation(s)
- Peina Zhou
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
- Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing, 210023, China
| | - Jingjie Dang
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
- Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing, 210023, China
| | - Zheng Jiang
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
- Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing, 210023, China
| | - Shilin Dai
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
- Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing, 210023, China
| | - Cheng Qu
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
- Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing, 210023, China.
| | - Qinan Wu
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
- Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing, 210023, China.
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing, 210023, China.
| |
Collapse
|
19
|
Yang B, Xu Y, Yu J, Wang Q, Fan Q, Zhao X, Qiao Y, Zhang Z, Zhou Q, Yin D, He M, He H. Salidroside pretreatment alleviates ferroptosis induced by myocardial ischemia/reperfusion through mitochondrial superoxide-dependent AMPKα2 activation. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 128:155365. [PMID: 38552436 DOI: 10.1016/j.phymed.2024.155365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 11/28/2023] [Accepted: 01/14/2024] [Indexed: 05/01/2024]
Abstract
BACKGROUND Ferroptosis, a form of regulated cell death (RCD) that relies on excessive reactive oxygen species (ROS) generation, Fe2+accumulation, abnormal lipid metabolism and is involved in various organ ischemia/reperfusion (I/R) injury, expecially in myocardium. Mitochondria are the powerhouses of eukaryotic cells and essential in regulating multiple RCD. However, the links between mitochondria and ferroptosis are still poorly understood. Salidroside (Sal), a natural phenylpropanoid glycoside isolated from Rhodiola rosea, has mult-bioactivities. However, the effects and mechanism in alleviating ferroptosis caused by myocardial I/R injury remains unclear. PURPOSE This study aimed to investigate whether pretreated with Sal could protect the myocardium against I/R damage and the underlying mechanisms. In particular, the relationship between Sal pretreatment, AMPKα2 activity, mitochondria and ROS generation was explored. STUDY DESIGN AND METHODS Firstly, A/R or I/R injury models were employed in H9c2 cells and Sprague-Dawley rats. And then the anti-ferroptotic effects and mechanism of Sal pretreatment was detected using multi-relevant indexes in H9c2 cells. Further, how does Sal pretreatment in AMPKα2 phosphorylation was explored. Finally, these results were validated by I/R injury in rats. RESULTS Similar to Ferrostatin-1 (a ferroptosis inhibitor) and MitoTEMPO, a mitochondrial free radical scavenger, Sal pretreatment effectively alleviated Fe2+ accumulation, redox disequilibrium and maintained mitochondrial energy production and function in I/R-induced myocardial injury, as demonstrated using multifunctional, enzymatic, and morphological indices. However, these effects were abolished by downregulation of AMPKα2 using an adenovirus, both in vivo and in vitro. Moreover, the results also provided a non-canonical mechanism that, under mild mitochondrial ROS generation, Sal pretreatment upregulated and phosphorylated AMPKα2, which enhanced mitochondrial complex I activity to activate innate adaptive responses and increase cellular tolerance to A/R injury. CONCLUSION Overall, our work highlighted mitochondria are of great impotance in myocardial I/R-induced ferroptosis and demonstrated that Sal pretreatment activated AMPKα2 against I/R injury, indicating that Sal could become a candidate phytochemical for the treatment of myocardial I/R injury.
Collapse
Affiliation(s)
- Bin Yang
- Jiangxi Provincial Key Laboratory of Basic Pharmacology, Nanchang University School of Pharmaceutical Science, Nanchang 330006, China
| | - Ying Xu
- Jiangxi Provincial Key Laboratory of Basic Pharmacology, Nanchang University School of Pharmaceutical Science, Nanchang 330006, China
| | - Jingzhi Yu
- Jiangxi Provincial Key Laboratory of Basic Pharmacology, Nanchang University School of Pharmaceutical Science, Nanchang 330006, China
| | - Qihao Wang
- Jiangxi Provincial Key Laboratory of Basic Pharmacology, Nanchang University School of Pharmaceutical Science, Nanchang 330006, China
| | - Qigui Fan
- Jiangxi Provincial Key Laboratory of Basic Pharmacology, Nanchang University School of Pharmaceutical Science, Nanchang 330006, China
| | - Xiaoyu Zhao
- Jiangxi Provincial Key Laboratory of Basic Pharmacology, Nanchang University School of Pharmaceutical Science, Nanchang 330006, China
| | - Yang Qiao
- Jiangxi Academy of Clinical Medical Sciences, the First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Zeyu Zhang
- Jiangxi Academy of Clinical Medical Sciences, the First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Qing Zhou
- Jiangxi Provincial Key Laboratory of Basic Pharmacology, Nanchang University School of Pharmaceutical Science, Nanchang 330006, China
| | - Dong Yin
- Jiangxi Provincial Key Laboratory of Basic Pharmacology, Nanchang University School of Pharmaceutical Science, Nanchang 330006, China
| | - Ming He
- Jiangxi Provincial Key Laboratory of Basic Pharmacology, Nanchang University School of Pharmaceutical Science, Nanchang 330006, China.
| | - Huan He
- Jiangxi Provincial Key Laboratory of Basic Pharmacology, Nanchang University School of Pharmaceutical Science, Nanchang 330006, China.
| |
Collapse
|
20
|
Sati P, Dhyani P, Sharma E, Attri DC, Jantwal A, Devi R, Calina D, Sharifi-Rad J. Gut Microbiota Targeted Approach by Natural Products in Diabetes Management: An Overview. Curr Nutr Rep 2024; 13:166-185. [PMID: 38498287 DOI: 10.1007/s13668-024-00523-1] [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] [Accepted: 02/16/2024] [Indexed: 03/20/2024]
Abstract
PURPOSE OF REVIEW This review delves into the complex interplay between obesity-induced gut microbiota dysbiosis and the progression of type 2 diabetes mellitus (T2DM), highlighting the potential of natural products in mitigating these effects. By integrating recent epidemiological data, we aim to provide a nuanced understanding of how obesity exacerbates T2DM through gut flora alterations. RECENT FINDINGS Advances in research have underscored the significance of bioactive ingredients in natural foods, capable of restoring gut microbiota balance, thus offering a promising approach to manage diabetes in the context of obesity. These findings build upon the traditional use of medicinal plants in diabetes treatment, suggesting a deeper exploration of their mechanisms of action. This comprehensive manuscript underscores the critical role of targeting gut microbiota dysbiosis in obesity-related T2DM management and by bridging traditional knowledge with current scientific evidence; we highlighted the need for continued research into natural products as a complementary strategy for comprehensive diabetes care.
Collapse
Affiliation(s)
- Priyanka Sati
- Department of Biotechnology, Kumaun University, Bhimtal, Uttarakhand, India
| | - Praveen Dhyani
- Institute for Integrated Natural Sciences, University of Koblenz, Koblenz, Germany
| | - Eshita Sharma
- Department of Biochemistry and Molecular Biology, Guru Nanak Dev University, Amritsar, Punjab, India
| | - Dharam Chand Attri
- Department of Botany, Central University of Jammu, Rahya-Suchani (Bagla), Jammu and Kashmir, India
| | - Arvind Jantwal
- Department of Pharmaceutical Sciences, Kumaun University, Bhimtal, Uttarakhand, India
| | - Rajni Devi
- Department of Microbiology, Punjab Agricultural University, Ludhiana-141004, Punjab, India
| | - Daniela Calina
- Department of Clinical Pharmacy, University of Medicine and Pharmacy of Craiova, 200349, Craiova, Romania.
| | | |
Collapse
|
21
|
Tao J, Gong Y, Chen S, Li W, Xie R, Zhang H, Chen N, Huang X, Li S. Dietary inclusion of Clostridium butyricum cultures alleviated impacts of high-carbohydrate diets in largemouth bass ( Micropterus salmoides). Br J Nutr 2024; 131:1308-1325. [PMID: 38073302 DOI: 10.1017/s0007114523002842] [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] [Indexed: 12/28/2023]
Abstract
A 60-d feeding trial was conducted to explore the potential regulatory effects of dietary Clostridium butyricum cultures (CBC) supplementation in high-carbohydrate diet (HCD) on carbohydrate utilisation, antioxidant capacity and intestinal microbiota of largemouth bass. Triplicate groups of largemouth bass (average weight 35·03 ± 0·04 g), with a destiny of twenty-eight individuals per tank, were fed low-carbohydrate diet and HCD supplemented with different concentration of CBC (0 %, 0·25 %, 0·50 % and 1·00 %). The results showed that dietary CBC inclusion alleviated the hepatic glycogen accumulation induced by HCD intake. Additionally, the expression of hepatic ampkα1 and insulin signaling pathway-related genes (ira, irb, irs, p13kr1 and akt1) increased linearly with dietary CBC inclusion, which might be associated with the activation of glycolysis-related genes (gk, pfkl and pk). Meanwhile, the expression of intestinal SCFA transport-related genes (ffar3 and mct1) was significantly increased with dietary CBC inclusion. In addition, the hepatic antioxidant capacity was improved with dietary CBC supplementation, as evidenced by linear decrease in malondialdehyde concentration and expression of keap1, and linear increase in antioxidant enzyme activities (total antioxidative capacity, total superoxide dismutase and catalase) and expression of antioxidant enzyme-related genes (nrf2, sod1, sod2 and cat). The analysis of bacterial 16S rRNA V3-4 region indicated that dietary CBC inclusion significantly reduced the enrichment of Firmicutes and potential pathogenic bacteria genus Mycoplasma but significantly elevated the relative abundance of Fusobacteria and Cetobacterium. In summary, dietary CBC inclusion improved carbohydrate utilization, antioxidant capacity and intestinal microbiota of largemouth bass fed HCD.
Collapse
Affiliation(s)
- Jiajie Tao
- Research Centre of the Ministry of Agriculture and Rural Affairs on Environmental Ecology and Fish Nutrition, Shanghai Ocean University, Shanghai, 201306, People's Republic of China
| | - Ye Gong
- Research Centre of the Ministry of Agriculture and Rural Affairs on Environmental Ecology and Fish Nutrition, Shanghai Ocean University, Shanghai, 201306, People's Republic of China
| | - Shiwen Chen
- Research Centre of the Ministry of Agriculture and Rural Affairs on Environmental Ecology and Fish Nutrition, Shanghai Ocean University, Shanghai, 201306, People's Republic of China
| | - Wenfei Li
- Research Centre of the Ministry of Agriculture and Rural Affairs on Environmental Ecology and Fish Nutrition, Shanghai Ocean University, Shanghai, 201306, People's Republic of China
| | - Ruitao Xie
- Key Laboratory of Aquatic, Livestock and Poultry Feed Science and Technology in South China, Ministry of Agriculture and Rural Affairs, Zhanjiang, People's Republic of China
| | - Haitao Zhang
- Key Laboratory of Aquatic, Livestock and Poultry Feed Science and Technology in South China, Ministry of Agriculture and Rural Affairs, Zhanjiang, People's Republic of China
| | - Naisong Chen
- Research Centre of the Ministry of Agriculture and Rural Affairs on Environmental Ecology and Fish Nutrition, Shanghai Ocean University, Shanghai, 201306, People's Republic of China
- National Demonstration Center on Experiment Teaching of Fisheries Science, Shanghai Ocean University, Shanghai, People's Republic of China
| | - Xuxiong Huang
- Research Centre of the Ministry of Agriculture and Rural Affairs on Environmental Ecology and Fish Nutrition, Shanghai Ocean University, Shanghai, 201306, People's Republic of China
- National Demonstration Center on Experiment Teaching of Fisheries Science, Shanghai Ocean University, Shanghai, People's Republic of China
| | - Songlin Li
- Research Centre of the Ministry of Agriculture and Rural Affairs on Environmental Ecology and Fish Nutrition, Shanghai Ocean University, Shanghai, 201306, People's Republic of China
- National Demonstration Center on Experiment Teaching of Fisheries Science, Shanghai Ocean University, Shanghai, People's Republic of China
| |
Collapse
|
22
|
Xia Y, Sun M, Huang H, Jin WL. Drug repurposing for cancer therapy. Signal Transduct Target Ther 2024; 9:92. [PMID: 38637540 PMCID: PMC11026526 DOI: 10.1038/s41392-024-01808-1] [Citation(s) in RCA: 82] [Impact Index Per Article: 82.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 03/05/2024] [Accepted: 03/19/2024] [Indexed: 04/20/2024] Open
Abstract
Cancer, a complex and multifactorial disease, presents a significant challenge to global health. Despite significant advances in surgical, radiotherapeutic and immunological approaches, which have improved cancer treatment outcomes, drug therapy continues to serve as a key therapeutic strategy. However, the clinical efficacy of drug therapy is often constrained by drug resistance and severe toxic side effects, and thus there remains a critical need to develop novel cancer therapeutics. One promising strategy that has received widespread attention in recent years is drug repurposing: the identification of new applications for existing, clinically approved drugs. Drug repurposing possesses several inherent advantages in the context of cancer treatment since repurposed drugs are typically cost-effective, proven to be safe, and can significantly expedite the drug development process due to their already established safety profiles. In light of this, the present review offers a comprehensive overview of the various methods employed in drug repurposing, specifically focusing on the repurposing of drugs to treat cancer. We describe the antitumor properties of candidate drugs, and discuss in detail how they target both the hallmarks of cancer in tumor cells and the surrounding tumor microenvironment. In addition, we examine the innovative strategy of integrating drug repurposing with nanotechnology to enhance topical drug delivery. We also emphasize the critical role that repurposed drugs can play when used as part of a combination therapy regimen. To conclude, we outline the challenges associated with repurposing drugs and consider the future prospects of these repurposed drugs transitioning into clinical application.
