1
|
Varadharajan V, Balu AK, Sinclair BJ, Perinbarajan GK, Jenifer A D, Ganesan Sudha H, Ramaswamy A, Venkidasamy B, Thiruvengadam M. Comprehensive analysis of Syzygium cumini L. pomace extract as an α-amylase inhibitor: In vitro inhibition, kinetics, and computational studies. Bioorg Chem 2025; 161:108498. [PMID: 40339502 DOI: 10.1016/j.bioorg.2025.108498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 04/12/2025] [Accepted: 04/19/2025] [Indexed: 05/10/2025]
Abstract
Type 2 diabetes mellitus (T2DM) is a widespread metabolic disorder characterized by impaired regulation of blood glucose levels. Jamun (Syzygium cumini L.) fruits and seeds have been traditionally used in Ayurveda to manage diabetes. While fruit and seed extracts have been extensively studied for their anti-α-amylase properties, pomace, a byproduct of juice extraction, remains under explored. This study investigated the α-amylase inhibitory potential of jamun pomace (JP) extract by using in vitro and in silico methods. Enzyme inhibition assays revealed an half-maximal inhibitory concentration (IC₅₀) value of 85.68 ± 5.22 μg/mL for the JP extract, comparable to acarbose (64.28 ± 7.15 μg/mL). The extract exhibited mixed-mode inhibition, whereas acarbose showed competitive mode inhibition. At 10 μg/mL, the Vmax of JP extract was half that of acarbose, demonstrating significant inhibition. GC-MS analysis identified 11 volatile compounds (R1-R11) in the JP extract. Density Functional Theory (DFT) and ADMET analyses confirmed the chemical reactivity of the volatiles, drug-like properties, and low toxicity. Molecular docking revealed a high binding score for R11 (-8.0 kcal/mol), similar to acarbose (-8.2 kcal/mol). Molecular dynamics simulations further demonstrated the stability of α-amylase complexes with R11, R3, and R8, with R11 showing the lowest binding energy (-28.75 ± 6.25 kcal/mol). These findings suggest that R11 and JP extracts hold promise as anti-diabetic agents. Utilizing JP extract as a nutraceutical offers the dual benefit of diabetes management and sustainable waste valorization in jamun juice production.
Collapse
Affiliation(s)
| | - Ashwath Kumar Balu
- Department of Biotechnology, PSG College of Technology, Peelamedu, Coimbatore, India; Department of Biotechnology, Indian Institute of Technology, Hyderabad, India
| | - Bruce Joshua Sinclair
- Departmet of Electronics and Communication Engineering, PSG College of Technology, Coimbatore, India
| | - Gopi Krishna Perinbarajan
- Departmet of Electronics and Communication Engineering, PSG College of Technology, Coimbatore, India
| | - Dharshini Jenifer A
- Department of Chemical Engineering, National Institute of Technology, Surathkal, Karnataka, India
| | | | - Arulvel Ramaswamy
- Department of Biotechnology, K S Rangasamy College of Technology, Tiruchengode, Namakkal District, Tamil Nadu 637 215, India
| | - Baskar Venkidasamy
- Centre for Biosciences and Biotechnology, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai 600077, Tamil Nadu, India.
| | - Muthu Thiruvengadam
- Department of Applied Bioscience, College of Life and Environmental Science, Konkuk University, Seoul, Republic of Korea.
| |
Collapse
|
2
|
Chopra C, Kukkar D, Bhatt P, Rajesh P, Kim KH. A review of deoxyribonucleic acid-based single-nucleotide polymorphisms in diabetic kidney disease among Asian populations: Challenges and future directions. Int J Biol Macromol 2025; 316:144407. [PMID: 40403785 DOI: 10.1016/j.ijbiomac.2025.144407] [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: 02/14/2025] [Revised: 05/15/2025] [Accepted: 05/18/2025] [Indexed: 05/24/2025]
Abstract
Diabetic kidney disease (DKD) is a persistent disorder that occurs as a result of long-term diabetes mellitus with genetic and environmental risk factors. The identification of DKD associated single-nucleotide polymorphisms (SNPs) is pivotal for patient screening. This manuscript briefly outlines the pathophysiology and the role of genetic factors in DKD expansion. It further discusses the utility of bioinformatic tools and laboratory techniques requried for the identification of DKD-specific SNPs along with integrated data analysis pipelines valuable to enhance the accuracy of genetic interpretation. A comparative analysis of various SNPs has also been made across diverse Asian populations in conjunction with environmental and lifestyle factors. The clinical relevance of SNPs in predicting DKD progression and stratifying patient risk is highlighted, with focus on gene-specific pathways and associated functional outcomes. The advances in genetic screening, gene-specific therapies, and microbiome-based therapy should help expand the utility of SNPs-based identification of DKD under diverse clinical settings. A structured clinical decision-making framework is proposed to support customized treatment based on SNP profiles. However, this domain has yet to gain widespread recognition with regard to variability in the effects of SNPs across diverse demographies, challenges in clinical translation, and ethical considerations in genetic testing.
Collapse
Affiliation(s)
- Chahat Chopra
- Department of Biotechnology, Chandigarh University, Gharuan, Mohali 140413, Punjab, India; University Centre for Research and Development, Chandigarh University, Gharuan, Mohali 140413, Punjab, India
| | - Deepak Kukkar
- Department of Biotechnology, Chandigarh University, Gharuan, Mohali 140413, Punjab, India; University Centre for Research and Development, Chandigarh University, Gharuan, Mohali 140413, Punjab, India.
| | - Poornima Bhatt
- Department of Biotechnology, Chandigarh University, Gharuan, Mohali 140413, Punjab, India; University Centre for Research and Development, Chandigarh University, Gharuan, Mohali 140413, Punjab, India
| | - Preeti Rajesh
- Department of Biotechnology, Brainware University, Ramkrishnapur Road, Barasat, Near Jagadighata Market, Kolkata, West Bengal 700125, India
| | - Ki-Hyun Kim
- Department of Civil and Environmental Engineering, Hanyang University, 222 Wangsimni-Ro, Seoul 04763, Republic of Korea.
| |
Collapse
|
3
|
Zhang Z, Wang J, Zhang Z, Gan Q, He Y, Chen D, Zhang Y, Zhao M. Systematic Exploration of Potential Toxicity Targets and Molecular Mechanisms of Emerging Short-Chain PFAS Substitutes: PFBA- and PFBS-Induced Hepatocellular Carcinoma Based on Toxicity Network Analysis, Machine Learning, and Biomimetic Calculations. J Appl Toxicol 2025. [PMID: 40448290 DOI: 10.1002/jat.4818] [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: 04/30/2025] [Revised: 05/09/2025] [Accepted: 05/16/2025] [Indexed: 06/02/2025]
Abstract
Perfluorobutanoic acid (PFBA) and perfluorobutanesulfonic acid (PFBS) are short-chain alternatives to traditional perfluoroalkyl and polyfluoroalkyl substances (PFASs). Long-term exposure to these pollutants is closely associated with hepatocellular carcinoma (HCC). However, the toxic targets and mechanisms underlying PFBA- and PFBS-induced HCC remain unclear. To address this knowledge gap, this study employed a multifaceted approach encompassing network toxicology, molecular docking, and molecular dynamic simulation. Thirty-six core targets associated with PFBA- and PFBS-induced HCC were identified, and 12 key genes were initially screened through network toxicity analysis. Subsequently, based on the TCGA and ICGC datasets, three classical algorithms were applied to screen key genes: PPARG, ESR1, and ALB. Further exploration of the HCC-related dataset from the GEO database identified six critical genes: PPARG, ESR1, CD36, ABCA1, ACACA, and ALB. Survival analysis and ROC analysis based on the TCGA dataset revealed and validated the strong association between the expression levels of key genes (PPARG, ESR1, and ACACA). Single-gene GSEA showed that these three key genes may induce HCC through multiple biological pathways via interfering with the normal growth and development of hepatocytes and promoting inflammation and cell proliferation. Ultimately, molecular dynamics demonstrated the strong binding affinities between PFBA, PFBS, and the three protein receptors, with the best stability and flexibility of the interaction between PFBS and PPARG. These findings provide insights into the theoretical foundation for applying network toxicology, molecular docking, and molecular dynamic simulations in environmental pollutant research.
