1
|
Zheng J, Zhang Y, Chen Y, Tian L. ChREBP drives fibroblast proliferation and promotes pulmonary fibrosis development. Cytokine 2025; 190:156906. [PMID: 40117836 DOI: 10.1016/j.cyto.2025.156906] [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: 10/09/2024] [Revised: 01/28/2025] [Accepted: 02/27/2025] [Indexed: 03/23/2025]
Abstract
OBJECTIVE The study aimed to investigate the role of carbohydrate-responsive element-binding protein (ChREBP) in the pathogenesis of pulmonary fibrosis (PF) by assessing its impact on fibrotic protein expression, fibroblast proliferation, and apoptosis in lung tissues. METHOD The PF model was established using bleomycin, and pathological changes in lung tissues were assessed through histopathological analysis. Expression levels of inflammatory markers and fibrotic proteins, including ChREBP, were measured using Western blot and ELISA. Additionally, human embryonic lung fibroblasts (MRC-5) were transfected with ChREBP overexpression or silencing vectors following TGF-β1 induction to examine changes in cellular behavior, including viability, apoptosis, and fibrotic protein expression. RESULTS The PF model group showed significant alveolar structural abnormalities and elevated levels of TNF-α, MMP-7 and TGF-β1. ChREBP expression was markedly increased in fibrotic tissues (P < 0.05). In vitro, ChREBP overexpression in MRC-5 cells enhanced fibrotic protein levels, increased cell viability, and reduced apoptosis rates. Conversely, silencing ChREBP reduced fibrotic protein expression, inhibited fibroblast proliferation, and increased apoptosis (P < 0.05). These findings suggest that ChREBP plays a key role in modulating fibrosis-related pathways in PF. CONCLUSIONS ChREBP is substantially upregulated in PF and plays a key role in promoting fibroblast proliferation and inhibiting apoptosis. These findings suggest that targeting ChREBP may present a novel therapeutic strategy for treating pulmonary fibrosis by modulating fibrotic and apoptotic pathways.
Collapse
Affiliation(s)
- Jian Zheng
- Department of Pulmonary and Critical Care Medicine, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, 250000, China
| | - Yang Zhang
- Department of Pulmonary and Critical Care Medicine, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, 250000, China
| | - Yan Chen
- Integrated Traditional Chinese and Western Medicine Ward, Shengli Oilfield Central Hospital, Dongying, Shandong 257000, China
| | - Li Tian
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, 250000, China; Department of Pulmonary and Critical Care Medicine, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, 250000, China.
| |
Collapse
|
2
|
Petricek KM, Kirchner M, Sommerfeld M, Stephanowitz H, Kiefer MF, Meng Y, Dittrich S, Dähnhardt HE, Mai K, Krause E, Mertins P, Wowro SJ, Schupp M. An acetylated Lysine Residue of Its Low-glucose Inhibitory Domain Controls Activity and Protein Interactions of ChREBP. J Mol Biol 2025; 437:169189. [PMID: 40339981 DOI: 10.1016/j.jmb.2025.169189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Revised: 04/17/2025] [Accepted: 05/01/2025] [Indexed: 05/10/2025]
Abstract
Carbohydrate response element-binding protein (ChREBP) is a transcription factor activated by glucose metabolites that orchestrates the expression of genes involved in glycolysis, de novo lipogenesis, and ATP homeostasis. Inadequate ChREBP activity impairs the cellular adaptations to glucose exposure and in humans associates with dyslipidemia, fatty liver disease, and type 2 diabetes. ChREBP activity is regulated by cytosolic-nuclear translocation involving its low-glucose inhibitory domain (LID). Whether this domain is targeted by post-translational lysine acetylation is unknown. Here we report a novel LID acetylation site that controls activity and protein interactions of ChREBP. Mutation of this residue increased glucose-induced activity and target gene expression of ChREBP. Mechanistically, mutant ChREBP protein showed more nuclear localization and enhanced genomic binding to a target promoter. Interactions with proteins that exhibit differential binding upon glucose exposure were attenuated by the mutation, demonstrating the importance of the LID in the formation of the protein interactome. Particularly interactions with 14-3-3 proteins, factors that regulate cytosolic/nuclear trafficking of ChREBP, were reduced, whereas interactions with proteins of the nucleosome remodeling deacetylase complex (NuRD) were increased. These molecular insights may shape new therapeutic strategies to target ChREBP activity and counteract metabolic diseases.
Collapse
Affiliation(s)
- Konstantin M Petricek
- Institute of Pharmacology, Max Rubner Center (MRC) for Cardiovascular Metabolic Renal Research, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany; Department of Endocrinology and Metabolism, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Zu Berlin, and Berlin Institute of Health, European Reference Network on Rare Endocrine Diseases (ENDO-ERN), Berlin, Germany
| | - Marieluise Kirchner
- Core Unit Proteomics, Berlin Institute of Health at Charité - Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Manuela Sommerfeld
- Institute of Pharmacology, Max Rubner Center (MRC) for Cardiovascular Metabolic Renal Research, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Heike Stephanowitz
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Berlin, Germany
| | - Marie F Kiefer
- Institute of Pharmacology, Max Rubner Center (MRC) for Cardiovascular Metabolic Renal Research, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Yueming Meng
- Institute of Pharmacology, Max Rubner Center (MRC) for Cardiovascular Metabolic Renal Research, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Sarah Dittrich
- Institute of Pharmacology, Max Rubner Center (MRC) for Cardiovascular Metabolic Renal Research, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Henriette E Dähnhardt
- Institute of Pharmacology, Max Rubner Center (MRC) for Cardiovascular Metabolic Renal Research, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Knut Mai
- Department of Endocrinology and Metabolism, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Zu Berlin, and Berlin Institute of Health, European Reference Network on Rare Endocrine Diseases (ENDO-ERN), Berlin, Germany; German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany; Department of Human Nutrition, German Institute of Human Nutrition Potsdam-Rehbrücke, Nuthetal, Germany
| | - Eberhard Krause
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Berlin, Germany
| | - Philipp Mertins
- Core Unit Proteomics, Berlin Institute of Health at Charité - Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Sylvia J Wowro
- Institute of Pharmacology, Max Rubner Center (MRC) for Cardiovascular Metabolic Renal Research, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Michael Schupp
- Institute of Pharmacology, Max Rubner Center (MRC) for Cardiovascular Metabolic Renal Research, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany.
| |
Collapse
|
3
|
Flores Monar GV, Sanchez Cruz C, Calderon Martinez E. Mindful Eating: A Deep Insight Into Fructose Metabolism and Its Effects on Appetite Regulation and Brain Function. J Nutr Metab 2025; 2025:5571686. [PMID: 40297675 PMCID: PMC12037248 DOI: 10.1155/jnme/5571686] [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: 09/30/2024] [Accepted: 03/25/2025] [Indexed: 04/30/2025] Open
Abstract
Fructose, a common sweetener in modern diets, has profound effects on both metabolism and brain function, primarily due to its distinct metabolic pathways. Unlike glucose, fructose bypasses critical regulatory steps in metabolism, particularly the phosphofructokinase-1 (PFK-1) feedback inhibition, leading to uncontrolled metabolism and increased fat storage. This review delves into the metabolic consequences of fructose consumption, including its limited role in directly stimulating insulin secretion, which affects satiety signaling and contributes to increased food intake. The small intestine initially helps metabolize ingested fructose, shielding the liver and brain from excessive exposure. However, when consumed in excess, particularly in diets high in processed foods, this protective mechanism becomes overwhelmed, contributing to metabolic disorders such as insulin resistance, obesity, and fatty liver disease. The review also explores fructose's impact on the brain, with a focus on the hippocampus, a key region for memory and learning. Chronic high fructose intake has been linked to mitochondrial dysfunction, increased production of reactive oxygen species (ROS), and neuroinflammation, all of which contribute to cognitive decline and impairments in memory and learning. Additionally, fructose-induced alterations in insulin signaling in the brain are associated with increased risk for neurodegenerative diseases. These findings underscore the potential long-term neurological consequences of excessive fructose intake and highlight the need for further human studies to assess the full spectrum of its effects on brain health. Addressing the rising consumption of fructose, particularly in processed foods, is essential for developing targeted strategies to mitigate its adverse metabolic and cognitive outcomes.
Collapse
Affiliation(s)
| | - Camila Sanchez Cruz
- Faculty of Medicine, Universidad Nacional Autónoma de México, Coyoacán, Ciudad de México, Mexico
| | | |
Collapse
|
4
|
Wang CM, Bai QF, Liu YJ, Lin J, Wei CC, Ma XH, Zhao JM, Zhu M, Chen YX, Shi YN, Shi JH, Zhang WJ. ChREBP mediates metabolic remodeling in FBP1-deficient liver. Am J Physiol Cell Physiol 2025; 328:C1234-C1246. [PMID: 40055186 DOI: 10.1152/ajpcell.00875.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 11/30/2024] [Accepted: 03/03/2025] [Indexed: 04/01/2025]
Abstract
The deficiency of fructose-1,6-bisphosphatase 1 (FBP1), a key enzyme of gluconeogenesis, causes fatty liver. However, its underlying mechanism and physiological significance are not fully understood. Here we demonstrate that carbohydrate response element-binding protein (ChREBP) mediates lipid metabolic remodeling and promotes progressive triglyceride accumulation against metabolic injury in adult FBP1-deficient liver. Inducible liver-specific deletion of Fbp1 gene caused progressive hepatomegaly and hepatic steatosis, with a marked increase in hepatic de novo lipogenesis (DNL) as well as a decrease in plasma β-hydroxybutyrate levels. Notably, FBP1 deficiency resulted in a persistent activation of ChREBP and its target genes involved in glycolysis, lipogenesis, and fatty acid oxidation, even under fasting conditions. Furthermore, liver-specific ChREBP disruption could markedly restore the phenotypes of enhanced DNL and triglyceride accumulation in FBP1-deficient liver but exacerbated its hepatomegaly and liver injury, which was associated with remarkable energy deficit, impaired mammalian target of rapamycin (mTOR) activation, and increased oxidative stress. Furthermore, metabolomics analysis revealed a robust elevation of phosphoenolpyruvate, phosphoglycerates, phospholipids, and ceramides caused by ChREBP deletion in FBP1-deficient liver. Put together, these results suggest that overactivation of ChREBP pathway mediates liver metabolic remodeling in the absence of FBP1, which contributes to the pathogenesis of progressive hepatic steatosis and provides a protection against liver injury. Thus, our findings point to a beneficial role of ChREBP in metabolic remodeling in the context of excessive gluconeogenic intermediates.NEW & NOTEWORTHY FBP1 deficiency in adulthood causes progressive hepatic steatosis due to the overactivation of ChREBP pathway, which enhances lipid synthesis and inhibits fat oxidation. ChREBP-mediated metabolic remodeling protects against liver injury caused by energy deficit and oxidative stress in FBP1-deficient liver.