Collapse
Affiliation(s)
- Ying Xia
- Center for Clinical Laboratories, The Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, PR China
- The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, 550001, PR China
- School of Clinical Laboratory Science, Guizhou Medical University, Guiyang, 550004, PR China
- Division of Gastroenterology and Hepatology, Department of Medicine and, Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Ming Sun
- Center for Clinical Laboratories, The Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, PR China
- School of Clinical Laboratory Science, Guizhou Medical University, Guiyang, 550004, PR China
| | - Hai Huang
- Center for Clinical Laboratories, The Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, PR China.
- School of Clinical Laboratory Science, Guizhou Medical University, Guiyang, 550004, PR China.
| | - Wei-Lin Jin
- Institute of Cancer Neuroscience, Medical Frontier Innovation Research Center, The First Hospital of Lanzhou University, The First Clinical Medical College of Lanzhou University, Lanzhou, 730000, PR China.
| |
Collapse
|
23
|
Sun Y, Wang Y, Lin Z, Zhang F, Zhang Y, Ren T, Wang L, Qiao Q, Shen M, Wang J, Song Y, Sun Y, Lin P. Irisin delays the onset of type 1 diabetes in NOD mice by enhancing intestinal barrier. Int J Biol Macromol 2024; 265:130857. [PMID: 38493812 DOI: 10.1016/j.ijbiomac.2024.130857] [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: 01/16/2024] [Revised: 03/11/2024] [Accepted: 03/12/2024] [Indexed: 03/19/2024]
Abstract
Type 1 diabetes (T1D), a complex autoimmune disease, is intricately linked to the gut's epithelial barrier function. Emerging evidence emphasizes the role of irisin, an exercise-related hormone, in preserving intestinal integrity. This study investigates whether irisin could delay T1D onset by enhancing the colon intestinal barrier. Impaired colon intestinal barriers were observed in newly diagnosed T1D patients and non-obese diabetic (NOD) mice, worsening with age and accompanied by islet inflammation. Using an LPS-induced colonic inflammation model, a dose-dependent impact of LPS on colon cells irisin expression, secretion, and barrier function was revealed. Exogenous irisin demonstrated remarkable effects, mitigating islet insulitis, enhancing energy expenditure, and alleviating autoimmune symptoms by reducing colon intestinal permeability. Single-cell RNA sequencing (scRNA-seq) highlighted irisin's positive impact on colon epithelial cell clusters, effectively restoring the intestinal barrier. Irisin also selectively modulated bacterial composition, averting potential bacterial translocation. Mechanistically, irisin enhanced colon intestinal barrier tight junction proteins through the AMPK/PI3K/AKT pathway, with FAM120A playing a crucial role. Irisin upregulated MUC3 expression, a protector against damage and inflammation. Harnessing irisin's exercise-mimicking properties suggests therapeutic potential in clinical settings for preventing T1D progression, offering valuable insights into fortifying the colon's intestinal barrier and managing autoimmune conditions associated with T1DM.
Collapse
Affiliation(s)
- Yujing Sun
- Department of Endocrine and Metabolic Diseases, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250012, China; Key Laboratory of Endocrine and Metabolic Diseases, Shandong Province Medicine and Health, Jinan, Shandong 250012, China; Jinan Clinical Research Center for Endocrine and Metabolic Disease, Jinan, Shandong 250012, China
| | - Yilin Wang
- Department of Orthopedics and Traumatology, Peking University People's Hospital, Beijing, China
| | - Ziang Lin
- Qilu Hospital of Shandong University, Jinan, 250012, Shandong Province, China
| | - Fuhua Zhang
- Department of Endocrine and Metabolic Diseases, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Yan Zhang
- Department of Gastroenterology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Tongxin Ren
- Department of Endocrine and Metabolic Diseases, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Lina Wang
- Department of Endocrine and Metabolic Diseases, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Qincheng Qiao
- Department of Endocrine and Metabolic Diseases, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Mengyang Shen
- Department of Endocrine and Metabolic Diseases, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250012, China; Key Laboratory of Endocrine and Metabolic Diseases, Shandong Province Medicine and Health, Jinan, Shandong 250012, China; Jinan Clinical Research Center for Endocrine and Metabolic Disease, Jinan, Shandong 250012, China
| | - Juncheng Wang
- Advanced Medical Research Institute, Meili Lake Translational Research Park, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Youchen Song
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Yu Sun
- Department of Endocrine and Metabolic Diseases, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250012, China; Key Laboratory of Endocrine and Metabolic Diseases, Shandong Province Medicine and Health, Jinan, Shandong 250012, China; Jinan Clinical Research Center for Endocrine and Metabolic Disease, Jinan, Shandong 250012, China.
| | - Peng Lin
- Department of Endocrine and Metabolic Diseases, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250012, China; Key Laboratory of Endocrine and Metabolic Diseases, Shandong Province Medicine and Health, Jinan, Shandong 250012, China; Jinan Clinical Research Center for Endocrine and Metabolic Disease, Jinan, Shandong 250012, China.
| |
Collapse
|
24
|
Zhang Y, Jiao X, Liu J, Feng G, Luo X, Zhang M, Zhang B, Huang L, Long Q. A new direction in Chinese herbal medicine ameliorates for type 2 diabetes mellitus: Focus on the potential of mitochondrial respiratory chain complexes. JOURNAL OF ETHNOPHARMACOLOGY 2024; 321:117484. [PMID: 38012971 DOI: 10.1016/j.jep.2023.117484] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 11/15/2023] [Accepted: 11/20/2023] [Indexed: 11/29/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Diabetes is a common chronic disease. Chinese herbal medicine (CHM) has a history of several thousand years in the treatment of diabetes, and active components with hypoglycemic effects extracted from various CHM, such as polysaccharides, flavonoids, terpenes, and steroidal saponins, have been widely used in the treatment of diabetes. AIM OF THE STUDY Research exploring the potential of various CHM compounds to regulate the mitochondrial respiratory chain complex to improve type 2 diabetes mellitus (T2DM). MATERIALS AND METHODS The literature data were primarily obtained from authoritative databases such as PubMed, CNKI, Wanfang, and others within the last decade. The main keywords used include "type 2 diabetes mellitus", "Chinese medicine", "Chinese herbal medicine", "mitochondrial respiratory chain complex", and "mitochondrial dysfunction". RESULTS Chinese herbal medicine primarily regulates the activity of mitochondrial respiratory chain complexes in various tissues such as liver, adipose tissue, skeletal muscle, pancreatic islets, and small intestine. It improves cellular energy metabolism through hypoglycemic, antioxidant, anti-inflammatory and lipid-modulating effects. Different components of CHM can regulate the same mitochondrial respiratory chain complexes, while the same components of a particular CHM can regulate different complex activities. The active components of CHM target different mitochondrial respiratory chain complexes, regulate their aberrant changes and effectively improve T2DM and its complications. CONCLUSION Chinese herbal medicine can modulate the function of mitochondrial respiratory chain complexes in various cell types and exert their hypoglycemic effects through various mechanisms. CHM has significant therapeutic potential in regulating mitochondrial respiratory chain complexes to improve T2DM, but further research is needed to explore the underlying mechanisms and conduct clinical trials to assess the safety and efficacy of these medications. This provides new perspectives and opportunities for personalized improvement and innovative developments in diabetes management.
Collapse
Affiliation(s)
- Yinghui Zhang
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine (Institute of Chinese Medicine), Guangdong Pharmaceutical University, Guangzhou, 510006, China; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education, Guangdong Pharmaceutical University, Guangzhou, 510006, China; Guangdong Provincial Key Laboratory of Chinese Medicine for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Xinyue Jiao
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine (Institute of Chinese Medicine), Guangdong Pharmaceutical University, Guangzhou, 510006, China; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education, Guangdong Pharmaceutical University, Guangzhou, 510006, China; Guangdong Provincial Key Laboratory of Chinese Medicine for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Jianying Liu
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine (Institute of Chinese Medicine), Guangdong Pharmaceutical University, Guangzhou, 510006, China; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education, Guangdong Pharmaceutical University, Guangzhou, 510006, China; Guangdong Provincial Key Laboratory of Chinese Medicine for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Gang Feng
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine (Institute of Chinese Medicine), Guangdong Pharmaceutical University, Guangzhou, 510006, China; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education, Guangdong Pharmaceutical University, Guangzhou, 510006, China; Guangdong Provincial Key Laboratory of Chinese Medicine for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Xia Luo
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine (Institute of Chinese Medicine), Guangdong Pharmaceutical University, Guangzhou, 510006, China; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education, Guangdong Pharmaceutical University, Guangzhou, 510006, China; Guangdong Provincial Key Laboratory of Chinese Medicine for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Mingyue Zhang
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine (Institute of Chinese Medicine), Guangdong Pharmaceutical University, Guangzhou, 510006, China; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education, Guangdong Pharmaceutical University, Guangzhou, 510006, China; Guangdong Provincial Key Laboratory of Chinese Medicine for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Binzhi Zhang
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine (Institute of Chinese Medicine), Guangdong Pharmaceutical University, Guangzhou, 510006, China; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education, Guangdong Pharmaceutical University, Guangzhou, 510006, China; Guangdong Provincial Key Laboratory of Chinese Medicine for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Lizhen Huang
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Qinqiang Long
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine (Institute of Chinese Medicine), Guangdong Pharmaceutical University, Guangzhou, 510006, China; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education, Guangdong Pharmaceutical University, Guangzhou, 510006, China; Guangdong Provincial Key Laboratory of Chinese Medicine for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou, 510006, China.
| |
Collapse
|
25
|
Wen D, Meng C, Feng Y, Shen L, Liu Y, Sun W, Chen G, Wu C. Syringaldehyde Exhibits Antibacterial and Antioxidant Activities against Mycobacterium marinum Infection. Microorganisms 2024; 12:348. [PMID: 38399751 PMCID: PMC10893232 DOI: 10.3390/microorganisms12020348] [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: 01/09/2024] [Revised: 01/27/2024] [Accepted: 01/31/2024] [Indexed: 02/25/2024] Open
Abstract
Tuberculosis (TB) is caused by infection with Mycobacterium tuberculosis (Mtb), which has a unique resistance to many antimicrobial agents. TB has emerged as a significant worldwide health issue because of the rise of multidrug-resistant strains causing drug-resistant TB (DR-TB). As a result, the development of new drugs or effective strategies is crucial for patients with TB. Mycobacterium marinum (Mm) and Mtb are both species of mycobacteria. In zebrafish, Mm proliferates and forms chronic granulomatous infections, which are similar to Mtb infections in lung tissue. Syringaldehyde (SA) is a member of the phenolic aldehyde family found in various plants. Here, we investigated its antioxidative and antibacterial properties in Mm-infected cells and zebrafish. Our results demonstrated that SA inhibits Mm-infected pulmonary epithelial cells and inhibits the proliferation of Mm in Mm-infected zebrafish, suggesting that SA provides an antibacterial effect during Mm infection. Further study demonstrated that supplementation with SA inhibits the production of malondialdehyde (MDA) and reactive oxygen species (ROS) and increases the levels of reduced glutathione (GSH) in Mm-infection-induced macrophages. SA inhibits the levels of MDA in Mm-infected zebrafish, suggesting that SA exerts antioxidative effects in vivo. Additionally, we found that SA promotes the expression of NRF2/HO-1/NQO-1 and the activation of the AMPK-α1/AKT/GSK-3β signaling pathway. In summary, our data demonstrated that SA exerts antioxidative and antibacterial effects during Mm infection both in vivo and in vitro and that the antioxidative effects of SA may be due to the regulation of NRF2/HO-1/NQO-1 and the AMPK-α1/AKT/GSK-3β signaling pathway.