Collapse
Affiliation(s)
- Zirui Zhang
- Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Jin Wang
- College of Computer Science, Chengdu University, Chengdu, Sichuan, China
| | - Zhongyi Zhang
- Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Qianrong Gan
- Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Yunliang He
- Institute of Traditional Chinese Medicine, Sichuan Academy of Chinese Medicine Sciences, Chengdu, Sichuan, China
| | - Donghui Chen
- Institute of Traditional Chinese Medicine, Sichuan Academy of Chinese Medicine Sciences, Chengdu, Sichuan, China
| | - Yong Zhang
- Institute of Traditional Chinese Medicine, Sichuan Academy of Chinese Medicine Sciences, Chengdu, Sichuan, China
| | - Mei Zhao
- Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| |
Collapse
|
4
|
Wang L, Huang J, Liu Z, Wang C. Decoding the Secrets of Odor-Active Compounds in Dark Tea. Compr Rev Food Sci Food Saf 2025; 24:e70206. [PMID: 40421835 DOI: 10.1111/1541-4337.70206] [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: 01/12/2025] [Revised: 04/23/2025] [Accepted: 05/07/2025] [Indexed: 05/28/2025]
Abstract
Dark tea has received growing attention in recent years due to its distinctive aroma and potential health benefits. The overall aroma profile of dark tea is determined by the combined effects of diverse odor-active compounds (OACs). However, comprehensive studies on these compounds remain limited. Beyond its role in sensory perception, could the distinctive aroma of dark tea also exert physiological functions? Existing research has primarily focused on the health benefits of non-volatile components in dark tea, such as tea polyphenols, tea polysaccharides, and theanine. However, the potential of OACs to influence physiological processes via olfactory receptors (ORs) remains underexplored in the literature. Accordingly, we systematically classify and comprehensively review OACs. Furthermore, we examine OAC-OR interactions, encompassing not only their roles in olfactory recognition and emotion regulation via nasal ORs but also their health benefits and potential therapeutic applications mediated by non-nasal ORs. Interestingly, our findings reveal that OACs follow a "structure-similarity-function-convergence" trend in both bioactivity and aromatic properties. Specifically, structurally similar OACs not only impart analogous aromatic profiles but may also share comparable physiological functions. This phenomenon provides new insights into the health potential of flavor compounds. This review not only enhances our understanding of OACs in dark tea but also systematically, and for the first time, explores the potential health-related functions mediated by interactions between OACs and ORs. It offers new insights for future integrative research on food flavor and health and may shed light on the synergistic role of odor and bioactive compounds in health regulation.
Collapse
Affiliation(s)
- Lianqing Wang
- Key Laboratory of Tea Science of Ministry of Education, Hunan Agricultural University, Changsha, Hunan, P. R. China
- National Research Center of Engineering and Technology for Utilization of Botanical Functional Ingredients, Changsha, Hunan, P. R. China
- Department of Food Nutrition and Health, The China Modern Agricultural Joint Graduate Institution, Zhengzhou, Henan, P. R. China
| | - Jianan Huang
- Key Laboratory of Tea Science of Ministry of Education, Hunan Agricultural University, Changsha, Hunan, P. R. China
- National Research Center of Engineering and Technology for Utilization of Botanical Functional Ingredients, Changsha, Hunan, P. R. China
- Yuelushan Laboratory, Changsha, Hunan, P. R. China
- National Key Laboratory for Tea Plant Germplasm Innovation and Resource Utilization, Hunan Agricultural University, Changsha, Hunan, P. R. China
- Key Laboratory for Evaluation and Utilization of Gene Resources of Horticultural Crops, Ministry of Agriculture and Rural Affairs of China, Hunan Agricultural University, Changsha, Hunan, P. R. China
| | - Zhonghua Liu
- Key Laboratory of Tea Science of Ministry of Education, Hunan Agricultural University, Changsha, Hunan, P. R. China
- National Research Center of Engineering and Technology for Utilization of Botanical Functional Ingredients, Changsha, Hunan, P. R. China
- Department of Food Nutrition and Health, The China Modern Agricultural Joint Graduate Institution, Zhengzhou, Henan, P. R. China
- Yuelushan Laboratory, Changsha, Hunan, P. R. China
- National Key Laboratory for Tea Plant Germplasm Innovation and Resource Utilization, Hunan Agricultural University, Changsha, Hunan, P. R. China
- Key Laboratory for Evaluation and Utilization of Gene Resources of Horticultural Crops, Ministry of Agriculture and Rural Affairs of China, Hunan Agricultural University, Changsha, Hunan, P. R. China
| | - Chao Wang
- Key Laboratory of Tea Science of Ministry of Education, Hunan Agricultural University, Changsha, Hunan, P. R. China
- National Research Center of Engineering and Technology for Utilization of Botanical Functional Ingredients, Changsha, Hunan, P. R. China
- Yuelushan Laboratory, Changsha, Hunan, P. R. China
- National Key Laboratory for Tea Plant Germplasm Innovation and Resource Utilization, Hunan Agricultural University, Changsha, Hunan, P. R. China
- Key Laboratory for Evaluation and Utilization of Gene Resources of Horticultural Crops, Ministry of Agriculture and Rural Affairs of China, Hunan Agricultural University, Changsha, Hunan, P. R. China
| |
Collapse
|
5
|
Ou X, Yu Z, Pan C, Zheng X, Li D, Qiao Z, Zheng X. Paeoniflorin: a review of its pharmacology, pharmacokinetics and toxicity in diabetes. Front Pharmacol 2025; 16:1551368. [PMID: 40260393 PMCID: PMC12009869 DOI: 10.3389/fphar.2025.1551368] [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: 12/25/2024] [Accepted: 03/19/2025] [Indexed: 04/23/2025] Open
Abstract
The escalating global prevalence of diabetes underscores the urgency of addressing its treatment and associated complications. Paeoniflorin, a monoterpenoid glycoside compound, has garnered substantial attention in recent years owing to its potential therapeutic efficacy in diabetes management. Thus, this study aims to systematically overview the pharmacological effects, pharmacokinetics and toxicity of paeoniflorin in diabetes. Plenty of evidences have verified that paeoniflorin improves diabetes and its complication through reducing blood sugar, enhancing insulin sensitivity, regulating gut microbiota and autophagy, restoration of mitochondrial function, regulation of lipid metabolism, anti-inflammation, anti-oxidative stress, inhibition of apoptosis, immune regulation and so on. Paeoniflorin possess the characteristics of rapid absorption, wide distribution, rapid metabolism and renal excretion. Meanwhile, toxicity studies have suggested that paeoniflorin has low acute toxicity, minimal subacute and chronic toxicity, and no genotoxic or mutational toxic effects. In conclusion, this paper systematically elucidates the potential therapeutic application and safety profile of paeoniflorin in diabetes management.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Xiaoyuan Zheng