Collapse
Affiliation(s)
- Chen-Ma Wang
- NHC Key Lab of Hormones and Development and Tianjin Key Lab of Metabolic Diseases, Tianjin Medical University Chu Hsien-I Memorial Hospital and Institute of Endocrinology, Tianjin, People's Republic of China
| | - Qiu-Fang Bai
- Department of Endocrinology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, People's Republic of China
| | - Ya-Jin Liu
- NHC Key Lab of Hormones and Development and Tianjin Key Lab of Metabolic Diseases, Tianjin Medical University Chu Hsien-I Memorial Hospital and Institute of Endocrinology, Tianjin, People's Republic of China
| | - Jie Lin
- National Key Laboratory of Immunity and Inflammation, Department of Pathophysiology, Naval Medical University, Shanghai, People's Republic of China
| | - Chun-Chun Wei
- National Key Laboratory of Immunity and Inflammation, Department of Pathophysiology, Naval Medical University, Shanghai, People's Republic of China
| | - Xian-Hua Ma
- National Key Laboratory of Immunity and Inflammation, Department of Pathophysiology, Naval Medical University, Shanghai, People's Republic of China
| | - Jia-Mu Zhao
- NHC Key Lab of Hormones and Development and Tianjin Key Lab of Metabolic Diseases, Tianjin Medical University Chu Hsien-I Memorial Hospital and Institute of Endocrinology, Tianjin, People's Republic of China
| | - Meng Zhu
- NHC Key Lab of Hormones and Development and Tianjin Key Lab of Metabolic Diseases, Tianjin Medical University Chu Hsien-I Memorial Hospital and Institute of Endocrinology, Tianjin, People's Republic of China
| | - Yu-Xia Chen
- National Key Laboratory of Immunity and Inflammation, Department of Pathophysiology, Naval Medical University, Shanghai, People's Republic of China
| | - Ya-Nan Shi
- NHC Key Lab of Hormones and Development and Tianjin Key Lab of Metabolic Diseases, Tianjin Medical University Chu Hsien-I Memorial Hospital and Institute of Endocrinology, Tianjin, People's Republic of China
| | - Jian-Hui Shi
- National Key Laboratory of Immunity and Inflammation, Department of Pathophysiology, Naval Medical University, Shanghai, People's Republic of China
| | - Weiping J Zhang
- NHC Key Lab of Hormones and Development and Tianjin Key Lab of Metabolic Diseases, Tianjin Medical University Chu Hsien-I Memorial Hospital and Institute of Endocrinology, Tianjin, People's Republic of China
- National Key Laboratory of Immunity and Inflammation, Department of Pathophysiology, Naval Medical University, Shanghai, People's Republic of China
| |
Collapse
|
5
|
Baya NA, Erdem IS, Venkatesh SS, Reibe S, Charles PD, Navarro-Guerrero E, Hill B, Lassen FH, Claussnitzer M, Palmer DS, Lindgren CM. Combining evidence from human genetic and functional screens to identify pathways altering obesity and fat distribution. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2024.09.19.24313913. [PMID: 39371160 PMCID: PMC11451655 DOI: 10.1101/2024.09.19.24313913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/08/2024]
Abstract
Overall adiposity and body fat distribution are heritable traits associated with altered risk of cardiometabolic disease and mortality. Performing rare variant (minor allele frequency<1%) association testing using exome-sequencing data from 402,375 participants in the UK Biobank (UKB) for nine overall and tissue-specific fat distribution traits, we identified 19 genes where putatively damaging rare variation associated with at least one trait (Bonferroni-adjusted P <1.58×10 -7 ) and 50 additional genes at FDR≤1% ( P ≤4.37×10 -5 ). These 69 genes exhibited significantly higher (one-sided t -test P =3.58×10 -18 ) common variant prioritisation scores than genes not significantly enriched for rare putatively damaging variation, with evidence of monotonic allelic series (dose-response relationships) among ultra-rare variants (minor allele count≤10) in 22 genes. Combining rare and common variation evidence, allelic series and longitudinal analysis, we selected 14 genes for CRISPR knockdown in human white adipose tissue cell lines. In three previously uncharacterised target genes, knockdown increased (two-sided t -test P <0.05) lipid accumulation, a cellular phenotype relevant for fat mass traits, compared to Cas9-empty negative controls: COL5A3 (fold change [FC]=1.72, P =0.0028), EXOC7 (FC=1.35, P =0.0096), and TRIP10 (FC=1.39, P =0.0157); furthermore, knockdown of PPARG (FC=0.25, P =5.52×10 -7 ) and SLTM (FC=0.51, P =1.91×10 -4 ) resulted in reduced lipid accumulation. Integrating across population-based genetic and in vitro functional evidence, we highlight therapeutic avenues for altering obesity and body fat distribution by modulating lipid accumulation.
Collapse
|
6
|
Karim M, Prey J, Willer F, Leiner H, Yasser M, Dombrowski F, Ribback S. Hepatic Deletion of Carbohydrate Response Element Binding Protein Impairs Hepatocarcinogenesis in a High-Fat Diet-Induced Mouse Model. Int J Mol Sci 2025; 26:2246. [PMID: 40076869 PMCID: PMC11900174 DOI: 10.3390/ijms26052246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 02/06/2025] [Accepted: 02/07/2025] [Indexed: 03/14/2025] Open
Abstract
The transcription factor carbohydrate response element binding protein (ChREBP) has emerged as a crucial regulator of hepatic glucose and lipid metabolism. The increased ChREBP activity involves the pro-oncogenic PI3K/AKT/mTOR signaling pathway that induces aberrant lipogenesis, thereby promoting hepatocellular carcinomas (HCC). However, the molecular pathogenesis of ChREBP-related hepatocarcinogenesis remains unexplored in the high-fat diet (HFD)-induced mouse model. Male C57BL/6J (WT) and liver-specific (L)-ChREBP-KO mice were maintained on either a HFD or a control diet for 12, 24, and 48 weeks, starting at the age of 4 weeks. At the end of the feeding period, mice were perfused, and liver tissues were formalin-fixed, paraffin-embedded, sectioned, and stained for histological and immunohistochemical analysis. Biochemical and gene expression analysis were conducted using serum and frozen liver tissue. Mice fed with HFD showed a significant increase (p < 0.05) in body weight from 8 weeks onwards compared to the control. WT and L-ChREBP-KO mice also demonstrated a significant increase (p < 0.05) in liver-to-body weight ratio in the 48-week HFD group. HFD mice exhibited a gradual rise in hepatic lipid accumulation over time, with 24-week mice showing a 20-30% increase in fat content, which further advanced to 80-100% fat accumulation at 48 weeks. Both dietary source and the increased expression of lipogenic pathways at transcriptional and protein levels induced steatosis and steatohepatitis in the HFD group. Moreover, WT mice on a HFD exhibited markedly higher inflammation compared to the L-ChREBP-KO mice. The enhanced lipogenesis, glycolysis, persistent inflammation, and activation of the AKT/mTOR pathway collectively resulted in significant metabolic disturbances, thereby promoting HCC development and progression in WT mice. In contrast, hepatic loss of ChREBP resulted in reduced hepatocyte proliferation in the HFD group, which significantly contributed to the impaired hepatocarcinogenesis and a reduced HCC occurrence in the L-ChREBP-KO mice. Our present study implicates that prolonged HFD feeding contributes to NAFLD/NASH, which in turn progresses to HCC development in WT mice. Collectively, hepatic ChREBP deletion ameliorates hepatic inflammation and metabolic alterations, thereby impairing NASH-driven hepatocarcinogenesis.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Silvia Ribback
- Institute of Pathology, University Medicine Greifswald, Friedrich-Loeffler-Str. 23e, 17475 Greifswald, Germany; (M.K.); (J.P.); (F.W.); (M.Y.); (F.D.)
| |
Collapse
|
7
|
Ghosh C, Kundu T, Pathak T, Saini S, Das N, Saini S, Sircar D, Kumar P, Roy P. Indian lychee honey ameliorates hepatic glucose uptake by regulating the ChREBP/Glut4 axis under insulin-resistant conditions. Food Funct 2025; 16:2031-2056. [PMID: 39963045 DOI: 10.1039/d4fo03900a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/04/2025]
Abstract
Many traditional treatments include honey owing to its magnificent health beneficiary effects. Recent studies have demonstrated the potent anti-diabetic activity of honey. However, its actual mechanism of action remains elusive. Moreover, being rich in sugar (75%-80%), its role in maintaining glucose homeostasis remains questionable. Although the polyphenol content of honey aids its hypoglycaemic activity, the small quantity of bioactive compounds in honey (0.5%-1.0%) may not be solely responsible for this. In the current study, an attempt was made to understand the role of Indian lychee honey (LyH) in regulating blood glucose levels under diabetic conditions. This study investigated whether LyH, although rich in sugars, can be used as an alternative to regulate glucose and lipid homeostasis under insulin-resistant conditions by regulating the ChREBP/Glut4 signalling pathway. This study was first performed in vitro in palmitic acid-induced insulin-resistant HepG2 cells. Various assays, such as FACS, GCMS, qRT-PCR, immunoblot and ChIP-qPCR, were performed to establish the anti-hyperglycaemic role of LyH in vitro. The in vitro results were subsequently confirmed in vivo using a high-fat diet-induced diabetic C57BL/6 mice model. The in vivo study was supported by several experiments, such as examining blood parameters, histopathology, double-immunohistochemistry and ELISA. Finally, the finding was validated by comparing it with a couple of GEO datasets from the NCBI database. This study found that LyH is an excellent choice for regulating blood sugar levels under diabetic conditions without significant harmful side effects. Moreover, LyH showed excellent hepatic glucose uptake activity in an insulin-independent manner. This activity is mainly governed by sugars as its main ingredient. LyH treatment also regulates hepatic lipid homeostasis by maintaining a balance between saturated and unsaturated fatty acids in insulin-resistant HepG2 cells. Further, sugar, when supplemented individually, caused severe inflammation, which was validated through histopathology, ELISA and IHC. Collectively, the findings of this study indicate that Indian LyH provides a better food matrix (the right proportion of sugars and different bioactive compounds), which significantly improves hyperglycemia and inflammation under diabetic conditions by regulating the hepatic ChREBP/Glut4 axis.
Collapse
Affiliation(s)
- Chandrachur Ghosh
- Molecular Endocrinology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee 247 667, Uttarakhand, India.
| | - Tathagata Kundu
- Molecular Endocrinology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee 247 667, Uttarakhand, India.
| | - Tiyasa Pathak
- Molecular Endocrinology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee 247 667, Uttarakhand, India.
| | - Saakshi Saini
- Molecular Endocrinology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee 247 667, Uttarakhand, India.
| | - Neeladrisingha Das
- Molecular Endocrinology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee 247 667, Uttarakhand, India.
| | - Surendra Saini
- Molecular Endocrinology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee 247 667, Uttarakhand, India.
| | - Debabrata Sircar
- Plant Molecular Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee 247 667, Uttarakhand, India
| | - Prabhat Kumar
- National Bee Board, DA & FW, Ministry of Agriculture and Farmers Welfare, B Wing, 2nd Floor, Janpath Bhawan, Janpath, New Delhi - 110 001, India
| | - Partha Roy
- Molecular Endocrinology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee 247 667, Uttarakhand, India.
- Center for Indian Knowledge Systems, Indian Institute of Technology Roorkee, Roorkee 247 667, Uttarakhand, India
| |
Collapse
|
8
|
Devnath HS, Medha MM, Islam MN, Biswas P, Oisay DS, Hossain A, Ema RS, Tareq MMI, Golder M, Hasan MN, Biswas B, Sadhu SK. Bioactive small compounds effectively inhibit ChREBP overexpression to treat NAFLD and T2DM: A computational drug development approach. Heliyon 2025; 11:e42477. [PMID: 40034298 PMCID: PMC11872590 DOI: 10.1016/j.heliyon.2025.e42477] [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: 09/03/2024] [Revised: 01/29/2025] [Accepted: 02/04/2025] [Indexed: 03/05/2025] Open
Abstract
A glucose-dependent carbohydrate-signaling gene regulator named Carbohydrate response element binding protein (ChREBP), has recently been discovered as a major metabolic regulator of enzymes involved in the progression of non-alcoholic fatty liver disease (NAFLD) and type-II diabetes mellitus (T2DM). As a result, this research is aimed to identify natural small molecules as drug candidates that target the ChREBP in order to counter aggressive NAFLD and T2DM. A comprehensive in silico drug design strategy was implemented to find possible inhibitors of the targeted protein. A site-specific molecular docking approach was used to screen 20 FDA approved anti-diabetic drugs and 494 phytochemicals from the natural sources against the ChREBP, and the top ten compounds were selected for further studies based on their binding affinities. The ADME and toxicity profiles of the selected ten drug compounds demonstrated their efficacy and safety. The result of the MD simulations of the protein-ligand complex structures indicated their stability and potential activity. A comprehensive data screening process following docking, ADMET properties, and MD simulation approaches, five compounds (dieckol, isocorilagin, stachyurin, stachysetin and thonningianin A) with favorable values against the targeted ChREBP were demonstrated which indicates their strong potential as promising and effective drug candidates for the treatment of NAFLD and T2DM.
Collapse
Affiliation(s)
| | | | - Md Naharul Islam
- Pharmacy Discipline, Khulna University, Khulna, 9208, Bangladesh
| | - Partha Biswas
- Laboratory of Pharmaceutical Biotechnology and Bioinformatics, Department of Genetic Engineering and Biotechnology, Jashore University of Science and Technology, Jashore, 7408, Bangladesh
| | | | - Arafat Hossain
- Biochemistry and Molecular Biology Department, Life Science Faculty, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalgonj, 8100, Bangladesh
| | | | - Md Mohaimenul Islam Tareq
- Laboratory of Pharmaceutical Biotechnology and Bioinformatics, Department of Genetic Engineering and Biotechnology, Jashore University of Science and Technology, Jashore, 7408, Bangladesh
| | - Mimi Golder
- Pharmacy Discipline, Khulna University, Khulna, 9208, Bangladesh
| | - Md Nazmul Hasan
- Laboratory of Pharmaceutical Biotechnology and Bioinformatics, Department of Genetic Engineering and Biotechnology, Jashore University of Science and Technology, Jashore, 7408, Bangladesh
| | - Biswajit Biswas
- Department of Pharmacy, Faculty of Biological Science and Technology, Jashore University of Science and Technology, Jashore, 7408, Bangladesh
| | | |
Collapse
|
9
|
An J, Astapova I, Zhang G, Cangelosi AL, Ilkayeva O, Marchuk H, Muehlbauer MJ, George T, Brozinick J, Herman MA, Newgard CB. Integration of metabolomic and transcriptomic analyses reveals regulatory functions of the ChREBP transcription factor in energy metabolism. Cell Rep 2025; 44:115278. [PMID: 39921857 DOI: 10.1016/j.celrep.2025.115278] [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: 09/24/2024] [Revised: 12/03/2024] [Accepted: 01/16/2025] [Indexed: 02/10/2025] Open
Abstract
The transcription factor carbohydrate response element binding protein (ChREBP) activates genes of glucose, fructose, and lipid metabolism in response to carbohydrate feeding. Integrated transcriptomic and metabolomic analyses in rats with GalNac-siRNA-mediated suppression of ChREBP expression in liver reveal other ChREBP functions. GalNac-siChREBP treatment reduces expression of genes involved in coenzyme A (CoA) biosynthesis, with lowering of CoA and short-chain acyl-CoA levels. Despite suppression of pyruvate kinase, pyruvate levels are maintained, possibly via increased expression of pyruvate and amino acid transporters. In addition, expression of multiple anaplerotic enzymes is decreased by GalNac-siChREBP treatment, affecting TCA cycle intermediates. Finally, GalNAc-siChREBP treatment suppresses late steps in purine and NAD synthesis, with increases in precursors and lowering of end products in both pathways. In sum, our study reveals functions of ChREBP beyond its canonical roles in carbohydrate and lipid metabolism to include regulation of substrate transport, mitochondrial function, and energy balance.