Collapse
Affiliation(s)
- Da Wen
- Institutes of Biomedical Sciences, Shanxi University, Taiyuan 030006, China
- Shanxi Provincial Key Laboratory of Medical Molecular Cell Biology, Shanxi University, Taiyuan 030006, China
| | - Chaoqun Meng
- Institutes of Biomedical Sciences, Shanxi University, Taiyuan 030006, China
- Shanxi Provincial Key Laboratory of Medical Molecular Cell Biology, Shanxi University, Taiyuan 030006, China
| | - Yazhi Feng
- Institutes of Biomedical Sciences, Shanxi University, Taiyuan 030006, China
- Shanxi Provincial Key Laboratory of Medical Molecular Cell Biology, Shanxi University, Taiyuan 030006, China
| | - Lin Shen
- Institutes of Biomedical Sciences, Shanxi University, Taiyuan 030006, China
- Shanxi Provincial Key Laboratory of Medical Molecular Cell Biology, Shanxi University, Taiyuan 030006, China
| | - Yiyao Liu
- Institutes of Biomedical Sciences, Shanxi University, Taiyuan 030006, China
- Shanxi Provincial Key Laboratory of Medical Molecular Cell Biology, Shanxi University, Taiyuan 030006, China
| | - Wei Sun
- Institutes of Biomedical Sciences, Shanxi University, Taiyuan 030006, China
- Shanxi Provincial Key Laboratory of Medical Molecular Cell Biology, Shanxi University, Taiyuan 030006, China
| | - Guangxin Chen
- Institutes of Biomedical Sciences, Shanxi University, Taiyuan 030006, China
- Shanxi Provincial Key Laboratory of Medical Molecular Cell Biology, Shanxi University, Taiyuan 030006, China
| | - Changxin Wu
- Institutes of Biomedical Sciences, Shanxi University, Taiyuan 030006, China
- Shanxi Provincial Key Laboratory of Medical Molecular Cell Biology, Shanxi University, Taiyuan 030006, China
- Shanxi Provincial Key Laboratory for Prevention and Treatment of Major Infectious Diseases, Shanxi University, Taiyuan 030006, China
| |
Collapse
|
26
|
Liu Q, Chen J, Zeng A, Song L. Pharmacological functions of salidroside in renal diseases: facts and perspectives. Front Pharmacol 2024; 14:1309598. [PMID: 38259279 PMCID: PMC10800390 DOI: 10.3389/fphar.2023.1309598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Accepted: 12/18/2023] [Indexed: 01/24/2024] Open
Abstract
Rhodiola rosea is a valuable functional medicinal plant widely utilized in China and other Asian countries for its anti-fatigue, anti-aging, and altitude sickness prevention properties. Salidroside, a most active constituent derived from Rhodiola rosea, exhibits potent antioxidative, hypoxia-resistant, anti-inflammatory, anticancer, and anti-aging effects that have garnered significant attention. The appreciation of the pharmacological role of salidroside has burgeoned over the last decade, making it a beneficial option for the prevention and treatment of multiple diseases, including atherosclerosis, Alzheimer's disease, Parkinson's disease, cardiovascular disease, and more. With its anti-aging and renoprotective effects, in parallel with the inhibition of oxidative stress and inflammation, salidroside holds promise as a potential therapeutic agent for kidney damage. This article provides an overview of the microinflammatory state in kidney disease and discuss the current therapeutic strategies, with a particular focus on highlighting the recent advancements in utilizing salidroside for renal disease. The potential mechanisms of action of salidroside are primarily associated with the regulation of gene and protein expression in glomerular endothelial cells, podocytes, renal tubule cells, renal mesangial cells and renal cell carcinoma cell, including TNF-α, TGF-β, IL-1β, IL-17A, IL-6, MCP-1, Bcl-2, VEGF, ECM protein, caspase-3, HIF-1α, BIM, as well as the modulation of AMPK/SIRT1, Nrf2/HO-1, Sirt1/PGC-1α, ROS/Src/Cav-1, Akt/GSK-3β, TXNIP-NLRP3, ERK1/2, TGF-β1/Smad2/3, PI3K/Akt, Wnt1/Wnt3a β-catenin, TLR4/NF-κB, MAPK, JAK2/STAT3, SIRT1/Nrf2 pathways. To the best of our knowledge, this review is the first to comprehensively cover the protective effects of salidroside on diverse renal diseases, and suggests that salidroside has great potential to be developed as a drug for the prevention and treatment of metabolic syndrome, cardiovascular and cerebrovascular diseases and renal complications.
Collapse
Affiliation(s)
- Qiong Liu
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Jianzhu Chen
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Anqi Zeng
- Translational Chinese Medicine Key Laboratory of Sichuan Province, Sichuan Academy of Chinese Medicine Sciences, Sichuan Institute for Translational Chinese Medicine, Chengdu, Sichuan, China
| | - Linjiang Song
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| |
Collapse
|
27
|
Li J, Chen K, Li X, Zhang X, Zhang L, Yang Q, Xia Y, Xie C, Wang X, Tong J, Shen Y. Mechanistic insights into the alterations and regulation of the AKT signaling pathway in diabetic retinopathy. Cell Death Discov 2023; 9:418. [PMID: 37978169 PMCID: PMC10656479 DOI: 10.1038/s41420-023-01717-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Revised: 11/05/2023] [Accepted: 11/08/2023] [Indexed: 11/19/2023] Open
Abstract
In the early stages of diabetic retinopathy (DR), diabetes-related hyperglycemia directly inhibits the AKT signaling pathway by increasing oxidative stress or inhibiting growth factor expression, which leads to retinal cell apoptosis, nerve proliferation and fundus microvascular disease. However, due to compensatory vascular hyperplasia in the late stage of DR, the vascular endothelial growth factor (VEGF)/phosphatidylinositol 3 kinase (PI3K)/AKT cascade is activated, resulting in opposite levels of AKT regulation compared with the early stage. Studies have shown that many factors, including insulin, insulin-like growth factor-1 (IGF-1), VEGF and others, can regulate the AKT pathway. Disruption of the insulin pathway decreases AKT activation. IGF-1 downregulation decreases the activation of AKT in DR, which abrogates the neuroprotective effect, upregulates VEGF expression and thus induces neovascularization. Although inhibiting VEGF is the main treatment for neovascularization in DR, excessive inhibition may lead to apoptosis in inner retinal neurons. AKT pathway substrates, including mammalian target of rapamycin (mTOR), forkhead box O (FOXO), glycogen synthase kinase-3 (GSK-3)/nuclear factor erythroid 2-related factor 2 (Nrf2), and nuclear factor kappa-B (NF-κB), are a research focus. mTOR inhibitors can delay or prevent retinal microangiopathy, whereas low mTOR activity can decrease retinal protein synthesis. Inactivated AKT fails to inhibit FOXO and thus causes apoptosis. The GSK-3/Nrf2 cascade regulates oxidation and inflammation in DR. NF-κB is activated in diabetic retinas and is involved in inflammation and apoptosis. Many pathways or vital activities, such as the Janus kinase (JAK)/signal transducer and activator of transcription (STAT) and mitogen-activated protein kinase (MAPK) signaling pathways, interact with the AKT pathway to influence DR development. Numerous regulatory methods can simultaneously impact the AKT pathway and other pathways, and it is essential to consider both the connections and interactions between these pathways. In this review, we summarize changes in the AKT signaling pathway in DR and targeted drugs based on these potential sites.
Collapse
Affiliation(s)
- Jiayuan Li
- Department of Ophthalmology, The First Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang, China
- Department of Cardiology, The Second Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang, China
| | - Kuangqi Chen
- Department of Ophthalmology, The First Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang, China
| | - Xiang Li
- Department of Ophthalmology, The First Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang, China
| | - Xuhong Zhang
- Department of Ophthalmology, The First Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang, China
| | - Liyue Zhang
- Department of Ophthalmology, The First Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang, China
| | - Qianjie Yang
- Department of Ophthalmology, The First Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang, China
| | - Yutong Xia
- Department of Ophthalmology, The First Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang, China
| | - Chen Xie
- Department of Ophthalmology, The First Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang, China
| | - Xiawei Wang
- Department of Ophthalmology, The First Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang, China
| | - Jianping Tong
- Department of Ophthalmology, The First Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang, China.
| | - Ye Shen
- Department of Ophthalmology, The First Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang, China.
| |
Collapse
|
28
|
Chang YR, Cheng WC, Hsiao YC, Su GW, Lin SJ, Wei YS, Chou HC, Lin HP, Lin GY, Chan HL. Links between oral microbiome and insulin resistance: Involvement of MAP kinase signaling pathway. Biochimie 2023; 214:134-144. [PMID: 37442534 DOI: 10.1016/j.biochi.2023.06.013] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 06/29/2023] [Accepted: 06/30/2023] [Indexed: 07/15/2023]
Abstract
Oral dysbiosis contributes to periodontitis and has implications for systemic diseases. Diabetes mellitus is a common metabolic disorder characterized by impaired glucose regulation. AMP-activated protein kinase (AMPK) plays a vital role in regulating glucose uptake and glycogenesis in the liver. This study aimed to investigate the association between periodontal bacteria and diabetes mellitus. A clinical trial was conducted to explore the association between oral bacteria and hyperglycemia. Additionally, we elucidated the molecular mechanisms by which periodontal bacteria cause insulin resistance. In the clinical trial, we discovered significant alterations in the expression levels of Fusobacterium nucleatum (Fn) and Tannerella forsythia (Tf) in patients with diabetes compared with healthy controls. Furthermore, Fn and Tf levels positively correlated with fasting blood glucose and glycated hemoglobin (HbA1C) levels. Moreover, we explored and elucidated the molecular mechanism by which Fusobacterium nucleatum culture filtrate (FNCF) induces cytokine release via the Toll-like receptor 2 (TLR2) signaling pathway in human gingival epithelial Smulow-Glickman (S-G) cells. This study investigated the effects of cytokines on insulin resistance pathways in liver cells. The use of an extracellular signal-regulated kinase (ERK) inhibitor (U0126) demonstrated that FNCF regulates the insulin receptor substrate 1 and protein kinase B (IRS1/AKT) signaling pathway, which affects key proteins involved in hepatic glycogen synthesis, including glycogen synthase kinase-3 beta (GSK3β) and glycogen synthase (GS), ultimately leading to insulin resistance. These findings suggest that ERK plays a crucial role in hepatocyte insulin resistance.