- Pharmacy Department, Chongqing Emergency Medical Center, Chongqing University Central Hospital, School of Medicine, Chongqing University, Chongqing, China
| |
Collapse
|
6
|
Faulkner JL. Prolonged obesity alters males' adipose profile: implications for disease? Physiol Genomics 2025; 57:254-255. [PMID: 39964952 PMCID: PMC12172062 DOI: 10.1152/physiolgenomics.00191.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 01/22/2025] [Accepted: 02/06/2025] [Indexed: 02/20/2025] Open
Affiliation(s)
- Jessica L Faulkner
- Department of Physiology and OBGYN, Medical College of Georgia at Augusta University, Augusta, Georgia, United States
| |
Collapse
|
7
|
Lin Y, Ni X, Zhu L, Lin Y, Peng C, Lei Z, Wang Y, Wang H, You X, Li J, Shen H, Wei J. Multi-miRNAs-Mediated Hepatic Lepr Axis Suppression: A Pparg-Dicer1 Pathway-Driven Mechanism in Spermatogenesis for the Intergenerational Transmission of Paternal Metabolic Syndrome. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2410831. [PMID: 39792613 PMCID: PMC11884570 DOI: 10.1002/advs.202410831] [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: 09/05/2024] [Revised: 12/14/2024] [Indexed: 01/12/2025]
Abstract
Bisphenol A (BPA) is an "environmental obesogen" and this study aims to investigate the intergenerational impacts of BPA-induced metabolic syndrome (MetS), specifically focusing on unraveling mechanisms. Exposure to BPA induces metabolic disorders in the paternal mice, which are then transmitted to offspring, leading to late-onset MetS. Mechanistically, BPA upregulates Srebf1, which in turn promotes the Pparg-dependent transcription of Dicer1 in spermatocytes, increasing the levels of multiple sperm microRNAs (miRNAs). Several of these miRNAs are highly expressed in a synchronized manner in liver of the offspring. miR149-5p, miR150-5p, and miR700-5p target a specific region in the Lepr 3'UTR, termed "SMITE" ("Several MiRNAs Targeting Elements"), to negatively regulate Lepr. These inherited anti-Lepr miRNAs, also referred to inherited anti-Lepr miRNAs (IAL-miRs), modulate hepatic steatosis, and insulin signaling through the Lepr regulatory Igfbp2, Egfr, and Ampk. Furthermore, IAL-miRs inhibit Ccnd1 not only via binding to "SMITE" but also via Lepr-Igfbp2 axis, which contribute to hepatocyte senescence. These pathological processes interact in a self-reinforcing cycle, worsening MetS in the paternal BPA-exposed offspring. The findings reveal mechanism wherein lipid metabolism reprogramming in spermatocytes-induced perturbations of sperm miRNAs, triggered by BPA, leads to intergenerational inheritance of paternal MetS through suppression of the hepatic Lepr axis in the offspring.
Collapse
Affiliation(s)
- Yi Lin
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, National Innovation Platform for Industry‐Education Integration in Vaccine ResearchSchool of Public HealthXiamen UniversityXiamen361102China
| | - Xiuye Ni
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, National Innovation Platform for Industry‐Education Integration in Vaccine ResearchSchool of Public HealthXiamen UniversityXiamen361102China
| | - Lin Zhu
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, National Innovation Platform for Industry‐Education Integration in Vaccine ResearchSchool of Public HealthXiamen UniversityXiamen361102China
| | - Yilong Lin
- Department of Basic Medical SciencesSchool of MedicineXiamen UniversityXiamen361102China
| | - Cai Peng
- Department of Basic Medical SciencesSchool of MedicineXiamen UniversityXiamen361102China
| | - Zhao Lei
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, National Innovation Platform for Industry‐Education Integration in Vaccine ResearchSchool of Public HealthXiamen UniversityXiamen361102China
| | - Yihui Wang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, National Innovation Platform for Industry‐Education Integration in Vaccine ResearchSchool of Public HealthXiamen UniversityXiamen361102China
| | - Huan Wang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, National Innovation Platform for Industry‐Education Integration in Vaccine ResearchSchool of Public HealthXiamen UniversityXiamen361102China
| | - Xiang You
- Department of Basic Medical SciencesSchool of MedicineXiamen UniversityXiamen361102China
| | - Juan Li
- Department of Basic Medical SciencesSchool of MedicineXiamen UniversityXiamen361102China
| | - Heqing Shen
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, National Innovation Platform for Industry‐Education Integration in Vaccine ResearchSchool of Public HealthXiamen UniversityXiamen361102China
| | - Jie Wei
- Department of Basic Medical SciencesSchool of MedicineXiamen UniversityXiamen361102China
| |
Collapse
|
8
|
Ma H, Lu Y, Chen W, Gao Z, Wu D, Chong Y, Wu J, Xi D, Deng W, Hong J. Multiple omics analysis reveals the regulation of SIRT4 on lipid deposition and metabolism during the differentiation of bovine preadipocytes. Genomics 2025; 117:111006. [PMID: 39875030 DOI: 10.1016/j.ygeno.2025.111006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 01/18/2025] [Accepted: 01/20/2025] [Indexed: 01/30/2025]
Abstract
The differentiation and lipid metabolism of preadipocytes are crucial processes in IMF deposition. Studies have demonstrated that SIRT4 plays essential roles in energy metabolism and redox homeostasis, with its expression being coordinately regulated by multiple transcription factors associated with energy and lipid metabolism. In this study, the findings of multiple omics analysis reveal that SIRT4 significantly up-regulates the expression of genes involved in adipogenesis and enhances the differentiation and lipid deposition of bovine preadipocytes. Furthermore, SIRT4 profoundly influences the expression pattern of metabolites by increasing the abundance of substances involved in lipid synthesis while decreasing those that promote lipid oxidative decomposition. Additionally, SIRT4 broadly up-regulates the expression levels of various lipid classes, including glycerolipids, glycerophospholipids, sphingolipids, and sterol lipids. These findings not only provide a theoretical basis for molecular breeding and genetic improvement in beef cattle, but also offer potential therapeutic approaches for energy homeostasis disorders and obesity.