Collapse
Affiliation(s)
- Jie An
- Sarah W. Stedman Nutrition and Metabolism Center & Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC, USA
| | - Inna Astapova
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Guofang Zhang
- Sarah W. Stedman Nutrition and Metabolism Center & Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC, USA; Division of Endocrinology, Metabolism and Nutrition, Department of Medicine, Duke University Medical Center, Durham, NC, USA
| | - Andrew L Cangelosi
- Sarah W. Stedman Nutrition and Metabolism Center & Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC, USA
| | - Olga Ilkayeva
- Sarah W. Stedman Nutrition and Metabolism Center & Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC, USA; Division of Endocrinology, Metabolism and Nutrition, Department of Medicine, Duke University Medical Center, Durham, NC, USA
| | - Hannah Marchuk
- Sarah W. Stedman Nutrition and Metabolism Center & Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC, USA
| | - Michael J Muehlbauer
- Sarah W. Stedman Nutrition and Metabolism Center & Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC, USA
| | - Tabitha George
- Sarah W. Stedman Nutrition and Metabolism Center & Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC, USA
| | | | - Mark A Herman
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Christopher B Newgard
- Sarah W. Stedman Nutrition and Metabolism Center & Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC, USA; Division of Endocrinology, Metabolism and Nutrition, Department of Medicine, Duke University Medical Center, Durham, NC, USA; Department of Pharmacology & Cancer Biology, Duke University Medical Center, Durham, NC, USA.
| |
Collapse
|
10
|
Deguchi K, Ushiroda C, Hidaka S, Tsuchida H, Yamamoto-Wada R, Seino Y, Suzuki A, Yabe D, Iizuka K. Chrebp Deletion and Mild Protein Restriction Additively Decrease Muscle and Bone Mass and Function. Nutrients 2025; 17:488. [PMID: 39940346 PMCID: PMC11819777 DOI: 10.3390/nu17030488] [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: 01/09/2025] [Revised: 01/19/2025] [Accepted: 01/22/2025] [Indexed: 02/16/2025] Open
Abstract
Background/Objectives: Carbohydrate and protein restriction are associated with sarcopenia and osteopenia, but the underlying mechanisms remain unclear. We aimed to determine whether mild protein restriction affects muscle and bone function in wild-type (WT) and homozygous carbohydrate response element binding protein (Chrebp) knockout (KO) mice. Methods: Eighteen-week-old male wild-type and homozygous carbohydrate response element binding protein (Chrebp) knockout (KO) mice were fed a control diet (20% protein) or a low-protein diet (15% protein) for 12 weeks. We estimated the muscle weight and limb grip strength as well as the bone mineral density, bone structure, and bone morphometry. Results: Chrebp deletion and a low-protein diet additively decreased body weight (WT control-KO low-protein: mean difference with 95% CI, 8.7 [6.3, 11.0], p < 0.0001) and epidydimal fat weight (1.0 [0.7, 1.2], p < 0.0001). Chrebp deletion and a low-protein diet additively decreased tibialis anterior muscle weight (0.03 [0.01, 0.05], p = 0.002) and limb grip strength (63.9 [37.4, 90.5], p < 0.0001) due to a decrease in insulin/insulin-like growth factor 1 mRNA and an increase in myostatin mRNA. In contrast, Chrebp deletion increased bone mineral density (BMD) (WT control-KO control: -6.1 [-1.0, -2.3], p = 0.0009), stiffness (-21.4 [-38.8, -4.1], p = 0.011), cancellous bone BV/TV (-6.517 [-10.99, -2.040], p = 0.003), and the number of trabeculae (-1.1 [-1.8, -0.5], p = 0.0008). However, in KO mice, protein restriction additively decreased BMD (KO control-KO low-protein: 8.1 [4.3, 11.9], p < 0.0001), bone stiffness (38.0 [21.3, 54.7], p < 0.0001), cancellous bone BV/TV (7.7 [3.3, 12.2], p = 0.006), and the number of trabeculae (1.2 [0.6, 1.9], p = 0.0004). The effects of mild protein restriction on bone formation parameters (osteoid volume (WT control-WT low-protein: -1.7 [-2.7, -0.7], p = 0.001) and the osteoid surface (-11.2 [-20.8, -1.5], p = 0.02) were observed only in wild-type (WT) mice. The levels of bone resorption markers, such as the number of osteoclasts on the surface, the number of osteoclasts, and surface erosion, did not differ between the groups. Conclusions: Both Chrebp deletion and protein restriction led to a decrease in muscle and bone function; therefore, an adequate intake of carbohydrates and proteins is important for maintaining muscle and bone mass and function. Further studies will be needed to elucidate the mechanisms by which ChREBP deletion and a low-protein diet cause osteosarcopenia.
Collapse
Affiliation(s)
- Kanako Deguchi
- Department of Clinical Nutrition, Fujita Health University, Toyoake 470-1192, Japan; (K.D.); (C.U.); (R.Y.-W.)
| | - Chihiro Ushiroda
- Department of Clinical Nutrition, Fujita Health University, Toyoake 470-1192, Japan; (K.D.); (C.U.); (R.Y.-W.)
| | - Shihomi Hidaka
- Department of Endocrinology, Diabetes and Metabolism, Fujita Health University, Toyoake 470-1192, Japan; (S.H.); (Y.S.); (A.S.)
| | - Hiromi Tsuchida
- Department of Diabetes, Endocrinology and Metabolism, Graduate School of Medicine, Gifu University, Gifu 501-1194, Japan; (H.T.); (D.Y.)
| | - Risako Yamamoto-Wada
- Department of Clinical Nutrition, Fujita Health University, Toyoake 470-1192, Japan; (K.D.); (C.U.); (R.Y.-W.)
| | - Yusuke Seino
- Department of Endocrinology, Diabetes and Metabolism, Fujita Health University, Toyoake 470-1192, Japan; (S.H.); (Y.S.); (A.S.)
| | - Atsushi Suzuki
- Department of Endocrinology, Diabetes and Metabolism, Fujita Health University, Toyoake 470-1192, Japan; (S.H.); (Y.S.); (A.S.)
| | - Daisuke Yabe
- Department of Diabetes, Endocrinology and Metabolism, Graduate School of Medicine, Gifu University, Gifu 501-1194, Japan; (H.T.); (D.Y.)
- Departments of Diabetes, Endocrinology and Nutrition, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan
| | - Katsumi Iizuka
- Department of Clinical Nutrition, Fujita Health University, Toyoake 470-1192, Japan; (K.D.); (C.U.); (R.Y.-W.)
- Food and Nutrition Service Department, Fujita Health University Hospital, Toyoake 470-1192, Japan
| |
Collapse
|
11
|
Tian Y, Gong J, He Z, Peng S, Huan Y, Cao H. Impact of protein intake from a caloric-restricted diet on liver lipid metabolism in overweight and obese rats of different sexes. Sci Rep 2025; 15:2340. [PMID: 39833384 PMCID: PMC11747403 DOI: 10.1038/s41598-025-86596-6] [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: 05/14/2024] [Accepted: 01/13/2025] [Indexed: 01/22/2025] Open
Abstract
In addition to being linked to an excess of lipid accumulation in the liver, being overweight or obese can also result in disorders of lipid metabolism. There is limited understanding regarding whether different levels of protein intake within an energy-restricted diet affect liver lipid metabolism in overweight and obese rats and whether these effects differ by gender, despite the fact that both high protein intake and calorie restriction can improve intrahepatic lipid. The purpose of this study is to explore the effects and mechanisms of different protein intakes within a calorie-restricted diet on liver lipid metabolism, and to investigate whether these effects exhibit gender differences. The Sprague-Dawley rats, which were half female and half male, were used to construct a rat model of overweight and obesity attributed to a high-fat diet. They were then split up into five groups: the normal control (NC) group, the model control (MC) group, the calorie-restricted low protein (LP) group, the calorie-restricted normal protein (NP) group, and the calorie-restricted high protein (HP) group. Body weight was measured weekly. Samples of plasma and liver were obtained after eight weeks and analyzed for glucose, triglycerides, cholesterol, and hormones in the plasma as well as the liver fat and factors involved in the liver's synthesis and degradation. For the male rats, compared to the HP group, the weight of liver fat in the LP and NP group was significantly higher (P < 0.05). However, for the female rats, there was no significant variation among the three calorie-restricted groups (P > 0.05). There was no significant variation in the concentration of total cholesterol (TC), very low density lipoprotein (VLDL), low density lipoprotein (LDL), and high density lipoprotein (HDL) among the three male calorie-restricted groups (P > 0.05), while the TC and VLDL concentrations in the female LP and NP group were significantly higher compared to those in the HP group (P < 0.05). Moreover, the trend of expression in the signaling pathways of adiponectin/phosphorylated AMP-activated protein kinase (p-AMPK) and adiponectin/peroxisome proliferators-activated receptor alpha (PPARα) in the liver was consistent with that of the liver fat content, and leptin acted in the same way as adiponectin. Compared with the three calorie-restricted groups, the expressions of nuclear sterol-regulatory element-binding protein-2 (nSREBP-2) and 3-hydroxy-3-methylglutaryl coenzyme A reductase (HMG-CoA reductase) involved in cholesterol synthesis and low-density lipoprotein receptor (LDLR) and cholesterol 7-alpha hydroxylase (CYP7A1) involved in cholesterol clearance in the MC group were significantly lower (P < 0.05). A 40% energy restriction can significantly reduce the body weight, body fat, liver fat, and the blood concentration of TG in both male and female overweight and obese rats, but it can significantly increase the blood concentration of TC in overweight and obese male rats. At the same time of 40% calorie restriction, increasing dietary protein intake to twice the normal protein intake has a stronger effect on promoting hepatic triglyceride oxidation and reducing liver fat content in the male overweight and obese rats by increasing the levels of adiponectin and leptin in the blood, and can also significantly reduce the plasma cholesterol concentration in the female overweight and obese rats through inhibiting cholesterol synthesis most likely by increasing glucagon level in the blood.
Collapse
Affiliation(s)
- Ying Tian
- Department of Nutrition and Food Hygiene, School of Public Health, Yangzhou University, Yangzhou, China.
| | - Jiawei Gong
- Department of Cuisine and Nutrition, School of Tourism and Cuisine, Yangzhou University, Yangzhou, China
| | - Zhiyan He
- Department of Cuisine and Nutrition, School of Tourism and Cuisine, Yangzhou University, Yangzhou, China
| | - Suwen Peng
- Department of Cuisine and Nutrition, School of Tourism and Cuisine, Yangzhou University, Yangzhou, China
| | - Yuping Huan
- Department of Cuisine and Nutrition, School of Tourism and Cuisine, Yangzhou University, Yangzhou, China
| | - Hongpeng Cao
- Department of Cuisine and Nutrition, School of Tourism and Cuisine, Yangzhou University, Yangzhou, China
| |
Collapse
|
12
|
da Silva FMO, Pimenta AM, Juvanhol LL, Hermsdorff HHM, Bressan J. Obesity Incidence According to Branched-Chain Amino Acid Intake and Plant-Based Diet Index Among Brazilian Adults: A Six-Year Follow-Up of the CUME Study. Nutrients 2025; 17:227. [PMID: 39861357 PMCID: PMC11767458 DOI: 10.3390/nu17020227] [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: 12/08/2024] [Revised: 01/03/2025] [Accepted: 01/07/2025] [Indexed: 01/27/2025] Open
Abstract
BACKGROUND Few studies have evaluated the impact of branched-chain amino acid (BCAA) intake on the risk of obesity in adults. The results are contradictory, and the causality has not been explored. This study assessed the association between BCAA intake and obesity incidence among Brazilian adults and investigated the potential moderating role of the plant-based index (PDI) in this relationship. METHODS A longitudinal study was conducted between 2016 and 2022, with 3090 participants (2043 women, 1047 men; mean age 34 years) from the Cohort of Universities of Minas Gerais (CUME) Study. Data were collected through an online questionnaire. The relationship between BCAA intake and obesity incidence was assessed using crude and adjusted Cox regression models. Restricted cubic spline analysis (RCS) was used to estimate the nonlinearity. The multiplicative interaction with PDI was tested. RESULTS The overall incidence of obesity was 192 cases (6.21%). The incidence was 16.4/1000 person-years in females; 21.8/1000 person-years in males; and 18.3/1000 person-years total, with a mean follow-up period of 3.4 years. Compared to the first tertile, the highest intake tertiles for BCAA (HR = 1.50, 95% CI = 1.03-2.18), isoleucine (HR = 1.52, 95% CI = 1.04-2.22), and leucine (HR = 1.51, 95% CI = 1.03-2.20) were independently associated with obesity risk. BCAA intake above 16 g/day increases the risk of obesity. CONCLUSIONS There was a positive association between the intake of BCAA, isoleucine, and leucine with the risk of obesity. The PDI accentuated the association between BCAA intake and obesity in both the lowest and highest quintiles.