Collapse
Affiliation(s)
- Yi-Ru Chang
- Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu, Taiwan; General Biologicals Corporation, Hsinchu, Taiwan
| | - Wen-Chi Cheng
- SDGs Teaching and Research Headquarters, Tzu Chi University, Hualien, Taiwan
| | - Ya-Chun Hsiao
- Department of Endocrinology and Metabolism, Hsinchu Mackay Memorial Hospital, Hsinchu, Taiwan
| | - Guan-Wei Su
- Dental Department of Hsinchu Mackay Memorial Hospital, Hsinchu, Taiwan
| | - Shan-Jen Lin
- Dental Department of Hsinchu Mackay Memorial Hospital, Hsinchu, Taiwan
| | - Yu-Shan Wei
- Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu, Taiwan
| | - Hsiu-Chuan Chou
- Institute of Analytical and Environmental Sciences, National Tsing Hua University, Hsinchu, Taiwan
| | | | - Guan-Yu Lin
- Institute of Molecular and Cellular Biology, National Tsing Hua University, Hsinchu, Taiwan
| | - Hong-Lin Chan
- Department of Medical Science and Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu, Taiwan.
| |
Collapse
|
29
|
Wan M, Dai J, Gan A, Wang J, Lin F, Zhang X, Lv X, Wu B, Yan T, Jia Y. A network pharmacology approach to investigate dehydrocostus lactone inhibits the proliferation and epithelial-mesenchymal transition of human gastric cancer cells via regulating the PI3K/Akt and extracellular signal-regulated kinases/mitogen-activated protein kinase signalling pathways. J Pharm Pharmacol 2023; 75:1344-1356. [PMID: 37403268 DOI: 10.1093/jpp/rgad065] [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: 03/16/2023] [Accepted: 06/22/2023] [Indexed: 07/06/2023]
Abstract
OBJECTIVES Dehydrocostus lactone (DHE), a sesquiterpene lactone, has been proven the significant inhibition of multiple cancer cells. However, there are limited reports on the activity of DHE in gastric cancer (GC). In this research, Network pharmacology predicted the anti-GC mechanism of DHE, and the prediction was verified by in-vitro experiments. METHODS Network pharmacology confirmed the major effect signalling pathway of DHE in treating GC. Cell viability assay, colony formation assay, wound healing assay, cell migration and invasion assay, apoptosis assay, western blot and real-time quantitative polymerase chain reaction verified the mechanism of DHE in GC cell lines. KEY FINDINGS The results showed that DHE inhibited the growth and metastasis of MGC803 and AGS GC cells. Mechanistically, the analysis results indicated that DHE significantly induced the apoptosis process by suppressing the PI3K/protein kinase B (Akt) signalling pathway, and inhibited epithelial-mesenchymal transition by suppressing the extracellular signal-regulated kinases (ERK)/MAPK signalling pathway. The Akt activator (SC79) inhibited DHE induced apoptosis, and DHE had similar effects with the ERK inhibitor (FR180204). CONCLUSIONS All results suggested that DHE was a potential natural chemotherapeutic drug in GC treatment.
Collapse
Affiliation(s)
- Meiqi Wan
- Faculty of Functional Food and Wine, Shenyang Pharmaceutical University, Shenyang, China
| | - Jun Dai
- Faculty of Functional Food and Wine, Shenyang Pharmaceutical University, Shenyang, China
| | - Anna Gan
- Faculty of Functional Food and Wine, Shenyang Pharmaceutical University, Shenyang, China
| | - Jinyu Wang
- Faculty of Functional Food and Wine, Shenyang Pharmaceutical University, Shenyang, China
| | - Fei Lin
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, China
| | - Xiaoying Zhang
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, China
| | - Xinyan Lv
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, China
| | - Bo Wu
- Faculty of Functional Food and Wine, Shenyang Pharmaceutical University, Shenyang, China
| | - Tingxu Yan
- Faculty of Functional Food and Wine, Shenyang Pharmaceutical University, Shenyang, China
| | - Ying Jia
- Faculty of Functional Food and Wine, Shenyang Pharmaceutical University, Shenyang, China
| |
Collapse
|
30
|
Zhu L, Fan X, Cao C, Li K, Hou W, Ci X. Xanthohumol protect against acetaminophen-induced hepatotoxicity via Nrf2 activation through the AMPK/Akt/GSK3β pathway. Biomed Pharmacother 2023; 165:115097. [PMID: 37406514 DOI: 10.1016/j.biopha.2023.115097] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 06/26/2023] [Accepted: 06/27/2023] [Indexed: 07/07/2023] Open
Abstract
OBJECTIVE Acetaminophen (APAP) is one of the world's popular and safe painkillers, and overdose can cause severe liver damage and even acute liver failure. The effect and mechanism of the xanthohumol on acetaminophen-induced hepatotoxicity remains unclear. METHODS The hepatoprotective effects of xanthohumol were studied using APAP-induced HepG2 cells and acute liver injury of mouse, seperately. RESULTS In vitro, xanthohumol inhibited H2O2- and acetaminophen-induced cytotoxicity and oxidative stress. Xanthohumol up-regulated the expression of Nrf2. Further mechanistic studies showed that xanthohumol triggered Nrf2 activation via the AMPK/Akt/GSK3β pathway to exert a cytoprotective effect. In vivo, xanthohumol significantly ameliorated acetaminophen-induced mortality, the elevation of ALT and AST, GSH depletion, MDA formation and histopathological changes. Xanthohumol effectively suppressed the phosphorylation and mitochondrial translocation of JNK, mitochondrial translocation of Bax, the activation o cytochrome c, AIF secretion and Caspase-3. In vivo, xanthohumol increased Nrf2 nuclear transcription and AMPK, Akt and GSK3β phosphorylation in vivo. In addition, whether xanthohumol protected against acetaminophen-induced liver injury in Nrf2 knockout mice has not been illustated. CONCLUSION Thus, xanthohumol exerted a hepatoprotective effect by inhibiting oxidative stress and mitochondrial dysfunction through the AMPK/Akt/GSK3β/Nrf2 antioxidant pathway.
Collapse
Affiliation(s)
- Laiyu Zhu
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, Jilin 130001, China
| | - Xiaoye Fan
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, Jilin 130001, China
| | - Chunyuan Cao
- Department of Hepatobiliary Pancreatic Surgery, Jilin Province People's Hospital, Changzhun, China
| | - Kailiang Li
- Department of Hepatobiliary Pancreatic Surgery, Jilin Province People's Hospital, Changzhun, China
| | - Wenli Hou
- Department of Cadre Ward, the First Hospital of Jilin University, 71 Xinmin Street, Chaoyang, Changchun, Jilin 130021, China.
| | - Xinxin Ci
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, Jilin 130001, China.
| |
Collapse
|
31
|
Qiu X, Wang J, Zhang N, Du T, Chen L, Xi H. Estradiol cypionate inhibits proliferation and promotes apoptosis of gastric cancer by regulating AKT ubiquitination. Biomed Pharmacother 2023; 165:115073. [PMID: 37392652 DOI: 10.1016/j.biopha.2023.115073] [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: 04/20/2023] [Revised: 06/14/2023] [Accepted: 06/23/2023] [Indexed: 07/03/2023] Open
Abstract
Gastric cancer is a common gastrointestinal malignancy worldwide, with a high mortality rate and poor prognosis. Multidrug resistance remains a major obstacle to successful treatment for patients. Hence, it is of great significance to develop novel therapies to potentiate the anti-tumor effect. In this study, we have investigated the effect of estradiol cypionate (ECP) on gastric cancer in vitro and vivo. Our data show that ECP inhibited the proliferation, promoted apoptosis, and caused G1/S phase arrest of gastric cancer cells. The mechanism by which ECP promoted apoptosis of gastric cancer cells was related to the downregulation of AKT protein expression caused by the increased ubiquitination modification levels of AKT, which finally inhibited the over-activation of the PI3K-AKT-mTOR signaling pathway. In vivo tumorigenesis experiments showed that ECP significantly inhibited the growth of gastric cancer cells, showing promise for clinical application. The above findings indicate that ECP inhibited the growth of gastric cancer and induced apoptosis through the PI3K /Akt/mTOR pathway. In summary, the efficacy showed in our data suggests that ECP is a promising anti-tumor compound for gastric cancer.
Collapse
Affiliation(s)
- Xiaochen Qiu
- Department of General Surgery, the First Medical Center of Chinese PLA General Hospital, Beijing 100853, China; Medical School of Chinese PLA, Beijing, China
| | - Juan Wang
- Department of Oncology, Dushu Lake Hospital Affiliated to Soochow University, Medical Center of Soochow University, Suzhou Dushu Lake Hospital, Suzhou, Jiangsu, 215123, China
| | - Nan Zhang
- Medical School of Chinese PLA, Beijing, China; Department of Gastrointestinal Surgery, Shanghai Fourth People's Hospital, School of Medicine, Tongi Uni-versuty, Shanghai 200434, China
| | - Tongde Du
- Suzhou Institute of Systems Medicine, No. 100 Chongwen Road, Suzhou City, Suzhou 215000, China.
| | - Lin Chen
- Department of General Surgery, the First Medical Center of Chinese PLA General Hospital, Beijing 100853, China; Medical School of Chinese PLA, Beijing, China.
| | - Hongqing Xi
- Department of General Surgery, the First Medical Center of Chinese PLA General Hospital, Beijing 100853, China; Medical School of Chinese PLA, Beijing, China.
| |
Collapse
|
32
|
Chen G, Wen D, Shen L, Feng Y, Xiong Q, Li P, Zhao Z. Cepharanthine Exerts Antioxidant and Anti-Inflammatory Effects in Lipopolysaccharide (LPS)-Induced Macrophages and DSS-Induced Colitis Mice. Molecules 2023; 28:6070. [PMID: 37630322 PMCID: PMC10458559 DOI: 10.3390/molecules28166070] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 08/10/2023] [Accepted: 08/10/2023] [Indexed: 08/27/2023] Open
Abstract
Cepharanthine (CEP), a biscoclaurine alkaloid extracted from Stephania cepharantha Hayata, has been widely used for the treatment of various acute and chronic diseases, including leukopenia, and snake bites. Here, our objective was to investigate the anti-oxidative stress and anti-inflammatory response effects of CEP in lipopolysaccharide (LPS)-induced macrophages as well as dextran sulfate sodium (DSS)-induced colitis mice. Our findings demonstrated that supplementation with CEP effectively mitigates body weight loss and elevation of disease activity index (DAI), reduces the malondialdehyde (MDA) content to 2.45 nM/mL while increasing the reduced glutathione (GSH) content to 35.53 μg/mL, inhibits inflammatory response, and maintains proper intestinal epithelium tight junctions in DSS-induced wild type (WT) mice. However, it failed to provide protective effects in DSS-induced transcription factor nuclear factor erythroid 2-related factor 2 (NRF2) knockout (NRF2-/-) mice. GSH content decreased to 10.85 μg/106 cells following LPS treatment, whereas supplementation with CEP increased the GSH content to 12.26 μg/106 cells. Moreover, CEP effectively attenuated ROS production in LPS-induced macrophages. Additionally, CEP exhibited inhibitory effects on pro-inflammatory cytokines and mediators in LPS-induced macrophages. Furthermore, we observed that supplementation with CEP promoted the expression of NRF2/heme oxygenase 1 (HO-1)/NADPH quinone oxidoreductase-1 (NQO-1) as well as the phosphorylation of the adenosine monophosphate-activated protein kinase alpha 1 (AMPK-α1)/protein kinase B (AKT)/glycogen synthase kinase-3 beta (GSK-3β) signaling pathway in macrophages while inhibiting the phosphorylation of the extracellular signal-regulated kinase (ERK)/c-Jun N-terminal kinase (JNK), and nuclear factor-kappa B p65 (NF-κB p65) signaling pathway in LPS-induced macrophages. Although CEP did not demonstrate inhibitory effects on oxidative stress or promote the expression of HO-1/NQO-1, it effectively activated the phosphorylation of the AMPK-α1/AKT/GSK-3β signaling pathway which is an upstream regulator of NRF2 in LPS-induced primary peritoneal macrophages from NRF2-/- mice. In summary, our findings suggest that CEP exerts protective effects against oxidative stress and inflammatory response by activating the AMPK-α1/AKT/GSK-3β/NRF2 signaling pathway while concurrently inhibiting the activation of mitogen activated protein kinases (MAPKs) and the NF-κB p65 signaling pathway. These results not only elucidate the mechanisms underlying CEP's protective effects on colon oxidative stress and inflammation but also provide evidence supporting NRF2 as a potential therapeutic target for IBD treatment.