Collapse
Affiliation(s)
- Hongming Ma
- Yunnan Provincial Key Laboratory of Animal Nutrition and Feed Science, College of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, Yunnan, China
| | - Ying Lu
- Yunnan Provincial Key Laboratory of Animal Nutrition and Feed Science, College of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, Yunnan, China
| | - Wei Chen
- Yunnan Provincial Key Laboratory of Animal Nutrition and Feed Science, College of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, Yunnan, China
| | - Zhendong Gao
- Yunnan Provincial Key Laboratory of Animal Nutrition and Feed Science, College of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, Yunnan, China
| | - Dongwang Wu
- Yunnan Provincial Key Laboratory of Animal Nutrition and Feed Science, College of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, Yunnan, China
| | - Yuqing Chong
- Yunnan Provincial Key Laboratory of Animal Nutrition and Feed Science, College of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, Yunnan, China
| | - Jiao Wu
- Yunnan Provincial Key Laboratory of Animal Nutrition and Feed Science, College of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, Yunnan, China
| | - Dongmei Xi
- Yunnan Provincial Key Laboratory of Animal Nutrition and Feed Science, College of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, Yunnan, China
| | - Weidong Deng
- Yunnan Provincial Key Laboratory of Animal Nutrition and Feed Science, College of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, Yunnan, China
| | - Jieyun Hong
- Yunnan Provincial Key Laboratory of Animal Nutrition and Feed Science, College of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, Yunnan, China.
| |
Collapse
|
9
|
Jiang L, Sun X, Wan Y, Qin Q, Xu M, Ma J, Zan L, Wang H. Transcriptome Reveals the Promoting Effect of Beta-Sitosterol on the Differentiation of Bovine Preadipocytes. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:3400-3412. [PMID: 39874185 DOI: 10.1021/acs.jafc.4c10452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/30/2025]
Abstract
Natural small molecule compounds play crucial roles in regulating fat deposition. Beta-sitosterol exhibits multiple biological activities such as cholesterol reduction and anticancer effects. However, its regulatory mechanism in the differentiation of bovine preadipocytes remains unclear. We identified potential associations of Beta-sitosterol with biological processes such as cholesterol regulation and lipid metabolism through the prediction of its targets. We utilized techniques such as Oil Red O staining, Western blotting, RNA-seq, and others to elucidate the promoting effect of Beta-sitosterol on the differentiation of bovine preadipocytes. Furthermore, reducing the expression of the most downregulated gene among differential expressed genes (DEGs), MGP, promotes the differentiation of bovine preadipocytes. After interfering with MGP, RNA-seq analysis on the sixth day of differentiation revealed that DEGs were most significantly enriched in the PPAR signaling pathway. In this pathway, the expression levels of genes related to adipocyte differentiation, including CD36, RXRα, RXRγ, FABP4, PLIN1, ADIPO, and CAP, were significantly upregulated (P < 0.01). Western blot and ELISA analysis on genes related to the PPAR signaling pathway showed that interfering with MGP increased the expression of proteins such as RXRα, indicating the possible activation of the PPAR signaling pathway. In summary, Beta-sitosterol may promote the differentiation of bovine preadipocytes by reducing the expression of MGP, thereby activating the PPAR signaling pathway.
Collapse
Affiliation(s)
- Lei Jiang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Xiaolei Sun
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Yuan Wan
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Qihua Qin
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Min Xu
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Jianqiang Ma
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Linsen Zan
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
- National Beef Cattle Improvement Centre, Yangling, Shaanxi 712100, China
| | - Hongbao Wang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
- National Beef Cattle Improvement Centre, Yangling, Shaanxi 712100, China
| |
Collapse
|
10
|
Tang R, Xie C, Zhang X. NOD1: a metabolic modulator. Front Endocrinol (Lausanne) 2025; 15:1484829. [PMID: 39906040 PMCID: PMC11790428 DOI: 10.3389/fendo.2024.1484829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Accepted: 12/30/2024] [Indexed: 02/06/2025] Open
Abstract
Nucleotide-binding oligomerization domain 1 (NOD1) is an intracellular pattern recognition receptor that detects injury signals and initiates inflammatory responses and host defense. Furthermore, NOD1 serves as a metabolic mediator by influencing the metabolism of various tissues, including adipose tissue, liver, cardiovascular tissue, pancreatic β cells, adrenal glands, and bones through diverse mechanisms. It has been discovered that activated NOD1 is associated with the pathological mechanisms of certain metabolic diseases. This review presents a comprehensive summary of the impact of NOD1 on tissue-specific metabolism.
Collapse
Affiliation(s)
- Ruobing Tang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Chunguang Xie
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Chengdu, Sichuan, China
- Department of Endocrinology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Xiyu Zhang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Chengdu, Sichuan, China
- Department of Endocrinology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| |
Collapse
|
11
|
Mandujano-Lázaro G, Torres-Rojas MF, Ramírez-Moreno E, Marchat LA. Virtual screening combined with molecular docking for the !identification of new anti-adipogenic compounds. Sci Prog 2025; 108:368504251320313. [PMID: 39936374 PMCID: PMC11815789 DOI: 10.1177/00368504251320313] [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: 02/13/2025]
Abstract
Obesity is an important risk factor for diabetes, cardiovascular diseases, and cancer, reducing the quality of life and expectancy of millions of people. Consequently, obesity has turned into one of the most health public problems worldwide, which highlights the urgent need for new and safe treatments. Obesity is mainly related to excessive fat accumulation; therefore, proteins participating in white adipose tissue increase and dysfunction are considered pertinent and attractive targets for developing new methods that can help with body weight control. In this context, virtual screening of libraries containing a large number of molecules represents a valuable strategy for the identification of potential anti-adipogenic compounds with reduced costs and time production. Here, we review the scientific literature about the prediction of new ligands of specific proteins through molecular docking and virtual screening of chemical libraries, with the aim of proposing new potential anti-adipogenic molecules. First, we present the targets related to adipogenesis and adipocyte functions that were selected for the following studies: PPARγ, Crif1, SIRT1, ERβ, PC1, FTO, Mss51, and FABP4. Then, we describe the obtention of new ligands according to the characteristics of the virtual screening approach, i.e. a structure-based drug design (SBDD) or a ligand-based drug design (LBDD). Finally, the critical analysis of these computational strategies and the corresponding results points out the necessity of combining computational and in vitro or in vivo assays for the identification of effective new anti-adipogenic molecules for obesity control. It also evidences that translating molecular docking and virtual screening results into successful drug candidates for adipogenesis and obesity control remains a huge challenge.