Collapse
Affiliation(s)
- Fernanda Maria Oliveira da Silva
- Laboratory of Energy Metabolism and Body Composition, Department of Nutrition and Health, Federal University of Viçosa, Viçosa 36570-900, Brazil; (F.M.O.d.S.); (H.H.M.H.)
- Laboratory of Clinical Analysis and Genomics, Department of Nutrition and Health, Federal University of Viçosa, Viçosa 36570-900, Brazil
| | | | | | - Helen Hermana Miranda Hermsdorff
- Laboratory of Energy Metabolism and Body Composition, Department of Nutrition and Health, Federal University of Viçosa, Viçosa 36570-900, Brazil; (F.M.O.d.S.); (H.H.M.H.)
- Laboratory of Clinical Analysis and Genomics, Department of Nutrition and Health, Federal University of Viçosa, Viçosa 36570-900, Brazil
| | - Josefina Bressan
- Laboratory of Energy Metabolism and Body Composition, Department of Nutrition and Health, Federal University of Viçosa, Viçosa 36570-900, Brazil; (F.M.O.d.S.); (H.H.M.H.)
- Laboratory of Clinical Analysis and Genomics, Department of Nutrition and Health, Federal University of Viçosa, Viçosa 36570-900, Brazil
| |
Collapse
|
13
|
Wu H, Yang ASP, Stelloo S, Roos FJM, te Morsche RHM, Verkerk AH, Luna-Velez MV, Wingens L, de Wilt JHW, Sauerwein RW, Mulder KW, van Heeringen SJ, Verstegen MMA, van der Laan LJW, Marks H, Bártfai R. Multi-omics analysis reveals distinct gene regulatory mechanisms between primary and organoid-derived human hepatocytes. Dis Model Mech 2025; 18:dmm050883. [PMID: 39878507 PMCID: PMC11810045 DOI: 10.1242/dmm.050883] [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: 05/07/2024] [Accepted: 11/25/2024] [Indexed: 01/31/2025] Open
Abstract
Hepatic organoid cultures are a powerful model to study liver development and diseases in vitro. However, hepatocyte-like cells differentiated from these organoids remain immature compared to primary human hepatocytes (PHHs), which are the benchmark in the field. Here, we applied integrative single-cell transcriptome and chromatin accessibility analysis to reveal gene regulatory mechanisms underlying these differences. We found that, in mature human hepatocytes, activator protein 1 (AP-1) factors co-occupy regulatory regions with hepatocyte-specific transcription factors, including HNF4A, suggesting their potential cooperation in governing hepatic gene expression. Comparative analysis identified distinct transcription factor sets that are specifically active in either PHHs or intrahepatic cholangiocyte organoid (ICO)-derived human hepatocytes. ELF3 was one of the factors uniquely expressed in ICO-derived hepatocytes, and its expression negatively correlated with hepatic marker gene expression. Functional analysis further revealed that ELF3 depletion increased the expression of key hepatic markers in ICO-derived hepatocytes. Our integrative analysis provides insights into the transcriptional regulatory networks of PHHs and hepatic organoids, thereby informing future strategies for developing improved hepatic models.
Collapse
Affiliation(s)
- Haoyu Wu
- Department of Molecular Biology, Faculty of Science, Radboud Institute for Molecular Life Science, Radboud University, Nijmegen 6525GA, The Netherlands
| | - Annie S. P. Yang
- Center for Infectious Diseases, Department of Medical Microbiology, Radboud University Medical Center, Nijmegen 6500HB, The Netherlands
| | - Suzan Stelloo
- Department of Molecular Biology, Faculty of Science, Radboud Institute for Molecular Life Science, Radboud University, Nijmegen 6525GA, The Netherlands
- Department of Molecular Biology, Faculty of Science, Radboud Institute for Molecular Life Science, Oncode Institute, Radboud University, Nijmegen 6525GA, The Netherlands
| | - Floris J. M. Roos
- Department of Surgery, Erasmus University Medical Center Transplant Institute, University Medical Center Rotterdam,Rotterdam 3000CA, TheNetherlands
| | - René H. M. te Morsche
- Department of Gastroenterology and Hepatology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen 6500HB, The Netherlands
| | - Anne H. Verkerk
- Department of Molecular Biology, Faculty of Science, Radboud Institute for Molecular Life Science, Radboud University, Nijmegen 6525GA, The Netherlands
| | - Maria V. Luna-Velez
- Department of Molecular Biology, Faculty of Science, Radboud Institute for Molecular Life Science, Radboud University, Nijmegen 6525GA, The Netherlands
- Department of Molecular Biology, Faculty of Science, Radboud Institute for Molecular Life Science, Oncode Institute, Radboud University, Nijmegen 6525GA, The Netherlands
| | - Laura Wingens
- Department of Molecular Developmental Biology, Faculty of Science, Radboud Institute for Molecular Life Sciences, Radboud University, Nijmegen 6525GA, The Netherlands
| | - Johannes H. W. de Wilt
- Department of Surgery, Radboud University Medical Center, Nijmegen 6500HB, The Netherlands
| | - Robert W. Sauerwein
- Center for Infectious Diseases, Department of Medical Microbiology, Radboud University Medical Center, Nijmegen 6500HB, The Netherlands
| | - Klaas W. Mulder
- Department of Molecular Developmental Biology, Faculty of Science, Radboud Institute for Molecular Life Sciences, Radboud University, Nijmegen 6525GA, The Netherlands
| | - Simon J. van Heeringen
- Department of Molecular Developmental Biology, Faculty of Science, Radboud Institute for Molecular Life Sciences, Radboud University, Nijmegen 6525GA, The Netherlands
| | - Monique M. A. Verstegen
- Department of Surgery, Erasmus University Medical Center Transplant Institute, University Medical Center Rotterdam,Rotterdam 3000CA, TheNetherlands
| | - Luc J. W. van der Laan
- Department of Surgery, Erasmus University Medical Center Transplant Institute, University Medical Center Rotterdam,Rotterdam 3000CA, TheNetherlands
| | - Hendrik Marks
- Department of Molecular Biology, Faculty of Science, Radboud Institute for Molecular Life Science, Radboud University, Nijmegen 6525GA, The Netherlands
| | - Richárd Bártfai
- Department of Molecular Biology, Faculty of Science, Radboud Institute for Molecular Life Science, Radboud University, Nijmegen 6525GA, The Netherlands
| |
Collapse
|
14
|
Deguchi K, Ushiroda C, Kamei Y, Kondo K, Tsuchida H, Seino Y, Yabe D, Suzuki A, Nagao S, Iizuka K. Glucose and Insulin Differently Regulate Gluconeogenic and Ureagenic Gene Expression. J Nutr Sci Vitaminol (Tokyo) 2025; 71:46-54. [PMID: 40024748 DOI: 10.3177/jnsv.71.46] [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: 03/04/2025]
Abstract
Glucose and insulin positively regulate glycolysis and lipogenesis through the activation of carbohydrate response element-binding protein (ChREBP) and sterol regulatory element-binding protein 1c (SREBP1c), but their respective roles in the regulation of gluconeogenic and ureagenic genes remain unclear. We compared the effects of the insulin antagonist S961 and Chrebp deletion on hepatic glycolytic, lipogenic, gluconeogenic, and ureagenic gene expression in mice. S961 markedly increased the plasma glucose, insulin, and 3-OH-butyrate concentrations and reduced the hepatic triglyceride content, but Chrebp deletion had no additive effect. We subsequently estimated the expression of genes involved in the pathways of glycolysis, gluconeogenesis, and lipogenesis. S961 potently decreased both Chrebp and Srebf1c, but Chrebp deletion weakly decreased Srebf1c mRNA expression. Both the S961 and Chrebp deletion caused decreases in glycolytic (Gck and Pklr) and lipogenic (Fasn, Scd1, Me1, Spot14, Elovl6) gene expression. S961 increased the expression of many gluconeogenic genes (G6pc, Fbp1, Aldob, Slc37a4, Pck), whereas Chrebp deletion reduced the expression of gluconeogenic genes other than Pck1. Finally, we checked the metabolites and gene expression in the ureagenesis pathway. S961 increased ureagenic gene (Arg1, Asl, Ass1, Cps1, Otc) expression, which was consistent with the metabolite data: there were reductions in the concentrations of glutamate and aspartate and increases in those of citrulline, ornithine, urea, and proline. However, Chrebp deletion had no additive effect on ureagenesis. In conclusion, insulin rather than glucose regulate ureagenic gene expression, whereas glucose and insulin regulate gluconegenic gene expression in opposite directions.
Collapse
Affiliation(s)
- Kanako Deguchi
- Department of Clinical Nutrition, Fujita Health University
| | | | - Yuka Kamei
- Advanced Medical Research Center for Animal Models of Human Diseases, Fujita Health University
| | | | - Hiromi Tsuchida
- Department of Diabetes, Endocrinology and Metabolism, Gifu University Graduate School of Medicine
| | - Yusuke Seino
- Department of Endocrinology, Diabetes and Metabolism, Fujita Health University
| | - Daisuke Yabe
- Department of Diabetes, Endocrinology and Metabolism, Gifu University Graduate School of Medicine
- Center for One Medicine Innovative Translational Research, Gifu University
| | - Atsushi Suzuki
- Department of Endocrinology, Diabetes and Metabolism, Fujita Health University
| | - Shizuko Nagao
- Advanced Medical Research Center for Animal Models of Human Diseases, Fujita Health University
| | - Katsumi Iizuka
- Department of Clinical Nutrition, Fujita Health University
- Food and Nutrition Service Department, Fujita Health University Hospital
| |
Collapse
|
15
|
Raza I, Sohail A, Muneer H, Fayyaz H, Uddin Z, Almars AI, Aggad WS, Almohaimeed HM, Ullah I. Viscosol Treatment Ameliorates Insulin-Mediated Regulation of Dyslipidemia, Hepatic Steatosis, and Lipid Metabolism by Targeting PTP1B in Type-2 Diabetic Mice Model. Int J Endocrinol 2024; 2024:3914332. [PMID: 39759127 PMCID: PMC11698613 DOI: 10.1155/ije/3914332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 08/06/2024] [Accepted: 11/25/2024] [Indexed: 01/07/2025] Open
Abstract
Type 2 diabetes mellitus (T2DM), a metabolic disorder, has the hallmarks of persistent hyperglycemia, insulin resistance, and dyslipidemia. Protein-tyrosine phosphatase 1B (PTP1B) was found to be overexpressed in many tissues in the case of T2DM and involved in the negative regulation of insulin signaling. So, PTP1B inhibition can act as a therapeutic target for T2DM. Numerous studies claimed the anti-inflammatory, hypoglycemic, hepatoprotective, and hypolipidemic activities of Dodonaea viscosa. Previously, we generated the high-fat diet (HFD)-low dose streptozotocin (STZ)-induced diabetic male mice model and treated it with a PTP1B inhibitor (5, 7-dihydroxy-3, 6-dimethoxy-2- (4-methoxy-3- (3-methyl-2-enyl) phenyl)-4H-chromen-4-one), isolated from Dodonaea viscosa. In the current study, we aimed to investigate the De novo lipogenesis, adipocyte differentiation, augmentation of lipoproteins clearance, fatty acid uptake, antilipolysis activity, and hepatic steatosis of PTP1B inhibition in adipose and liver tissues of the HFD-STZ-induced diabetic mice model. We found the retrieval of normal morphology of adipocytes and hepatocytes in the compound-treated group. The biochemical parameters showed the gradual reduction of LDL, VLDL, TC, and TG in the serum of the compound-treated group. To further test our hypothesis, real-time PCR was performed, and data revealed the reduction of PTP1B and other inflammatory markers in both tissues, showing enhanced expression of insulin signaling markers (INSR, IRS1, IRS2, and PI3K). Our compound upregulated the adipogenic (PPARγ), lipogenic (SREBP1c, FAS, ACC, and DGAT2), lipoprotein clearance (LPL, LDLR, and VLDLR), fatty acid uptake (CD36 and FATP1), and lipid droplet forming (FSP27 and perilipin-1) markers expressions in adipocytes and downregulated in hepatocytes. Furthermore, we found elevated cholesterol efflux (in adipose and liver) and decreased lipolysis in adipocytes and elevated in hepatocytes. Hence, we can conclude that our compound protects the adipocytes from abrupt lipolysis and stimulates adipocyte differentiation. In addition, it plays a hepatic protective role by shifting clearance and uptake of lipoproteins and fatty acids to the peripheral tissues and retrieving the fatty liver condition.