Collapse
Affiliation(s)
- Guangxin Chen
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
- State Key Laboratory of Biological Feed, Ministry of Agriculture and Rural Afairs, Boen Biotechnology Co., Ltd., Ganzhou 341000, China
- Institute of Biomedical Sciences, Shanxi University, Taiyuan 030006, China; (D.W.); (L.S.); (Y.F.); (Q.X.); (P.L.)
| | - Da Wen
- Institute of Biomedical Sciences, Shanxi University, Taiyuan 030006, China; (D.W.); (L.S.); (Y.F.); (Q.X.); (P.L.)
| | - Lin Shen
- Institute of Biomedical Sciences, Shanxi University, Taiyuan 030006, China; (D.W.); (L.S.); (Y.F.); (Q.X.); (P.L.)
| | - Yazhi Feng
- Institute of Biomedical Sciences, Shanxi University, Taiyuan 030006, China; (D.W.); (L.S.); (Y.F.); (Q.X.); (P.L.)
| | - Qiuhong Xiong
- Institute of Biomedical Sciences, Shanxi University, Taiyuan 030006, China; (D.W.); (L.S.); (Y.F.); (Q.X.); (P.L.)
| | - Ping Li
- Institute of Biomedical Sciences, Shanxi University, Taiyuan 030006, China; (D.W.); (L.S.); (Y.F.); (Q.X.); (P.L.)
| | - Zhonghua Zhao
- Institute of Biomedical Sciences, Shanxi University, Taiyuan 030006, China; (D.W.); (L.S.); (Y.F.); (Q.X.); (P.L.)
| |
Collapse
|
33
|
Zhu X, Ren T, Xiong Q, Lin Z, Lin X, Lin G. Salidroside alleviates diet-induced obesity and insulin resistance by activating Nrf2/ARE pathway and enhancing the thermogenesis of adipose tissues. Food Sci Nutr 2023; 11:4735-4744. [PMID: 37576042 PMCID: PMC10420790 DOI: 10.1002/fsn3.3450] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 04/26/2023] [Accepted: 05/02/2023] [Indexed: 08/15/2023] Open
Abstract
Recent reports suggest that salidroside protects cardiomyocytes from oxidative injury and stimulates glucose uptake by skeletal muscle cells. Despite these findings, the therapeutic potential of salidroside in the treatment of obesity and insulin resistance remains uncertain and requires further investigation. In the present study, the treatment effect of salidroside on the onset and development of the obese phenotype and insulin resistance as well as the underlying mechanisms was investigated using long-term high-fat diet-induced obese mice supplemented with salidroside. We used biochemical kits to determine serum biochemical parameters (including triacylglycerol, total cholesterol, high-density lipoprotein cholesterol, low-density lipoprotein cholesterol, fasting glucose, and insulin). The results show that salidroside-supplemented animals showed better glucose tolerance and insulin sensitivity, decreased blood lipids, and weight gain (p < .05). Protein expression of p-Nrf2 and Nrf2 was analyzed by western blotting, and the mRNA levels of thermogenic-related genes (Ucp1, Pgc1a, Prdm16, and Cidea) were detected by quantitative RT-PCR. The results show an improvement in lipid peroxidation and Nrf2/ARE signaling, as well as an increased expression of the Ucp1, Pgc1a, Prdm16, and Cidea (p < .05). Our evidence suggests that salidroside alleviates diet-induced obesity and insulin resistance potentially by activating Nrf2/ARE pathway and enhancing the thermogenesis of adipose tissues. This induction represents a potential technique for the management of comorbidities related to obesity and its prevention.
Collapse
Affiliation(s)
- Xiaozhen Zhu
- Department of PharmacyThe Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical UniversityWenzhouChina
| | - Ting Ren
- College of Life and Environmental ScienceWenzhou UniversityWenzhouChina
| | - Qiushuang Xiong
- Department of PharmacyThe Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical UniversityWenzhouChina
| | - Zhengfeng Lin
- Department of PharmacyThe Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical UniversityWenzhouChina
| | - Xiaoxiao Lin
- Department of PharmacyThe Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical UniversityWenzhouChina
| | - Guangyong Lin
- Department of PharmacyThe Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical UniversityWenzhouChina
| |
Collapse
|
34
|
Liu J, Cai J, Fan P, Dong X, Zhang N, Tai J, Cao Y. Salidroside protects mice from high-fat diet-induced obesity by modulating the gut microbiota. Int Immunopharmacol 2023; 120:110278. [PMID: 37192552 DOI: 10.1016/j.intimp.2023.110278] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 04/21/2023] [Accepted: 04/30/2023] [Indexed: 05/18/2023]
Abstract
Obesity is a systemic disease with multisystem inflammation associated with gut dysbiosis. Salidroside (SAL) which is a major glycoside extracted from Rhodiola rosea L. has a wide range of pharmacological effects, but the role of gut microbiota in the protective effects of SAL on obesity has not been studied. Herein, we aim to explore whether SAL could ameliorate high-fat diet (HFD)-induced obesity in mice by modulating microbiota. Results showed that oral treatment with SAL alleviated HFD-induced obesity in mice as evidenced by body weight and fat weight. SAL supplementation effectively attenuated fat accumulation, lipid synthesis genes expression, liver inflammation, and metabolic endotoxemia. In addition, SAL treatment alleviated intestinal damage and increased the expression of mucin protein (Mucin-2) and tight junction (TJ) proteins (Occludin and Zonula Occludens-1). 16S rRNA sequencing analysis revealed that the gut microbiota of obese mice was also partly improved by SAL via restoring the microbial community structure and diversity. A fecal microbiota transplantation (FMT) study was designed to verify the causality. Compared with fecal transplantation (FM) from the HFD-treated mice, FM from the SAL-treated mice significantly mitigate the symptoms of obese mice, including decreasing body weight, fat accumulation, and attenuating pathological damage in the gut. Thus, SAL could be a remarkable candidate to prevent obesity.
Collapse
Affiliation(s)
- Jiuxi Liu
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun 130062, People's Republic of China; Guangzhou Institute of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, People's Republic of China
| | - Jiapei Cai
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun 130062, People's Republic of China
| | - Peng Fan
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun 130062, People's Republic of China
| | - Xue Dong
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun 130062, People's Republic of China
| | - Naisheng Zhang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun 130062, People's Republic of China
| | - Jiandong Tai
- Department of Colorectal and Anal Surgery, The First Hospital of Jilin University, Changchun 130021, People's Republic of China.
| | - Yongguo Cao
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun 130062, People's Republic of China; Key Laboratory for Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun 130062, People's Republic of China.
| |
Collapse
|
35
|
Liu Y, Liu Q, Zhang Z, Yang Y, Zhou Y, Yan H, Wang X, Li X, Zhao J, Hu J, Yang S, Tian Y, Yao Y, Qiu Z, Song Y, Yang Y. The regulatory role of PI3K in ageing-related diseases. Ageing Res Rev 2023; 88:101963. [PMID: 37245633 DOI: 10.1016/j.arr.2023.101963] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 05/23/2023] [Accepted: 05/25/2023] [Indexed: 05/30/2023]
Abstract
Ageing is a physiological/pathological process accompanied by the progressive damage of cell function, triggering various ageing-related disorders. Phosphatidylinositol 3-kinase (PI3K), which serves as one of the central regulators of ageing, is closely associated with cellular characteristics or molecular features, such as genome instability, telomere erosion, epigenetic alterations, and mitochondrial dysfunction. In this review, the PI3K signalling pathway was firstly thoroughly explained. The link between ageing pathogenesis and the PI3K signalling pathway was then summarized. Finally, the key regulatory roles of PI3K in ageing-related illnesses were investigated and stressed. In summary, we revealed that drug development and clinical application targeting PI3K is one of the focal points for delaying ageing and treating ageing-related diseases in the future.
Collapse
Affiliation(s)
- Yanqing Liu
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an 710069, China
| | - Qiong Liu
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an 710069, China
| | - Zhe Zhang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an 710069, China
| | - Yaru Yang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an 710069, China
| | - Yazhe Zhou
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an 710069, China
| | - Huanle Yan
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an 710069, China
| | - Xin Wang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an 710069, China
| | - Xiaoru Li
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an 710069, China
| | - Jing Zhao
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an 710069, China
| | - Jingyan Hu
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an 710069, China
| | - Shulin Yang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an 710069, China
| | - Yifan Tian
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an 710069, China
| | - Yu Yao
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an 710069, China
| | - Zhenye Qiu
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an 710069, China
| | - Yanbin Song
- Department of Cardiology, Affiliated Hospital, Yan'an University, 43 North Street, Yan'an 716000, China.
| | - Yang Yang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an 710069, China.
| |
Collapse
|
36
|
Li B, Guo J, Zhou X, Li W, Wang N, Cao R, Cui S. The emerging role of pyroptosis in neuropathic pain. Int Immunopharmacol 2023; 121:110562. [PMID: 37364324 DOI: 10.1016/j.intimp.2023.110562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 06/10/2023] [Accepted: 06/22/2023] [Indexed: 06/28/2023]
Abstract
Neuropathic pain caused by somatosensory system injuries is notoriously difficult to treat. Previous research has shown that neuroinflammation and cell death have been implicated in the pathophysiology of neuropathic pain. Pyroptosis is a form of programmed cell death associated with inflammatory processes, as it can enhance or sustain the inflammatory response by releasing pro-inflammatory cytokines. This review presents the current knowledge on pyroptosis and its underlying mechanisms, including the canonical and noncanonical pathways. Moreover, we discuss recent findings on the role of pyroptosis in neuropathic pain and its potential as a therapeutic target. In conclusion, this review highlights the potential significance of pyroptosis as a promising target for developing innovative therapies to treat neuropathic pain.