Collapse
Affiliation(s)
- Gilberto Mandujano-Lázaro
- Laboratorio de Biomedicina Molecular 2, ENMH, Instituto Politécnico Nacional, Ciudad de México, México
| | - María F Torres-Rojas
- Laboratorio de Biomedicina Molecular 2, ENMH, Instituto Politécnico Nacional, Ciudad de México, México
| | - Esther Ramírez-Moreno
- Laboratorio de Biomedicina Molecular 2, ENMH, Instituto Politécnico Nacional, Ciudad de México, México
| | - Laurence A Marchat
- Laboratorio de Biomedicina Molecular 2, ENMH, Instituto Politécnico Nacional, Ciudad de México, México
| |
Collapse
|
12
|
Ghasemzadeh Rahbardar M, Fazeli Kakhki H, Hosseinzadeh H. Ziziphus jujuba (Jujube) in Metabolic Syndrome: From Traditional Medicine to Scientific Validation. Curr Nutr Rep 2024; 13:845-866. [PMID: 39354208 DOI: 10.1007/s13668-024-00581-5] [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: 09/13/2024] [Indexed: 10/03/2024]
Abstract
PURPOSE OF REVIEW This review evaluates the therapeutic potential of Ziziphus jujuba and its main components in managing complications of metabolic syndrome, including diabetes, dyslipidemia, obesity, and hypertension. RECENT FINDINGS The reviewed studies provide evidence supporting the use of Z. jujuba and its main components (lupeol and betulinic acid) as natural treatments for complications of metabolic syndrome. These substances enhance glucose uptake through the activation of signaling pathways such as phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt), reduce hepatic glucose synthesis, and increase glucose uptake by adipocytes and skeletal muscle cells. They also improve insulin sensitivity by modulating AMP-activated protein kinase (AMPK) activity and regulating insulin signaling proteins and glucose transporters. In the field of dyslipidemia, they inhibit triglyceride synthesis, lipid accumulation, and adipogenic enzymes, while influencing key signaling pathways involved in adipogenesis. Z. jujuba and its constituents demonstrate anti-adipogenic effects, inhibiting lipid accumulation and modulating adipogenic enzymes and transcription factors. They also exhibit positive effects on endothelial function and vascular health by enhancing endothelial nitric oxide synthase (eNOS) expression, NO production, and antioxidant enzyme activity. Z. jujuba, lupeol, and betulinic acid hold promise as natural treatments for complications of metabolic syndrome. They improve glucose metabolism, insulin sensitivity, and lipid profiles while exerting anti-adipogenic effects and enhancing endothelial function. However, further research is needed to elucidate the mechanisms and confirm their efficacy in clinical trials. These natural compounds offer potential as alternative therapies for metabolic disorders and contribute to the growing body of evidence supporting the use of natural medicines in their management.
Collapse
Affiliation(s)
| | - Homa Fazeli Kakhki
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hossein Hosseinzadeh
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
13
|
Li Y, Xu K, Zhou A, Xu Z, Wu J, Peng X, Mei S, Chen H. Integrative Transcriptomics and Proteomics Analysis Reveals THRSP's Role in Lipid Metabolism. Genes (Basel) 2024; 15:1562. [PMID: 39766829 PMCID: PMC11675175 DOI: 10.3390/genes15121562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 11/27/2024] [Accepted: 11/28/2024] [Indexed: 01/11/2025] Open
Abstract
Background/Objectives: Abnormalities in lipid metabolism and endoplasmic reticulum (ER) stress are strongly associated with the development of a multitude of pathological conditions, including nonalcoholic fatty liver disease (NAFLD), diabetes mellitus, and obesity. Previous studies have indicated a potential connection between thyroid hormone responsive (THRSP) and lipid metabolism and that ER stress may participate in the synthesis of key regulators of adipogenesis. However, the specific mechanisms remain to be investigated. Methods: In this study, we explored the roles of THRSP in lipid metabolism by interfering with THRSP gene expression in mouse mesenchymal stem cells, comparing the effects on adipogenesis between control and interfered groups, and by combining transcriptomic and proteomic analysis. Results: Our results showed that the number of lipid droplets was significantly reduced after interfering with THRSP, and the expression levels of key regulators of adipogenesis, such as LPL, FABP4, PLIN1, and CIDEC, were significantly downregulated. Both transcriptomic and proteomic results showed that the differential genes (proteins) were enriched in the processes of lipolytic regulation, ER stress, cholesterol metabolism, sphingolipid metabolism, PPAR signaling pathway, and glycerophospholipid metabolism. The ER stress marker gene, ATF6, was the most significantly downregulated transcription factor. In addition, RT-qPCR validation indicated that the expression levels of PPAR signaling pathway gene SCD1; key genes of lipid droplet generation including LIPE, DGAT1, and AGPAT2; and ER stress marker gene ATF6 were significantly downregulated. Conclusions: These suggest that THRSP is involved in regulating ER stress and the PPAR signaling pathway, which is closely related to lipid synthesis and metabolism. Interfering with the expression of THRSP may be helpful in ameliorating the occurrence of diseases related to abnormalities in lipid metabolism.
Collapse
Affiliation(s)
- Yujie Li
- Laboratory of Genetic Breeding, Reproduction and Precision Livestock Farming, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan 430023, China; (Y.L.); (K.X.); (A.Z.)
- Hubei Provincial Center of Technology Innovation for Domestic Animal Breeding, Wuhan Polytechnic University, Wuhan 430023, China
- Key Laboratory of Animal Embryo Engineering and Molecular Breeding of Hubei Province, Wuhan 430064, China; (Z.X.); (J.W.); (X.P.)
| | - Ke Xu
- Laboratory of Genetic Breeding, Reproduction and Precision Livestock Farming, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan 430023, China; (Y.L.); (K.X.); (A.Z.)
- Hubei Provincial Center of Technology Innovation for Domestic Animal Breeding, Wuhan Polytechnic University, Wuhan 430023, China
| | - Ao Zhou
- Laboratory of Genetic Breeding, Reproduction and Precision Livestock Farming, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan 430023, China; (Y.L.); (K.X.); (A.Z.)
- Hubei Provincial Center of Technology Innovation for Domestic Animal Breeding, Wuhan Polytechnic University, Wuhan 430023, China
| | - Zhong Xu
- Key Laboratory of Animal Embryo Engineering and Molecular Breeding of Hubei Province, Wuhan 430064, China; (Z.X.); (J.W.); (X.P.)
| | - Junjing Wu
- Key Laboratory of Animal Embryo Engineering and Molecular Breeding of Hubei Province, Wuhan 430064, China; (Z.X.); (J.W.); (X.P.)
| | - Xianwen Peng
- Key Laboratory of Animal Embryo Engineering and Molecular Breeding of Hubei Province, Wuhan 430064, China; (Z.X.); (J.W.); (X.P.)
| | - Shuqi Mei
- Key Laboratory of Animal Embryo Engineering and Molecular Breeding of Hubei Province, Wuhan 430064, China; (Z.X.); (J.W.); (X.P.)
| | - Hongbo Chen
- Laboratory of Genetic Breeding, Reproduction and Precision Livestock Farming, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan 430023, China; (Y.L.); (K.X.); (A.Z.)
- Hubei Provincial Center of Technology Innovation for Domestic Animal Breeding, Wuhan Polytechnic University, Wuhan 430023, China
| |
Collapse
|
14
|
Li D, Han H, Sun Y, Zhang H, Yoshitomi R, Kaul SC, Wadhwa R. Molecular Insights into the Inhibition of Lipid Accumulation in Hepatocytes by Unique Extracts of Ashwagandha. Int J Mol Sci 2024; 25:12256. [PMID: 39596320 PMCID: PMC11594306 DOI: 10.3390/ijms252212256] [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: 09/23/2024] [Revised: 11/01/2024] [Accepted: 11/05/2024] [Indexed: 11/28/2024] Open
Abstract
We investigated the effect of purified withanolides and extracts derived from Ashwagandha on steatosis, the abnormal accumulation of fat that can lead to non-alcoholic fatty liver disease (NAFLD). Collaborator of ARF (CARF, also known as CDKN2AIP, a protein that regulates hepatic lipid metabolism, fat buildup, and liver damage) was used as an indicator. Six withanolides (Withaferin A, Withanone, Withanolide B, Withanoside IV, Withanoside V, and Withanostraminolide-12 deoxy) reversed the decrease in CARF caused by exposure to free fatty acids (FFAs) in liver-derived cells (HepG2 hepatocytes). After analyzing the effects of these withanolides on CARF mRNA and protein levels, FFA accumulation, protein aggregation, and oxidative and DNA damage stresses, we selected Withaferin A and Withanone for molecular analyses. Using the palmitic-acid-induced fatty acid accumulation stress model in Huh7 cells, we found a significant reduction in the activity of the key regulators of lipogenesis pathways, including sterol regulatory element-binding protein-1c (SREBP-1c), fatty acid synthase (FASN), and peroxisome proliferator-activated receptors (PPARγ and PPARα). This in vitro study suggests that low, non-toxic doses of Withaferin A, Withanone, or Ashwagandha extracts containing these withanolides possess anti-steatosis and antioxidative-stress properties. Further in vivo and clinical studies are required to investigate the therapeutic potential of these Ashwagandha-derived bioactive ingredients for NAFLD.