Collapse
Affiliation(s)
- Idrees Raza
- Department of Biochemistry, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad 45320, Pakistan
- Department of Biochemistry & Biotechnology, FVAS, Muhammad Nawaz Shareef University of Agriculture, Multan, Pakistan
| | - Aamir Sohail
- Department of Biochemistry, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad 45320, Pakistan
| | - Hamza Muneer
- Department of Biochemistry, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad 45320, Pakistan
- Department of Biochemistry & Biotechnology, FVAS, Muhammad Nawaz Shareef University of Agriculture, Multan, Pakistan
| | - Hajra Fayyaz
- Department of Biochemistry, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad 45320, Pakistan
| | - Zia Uddin
- Department of Pharmacy, COMSATS University Islamabad, Abbottabad Campus, Abbottabad 22060, Khyber Pakhtunkhwa, Pakistan
| | - Amany I. Almars
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Waheeb S. Aggad
- Division of Anatomy, Department of Basic Medical Sciences, College of Medicine, University of Jeddah, P.O. Box 8304, Jeddah 23234, Saudi Arabia
| | - Hailah M. Almohaimeed
- Department of Basic Science, College of Medicine, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia
| | - Imran Ullah
- Department of Biochemistry, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad 45320, Pakistan
| |
Collapse
|
16
|
Misceo D, Mocciaro G, D'Amore S, Vacca M. Diverting hepatic lipid fluxes with lifestyles revision and pharmacological interventions as a strategy to tackle steatotic liver disease (SLD) and hepatocellular carcinoma (HCC). Nutr Metab (Lond) 2024; 21:112. [PMID: 39716321 DOI: 10.1186/s12986-024-00871-3] [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: 08/22/2024] [Accepted: 11/13/2024] [Indexed: 12/25/2024] Open
Abstract
Steatotic liver disease (SLD) and Hepatocellular Carcinoma (HCC) are characterised by a substantial rewiring of lipid fluxes caused by systemic metabolic unbalances and/or disrupted intracellular metabolic pathways. SLD is a direct consequence of the interaction between genetic predisposition and a chronic positive energy balance affecting whole-body energy homeostasis and the function of metabolically-competent organs. In this review, we discuss how the impairment of the cross-talk between peripheral organs and the liver stalls glucose and lipid metabolism, leading to unbalances in hepatic lipid fluxes that promote hepatic fat accumulation. We also describe how prolonged metabolic stress builds up toxic lipid species in the liver, and how lipotoxicity and metabolic disturbances drive disease progression by promoting a chronic activation of wound healing, leading to fibrosis and HCC. Last, we provide a critical overview of current state of the art (pre-clinical and clinical evidence) regarding mechanisms of action and therapeutic efficacy of candidate SLD treatment options, and their potential to interfere with SLD/HCC pathophysiology by diverting lipids away from the liver therefore improving metabolic health.
Collapse
Affiliation(s)
- Davide Misceo
- Department of Interdisciplinary Medicine, Clinica Medica "C. Frugoni", "Aldo Moro" University of Bari, Piazza Giulio Cesare 11, 70124, Bari, Italy
| | - Gabriele Mocciaro
- Roger Williams Institute of Liver Studies, Foundation for Liver Research, London, SE5 9NT, UK
| | - Simona D'Amore
- Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), Clinica Medica "G. Baccelli", "Aldo Moro" University of Bari, 70124, Bari, Italy.
| | - Michele Vacca
- Department of Interdisciplinary Medicine, Clinica Medica "C. Frugoni", "Aldo Moro" University of Bari, Piazza Giulio Cesare 11, 70124, Bari, Italy.
- Roger Williams Institute of Liver Studies, Foundation for Liver Research, London, SE5 9NT, UK.
| |
Collapse
|
17
|
Xu X, Mendoza A, Krumm CS, Su S, Acuña M, Bare CJ, Holman CD, Cortopassi M, Nicholls HT, Dartigue V, Hollenberg AN, Lee AH, Hagen SJ, Cohen DE. ChREBP-mediated up-regulation of Them1 coordinates thermogenesis with glycolysis and lipogenesis in response to chronic stress. Sci Signal 2024; 17:eadk7971. [PMID: 39626011 PMCID: PMC11817722 DOI: 10.1126/scisignal.adk7971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Revised: 08/15/2024] [Accepted: 11/12/2024] [Indexed: 02/06/2025]
Abstract
Activation of thermogenic brown adipose tissue (BAT) and inducible beige adipose tissue (BeAT) is triggered by environmental or metabolic stimuli, including cold ambient temperatures and nutrient stress. Thioesterase superfamily member 1 (Them1), a long-chain fatty acyl-CoA thioesterase that is enriched in BAT, suppresses acute cold-induced thermogenesis. Here, we demonstrate that Them1 expression was induced in BAT and BeAT by the carbohydrate response element binding protein (ChREBP) in response to chronic cold exposure or to the activation of the integrated stress response (ISR) by nutrient excess. Under either condition, Them1 suppressed energy expenditure. Consequently, mice lacking Them1 in BAT and BeAT exhibited resistance to obesity and glucose intolerance induced by feeding with a high-fat diet. During chronic cold exposure or ISR activation, Them1 accumulated in the nucleus, where it interacted with ChREBP and reduced the expression of its target genes, including those encoding enzymes that mediate glycolysis and de novo lipogenesis. These findings demonstrate that in response to chronic cold- or nutrient-induced stress, the induction of Them1 by ChREBP limits thermogenesis while coordinately reducing glucose utilization and lipid biosynthesis through its distinct cytoplasmic and nuclear activities. Targeted inhibition of Them1 could be a potential therapeutic approach to increase the activity of BAT and BeAT to enhance energy expenditure in the management of obesity-associated metabolic disorders.
Collapse
Affiliation(s)
- Xu Xu
- Division of Gastroenterology and Hepatology, Joan & Sanford I. Weill Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
- Division of Surgical Sciences, Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | - Arturo Mendoza
- Division of Endocrinology, Diabetes and Metabolism, Joan & Sanford I. Weill Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Christopher S. Krumm
- Division of Gastroenterology and Hepatology, Joan & Sanford I. Weill Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Shi Su
- Division of Gastroenterology, Hepatology and Endoscopy, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Mariana Acuña
- Division of Gastroenterology and Hepatology, Joan & Sanford I. Weill Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
- Division of Gastroenterology, Hepatology and Endoscopy, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Curtis J. Bare
- Division of Gastroenterology and Hepatology, Joan & Sanford I. Weill Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Corey D. Holman
- Division of Gastroenterology and Hepatology, Joan & Sanford I. Weill Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Marissa Cortopassi
- Division of Gastroenterology and Hepatology, Joan & Sanford I. Weill Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Hayley T. Nicholls
- Division of Gastroenterology and Hepatology, Joan & Sanford I. Weill Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Vincent Dartigue
- Division of Gastroenterology and Hepatology, Joan & Sanford I. Weill Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
- Division of Gastroenterology, Hepatology and Endoscopy, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Anthony N. Hollenberg
- Division of Endocrinology, Diabetes and Metabolism, Joan & Sanford I. Weill Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Ann-Hwee Lee
- Department of Pathology & Laboratory Medicine, Weill Cornell Medicine, New York, NY, 10065, USA
- Present address: Regeneron Pharmaceuticals, Tarrytown, NY 10591, USA
| | - Susan J. Hagen
- Division of Surgical Sciences, Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | - David E. Cohen
- Division of Gastroenterology and Hepatology, Joan & Sanford I. Weill Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
- Division of Gastroenterology, Hepatology and Endoscopy, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- Lead contact
| |
Collapse
|
18
|
Small L, Nguyen TV, Larance M, Saunders DN, Hoy AJ, Schmitz-Peiffer C, Cooney GJ, Brandon AE. Liver proteomics identifies a disconnect between proteins associated with de novo lipogenesis and triglyceride storage. J Lipid Res 2024; 65:100687. [PMID: 39490929 PMCID: PMC11626007 DOI: 10.1016/j.jlr.2024.100687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 10/17/2024] [Accepted: 10/21/2024] [Indexed: 11/05/2024] Open
Abstract
De novo lipogenesis (DNL) has been implicated in the development and progression of liver steatosis. Hepatic DNL is strongly influenced by dietary macronutrient composition with diets high in carbohydrate increasing DNL while diets high in fat decrease DNL. The enzymes in the core DNL pathway have been well characterized; however, less is known about other liver proteins that play accessory or regulatory roles. In the current study, we associate measured rates of hepatic DNL and fat content with liver proteomic analysis in mice to identify known and unknown proteins that may have a role in DNL. Male mice were fed either a standard chow diet, a semipurified high starch or high-fat diet. Both semipurified diets resulted in increased body weight, fat mass, and liver triglyceride content compared to chow controls, and hepatic DNL was increased in the high starch and decreased in high fat-fed mice. Proteomic analysis identified novel proteins associated with DNL that are involved in taurine metabolism, suggesting a link between these pathways. There was no relationship between proteins that associated with DNL and those associated with liver triglyceride content. Further analysis identified proteins that are differentially regulated when comparing a nonpurified chow diet to either of the semipurified diets, which provide a set of proteins that are influenced by dietary complexity. Finally, we compared the liver proteome between 4- and 30-week diet-fed mice and found remarkable similarity suggesting that metabolic remodeling of the liver occurs rapidly in response to differing dietary components. Together, these findings highlight novel proteins associated with hepatic DNL independently of liver fat content and suggest rapid liver metabolic remodeling in response to dietary composition changes.
Collapse
Affiliation(s)
- Lewin Small
- School of Life and Environmental Sciences, Charles Perkins Centre, Faculty of Science, The University of Sydney, Sydney, NSW, Australia; Garvan Institute, Sydney, NSW, Australia.
| | | | - Mark Larance
- School of Medical Sciences, Charles Perkins Centre, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Darren N Saunders
- School of Medical Sciences, Charles Perkins Centre, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Andrew J Hoy
- School of Medical Sciences, Charles Perkins Centre, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Carsten Schmitz-Peiffer
- School of Life and Environmental Sciences, Charles Perkins Centre, Faculty of Science, The University of Sydney, Sydney, NSW, Australia; Garvan Institute, Sydney, NSW, Australia
| | - Gregory J Cooney
- Garvan Institute, Sydney, NSW, Australia; School of Medical Sciences, Charles Perkins Centre, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Amanda E Brandon
- School of Life and Environmental Sciences, Charles Perkins Centre, Faculty of Science, The University of Sydney, Sydney, NSW, Australia; Garvan Institute, Sydney, NSW, Australia.
| |
Collapse
|
19
|
Cheng SS, Mody AC, Woo CM. Opportunities for Therapeutic Modulation of O-GlcNAc. Chem Rev 2024; 124:12918-13019. [PMID: 39509538 DOI: 10.1021/acs.chemrev.4c00417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2024]
Abstract
O-Linked β-N-acetylglucosamine (O-GlcNAc) is an essential, dynamic monosaccharide post-translational modification (PTM) found on serine and threonine residues of thousands of nucleocytoplasmic proteins. The installation and removal of O-GlcNAc is controlled by a single pair of enzymes, O-GlcNAc transferase (OGT) and O-GlcNAcase (OGA), respectively. Since its discovery four decades ago, O-GlcNAc has been found on diverse classes of proteins, playing important functional roles in many cellular processes. Dysregulation of O-GlcNAc homeostasis has been implicated in the pathogenesis of disease, including neurodegeneration, X-linked intellectual disability (XLID), cancer, diabetes, and immunological disorders. These foundational studies of O-GlcNAc in disease biology have motivated efforts to target O-GlcNAc therapeutically, with multiple clinical candidates under evaluation. In this review, we describe the characterization and biochemistry of OGT and OGA, cellular O-GlcNAc regulation, development of OGT and OGA inhibitors, O-GlcNAc in pathophysiology, clinical progress of O-GlcNAc modulators, and emerging opportunities for targeting O-GlcNAc. This comprehensive resource should motivate further study into O-GlcNAc function and inspire strategies for therapeutic modulation of O-GlcNAc.