Collapse
Affiliation(s)
- Baolong Li
- Department of Hand and Foot Surgery, The Third Bethune Hospital of Jilin University, Changchun, China; Key Laboratory of Peripheral Nerve Injury and Regeneration of Jilin Province, Changchun, China
| | - Jin Guo
- Department of Hand and Foot Surgery, The Third Bethune Hospital of Jilin University, Changchun, China; Key Laboratory of Peripheral Nerve Injury and Regeneration of Jilin Province, Changchun, China
| | - Xiongyao Zhou
- Department of Hand and Foot Surgery, The Third Bethune Hospital of Jilin University, Changchun, China; Key Laboratory of Peripheral Nerve Injury and Regeneration of Jilin Province, Changchun, China
| | - Weizhen Li
- Department of Hand and Foot Surgery, The Third Bethune Hospital of Jilin University, Changchun, China; Key Laboratory of Peripheral Nerve Injury and Regeneration of Jilin Province, Changchun, China
| | - Ningning Wang
- Department of Hand and Foot Surgery, The Third Bethune Hospital of Jilin University, Changchun, China; Key Laboratory of Peripheral Nerve Injury and Regeneration of Jilin Province, Changchun, China
| | - Rangjuan Cao
- Department of Hand and Foot Surgery, The Third Bethune Hospital of Jilin University, Changchun, China; Key Laboratory of Peripheral Nerve Injury and Regeneration of Jilin Province, Changchun, China.
| | - Shusen Cui
- Department of Hand and Foot Surgery, The Third Bethune Hospital of Jilin University, Changchun, China; Key Laboratory of Peripheral Nerve Injury and Regeneration of Jilin Province, Changchun, China.
| |
Collapse
|
37
|
Ge Y, Bruno M, Nash MS, Coates NH, Chorley BN, Cave MC, Beier JI. Vinyl chloride enhances high-fat diet-induced proteome alterations in the mouse pancreas related to metabolic dysfunction. Toxicol Sci 2023; 193:103-114. [PMID: 36892438 PMCID: PMC10176240 DOI: 10.1093/toxsci/kfad024] [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] [Indexed: 03/10/2023] Open
Abstract
Alterations in physiological processes in pancreas have been associated with various metabolic dysfunctions and can result from environmental exposures, such as chemicals and diet. It was reported that environmental vinyl chloride (VC) exposure, a common industrial organochlorine and environmental pollutant, significantly exacerbated metabolic-related phenotypes in mice fed concurrently with high-fat diet (HFD) but not low-fat diet (LFD). However, little is known about the role of the pancreas in this interplay, especially at a proteomic level. The present study was undertaken to examine the protein responses to VC exposure in pancreas tissues of C57BL/6J mice fed LFD or HFD, with focus on the investigation of protein expression and/or phosphorylation levels of key protein biomarkers of carbohydrate, lipid, and energy metabolism, oxidative stress and detoxification, insulin secretion and regulation, cell growth, development, and communication, immunological responses and inflammation, and biomarkers of pancreatic diseases and cancers. We found that the protein alterations may indicate diet-mediated susceptibility in mouse pancreas induced by HFD to concurrent exposure of low levels of inhaled VC. These proteome biomarkers may lead to a better understanding of pancreas-mediated adaptive or adverse response and susceptibility to metabolic disease.
Collapse
Affiliation(s)
- Yue Ge
- Center for Computational Toxicology and Exposure, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina 27711, USA
| | - Maribel Bruno
- Center for Computational Toxicology and Exposure, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina 27711, USA
| | - Maliha S Nash
- Center for Public Health and Environmental Assessment, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina 27711, USA
| | - Najwa Haykal Coates
- Center for Public Health and Environmental Assessment, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina 27711, USA
| | - Brian N Chorley
- Center for Computational Toxicology and Exposure, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina 27711, USA
| | - Matthew C Cave
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, Kentucky 40202, USA
| | - Juliane I Beier
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, USA
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, USA
- Pittsburgh Liver Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, USA
| |
Collapse
|
38
|
Guo Z, Liu FY, Yang D, Wang MY, Li CF, Tang N, Ma SQ, An P, Yang Z, Tang QZ. Salidroside ameliorates pathological cardiac hypertrophy via TLR4-TAK1-dependent signaling. Phytother Res 2023; 37:1839-1849. [PMID: 36512326 DOI: 10.1002/ptr.7701] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 11/09/2022] [Accepted: 11/19/2022] [Indexed: 12/14/2022]
Abstract
Salidroside, a prominent active ingredient in traditional Chinese medicines, is garnering increased attention because of its unique pharmacological effects against ischemic heart disease via MAPK signaling, which plays a critical role in regulating the evolution of ventricular hypertrophy. However, the function of Salidroside on myocardial hypertrophy has not yet been elucidated. C57BL/6 mice were subjected to transverse aortic constriction (TAC), and treated with Salidroside (100 mg kg-1 day-1 ) by oral gavage for 3 weeks starting 1 week after surgery. Four weeks after TAC surgery, the mice were subjected to echocardiography and then sacrificed to harvest the hearts for analysis. For in vitro study, neonatal rat cardiomyocytes were used to validate the protective effects of Salidroside in response to Angiotensin II (Ang II, 1 μM) stimulation. Here, we proved that Salidroside dramatically inhibited hypertrophic reactions generated by pressure overload and isoproterenol (ISO) injection. Salidroside prevented the activation of the TAK1-JNK/p38 axis. Salidroside pretreatment of TAK1-inhibited cardiomyocytes shows no additional attenuation of Ang II-induced cardiomyocytes hypertrophy and signaling pathway activation. The overexpression of constitutively active TAK1 removed the protective effects of Salidroside on myocardial hypertrophy. TAC-induced increase of TLR4 protein expression was reduced considerably in the Salidroside treated mice. Transient transfection of small interfering RNA targeting TLR4 (siTLR4) in cardiomyocytes did not further decrease the activation of the TAK1/JNK-p38 axis. In conclusion, Salidroside functioned as a TLR4 inhibitor and displayed anti-hypertrophic action via the TAK1/JNK-p38 pathway.
Collapse
Affiliation(s)
- Zhen Guo
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, People's Republic of China
| | - Fang-Yuan Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, People's Republic of China
| | - Dan Yang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, People's Republic of China
| | - Ming-Yu Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, People's Republic of China
| | - Chen-Fei Li
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, People's Republic of China
| | - Nan Tang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, People's Republic of China
| | - Shu-Qing Ma
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, People's Republic of China
| | - Peng An
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, People's Republic of China
| | - Zheng Yang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, People's Republic of China
| | - Qi-Zhu Tang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, People's Republic of China
| |
Collapse
|
39
|
Wang W, Yang W, Dai Y, Liu J, Chen ZY. Production of Food-Derived Bioactive Peptides with Potential Application in the Management of Diabetes and Obesity: A Review. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:5917-5943. [PMID: 37027889 PMCID: PMC11966776 DOI: 10.1021/acs.jafc.2c08835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 03/12/2023] [Accepted: 03/14/2023] [Indexed: 06/19/2023]
Abstract
The prevalence of diabetes mellitus and obesity is increasing worldwide. Bioactive peptides are naturally present in foods or in food-derived proteins. Recent research has shown that these bioactive peptides have an array of possible health benefits in the management of diabetes and obesity. First, this review will summarize the top-down and bottom-up production methods of the bioactive peptides from different protein sources. Second, the digestibility, bioavailability, and metabolic fate of the bioactive peptides are discussed. Last, the present review will discuss and explore the mechanisms by which these bioactive peptides help against obesity and diabetes based on in vitro and in vivo studies. Although several clinical studies have demonstrated that bioactive peptides are beneficial in alleviating diabetes and obesity, more double-blind randomized controlled trials are needed in the future. This review has provided novel insights into the potential of food-derived bioactive peptides as functional foods or nutraceuticals to manage obesity and diabetes.
Collapse
Affiliation(s)
- Weiwei Wang
- College
of Food Science and Engineering, Nanjing
University of Finance and Economics/Collaborative Innovation Center
for Modern Grain Circulation and Safety, Nanjing 210023, China
| | - Wenjian Yang
- College
of Food Science and Engineering, Nanjing
University of Finance and Economics/Collaborative Innovation Center
for Modern Grain Circulation and Safety, Nanjing 210023, China
| | - Yi Dai
- College
of Food Science and Engineering, Nanjing
University of Finance and Economics/Collaborative Innovation Center
for Modern Grain Circulation and Safety, Nanjing 210023, China
| | - Jianhui Liu
- College
of Food Science and Engineering, Nanjing
University of Finance and Economics/Collaborative Innovation Center
for Modern Grain Circulation and Safety, Nanjing 210023, China
| | - Zhen-Yu Chen
- Food
& Nutritional Sciences Programme, School of Life Sciences, The Chinese University of Hong Kong, Shatin, NT, Hong Kong, China
| |
Collapse
|
40
|
Fei SF, Tong DB, Jia F. Antiatherosclerotic Effect and Molecular Mechanism of Salidroside. Rev Cardiovasc Med 2023; 24:97. [PMID: 39076283 PMCID: PMC11273014 DOI: 10.31083/j.rcm2404097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 11/25/2022] [Accepted: 12/12/2022] [Indexed: 07/31/2024] Open
Abstract
Atherosclerotic cardiovascular disease is currently the leading cause of death worldwide. Its pathophysiological basis includes endothelial dysfunction, macrophage activation, vascular smooth muscle cell (VSMC) proliferation, lipid metabolism, platelet aggregation, and changes in the gut microbiota. Salidroside has beneficial effects on atherosclerosis through multiple pathways. In this review, we present studies on the regulatory effect of salidroside on atherosclerosis. Furthermore, we report the protective effects of salidroside against atherosclerosis by ameliorating endothelial dysfunction, suppressing macrophage activation and polarization, inhibiting VSMC proliferation, adjusting lipid metabolism, attenuating platelet aggregation, and modulating the gut microbiota. This review provides further understanding of the molecular mechanism of salidroside and new ideas for atherosclerosis management.
Collapse
Affiliation(s)
- Si-Fan Fei
- Department of Cardiovascular Medicine, The First People's Hospital of Changzhou, The Third Affiliated Hospital of Soochow University, 213000 Changzhou, Jiangsu, China
| | - De-Bing Tong
- Department of Cardiovascular Medicine, The First People's Hospital of Changzhou, The Third Affiliated Hospital of Soochow University, 213000 Changzhou, Jiangsu, China
| | - Fang Jia
- Department of Cardiovascular Medicine, The First People's Hospital of Changzhou, The Third Affiliated Hospital of Soochow University, 213000 Changzhou, Jiangsu, China
| |
Collapse
|
41
|
Wang Q, Yang X, Ma J, Xie X, Sun Y, Chang X, Bi H, Xue H, Qin Z. PI3K/AKT pathway promotes keloid fibroblasts proliferation by enhancing glycolysis under hypoxia. Wound Repair Regen 2023; 31:139-155. [PMID: 36571288 DOI: 10.1111/wrr.13067] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 11/30/2022] [Accepted: 12/20/2022] [Indexed: 12/27/2022]
Abstract
Our previous study demonstrated altered glucose metabolism and enhanced phosphorylation of the PI3K/AKT pathway in keloid fibroblasts (KFb) under hypoxic conditions. However, whether the PI3K/AKT pathway influences KFb cell function by regulating glucose metabolism under hypoxic conditions remains unclear. Here, we show that when PI3K/AKT pathway was inactivated with LY294002, the protein expression of glycolytic enzymes decreased, while the amount of mitochondria and mitochondrial membrane potential increased. The key parameters of extracellular acidification rate markedly diminished, and those of oxygen consumption rate significantly increased after inhibition of the PI3K/AKT pathway. When the PI3K/AKT pathway was suppressed, the levels of reactive oxygen species (ROS) and mitochondrial ROS (mitoROS) were significantly increased. Meanwhile, cell proliferation, migration and invasion were inhibited, and apoptosis was increased when the PI3K/AKT pathway was blocked. Additionally, cell proliferation was compromised when KFb were treated with both SC79 (an activator of the PI3K/AKT pathway) and 2-deoxy-d-glucose (an inhibitor of glycolysis), compared with the SC79 group. Moreover, a positive feedback mechanism was demonstrated between the PI3K/AKT pathway and hypoxia-inducible factor-1α (HIF-1α). Our data collectively demonstrated that the PI3K/AKT pathway promotes proliferation and inhibits apoptosis in KFb under hypoxia by regulating glycolysis, indicating that the PI3K/AKT signalling pathway could be a therapeutic target for keloids.