Collapse
Affiliation(s)
- Dongyang Li
- AIST-INDIA DAILAB, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba 305-8565, Japan (H.Z.); (S.C.K.)
| | - Hanlin Han
- AIST-INDIA DAILAB, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba 305-8565, Japan (H.Z.); (S.C.K.)
- Graduate School of Science and Technology, University of Tsukuba, 1-1-1 Tennodai, Tsukuba 305-8577, Japan
| | - Yixin Sun
- AIST-INDIA DAILAB, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba 305-8565, Japan (H.Z.); (S.C.K.)
- Graduate School of Science and Technology, University of Tsukuba, 1-1-1 Tennodai, Tsukuba 305-8577, Japan
| | - Huayue Zhang
- AIST-INDIA DAILAB, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba 305-8565, Japan (H.Z.); (S.C.K.)
- Graduate School of Science and Technology, University of Tsukuba, 1-1-1 Tennodai, Tsukuba 305-8577, Japan
| | - Ren Yoshitomi
- AIST-INDIA DAILAB, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba 305-8565, Japan (H.Z.); (S.C.K.)
| | - Sunil C. Kaul
- AIST-INDIA DAILAB, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba 305-8565, Japan (H.Z.); (S.C.K.)
| | - Renu Wadhwa
- AIST-INDIA DAILAB, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba 305-8565, Japan (H.Z.); (S.C.K.)
- Graduate School of Science and Technology, University of Tsukuba, 1-1-1 Tennodai, Tsukuba 305-8577, Japan
| |
Collapse
|
15
|
Tian Q, Wang M, Wang X, Lei Z, Ahmad O, Chen D, Zheng W, Shen P, Yang N. Identification of an alternative ligand-binding pocket in peroxisome proliferator-activated receptor gamma and its correlated selective agonist for promoting beige adipocyte differentiation. MedComm (Beijing) 2024; 5:e650. [PMID: 38988496 PMCID: PMC11233932 DOI: 10.1002/mco2.650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Revised: 06/09/2024] [Accepted: 06/12/2024] [Indexed: 07/12/2024] Open
Abstract
The pharmacological activation of peroxisome proliferator-activated receptor gamma (PPARγ) is a convenient and promising strategy for promoting beige adipocyte biogenesis to combat obesity-related metabolic disorders. However, thiazolidinediones (TZDs), the full agonists of PPARγ exhibit severe side effects in animal models and in clinical settings. Therefore, the development of efficient and safe PPARγ modulators for the treatment of metabolic diseases is emerging. In this study, using comprehensive methods, we report a previously unidentified ligand-binding pocket (LBP) in PPARγ and link it to beige adipocyte differentiation. Further virtual screening of 4097 natural compounds based on this novel LBP revealed that saikosaponin A (NJT-2), a terpenoid compound, can bind to PPARγ to induce coactivator recruitment and effectively activate PPARγ-mediated transcription of the beige adipocyte program. In a mouse model, NJT-2 administration efficiently promoted beige adipocyte biogenesis and improved obesity-associated metabolic dysfunction, with significantly fewer adverse effects than those observed with TZD. Our results not only provide an advanced molecular insight into the structural ligand-binding details in PPARγ, but also develop a linked selective and safe agonist for obesity treatment.
Collapse
Affiliation(s)
- Qiang Tian
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Urology The Affiliated Nanjing Drum Tower Hospital The Affiliated Hospital of Nanjing University Medical School School of Life Sciences Nanjing University Nanjing China
- Shenzhen Research Institute of Nanjing University Shenzhen China
| | - Miaohua Wang
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Urology The Affiliated Nanjing Drum Tower Hospital The Affiliated Hospital of Nanjing University Medical School School of Life Sciences Nanjing University Nanjing China
| | - Xueting Wang
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Urology The Affiliated Nanjing Drum Tower Hospital The Affiliated Hospital of Nanjing University Medical School School of Life Sciences Nanjing University Nanjing China
| | - Zhenli Lei
- School of Pharmaceutical Sciences Wenzhou Medical University Wenzhou China
| | - Owais Ahmad
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Urology The Affiliated Nanjing Drum Tower Hospital The Affiliated Hospital of Nanjing University Medical School School of Life Sciences Nanjing University Nanjing China
| | - Dianhua Chen
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Urology The Affiliated Nanjing Drum Tower Hospital The Affiliated Hospital of Nanjing University Medical School School of Life Sciences Nanjing University Nanjing China
| | - Wei Zheng
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Urology The Affiliated Nanjing Drum Tower Hospital The Affiliated Hospital of Nanjing University Medical School School of Life Sciences Nanjing University Nanjing China
| | - Pingping Shen
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Urology The Affiliated Nanjing Drum Tower Hospital The Affiliated Hospital of Nanjing University Medical School School of Life Sciences Nanjing University Nanjing China
- Shenzhen Research Institute of Nanjing University Shenzhen China
| | - Nanfei Yang
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Urology The Affiliated Nanjing Drum Tower Hospital The Affiliated Hospital of Nanjing University Medical School School of Life Sciences Nanjing University Nanjing China
- Shenzhen Research Institute of Nanjing University Shenzhen China
- School of Pharmaceutical Sciences Wenzhou Medical University Wenzhou China
| |
Collapse
|
16
|
Kuang DD, Li XY, Qian XP, Zhang T, Deng YY, Li QM, Luo JP, Zha XQ. Tea Polysaccharide Ameliorates High-Fat Diet-Induced Renal Tubular Ectopic Lipid Deposition via Regulating the Dynamic Balance of Lipogenesis and Lipolysis. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:12582-12595. [PMID: 38788215 DOI: 10.1021/acs.jafc.4c02606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/26/2024]
Abstract
Renal tubular ectopic lipid deposition (ELD) plays a significant role in the development of chronic kidney disease, posing a great threat to human health. The present work aimed to explore the intervention effect and potential molecular mechanism of a purified tea polysaccharide (TPS3A) on renal tubular ELD. The results demonstrated that TPS3A effectively improved kidney function and slowed the progression of tubulointerstitial fibrosis in high-fat-diet (HFD)-exposed ApoE-/- mice. Additionally, TPS3A notably suppressed lipogenesis and enhanced lipolysis, as shown by the downregulation of lipogenesis markers (SREBP-1 and FAS) and the upregulation of lipolysis markers (HSL and ATGL), thereby reducing renal tubular ELD in HFD-fed ApoE-/- mice and palmitic-acid-stimulated HK-2 cells. The AMPK-SIRT1-FoxO1 axis is a core signal pathway in regulating lipid deposition. Consistently, TPS3A significantly increased the levels of phosphorylated-AMPK, SIRT1, and deacetylation of Ac-FoxO1. However, these effects of TPS3A on lipogenesis and lipolysis were abolished by AMPK siRNA, SIRT1 siRNA, and FoxO1 inhibitor, resulting in exacerbated lipid deposition. Taken together, TPS3A shows promise in ameliorating renal tubular ELD by inhibiting lipogenesis and promoting lipolysis through the AMPK-SIRT1-FoxO1 signaling pathway.