Collapse
Affiliation(s)
- Steven S Cheng
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, Massachusetts 02138, United States
| | - Alison C Mody
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, Massachusetts 02138, United States
| | - Christina M Woo
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, Massachusetts 02138, United States
- Affiliate member of the Broad Institute, Cambridge, Massachusetts 02142, United States
| |
Collapse
|
20
|
Liu Y, Fan Y, Liu J, Liu X, Li X, Hu J. Application and mechanism of Chinese herb medicine in the treatment of non-alcoholic fatty liver disease. Front Pharmacol 2024; 15:1499602. [PMID: 39605910 PMCID: PMC11598537 DOI: 10.3389/fphar.2024.1499602] [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: 09/21/2024] [Accepted: 10/30/2024] [Indexed: 11/29/2024] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a chronic liver condition closely associated with metabolic syndrome, with its incidence rate continuously rising globally. Recent studies have shown that the development of NAFLD is associated with insulin resistance, lipid metabolism disorder, oxidative stress and endoplasmic reticulum stress. Therapeutic strategies for NAFLD include lifestyle modifications, pharmacological treatments, and emerging biological therapies; however, there is currently no specific drug to treat NAFLD. However Chinese herb medicine (CHM) has shown potential in the treatment of NAFLD due to its unique therapeutic concepts and methods for centuries in China. This review aims to summarize the pathogenesis of NAFLD and some CHMs that have been shown to have therapeutic effects on NAFLD, thus enriching the scientific connotation of TCM theories and facilitating the exploration of TCM in the treatment of NAFLD.
Collapse
Affiliation(s)
- Yuqiao Liu
- College of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yue Fan
- College of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jibin Liu
- College of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiyang Liu
- College of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiuyan Li
- College of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jingqing Hu
- College of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Xin-Huangpu Joint Innovation Institute of Chinese Medicine, Guangzhou, China
| |
Collapse
|
21
|
Hansen K, Peters K, Burkert CK, Brose E, Calvisi DF, Ehricke K, Engeler M, Knuth E, Kröger N, Lohr A, Prey J, Sonke J, Vakeel P, Wladasch J, Zimmer J, Dombrowski F, Ribback S. Knockout of the Carbohydrate Responsive Element Binding Protein Enhances Proliferation and Tumorigenesis in Renal Tubules of Mice. Int J Mol Sci 2024; 25:11438. [PMID: 39518998 PMCID: PMC11545909 DOI: 10.3390/ijms252111438] [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: 09/10/2024] [Revised: 10/10/2024] [Accepted: 10/23/2024] [Indexed: 11/16/2024] Open
Abstract
Glycogen-storing so-called clear cell kidney tubules (CCTs), precursor lesions of renal cell carcinoma, have been described in diabetic rats and in humans. The lesions show upregulation of the Akt/mTOR-pathway and the related transcription factor carbohydrate responsive element binding protein (ChREBP), which is supposedly pro-oncogenic. We investigated the effect of ChREBP-knockout on nephrocarcinogenesis in streptozotocin-induced diabetic and normoglycemic mice. Diabetic, but not non-diabetic mice, showed CCTs at 3, 6 and 12 months of age. Glycogenosis was confirmed by periodic acid schiff reaction and transmission electron microscopy. CCTs in ChREBP-knockout mice consisted of larger cells and occurred more frequently compared to wildtype mice. Progression towards kidney tumors was observed in both diabetic groups but occurred earlier in ChREBP-knockout mice. Proliferative activity assessed by BrdU-labeling was lower in 1-week-old but higher in 12-month-old diabetic ChREBP-knockout mice. Surprisingly, renal neoplasms occurred spontaneously in non-diabetic ChREBP-knockout, but not non-diabetic wildtype mice, indicating an unexpected tumor-suppressive function of ChREBP. Immunohistochemistry showed upregulated glycolysis and lipogenesis, along with activated Akt/mTOR-signaling in tumors of ChREBP-knockout groups. Immunohistochemistry of human clear cell renal cell carcinomas revealed reduced ChREBP expression compared to normal kidney tissue. However, the molecular mechanisms by which loss of ChREBP might facilitate tumorigenesis require further investigation.
Collapse
Affiliation(s)
- Kerrin Hansen
- Institut für Pathologie, Universitaetsmedizin Greifswald, DE-17489 Greifswald, Germany; (K.P.); (M.E.); (J.P.); (P.V.); (J.W.); (F.D.); (S.R.)
| | - Kristin Peters
- Institut für Pathologie, Universitaetsmedizin Greifswald, DE-17489 Greifswald, Germany; (K.P.); (M.E.); (J.P.); (P.V.); (J.W.); (F.D.); (S.R.)
| | - Christian K. Burkert
- Institut für Pathologie, Universitaetsmedizin Greifswald, DE-17489 Greifswald, Germany; (K.P.); (M.E.); (J.P.); (P.V.); (J.W.); (F.D.); (S.R.)
| | - Eric Brose
- Institut für Pathologie, Universitaetsmedizin Greifswald, DE-17489 Greifswald, Germany; (K.P.); (M.E.); (J.P.); (P.V.); (J.W.); (F.D.); (S.R.)
| | - Diego F. Calvisi
- Institut für Pathologie, Universität Regensburg, DE-93053 Regensburg, Germany;
| | - Katrina Ehricke
- Institut für Pathologie, Universitaetsmedizin Greifswald, DE-17489 Greifswald, Germany; (K.P.); (M.E.); (J.P.); (P.V.); (J.W.); (F.D.); (S.R.)
| | - Maren Engeler
- Institut für Pathologie, Universitaetsmedizin Greifswald, DE-17489 Greifswald, Germany; (K.P.); (M.E.); (J.P.); (P.V.); (J.W.); (F.D.); (S.R.)
| | - Elisa Knuth
- Institut für Pathologie, Universitaetsmedizin Greifswald, DE-17489 Greifswald, Germany; (K.P.); (M.E.); (J.P.); (P.V.); (J.W.); (F.D.); (S.R.)
| | - Nils Kröger
- Klinik und Poliklinik für Urologie, Universitaetsmedizin Greifswald, DE-17489 Greifswald, Germany
| | - Andrea Lohr
- Institut für Pathologie, Universitaetsmedizin Greifswald, DE-17489 Greifswald, Germany; (K.P.); (M.E.); (J.P.); (P.V.); (J.W.); (F.D.); (S.R.)
| | - Jessica Prey
- Institut für Pathologie, Universitaetsmedizin Greifswald, DE-17489 Greifswald, Germany; (K.P.); (M.E.); (J.P.); (P.V.); (J.W.); (F.D.); (S.R.)
| | - Jenny Sonke
- Institut für Pathologie, Universitaetsmedizin Greifswald, DE-17489 Greifswald, Germany; (K.P.); (M.E.); (J.P.); (P.V.); (J.W.); (F.D.); (S.R.)
| | - Padmanabhan Vakeel
- Institut für Pathologie, Universitaetsmedizin Greifswald, DE-17489 Greifswald, Germany; (K.P.); (M.E.); (J.P.); (P.V.); (J.W.); (F.D.); (S.R.)
| | - Juliane Wladasch
- Institut für Pathologie, Universitaetsmedizin Greifswald, DE-17489 Greifswald, Germany; (K.P.); (M.E.); (J.P.); (P.V.); (J.W.); (F.D.); (S.R.)
| | - Jenny Zimmer
- Institut für Pathologie, Universitaetsmedizin Greifswald, DE-17489 Greifswald, Germany; (K.P.); (M.E.); (J.P.); (P.V.); (J.W.); (F.D.); (S.R.)
| | - Frank Dombrowski
- Institut für Pathologie, Universitaetsmedizin Greifswald, DE-17489 Greifswald, Germany; (K.P.); (M.E.); (J.P.); (P.V.); (J.W.); (F.D.); (S.R.)
| | - Silvia Ribback
- Institut für Pathologie, Universitaetsmedizin Greifswald, DE-17489 Greifswald, Germany; (K.P.); (M.E.); (J.P.); (P.V.); (J.W.); (F.D.); (S.R.)
| |
Collapse
|
22
|
Li T, Chiang JYL. Bile Acid Signaling in Metabolic and Inflammatory Diseases and Drug Development. Pharmacol Rev 2024; 76:1221-1253. [PMID: 38977324 PMCID: PMC11549937 DOI: 10.1124/pharmrev.124.000978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 06/26/2024] [Accepted: 06/28/2024] [Indexed: 07/10/2024] Open
Abstract
Bile acids are the end products of cholesterol catabolism. Hepatic bile acid synthesis accounts for a major fraction of daily cholesterol turnover in humans. Biliary secretion of bile acids generates bile flow and facilitates biliary secretion of lipids, endogenous metabolites, and xenobiotics. In intestine, bile acids facilitate the digestion and absorption of dietary lipids and fat-soluble vitamins. Through activation of nuclear receptors and G protein-coupled receptors and interaction with gut microbiome, bile acids critically regulate host metabolism and innate and adaptive immunity and are involved in the pathogenesis of cholestasis, metabolic dysfunction-associated steatotic liver disease, alcohol-associated liver disease, type-2 diabetes, and inflammatory bowel diseases. Bile acids and their derivatives have been developed as potential therapeutic agents for treating chronic metabolic and inflammatory liver diseases and gastrointestinal disorders. SIGNIFICANCE STATEMENT: Bile acids facilitate biliary cholesterol solubilization and dietary lipid absorption, regulate host metabolism and immunity, and modulate gut microbiome. Targeting bile acid metabolism and signaling holds promise for treating metabolic and inflammatory diseases.
Collapse
Affiliation(s)
- Tiangang Li
- Department of Biochemistry and Physiology, Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma (T.L.); and Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, Ohio (J.Y.L.C.)
| | - John Y L Chiang
- Department of Biochemistry and Physiology, Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma (T.L.); and Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, Ohio (J.Y.L.C.)
| |
Collapse
|
23
|
An J, Astapova I, Zhang G, Cangelosi AL, Ilkayeva O, Marchuk H, Muehlbauer MJ, George T, Brozinick J, Herman MA, Newgard CB. Integration of metabolomic and transcriptomic analyses reveals novel regulatory functions of the ChREBP transcription factor in energy metabolism. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.17.613577. [PMID: 39345566 PMCID: PMC11429843 DOI: 10.1101/2024.09.17.613577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Carbohydrate Response Element-Binding Protein (ChREBP) is a transcription factor that activates key genes involved in glucose, fructose, and lipid metabolism in response to carbohydrate feeding, but its other potential roles in metabolic homeostasis have not been as well studied. We used liver-selective GalNAc-siRNA technology to suppress expression of ChREBP in rats fed a high fat/high sucrose diet and characterized hepatic and systemic responses by integrating transcriptomic and metabolomic analyses. GalNAc-siChREBP-treated rats had lower levels of multiple short-chain acyl CoA metabolites compared to rats treated with GalNAc-siCtrl containing a non-targeting siRNA sequence. These changes were related to a sharp decrease in free CoA levels in GalNAc-siChREBP treated-rats, accompanied by lower expression of transcripts encoding enzymes and transporters involved in CoA biosynthesis. These activities of ChREBP likely contribute to its complex effects on hepatic lipid and energy metabolism. While core enzymes of fatty acid (FA) oxidation are induced by ChREBP knockdown, accumulation of liver acylcarnitines and circulating ketones indicate diversion of acetyl CoA to ketone production rather than complete oxidation in the TCA cycle. Despite strong suppression of pyruvate kinase and activation of pyruvate dehydrogenase, pyruvate levels were maintained, likely via increased expression of pyruvate transporters, and decreased expression of lactate dehydrogenase and alanine transaminase. GalNAc-siChREBP treatment increased hepatic citrate and isocitrate levels while decreasing levels of distal TCA cycle intermediates. The drop in free CoA levels, needed for the 2-ketoglutarate dehydrogenase reaction, as well as a decrease in transcripts encoding the anaplerotic enzymes pyruvate carboxylase, glutamate dehydrogenase, and aspartate transaminase likely contributed to these effects. GalNAc-siChREBP treatment caused striking increases in PRPP and ZMP/AICAR levels, and decreases in GMP, IMP, AMP, NaNM, NAD(P), and NAD(P)H levels, accompanied by reduced expression of enzymes that catalyze late steps in purine and NAD synthesis. ChREBP suppression also increased expression of a set of plasma membrane amino acid transporters, possibly as an attempt to replenish TCA cycle intermediates. In sum, combining transcriptomic and metabolomic analyses has revealed regulatory functions of ChREBP that go well beyond its canonical roles in control of carbohydrate and lipid metabolism to now include mitochondrial metabolism and cellular energy balance.