Collapse
Affiliation(s)
- Qifei Wang
- Department of Plastic Surgery, Peking University Third Hospital, Beijing, China
| | - Xin Yang
- Department of Plastic Surgery, Peking University Third Hospital, Beijing, China
| | - Jianxun Ma
- Department of Plastic Surgery, Peking University Third Hospital, Beijing, China
| | - Xiang Xie
- Department of Plastic Surgery, Peking University Third Hospital, Beijing, China
| | - Yimou Sun
- Department of Plastic Surgery, Peking University Third Hospital, Beijing, China
| | - Xu Chang
- Department of Plastic Surgery, Peking University Third Hospital, Beijing, China
| | - Hongsen Bi
- Department of Plastic Surgery, Peking University Third Hospital, Beijing, China
| | - Hongyu Xue
- Department of Plastic Surgery, Peking University Third Hospital, Beijing, China
| | - Zelian Qin
- Department of Plastic Surgery, Peking University Third Hospital, Beijing, China
| |
Collapse
|
42
|
Ahmed SA, Sarma P, Barge SR, Swargiary D, Devi GS, Borah JC. Xanthosine, a purine glycoside mediates hepatic glucose homeostasis through inhibition of gluconeogenesis and activation of glycogenesis via regulating the AMPK/ FoxO1/AKT/GSK3β signaling cascade. Chem Biol Interact 2023; 371:110347. [PMID: 36627075 DOI: 10.1016/j.cbi.2023.110347] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 12/22/2022] [Accepted: 01/06/2023] [Indexed: 01/09/2023]
Abstract
Type 2 Diabetes Mellitus (T2DM) is characterized by hepatic insulin resistance, which results in increased glucose production and reduced glycogen storage in the liver. There is no previous study in the literature that has explored the role of Xanthosine in hepatic insulin resistance. Moreover, mechanistic explanation for the beneficial effects of Xanthosine in lowering glucose production in diabetes is yet to be determined. This study for the first time investigated the beneficial effects of Tribulus terrestris (TT) and its active constituent, Xanthosine on gluconeogenesis and glycogenesis in Free Fatty Acid (FFA)-induced CC1 hepatocytes and streptozotocin (STZ)-induced Wistar rats. Xanthosine enhanced glucose uptake and decreased glucose production through phosphorylation of AMP-activated protein kinase (AMPK) and forkhead box transcription factor O1 (FoxO1), and downregulation of two rate limiting enzymes of gluconeogenesis, phosphoenolpyruvate carboxykinase (PEPCK) and glucose-6-phosphatase (G6Pase) expression in FFA-induced CC1 cells. Xanthosine also prevented FFA-induced decreases in the phosphorylation of AKT/Protein kinase B, glycogen synthase kinase-3β (GSK3β), and increased glycogen synthase (GS) phosphorylation to increase the glycogen content in the hepatocytes. Moreover, in STZ-induced diabetic rats, oral administration of TT n-butanol fraction (TTBF) enriched with compound Xanthosine (10, 50 & 100 mg/kg body weight) improved insulin sensitivity, reduced fasting blood glucose levels, improved glucose homeostasis by reducing gluconeogenesis via AMPK/FoxO1-mediated PEPCK and G6Pase down-regulation and increasing glycogenesis via AKT/GSK3β-mediated GS activation. Overall, Xanthosine may be developed further for treating insulin resistance and hyperglycemia in T2DM.
Collapse
Affiliation(s)
- Semim Akhtar Ahmed
- Chemical Biology Laboratory 1, Life Sciences Division, Institute of Advanced Study in Science & Technology, Guwahati, 781035, Assam, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Pranamika Sarma
- Chemical Biology Laboratory 1, Life Sciences Division, Institute of Advanced Study in Science & Technology, Guwahati, 781035, Assam, India
| | - Sagar Ramrao Barge
- Chemical Biology Laboratory 1, Life Sciences Division, Institute of Advanced Study in Science & Technology, Guwahati, 781035, Assam, India
| | - Deepsikha Swargiary
- Chemical Biology Laboratory 1, Life Sciences Division, Institute of Advanced Study in Science & Technology, Guwahati, 781035, Assam, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Gurumayum Shalini Devi
- Chemical Biology Laboratory 1, Life Sciences Division, Institute of Advanced Study in Science & Technology, Guwahati, 781035, Assam, India
| | - Jagat C Borah
- Chemical Biology Laboratory 1, Life Sciences Division, Institute of Advanced Study in Science & Technology, Guwahati, 781035, Assam, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India.
| |
Collapse
|
43
|
Zhou J, Yan S, Guo X, Gao Y, Chen S, Li X, Zhang Y, Wang Q, Zheng T, Chen L. Salidroside protects pancreatic β-cells against pyroptosis by regulating the NLRP3/GSDMD pathway in diabetic conditions. Int Immunopharmacol 2023; 114:109543. [PMID: 36508922 DOI: 10.1016/j.intimp.2022.109543] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 11/30/2022] [Accepted: 12/01/2022] [Indexed: 12/13/2022]
Abstract
The NACHT, LRP, and PYD domains-containing protein 3 (NLRP3) inflammasome-evoked chronic inflammation is involved in the pathogenesis of diabetes mellitus (DM), and the NLRP3/gasdermin D (GSDMD)-mediated canonical pathway of pyroptosis leads to the loss of pancreatic β-cells and failure of pancreatic function in DM. A previous study demonstrated that salidroside (SAL) alleviates the pathological hyperplasia of pancreatic β-cells in db/db mice. However, it is not clear whether the NLRP3/GSDMD pathway-mediated pyroptosis can be regulated by SAL. In addition, the action of SAL on pancreatic β-cells in DM remains poorly understood. Thus, this study aimed to investigate the effects and underlying mechanisms of SAL on pancreatic β-cell pyroptosis. Rat insulinoma (INS-1) cells were cultured in a medium containing either high glucose (HG) or HG plus high insulin (HG-HI), and the effects of SAL on cell viability, AMP-activated protein kinase (AMPK) activity, reactive oxygen species (ROS) generation, NLRP3/GSDMD activation, and pyroptotic body formation were assessed. Streptozocin-induced DM mice were used to further investigate the effects of SAL on pancreatic pyroptosis. The results revealed aberrances on cell viability, AMPK activity, ROS generation, NLRP3/GSDMD activation, and pyroptotic body formation in HG- and HG-HI-exposed INS-1 cells; these abnormal effects were corrected by SAL in both a concentration- and AMPK-dependent manner. Moreover, SAL administration activated AMPK, suppressed NLRP3/GSDMD signaling, and protected pancreatic β-cells against pyroptosis in DM mice. These findings suggest that SAL promotes AMPK activation to suppress NLRP3/GSDMD-related pyroptosis in pancreatic β-cells under DM conditions.
Collapse
Affiliation(s)
- Jun Zhou
- School of Pharmaceutical Sciences, Hubei University of Medicine, Shiyan, Hubei, China; Institute of Wudang Traditional Chinese Medicine, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Shan Yan
- School of Pharmaceutical Sciences, Hubei University of Medicine, Shiyan, Hubei, China; Institute of Wudang Traditional Chinese Medicine, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Xu Guo
- School of Pharmaceutical Sciences, Hubei University of Medicine, Shiyan, Hubei, China; Institute of Wudang Traditional Chinese Medicine, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Yanguo Gao
- School of Pharmaceutical Sciences, Hubei University of Medicine, Shiyan, Hubei, China; Institute of Wudang Traditional Chinese Medicine, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Shiqi Chen
- School of Pharmaceutical Sciences, Hubei University of Medicine, Shiyan, Hubei, China; Institute of Wudang Traditional Chinese Medicine, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Xiaohan Li
- School of Pharmaceutical Sciences, Hubei University of Medicine, Shiyan, Hubei, China; Institute of Wudang Traditional Chinese Medicine, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Yonghong Zhang
- School of Pharmaceutical Sciences, Hubei University of Medicine, Shiyan, Hubei, China; Institute of Wudang Traditional Chinese Medicine, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Qibin Wang
- School of Pharmaceutical Sciences, Hubei University of Medicine, Shiyan, Hubei, China; Institute of Wudang Traditional Chinese Medicine, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China; Department of Pharmacy, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Tao Zheng
- School of Pharmaceutical Sciences, Hubei University of Medicine, Shiyan, Hubei, China; Institute of Wudang Traditional Chinese Medicine, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China; Department of Pharmacy, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China.
| | - Li Chen
- School of Pharmaceutical Sciences, Hubei University of Medicine, Shiyan, Hubei, China; Institute of Wudang Traditional Chinese Medicine, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China; Department of Pharmacy, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China.
| |
Collapse
|
44
|
Jiang B, Feng L, Yang T, Guo W, Li Y, Wang T, Liu C, Su H. Combination of chloroquine diphosphate and salidroside induces human liver cell apoptosis via regulation of mitochondrial dysfunction and autophagy. Mol Med Rep 2022; 27:37. [PMID: 36579660 PMCID: PMC9827261 DOI: 10.3892/mmr.2022.12924] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 11/24/2022] [Indexed: 12/28/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is the leading cause of cancer‑associated death in the world. Chemotherapy remains the primary treatment method for HCC. Despite advances in chemotherapy and modalities, recurrence and resistance limit therapeutic success. Salidroside (Sal), a bioactive component extracted from the rhizome of Rhodiola rosea L, exhibits a spectrum of biological activities including antitumor effects. In the present study, it was demonstrated that Sal could induce apoptosis and autophagy of 97H cells by using CCK‑8 assay, transmission electron microscopy (TEM), Hoechst33342 staining, MDC staining, western blotting. Pretreatment with Sal enhanced apoptosis and autophagy via upregulation of expression levels of Bax, Caspase‑3, Caspase‑9, light chain (LC)3‑II and Beclin‑1 proteins and downregulation of expression levels of Bcl‑2, LC3‑I and p62 protein in 97H cells. Furthermore, Sal was demonstrated to inhibit activation of the PI3K/Akt/mTOR signaling pathway and, when combined with autophagy inhibitor chloroquine diphosphate (CQ), increased phosphorylation of PI3K, Akt and mTOR proteins. The combined treatment with Sal and CQ not only decreased Sal‑induced autophagy, but also accelerated Sal‑induced apoptosis. Therefore, Sal‑induced autophagy might serve a role as a defense mechanism in human liver cancer cells and its inhibition may be a promising strategy for the adjuvant chemotherapy of liver cancer.
Collapse
Affiliation(s)
- Bing Jiang
- School of Basic Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou, Gansu 730000, P.R. China
| | - Longfei Feng
- School of Basic Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou, Gansu 730000, P.R. China
| | - Tao Yang
- School of Basic Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou, Gansu 730000, P.R. China
| | - Wenjing Guo
- School of Basic Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou, Gansu 730000, P.R. China
| | - Yangyang Li
- School of Basic Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou, Gansu 730000, P.R. China
| | - Tao Wang
- Translational Medicine Research Center, Gansu Provincial Academic Institute for Medical Research, Gansu Provincial Cancer Hospital, Lanzhou, Gansu 730050, P.R. China
| | - Chengguang Liu
- Clinical College of Integrated Chinese and Western Medicine, Guangxi University of Traditional Chinese Medicine, Nanning, Guangxi 530200, P.R. China
| | - Haixiang Su
- School of Basic Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou, Gansu 730000, P.R. China,Translational Medicine Research Center, Gansu Provincial Academic Institute for Medical Research, Gansu Provincial Cancer Hospital, Lanzhou, Gansu 730050, P.R. China,Correspondence to: Professor Haixiang Su, Translational Medicine Research Center, Gansu Provincial Academic Institute for Medical Research, Gansu Provincial Cancer Hospital, 2 Xiaoxihu East Street, Qilihe, Lanzhou, Gansu 730050, P.R. China, E-mail:
| |
Collapse
|
45
|
Pei D, Tian S, Bao Y, Zhang J, Xu D, Piao M. Protective effect of salidroside on streptozotocin-induced diabetic nephropathy by inhibiting oxidative stress and inflammation in rats via the Akt/GSK-3β signalling pathway. PHARMACEUTICAL BIOLOGY 2022; 60:1732-1738. [PMID: 36086879 PMCID: PMC9467606 DOI: 10.1080/13880209.2022.2116055] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 06/30/2022] [Accepted: 08/11/2022] [Indexed: 06/15/2023]
Abstract
CONTEXT Salidroside (SAL), one of the major glycosides isolated from the roots of Rhodiola rosea L. (Crassulaceae), has anti-inflammatory, antioxidant, and antidiabetic properties. OBJECTIVE Our study assessed whether SAL exerts a protective effect on streptozotocin (STZ)-induced diabetic nephropathy (DN) in rats via the Akt/GSK-3β signalling pathway. MATERIALS AND METHODS Adult male Wistar rats were divided into three groups (n = 8): normal control, DN + vehicle, and DN + SAL. SAL (50 mg/kg/day, oral) was administered for 8 weeks. Biochemical and histopathologic examinations were performed to evaluate the therapeutic effects of SAL on oxidative stress, inflammation, renal function, and apoptosis. RESULTS SAL induced rats demonstrated ameliorated levels of FBG (20.53 ± 0.72 mmol/L vs. 26.02 ± 1.44 mmol/L), urine albumin excretion (27.00 ± 1.46 mmol/L vs. 41.00 ± 1.59 mmol/L), blood urea nitrogen (14.42 ± 0.70 mmol/L vs. 17.77 ± 0.72 mmol/L), and serum creatinine (112.80 ± 6.98 mmol/L vs. 159.00 ± 3.81 mmol/L) compared to normal control rats, along with the alleviation of renal pathologic changes by improving the irregular shape of glomeruli tissues. Biochemical analysis showed that SAL-treated animals displayed suppressed levels of serum inflammatory cytokines and kidney oxidative stress markers and attenuated apoptotic characteristics. Moreover, it increased the phosphorylation levels of Akt and GSK-3β in kidneys. DISCUSSION AND CONCLUSION The present study validated the involvement of the Akt/GSK-3β signalling pathway in renal improvement. These findings can form the basis to investigate the protective effect of SAL in DN in clinical trials.