Collapse
Affiliation(s)
- Dan-Dan Kuang
- Engineering Research Centre of Bioprocess of Ministry of Education, School of Food and Biological Engineering, Hefei University of Technology, No. 193 Tunxi Road, Hefei 230009, People's Republic of China
- School of Food and Biological Engineering, Hefei University of Technology, No. 193 Tunxi Road, Hefei 230009, People's Republic of China
| | - Xue-Ying Li
- Engineering Research Centre of Bioprocess of Ministry of Education, School of Food and Biological Engineering, Hefei University of Technology, No. 193 Tunxi Road, Hefei 230009, People's Republic of China
- School of Food and Biological Engineering, Hefei University of Technology, No. 193 Tunxi Road, Hefei 230009, People's Republic of China
| | - Xin-Ping Qian
- Engineering Research Centre of Bioprocess of Ministry of Education, School of Food and Biological Engineering, Hefei University of Technology, No. 193 Tunxi Road, Hefei 230009, People's Republic of China
- School of Food and Biological Engineering, Hefei University of Technology, No. 193 Tunxi Road, Hefei 230009, People's Republic of China
| | - Ting Zhang
- Engineering Research Centre of Bioprocess of Ministry of Education, School of Food and Biological Engineering, Hefei University of Technology, No. 193 Tunxi Road, Hefei 230009, People's Republic of China
- School of Food and Biological Engineering, Hefei University of Technology, No. 193 Tunxi Road, Hefei 230009, People's Republic of China
| | - Yuan-Yuan Deng
- Sericultural and Agri-Food Research Institute, Guangdong Academy of Agricultural Sciences, Guangzhou 510610, People's Republic of China
| | - Qiang-Ming Li
- Engineering Research Centre of Bioprocess of Ministry of Education, School of Food and Biological Engineering, Hefei University of Technology, No. 193 Tunxi Road, Hefei 230009, People's Republic of China
- School of Food and Biological Engineering, Hefei University of Technology, No. 193 Tunxi Road, Hefei 230009, People's Republic of China
| | - Jian-Ping Luo
- Engineering Research Centre of Bioprocess of Ministry of Education, School of Food and Biological Engineering, Hefei University of Technology, No. 193 Tunxi Road, Hefei 230009, People's Republic of China
- School of Food and Biological Engineering, Hefei University of Technology, No. 193 Tunxi Road, Hefei 230009, People's Republic of China
| | - Xue-Qiang Zha
- Engineering Research Centre of Bioprocess of Ministry of Education, School of Food and Biological Engineering, Hefei University of Technology, No. 193 Tunxi Road, Hefei 230009, People's Republic of China
- School of Food and Biological Engineering, Hefei University of Technology, No. 193 Tunxi Road, Hefei 230009, People's Republic of China
| |
Collapse
|
17
|
Martins BC, da Silva Ribeiro M, Teixeira AVS, Peixoto TC, Lisboa PC, Martins FF, Souza-Mello V, Daleprane JB. Consumption of interesterified palm oil leads inflammation of white adipose tissue and triggers metabolic disturbances in mice on a high-fat diet. Sci Rep 2024; 14:12530. [PMID: 38822155 PMCID: PMC11143230 DOI: 10.1038/s41598-024-63488-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 05/29/2024] [Indexed: 06/02/2024] Open
Abstract
Growing obesity is linked to shifts in dietary patterns, particularly the increased intake of ultra-processed high-fat foods. This study aimed to evaluate the effects of interesterified palm oil consumption on glucose homeostasis, adipose tissue remodeling, and hepatic lipogenesis in C57BL/6 mice fed a high-fat diet. Sixty C57BL/6 mice were divided into four groups (n = 15): the control group (C) fed a standard diet (4% soybean oil), the high-fat group (HF) (23.8% lard), the high palm oil fat group (HFP) (23.8% palm oil), and the high interesterified palm fat group (HFI) (23.8% interesterified palm oil) for 8 weeks (all groups received 50% energy from lipids). The HFI group exhibited higher body mass than the HF group (+ 11%, P < 0.05), which was attributed to an increased percentage of fat mass. Plasma concentrations of IL-6, insulin, and HOMA-IR were also elevated in the HFI group. Both the HFP and HFI groups showed hypertrophied adipocytes and pancreatic islets, increased alpha and beta cell masses, hepatic steatosis, low expression of genes related to beta-oxidation, and upregulated lipogenesis. In conclusion, the consumption of interesterified palm oil alters inflammatory and glucose profiles.
Collapse
Affiliation(s)
- Bruna Cadete Martins
- Laboratory for Studies of Interactions Between Nutrition and Genetics, LEING, Department of Basic and Experimental Nutrition, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Mayara da Silva Ribeiro
- Laboratory for Studies of Interactions Between Nutrition and Genetics, LEING, Department of Basic and Experimental Nutrition, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Ananda Vitoria Silva Teixeira
- Laboratory for Studies of Interactions Between Nutrition and Genetics, LEING, Department of Basic and Experimental Nutrition, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Thamara Cherem Peixoto
- Laboratory for Studies of Interactions Between Nutrition and Genetics, LEING, Department of Basic and Experimental Nutrition, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Patrícia Cristina Lisboa
- Laboratory of Endocrine Physiology, Department of Physiological Sciences, State University of Rio de Janeiro, Rio de Janeiro, RJ, 20551-030, Brazil
| | - Fabiane Ferreira Martins
- Laboratory for Studies of Interactions Between Nutrition and Genetics, LEING, Department of Basic and Experimental Nutrition, Rio de Janeiro State University, Rio de Janeiro, Brazil
- Department of Morphology, Federal University of Rio Grande Do Norte, Rio Grande do Norte, Brazil
| | - Vanessa Souza-Mello
- Laboratory of Morphometry, Metabolism and Cardiovascular Diseases, Biomedical Center, Institute of Biology, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Julio Beltrame Daleprane
- Laboratory for Studies of Interactions Between Nutrition and Genetics, LEING, Department of Basic and Experimental Nutrition, Rio de Janeiro State University, Rio de Janeiro, Brazil.