Collapse
Affiliation(s)
- Jie An
- Sarah W. Stedman Nutrition and Metabolism Center & Duke Molecular Physiology Institute, Duke University Medical Center
| | - Inna Astapova
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Baylor College of Medicine
| | - Guofang Zhang
- Sarah W. Stedman Nutrition and Metabolism Center & Duke Molecular Physiology Institute, Duke University Medical Center
- Division of Endocrinology, Metabolism and Nutrition, Department of Medicine, Duke University Medical Center
| | - Andrew L. Cangelosi
- Sarah W. Stedman Nutrition and Metabolism Center & Duke Molecular Physiology Institute, Duke University Medical Center
| | - Olga Ilkayeva
- Sarah W. Stedman Nutrition and Metabolism Center & Duke Molecular Physiology Institute, Duke University Medical Center
- Division of Endocrinology, Metabolism and Nutrition, Department of Medicine, Duke University Medical Center
| | - Hannah Marchuk
- Sarah W. Stedman Nutrition and Metabolism Center & Duke Molecular Physiology Institute, Duke University Medical Center
| | - Michael J. Muehlbauer
- Sarah W. Stedman Nutrition and Metabolism Center & Duke Molecular Physiology Institute, Duke University Medical Center
| | - Tabitha George
- Sarah W. Stedman Nutrition and Metabolism Center & Duke Molecular Physiology Institute, Duke University Medical Center
| | | | - Mark A. Herman
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Baylor College of Medicine
| | - Christopher B. Newgard
- Sarah W. Stedman Nutrition and Metabolism Center & Duke Molecular Physiology Institute, Duke University Medical Center
- Division of Endocrinology, Metabolism and Nutrition, Department of Medicine, Duke University Medical Center
- Department of Pharmacology & Cancer Biology, Duke University Medical Center
| |
Collapse
|
24
|
Wang H, Stevens T, Lu J, Roberts A, Van't Land C, Muzumdar R, Gong Z, Vockley J, Prochownik EV. Body-Wide Inactivation of the Myc-Like Mlx Transcription Factor Network Accelerates Aging and Increases the Lifetime Cancer Incidence. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2401593. [PMID: 38976573 PMCID: PMC11425880 DOI: 10.1002/advs.202401593] [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: 02/14/2024] [Revised: 06/08/2024] [Indexed: 07/10/2024]
Abstract
The "Mlx" and "Myc" transcription factor networks cross-communicate and share many common gene targets. Myc's activity depends upon its heterodimerization with Max, whereas the Mlx Network requires that the Max-like factor Mlx associate with the Myc-like factors MondoA or ChREBP. The current work demonstrates that body-wide Mlx inactivation, like that of Myc, accelerates numerous aging-related phenotypes pertaining to body habitus and metabolism. The deregulation of numerous aging-related Myc target gene sets is also accelerated. Among other functions, these gene sets often regulate ribosomal and mitochondrial structure and function, genomic stability, and aging. Whereas "MycKO" mice have an extended lifespan because of a lower cancer incidence, "MlxKO" mice have normal lifespans and a higher cancer incidence. Like Myc, the expression of Mlx, MondoA, and ChREBP and their control over their target genes deteriorate with age in both mice and humans. Collectively, these findings underscore the importance of lifelong and balanced cross-talk between the two networks to maintain proper function and regulation of the many factors that can affect normal aging.
Collapse
Affiliation(s)
- Huabo Wang
- Division of Hematology/OncologyUPMC Children's Hospital of PittsburghPittsburghPA15201USA
| | - Taylor Stevens
- Division of Hematology/OncologyUPMC Children's Hospital of PittsburghPittsburghPA15201USA
| | - Jie Lu
- Division of Hematology/OncologyUPMC Children's Hospital of PittsburghPittsburghPA15201USA
| | - Alexander Roberts
- Division of Hematology/OncologyUPMC Children's Hospital of PittsburghPittsburghPA15201USA
| | - Clinton Van't Land
- Division of Medical GeneticsUPMC Children's Hospital of PittsburghPittsburghPA15201USA
| | - Radhika Muzumdar
- Division of EndocrinologyUPMC Children's Hospital of PittsburghPittsburghPA15201USA
| | - Zhenwei Gong
- Division of EndocrinologyUPMC Children's Hospital of PittsburghPittsburghPA15201USA
| | - Jerry Vockley
- Division of Medical GeneticsUPMC Children's Hospital of PittsburghPittsburghPA15201USA
| | - Edward V. Prochownik
- Division of Hematology/OncologyUPMC Children's Hospital of PittsburghPittsburghPA15201USA
- The Department of Microbiology and Molecular GeneticsUPMCPittsburghPA15201USA
- The Hillman Cancer Center of UPMC5115 Centre AvePittsburghPA15232USA
- The Pittsburgh Liver Research CenterUPMCPittsburghPA15224USA
| |
Collapse
|
25
|
Buziau AM, Oosterveer MH, Wouters K, Bos T, Tolan DR, Agius L, Ford BE, Cassiman D, Stehouwer CDA, Schalkwijk CG, Brouwers MCGJ. Hepatic glucokinase regulatory protein and carbohydrate response element binding protein attenuation reduce de novo lipogenesis but do not mitigate intrahepatic triglyceride accumulation in Aldob deficiency. Mol Metab 2024; 87:101984. [PMID: 38972375 PMCID: PMC11300931 DOI: 10.1016/j.molmet.2024.101984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 06/15/2024] [Accepted: 07/02/2024] [Indexed: 07/09/2024] Open
Abstract
OBJECTIVE Stable isotope studies have shown that hepatic de novo lipogenesis (DNL) plays an important role in the pathogenesis of intrahepatic lipid (IHL) deposition. Furthermore, previous research has demonstrated that fructose 1-phosphate (F1P) not only serves as a substrate for DNL, but also acts as a signalling metabolite that stimulates DNL from glucose. The aim of this study was to elucidate the mediators of F1P-stimulated DNL, with special focus on two key regulators of intrahepatic glucose metabolism, i.e., glucokinase regulatory protein (GKRP) and carbohydrate response element binding protein (ChREBP). METHODS Aldolase B deficient mice (Aldob-/-), characterized by hepatocellular F1P accumulation, enhanced DNL, and hepatic steatosis, were either crossed with GKRP deficient mice (Gckr-/-) or treated with short hairpin RNAs directed against hepatic ChREBP. RESULTS Aldob-/- mice showed higher rates of de novo palmitate synthesis from glucose when compared to wildtype mice (p < 0.001). Gckr knockout reduced de novo palmitate synthesis in Aldob-/- mice (p = 0.017), without affecting the hepatic mRNA expression of enzymes involved in DNL. In contrast, hepatic ChREBP knockdown normalized the hepatic mRNA expression levels of enzymes involved in DNL and reduced fractional DNL in Aldob-/- mice (p < 0.05). Of interest, despite downregulation of DNL in response to Gckr and ChREBP attenuation, no reduction in intrahepatic triglyceride levels was observed. CONCLUSIONS Both GKRP and ChREBP mediate F1P-stimulated DNL in aldolase B deficient mice. Further studies are needed to unravel the role of GKRP and hepatic ChREBP in regulating IHL accumulation in aldolase B deficiency.
Collapse
Affiliation(s)
- Amée M Buziau
- Department of Internal Medicine, Division of Endocrinology and Metabolic Disease, Maastricht University Medical Center+, Maastricht, the Netherlands; Department of Internal Medicine, CARIM, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, the Netherlands
| | - Maaike H Oosterveer
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands; Department of Laboratory Medicine, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Kristiaan Wouters
- Department of Internal Medicine, Division of Endocrinology and Metabolic Disease, Maastricht University Medical Center+, Maastricht, the Netherlands; Department of Internal Medicine, CARIM, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, the Netherlands
| | - Trijnie Bos
- Department of Laboratory Medicine, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Dean R Tolan
- Department of Biology, Boston University, Boston, MA, USA
| | - Loranne Agius
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
| | - Brian E Ford
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
| | - David Cassiman
- Department of Gastroenterology-Hepatology and Metabolic Center, University Hospital Leuven, Leuven, Belgium
| | - Coen D A Stehouwer
- Department of Internal Medicine, CARIM, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht University Medical Center+, Maastricht, the Netherlands
| | - Casper G Schalkwijk
- Department of Internal Medicine, Division of Endocrinology and Metabolic Disease, Maastricht University Medical Center+, Maastricht, the Netherlands; Department of Internal Medicine, CARIM, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, the Netherlands
| | - Martijn C G J Brouwers
- Department of Internal Medicine, Division of Endocrinology and Metabolic Disease, Maastricht University Medical Center+, Maastricht, the Netherlands; Department of Internal Medicine, CARIM, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, the Netherlands; Department of Internal Medicine, CARIM, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht University Medical Center+, Maastricht, the Netherlands.
| |
Collapse
|
26
|
Wang P, Xiao H, Wu T, Fu Q, Song X, Zhao Y, Li Y, Huang J, Song Z. Activation of skeletal carbohydrate-response element binding protein (ChREBP)-mediated de novo lipogenesis increases intramuscular fat content in chickens. ANIMAL NUTRITION (ZHONGGUO XU MU SHOU YI XUE HUI) 2024; 18:107-118. [PMID: 39091296 PMCID: PMC11292260 DOI: 10.1016/j.aninu.2024.04.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Revised: 04/10/2024] [Accepted: 04/10/2024] [Indexed: 08/04/2024]
Abstract
The intracellular lipids in muscle cells of farm animals play a crucial role in determining the overall intramuscular fat (IMF) content, which has a positive impact on meat quality. However, the mechanisms underlying the deposition of lipids in muscle cells of farm animals are not yet fully understood. The purpose of this study was to determine the roles of carbohydrate-response element binding protein (ChREBP) and fructose in IMF deposition of chickens. For virus-mediated ChREBP overexpression in tibialis anterior (TA) muscle of chickens, seven 5-d-old male yellow-feather chickens were used. At 10 d after virus injection, the chickens were slaughtered to obtain TA muscles for analysis. For fructose administration trial, sixty 9-wk-old male yellow-feather chickens were randomly divided into 2 groups, with 6 replicates per group and 5 chickens per replicate. The chickens were fed either a basal diet or a basal diet supplemented with 10% fructose (purity ≥ 99%). At 4 wk later, the chickens were slaughtered, and breast and thigh muscles were collected for analysis. The results showed that the skeletal ChREBP mRNA levels were positively associated with IMF content in multiple species, including the chickens, pigs, and mice (P < 0.05). ChREBP overexpression increased lipid accumulation in both muscle cells in vitro and the TA muscles of mice and chickens in vivo (P < 0.05), by activation of the de novo lipogenesis (DNL) pathway. Moreover, activation of ChREBP by dietary fructose administration also resulted in increased IMF content in mice and notably chickens (P < 0.05). Furthermore, the lipidomics analysis revealed that ChREBP activation altered the lipid composition of chicken IMF and tented to improve the flavor profile of the meat. In conclusion, this study found that ChREBP plays a pivotal role in mediating the deposition of fat in chicken muscles in response to fructose-rich diets, which provides a novel strategy for improving meat quality in the livestock industry.
Collapse
Affiliation(s)
- Peng Wang
- Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, College of Animal Science and Technology, Guangxi University, Nanning, Guangxi 530004, China
| | - Haihan Xiao
- Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, College of Animal Science and Technology, Guangxi University, Nanning, Guangxi 530004, China
| | - Tian Wu
- Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, College of Animal Science and Technology, Guangxi University, Nanning, Guangxi 530004, China
| | - Qinghua Fu
- Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, College of Animal Science and Technology, Guangxi University, Nanning, Guangxi 530004, China
| | - Xudong Song
- Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, College of Animal Science and Technology, Guangxi University, Nanning, Guangxi 530004, China
| | - Yameng Zhao
- Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, College of Animal Science and Technology, Guangxi University, Nanning, Guangxi 530004, China
| | - Yan Li
- Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, College of Animal Science and Technology, Guangxi University, Nanning, Guangxi 530004, China
| | - Jieping Huang
- Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, College of Animal Science and Technology, Guangxi University, Nanning, Guangxi 530004, China
| | - Ziyi Song
- Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, College of Animal Science and Technology, Guangxi University, Nanning, Guangxi 530004, China
| |
Collapse
|
27
|
Algül KF, Şekerler T, Şen A, Gülmez G, Şener A. Effects of Crataegus orientalis fruit extract on lipid accumulation in oleic acid-induced HepG2 cells. ADVANCES IN TRADITIONAL MEDICINE 2024; 24:923-933. [DOI: 10.1007/s13596-024-00750-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 04/08/2024] [Indexed: 01/03/2025]
|
28
|
Baek S, Seo DS, Kang J, Ahmad Y, Park S, Joo S, Kim K, Jang Y. ChREBP plays a pivotal role in the nutrient-mediated regulation of metabolic gene expression in brown adipose tissue. Life Sci 2024; 351:122843. [PMID: 38880168 DOI: 10.1016/j.lfs.2024.122843] [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: 02/02/2024] [Revised: 05/27/2024] [Accepted: 06/12/2024] [Indexed: 06/18/2024]
Abstract
AIMS Carbohydrate-responsive element-binding protein (ChREBP) is a transcription factor that regulates several metabolic genes, including the lipogenic enzymes necessary for the metabolic conversion of carbohydrates into lipids. Although the crucial role of ChREBP in the liver, the primary site of de novo lipogenesis, has been studied, its functional role in adipose tissues, particularly brown adipose tissue (BAT), remains unclear. In this study, we investigated the role of ChREBP in BAT under conditions of a high-carbohydrate diet (HCD) and ketogenic diet (KD), represented by extremely low carbohydrate intake. MAIN METHODS Using an adeno-associated virus and Cas9 knock-in mice, we rapidly generated Chrebp brown adipocyte-specific knock-out (B-KO) mice, bypassing the necessity for prolonged breeding by using the Cre-Lox system. KEY FINDINGS We demonstrated that ChREBP is essential for glucose metabolism and lipogenic gene expression in BAT under HCD conditions in Chrebp B-KO mice. After nutrient intake, Chrebp B-KO attenuated the KD-induced expression of several inflammatory genes in BAT. SIGNIFICANCE Our results indicated that ChREBP, a nutrient-sensing regulator, is indispensable for expressing a diverse range of metabolic genes in BAT.