Collapse
Affiliation(s)
- Delong Pei
- Department of Urology, Affiliated Hospital of Yanbian University, Yanji, Jilin Province, China
| | - Shengri Tian
- Department of Urology, Affiliated Hospital of Yanbian University, Yanji, Jilin Province, China
| | - Yanqiu Bao
- Center of Morphological Experiment, Medical College of Yanbian University, Yanji, Jilin Province, China
| | - Jun Zhang
- Center of Morphological Experiment, Medical College of Yanbian University, Yanji, Jilin Province, China
| | - Dongyuan Xu
- Center of Morphological Experiment, Medical College of Yanbian University, Yanji, Jilin Province, China
| | - Minhu Piao
- Department of Urology, Affiliated Hospital of Yanbian University, Yanji, Jilin Province, China
| |
Collapse
|
46
|
Park JE, Han JS. HM-chromanone reverses the blockade of insulin signaling induced by high glucose levels in human HepG2 cells. Eur J Pharmacol 2022; 937:175358. [DOI: 10.1016/j.ejphar.2022.175358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 10/05/2022] [Accepted: 10/28/2022] [Indexed: 11/17/2022]
|
47
|
Tian S, Zhao H, Song H. Shared signaling pathways and targeted therapy by natural bioactive compounds for obesity and type 2 diabetes. Crit Rev Food Sci Nutr 2022; 64:5039-5056. [PMID: 36397728 DOI: 10.1080/10408398.2022.2148090] [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] [Indexed: 11/19/2022]
Abstract
Epidemiological evidence showed that patients suffering from obesity and T2DM are significantly at higher risk for chronic low-grade inflammation, oxidative stress, nonalcoholic fatty liver (NAFLD) and intestinal flora imbalance. Increasing evidence of pathological characteristics illustrates that some common signaling pathways participate in the occurrence, progression, treatment, and prevention of obesity and T2DM. These signaling pathways contain the pivotal players in glucose and lipid metabolism, e.g., AMPK, PI3K/AKT, FGF21, Hedgehog, Notch, and WNT; the inflammation response, for instance, Nrf2, MAPK, NF- kB, and JAK/STAT. Bioactive compounds from plants have emerged as key food components related to healthy status and disease prevention. They can act as signaling molecules to initiate or mediate signaling transduction that regulates cell function and homeostasis to repair and re-functionalize the damaged tissues and organs. Therefore, it is crucial to continuously investigate bioactive compounds as sources of new pharmaceuticals for obesity and T2DM. This review provides comprehensive information of the commonly shared signaling pathways between obesity and T2DM, and we also summarize the therapeutic bioactive compounds that may serve as anti-obesity and/or anti-diabetes therapeutics by regulating these associated pathways, which contribute to improving glucose and lipid metabolism, attenuating inflammation.
Collapse
Affiliation(s)
- Shuhua Tian
- College of Food Science and Engineering, Nanjing University of Finance and Economics/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing, China
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Haizhen Zhao
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Haizhao Song
- College of Food Science and Engineering, Nanjing University of Finance and Economics/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing, China
| |
Collapse
|
48
|
Research progress of targeting NLRP3 inflammasome in peripheral nerve injury and pain. Int Immunopharmacol 2022; 110:109026. [DOI: 10.1016/j.intimp.2022.109026] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 06/25/2022] [Accepted: 06/30/2022] [Indexed: 01/08/2023]
|
49
|
Yan S, Lu W, Zhou J, Guo X, Li J, Cheng H, Zhu X, Zhao Y, Duan M, Yang H, Zhang Y, Wang Q, Chen L, Zheng T. Aqueous extract of Scrophularia ningpoensis improves insulin sensitivity through AMPK-mediated inhibition of the NLRP3 inflammasome. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 104:154308. [PMID: 35792447 DOI: 10.1016/j.phymed.2022.154308] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 06/02/2022] [Accepted: 06/27/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND Scrophularia ningpoensis Hemsl. is a commonly used medicinal plant in China for the treatment of diabetes mellitus (DM), but its mechanism of action remains poorly described. Type 2 diabetes mellitus (T2DM) accounts for > 90% of all DM cases and is characterized by insulin resistance. PURPOSE The aim of this study was to investigate whether the insulin sensitivity can be improved by treatment with aqueous extract of S. ningpoensis (AESN) and further explore its mechanism(s) of activity. METHODS Primary mouse hepatocytes and human HepG2 hepatocytes were used to investigate the effects of AESN on cell viability, AMP-activated protein kinase (AMPK) activation and glucose output under normal culture conditions. To mimic hyperglycemia and insulin resistance in vitro, hepatocytes were exposed to high glucose (HG), and the influences of AESN on AMPK phosphorylation, NLRP3 inflammation activation, insulin signaling, lipid accumulation and glucose output were investigated. Increasing doses of AESN (50, 100 and 200 mg/kg/day) were administered by gavage to db/db mice for 8 weeks, and then biochemical analysis and histopathological examinations were performed. RESULTS AESN significantly activated AMPK and inhibited glucose output in hepatocytes, but did not impact cell viability under normal culture conditions. Moreover, in HG-treated hepatocytes, AESN protected against aberrant AMPK activity, NLRP3 inflammasome activation, insulin signaling, and lipid accumulation. AMPK inhibition abolished the regulatory effects of AESN on the NLRP3 inflammasome, insulin signaling, lipid accumulation, and glucose output of hepatocytes following HG exposure. Furthermore, AESN administration reduced blood glucose and serum insulin levels, improved lipid profiles and insulin resistance, and corrected the aberrant AMPK activity and NLRP3 inflammasome activation in liver tissues. CONCLUSION AESN improves insulin sensitivity via AMPK-mediated NLRP3 inflammasome inhibition.
Collapse
Affiliation(s)
- Shan Yan
- Institute of Wudang Traditional Chinese Medicine, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China; Department of Pharmacy, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China; College of Pharmacy, Hubei University of Medicine, Shiyan, Hubei, China
| | - Wei Lu
- Department of Pharmacy, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Jun Zhou
- Institute of Wudang Traditional Chinese Medicine, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China; Department of Pharmacy, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China; College of Pharmacy, Hubei University of Medicine, Shiyan, Hubei, China
| | - Xu Guo
- Institute of Wudang Traditional Chinese Medicine, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China; Department of Pharmacy, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China; College of Pharmacy, Hubei University of Medicine, Shiyan, Hubei, China
| | - Juyi Li
- Department of Pharmacy, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Hongbo Cheng
- College of Pharmacy, Hubei University of Medicine, Shiyan, Hubei, China
| | - Xiaoyan Zhu
- College of Pharmacy, Hubei University of Medicine, Shiyan, Hubei, China
| | - Yan Zhao
- Institute of Wudang Traditional Chinese Medicine, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China; Department of Pharmacy, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China; College of Pharmacy, Hubei University of Medicine, Shiyan, Hubei, China
| | - Mingzhu Duan
- College of Pharmacy, Hubei University of Medicine, Shiyan, Hubei, China
| | - Hongxu Yang
- College of Pharmacy, Hubei University of Medicine, Shiyan, Hubei, China
| | - Yonghong Zhang
- Department of Pharmacy, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Qibin Wang
- Department of Pharmacy, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China; College of Pharmacy, Hubei University of Medicine, Shiyan, Hubei, China
| | - Li Chen
- Institute of Wudang Traditional Chinese Medicine, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China; Department of Pharmacy, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China; College of Pharmacy, Hubei University of Medicine, Shiyan, Hubei, China,.
| | - Tao Zheng
- Institute of Wudang Traditional Chinese Medicine, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China; Department of Pharmacy, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China; College of Pharmacy, Hubei University of Medicine, Shiyan, Hubei, China,.
| |
Collapse
|
50
|
Mansour SZ, Moustafa EM, Moawed FSM. Modulation of endoplasmic reticulum stress via sulforaphane-mediated AMPK upregulation against nonalcoholic fatty liver disease in rats. Cell Stress Chaperones 2022; 27:499-511. [PMID: 35779187 PMCID: PMC9485504 DOI: 10.1007/s12192-022-01286-w] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 06/14/2022] [Accepted: 06/17/2022] [Indexed: 01/24/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a major health concern. Endoplasmic reticulum (ER) stress, inflammation, and metabolic dysfunctions may be targeted to prevent the progress of nonalcoholic fatty liver disease. Sulforaphane (SFN), a sulfur-containing compound that is abundant in broccoli florets, seeds, and sprouts, has been reported to have beneficial effects on attenuating metabolic diseases. In light of this, the present study was designed to elucidate the mechanisms by which SFN ameliorated ER stress, inflammation, lipid metabolism, and insulin resistance - induced by a high-fat diet and ionizing radiation (IR) in rats. In our study, the rats were randomly divided into five groups: control, HFD, HFD + SFN, HFD + IR, and HFD + IR + SFN groups. After the last administration of SFN, liver and blood samples were taken. As a result, the lipid profile, liver enzymes, glucose, insulin, IL-1β, adipokines (leptin and resistin), and PI3K/AKT protein levels, as well as the mRNA gene expression of ER stress markers (IRE-1, sXBP-1, PERK, ATF4, and CHOP), fatty acid synthase (FAS), peroxisome proliferator-activated receptor-α (PPAR-α). Interestingly, SFN treatment modulated the levels of proinflammatory cytokine including IL-1β, metabolic indices (lipid profile, glucose, insulin, and adipokines), and ER stress markers in HFD and HFD + IR groups. SFN also increases the expression of PPAR-α and AMPK genes in the livers of HFD and HFD + IR groups. Meanwhile, the gene expression of FAS and CHOP was significantly attenuated in the SFN-treated groups. Our results clearly show that SFN inhibits liver toxicity induced by HFD and IR by ameliorating the ER stress events in the liver tissue through the upregulation of AMPK and PPAR-α accompanied by downregulation of FAS and CHOP gene expression.
Collapse
Affiliation(s)
- Somaya Z Mansour
- Radiation Biology Research, National Center for Radiation Research and Technology, Atomic Energy Authority, Cairo, Egypt
| | - Enas M Moustafa
- Radiation Biology Research, National Center for Radiation Research and Technology, Atomic Energy Authority, Cairo, Egypt
| | - Fatma S M Moawed
- Health Radiation Research, National Center for Radiation Research and Technology, Egyptian Atomic Energy Authority, Cairo, Egypt.
| |
Collapse
|