| |
Collapse
|
18
|
Barbalho SM, de Alvares Goulart R, Minniti G, Bechara MD, de Castro MVM, Dias JA, Laurindo LF. Unraveling the rationale and conducting a comprehensive assessment of KD025 (Belumosudil) as a candidate drug for inhibiting adipogenic differentiation-a systematic review. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:2681-2699. [PMID: 37966572 DOI: 10.1007/s00210-023-02834-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 11/02/2023] [Indexed: 11/16/2023]
Abstract
Rho-associated kinases (ROCKs) are crucial during the adipocyte differentiation process. KD025 (Belumosudil) is a newly developed inhibitor that selectively targets ROCK2. It has exhibited consistent efficacy in impeding adipogenesis across a spectrum of in vitro models of adipogenic differentiation. Given the novelty of this treatment, a comprehensive systematic review has not been conducted yet. This systematic review aims to fill this knowledge void by providing readers with an extensive examination of the rationale behind KD025 and its impacts on adipogenesis. Preclinical evidence was gathered owing to the absence of clinical trials. The Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines were followed, and the study's quality was assessed using the Joanna Briggs Institute (JBI) Checklist Critical Appraisal Tool for Systematic Reviews. In various in vitro models, such as 3T3-L1 cells, human orbital fibroblasts, and human adipose-derived stem cells, KD025 demonstrated potent anti-adipogenic actions. At a molecular level, KD025 had significant effects, including decreasing fibronectin (Fn) expression, inhibiting ROCK2 and CK2 activity, suppressing lipid droplet formation, and reducing the expression of proadipogenic genes peroxisome proliferator-activated receptor gamma (PPARγ) and CCAAT/enhancer-binding protein α (C/EBPα). Additionally, KD025 resulted in the suppression of fatty acid-binding protein 4 (FABP4 or AP2) expression, a decrease in sterol regulatory element binding protein 1c (SREBP-1c) and Glut-4 expression. Emphasis must be placed on the fact that while KD025 shows potential in preclinical studies and experimental models, extensive research is crucial to assess its efficacy, safety, and potential therapeutic applications thoroughly and directly in human subjects.
Collapse
Affiliation(s)
- Sandra Maria Barbalho
- Department of Biochemistry and Pharmacology, School of Medicine, Universidade de Marília (UNIMAR), Marília, São Paulo, 17525-902, Brazil
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, School of Medicine, Universidade de Marília (UNIMAR), Marília, São Paulo, 17525-902, Brazil
- Department of Biochemistry and Nutrition, School of Food and Technology of Marília (FATEC), Marília, São Paulo, 17500-000, Brazil
| | - Ricardo de Alvares Goulart
- Department of Biochemistry and Pharmacology, School of Medicine, Universidade de Marília (UNIMAR), Marília, São Paulo, 17525-902, Brazil
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, School of Medicine, Universidade de Marília (UNIMAR), Marília, São Paulo, 17525-902, Brazil
| | - Giulia Minniti
- Department of Biochemistry and Pharmacology, School of Medicine, Universidade de Marília (UNIMAR), Marília, São Paulo, 17525-902, Brazil
| | - Marcelo Dib Bechara
- Department of Biochemistry and Pharmacology, School of Medicine, Universidade de Marília (UNIMAR), Marília, São Paulo, 17525-902, Brazil
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, School of Medicine, Universidade de Marília (UNIMAR), Marília, São Paulo, 17525-902, Brazil
| | - Marcela Vialogo Marques de Castro
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, School of Medicine, Universidade de Marília (UNIMAR), Marília, São Paulo, 17525-902, Brazil
| | - Jefferson Aparecido Dias
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, School of Medicine, Universidade de Marília (UNIMAR), Marília, São Paulo, 17525-902, Brazil
| | - Lucas Fornari Laurindo
- Department of Biochemistry and Pharmacology, School of Medicine, Universidade de Marília (UNIMAR), Marília, São Paulo, 17525-902, Brazil.
- Department of Biochemistry and Pharmacology, School of Medicine, Faculdade de Medicina de Marília (FAMEMA), Marília, São Paulo, 17519-030, Brazil.
| |
Collapse
|
19
|
Yu C, Wang D, Shen C, Luo Z, Zhang H, Zhang J, Xu W, Xu J. Microbe-derived Antioxidants Enhance Lipid Synthesis by Regulating the Hepatic AMPKα-SREBP1c Pathway in Weanling Piglets. J Nutr 2024; 154:1101-1108. [PMID: 38340959 DOI: 10.1016/j.tjnut.2024.02.002] [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: 11/19/2023] [Revised: 02/01/2024] [Accepted: 02/01/2024] [Indexed: 02/12/2024] Open
Abstract
BACKGROUND Weaning usually causes low feed intake and weight loss in piglets, which mobilizes lipid to energize. The microbe-derived antioxidants (MAs) exhibit great potential in antioxidation, anti-inflammation, and metabolic regulation. OBJECTIVES We aimed to investigate the changes of lipid metabolism postweaning and effects of MA on growth performance and hepatic lipid metabolism in weanling piglets. METHODS In the first experiment, piglets weaned at 21 d of age were slaughtered on weaning day (d0), 4 (d4), and 14 (d14) postweaning (6 piglets per day). In the second experiment, piglets were divided into 2 groups, receiving MA (MA) and saline gavage (CON), respectively. All piglets were weaned at 21 d of age and 6 piglets from each group were slaughtered at 25 d of age. RESULTS In experiment 1, the serum triglyceride, total cholesterol (TC), and LDL cholesterol on d4 and d14 declined significantly compared with d0 (P < 0.05). The serum leptin on d0 was higher than that on d4 and d14 (P < 0.05). The serum ghrelin kept increasing from d0 to d14 (P < 0.05). The hepatic hormone-sensitive lipase and adipose triglyceride lipase first increased from d0 to d4 and then decreased from d4 to d14 (P < 0.05). In experiment 2, the average daily gain and average daily feed intake from 21 to 25 d of age increased in the MA group compared with the CON group (P < 0.05). The serum TC, hepatic TC, and glucose of MA group showed a significant increase than that of the CON group (P < 0.05). The expression of SCD1, ACAT2, and PPARγ were upregulated in the MA group (P < 0.05). Contrary to the decreased expression of phosphorylation of adenosine 5'-monophosphate-activated protein kinase alfa subunit (Thr172), the nuclear sterol regulatory element-binding protein 1c, fatty acid synthase, and peroxisome proliferator-activated receptor gamma of MA group increased than that of CON group (P < 0.05). CONCLUSIONS Weaning promoted hepatic lipolysis and MA could enhance lipid synthesis by regulating adenosine 5'-monophosphate-activated protein kinase alfa subunit-sterol regulatory element-binding protein 1c pathway, thus improving growth performance of weanling piglets.
Collapse
Affiliation(s)
- Chengbing Yu
- Shanghai Key Laboratory of Veterinary and Biotechnology, Department of Animal Science, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, China
| | - Di Wang
- Shanghai Key Laboratory of Veterinary and Biotechnology, Department of Animal Science, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, China
| | - Cheng Shen
- Shanghai Key Laboratory of Veterinary and Biotechnology, Department of Animal Science, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, China
| | - Zhen Luo
- Shanghai Key Laboratory of Veterinary and Biotechnology, Department of Animal Science, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, China
| | - Hongcai Zhang
- Shanghai Key Laboratory of Veterinary and Biotechnology, Department of Animal Science, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, China
| | - Jing Zhang
- Shanghai Key Laboratory of Veterinary and Biotechnology, Department of Animal Science, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, China
| | - Weina Xu
- Shanghai Key Laboratory of Veterinary and Biotechnology, Department of Animal Science, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, China
| | - Jianxiong Xu
- Shanghai Key Laboratory of Veterinary and Biotechnology, Department of Animal Science, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|