Collapse
Affiliation(s)
- Seungwoo Baek
- Department of Biology and Chemistry, Changwon National University, Changwon, Republic of Korea
| | - Dong Soo Seo
- Department of Biology and Chemistry, Changwon National University, Changwon, Republic of Korea
| | - Jaehyeon Kang
- Department of Biology and Chemistry, Changwon National University, Changwon, Republic of Korea
| | - Yusra Ahmad
- Department of Biology and Chemistry, Changwon National University, Changwon, Republic of Korea
| | - Sungjun Park
- Department of Biology and Chemistry, Changwon National University, Changwon, Republic of Korea
| | - Sungmin Joo
- Department of Biology and Chemistry, Changwon National University, Changwon, Republic of Korea
| | - KyeongJin Kim
- Department of Biomedical Sciences, College of Medicine, Inha University, Incheon, Republic of Korea
| | - Younghoon Jang
- Department of Biology and Chemistry, Changwon National University, Changwon, Republic of Korea.
| |
Collapse
|
29
|
Tsukamoto R, Watanabe K, Kodaka M, Iwase M, Sakiyama H, Inoue Y, Suzuki T, Yamamoto Y, Shimizu M, Sato R, Inoue J. HNF4α is required for Tkfc promoter activation by ChREBP. Biosci Biotechnol Biochem 2024; 88:941-947. [PMID: 38782732 DOI: 10.1093/bbb/zbae067] [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: 04/03/2024] [Accepted: 05/09/2024] [Indexed: 05/25/2024]
Abstract
Triokinase/FMN cyclase (Tkfc) is involved in fructose metabolism and is responsible for the phosphorylation of glyceraldehyde to glyceraldehyde-3-phosphate. In this study, we showed that refeeding induced hepatic expression of Tkfc in mice. Luciferase reporter gene assays using the Tkfc promoter revealed the existence of 2 hepatocyte nuclear factor 4α (HNF4α)-responsive elements (HNF4RE1 and HNF4RE2) and 1 carbohydrate-responsive element-binding protein (ChREBP)-responsive element (ChoRE1). Deletion and mutation of HNF4RE1 and HNF4RE2 or ChoRE1 abolished HNF4α and ChREBP responsiveness, respectively. HNF4α and ChREBP synergistically stimulated Tkfc promoter activity. ChoRE1 mutation attenuated but maintained HNF4α responsiveness, whereas HNF4RE1 and HNF4RE2 mutations abolished ChREBP responsiveness. Moreover, Tkfc promoter activity stimulation by ChREBP was attenuated upon HNF4α knockdown. Furthermore, Tkfc expression was decreased in the livers of ChREBP-/- and liver-specific HNF4-/- (Hnf4αΔHep) mice. Altogether, our data indicate that Tkfc is a target gene of ChREBP and HNF4α, and Tkfc promoter activity stimulation by ChREBP requires HNF4α.
Collapse
Affiliation(s)
- Rena Tsukamoto
- D epartment of Agricultural Chemistry, Faculty of Applied Biosciences, Tokyo University of Agriculture, Tokyo, Japan
| | - Kyoko Watanabe
- D epartment of Agricultural Chemistry, Faculty of Applied Biosciences, Tokyo University of Agriculture, Tokyo, Japan
| | - Manami Kodaka
- D epartment of Agricultural Chemistry, Faculty of Applied Biosciences, Tokyo University of Agriculture, Tokyo, Japan
| | - Masamori Iwase
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | | | - Yusuke Inoue
- Division of Molecular Science, Graduate School of Science and Technology, Gunma University, Gunma, Japan
| | - Tsukasa Suzuki
- D epartment of Agricultural Chemistry, Faculty of Applied Biosciences, Tokyo University of Agriculture, Tokyo, Japan
| | - Yuji Yamamoto
- D epartment of Agricultural Chemistry, Faculty of Applied Biosciences, Tokyo University of Agriculture, Tokyo, Japan
| | - Makoto Shimizu
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Ryuichiro Sato
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Jun Inoue
- D epartment of Agricultural Chemistry, Faculty of Applied Biosciences, Tokyo University of Agriculture, Tokyo, Japan
| |
Collapse
|
30
|
Lee JA, Gu MJ, Lee YR, Kim Y, Choi I, Kim D, Ha SK. Lindera obtusiloba Blume Alleviates Non-Alcoholic Fatty Liver Disease Promoted by N ε-(carboxymethyl)lysine. Nutrients 2024; 16:2330. [PMID: 39064772 PMCID: PMC11280000 DOI: 10.3390/nu16142330] [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: 06/27/2024] [Revised: 07/15/2024] [Accepted: 07/17/2024] [Indexed: 07/28/2024] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a major issue because it is closely associated with metabolic diseases. Advanced glycation end products (AGEs) are implicated as risk factors for steatosis during NAFLD progression. AGEs influence NAFLD progression through a receptor-independent pathway involving AGE cross-link formation and a receptor-dependent pathway that binds to receptors like receptors for advanced glycation end products (RAGE). The objectives of this study are to examine the effect of Lindera obtusiloba Blume (LO) on NAFLD promoted by Nε-(carboxymethyl)lysine (CML), one of the most common dietary AGEs. The anti-glycation effects of LO were evaluated by inhibiting the AGEs formation and AGEs-collagen cross-links breaking. The efficacy of LO against NAFLD promoted by CML was assessed using both in vitro and in vivo models. NAFLD was induced in mice by feeding a high-fat diet and orally administering CML over a period of 12 weeks, and the effects of LO on lipid metabolism and its regulatory mechanisms were investigated. LO showed the effect of inhibited AGEs formation and breakage, and collagen cross-linking. Fed a high-fat diet with administered CML by gavage, LO administration resulted in a reduction in body weight, fat mass, serum triglycerides, total cholesterol, and low-density lipoprotein cholesterol levels. LO reduced hepatic CML accumulation and RAGE expression in mice fed a high-fat diet and orally administered CML. LO alleviated hepatic steatosis accompanied by lipid accumulation and histological damage by suppressing the expression of sterol regulatory element-binding protein 1c, carbohydrate response element binding protein, fatty acid synthase, stearoyl-CoA desaturase1, tumor necrosis factor-α, and interleukin-1β. LO alleviated the MAPK/NF-κB expression by attenuating CML and RAGE expression. Taken together, our results demonstrate that LO alleviates the progression of NAFLD by lowering the levels of AGEs by downregulating CML/RAGE expression.
Collapse
Affiliation(s)
- Jin-Ah Lee
- Korea Food Research Institute, 245, Nongsaengmyeong-ro, Iseo-myeon, Wanju-gun 55365, Republic of Korea; (J.-A.L.); (M.J.G.); (Y.R.L.); (Y.K.); (I.C.)
| | - Min Ji Gu
- Korea Food Research Institute, 245, Nongsaengmyeong-ro, Iseo-myeon, Wanju-gun 55365, Republic of Korea; (J.-A.L.); (M.J.G.); (Y.R.L.); (Y.K.); (I.C.)
| | - Yu Ra Lee
- Korea Food Research Institute, 245, Nongsaengmyeong-ro, Iseo-myeon, Wanju-gun 55365, Republic of Korea; (J.-A.L.); (M.J.G.); (Y.R.L.); (Y.K.); (I.C.)
| | - Yoonsook Kim
- Korea Food Research Institute, 245, Nongsaengmyeong-ro, Iseo-myeon, Wanju-gun 55365, Republic of Korea; (J.-A.L.); (M.J.G.); (Y.R.L.); (Y.K.); (I.C.)
| | - Inwook Choi
- Korea Food Research Institute, 245, Nongsaengmyeong-ro, Iseo-myeon, Wanju-gun 55365, Republic of Korea; (J.-A.L.); (M.J.G.); (Y.R.L.); (Y.K.); (I.C.)
| | - Donghwan Kim
- Korea Food Research Institute, 245, Nongsaengmyeong-ro, Iseo-myeon, Wanju-gun 55365, Republic of Korea; (J.-A.L.); (M.J.G.); (Y.R.L.); (Y.K.); (I.C.)
| | - Sang Keun Ha
- Korea Food Research Institute, 245, Nongsaengmyeong-ro, Iseo-myeon, Wanju-gun 55365, Republic of Korea; (J.-A.L.); (M.J.G.); (Y.R.L.); (Y.K.); (I.C.)
- Division of Food Biotechnology, University of Science and Technology, Daejeon 34113, Republic of Korea
| |
Collapse
|
31
|
Jamerson LE, Bradshaw PC. The Roles of White Adipose Tissue and Liver NADPH in Dietary Restriction-Induced Longevity. Antioxidants (Basel) 2024; 13:820. [PMID: 39061889 PMCID: PMC11273496 DOI: 10.3390/antiox13070820] [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: 05/29/2024] [Revised: 07/01/2024] [Accepted: 07/03/2024] [Indexed: 07/28/2024] Open
Abstract
Dietary restriction (DR) protocols frequently employ intermittent fasting. Following a period of fasting, meal consumption increases lipogenic gene expression, including that of NADPH-generating enzymes that fuel lipogenesis in white adipose tissue (WAT) through the induction of transcriptional regulators SREBP-1c and CHREBP. SREBP-1c knockout mice, unlike controls, did not show an extended lifespan on the DR diet. WAT cytoplasmic NADPH is generated by both malic enzyme 1 (ME1) and the pentose phosphate pathway (PPP), while liver cytoplasmic NADPH is primarily synthesized by folate cycle enzymes provided one-carbon units through serine catabolism. During the daily fasting period of the DR diet, fatty acids are released from WAT and are transported to peripheral tissues, where they are used for beta-oxidation and for phospholipid and lipid droplet synthesis, where monounsaturated fatty acids (MUFAs) may activate Nrf1 and inhibit ferroptosis to promote longevity. Decreased WAT NADPH from PPP gene knockout stimulated the browning of WAT and protected from a high-fat diet, while high levels of NADPH-generating enzymes in WAT and macrophages are linked to obesity. But oscillations in WAT [NADPH]/[NADP+] from feeding and fasting cycles may play an important role in maintaining metabolic plasticity to drive longevity. Studies measuring the WAT malate/pyruvate as a proxy for the cytoplasmic [NADPH]/[NADP+], as well as studies using fluorescent biosensors expressed in the WAT of animal models to monitor the changes in cytoplasmic [NADPH]/[NADP+], are needed during ad libitum and DR diets to determine the changes that are associated with longevity.
Collapse
Affiliation(s)
| | - Patrick C. Bradshaw
- Department of Biomedical Sciences, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA
| |
Collapse
|
32
|
Zhang D, Zhao Y, Zhang G, Lank D, Cooke S, Wang S, Nuotio-Antar A, Tong X, Yin L. Suppression of hepatic ChREBP⍺-CYP2C50 axis-driven fatty acid oxidation sensitizes mice to diet-induced MASLD/MASH. Mol Metab 2024; 85:101957. [PMID: 38740087 PMCID: PMC11145360 DOI: 10.1016/j.molmet.2024.101957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Revised: 05/03/2024] [Accepted: 05/09/2024] [Indexed: 05/16/2024] Open
Abstract
OBJECTIVES Compromised hepatic fatty acid oxidation (FAO) has been observed in human MASH patients and animal models of MASLD/MASH. It remains poorly understood how and when the hepatic FAO pathway is suppressed during the progression of MASLD towards MASH. Hepatic ChREBP⍺ is a classical lipogenic transcription factor that responds to the intake of dietary sugars. METHODS We examined its role in regulating hepatocyte fatty acid oxidation (FAO) and the impact of hepatic Chrebpa deficiency on sensitivity to diet-induced MASLD/MASH in mice. RESULTS We discovered that hepatocyte ChREBP⍺ is both necessary and sufficient to maintain FAO in a cell-autonomous manner independently of its DNA-binding activity. Supplementation of synthetic PPAR⍺/δ agonist is sufficient to restore FAO in Chrebp-/- primary mouse hepatocytes. Hepatic ChREBP⍺ was decreased in mouse models of diet-induced MAFSLD/MASH and in patients with MASH. Hepatocyte-specific Chrebp⍺ knockout impaired FAO, aggravated liver steatosis and inflammation, leading to early-onset fibrosis in response to diet-induced MASH. Conversely, liver overexpression of ChREBP⍺-WT or its non-lipogenic mutant enhanced FAO, reduced lipid deposition, and alleviated liver injury, inflammation, and fibrosis. RNA-seq analysis identified the CYP450 epoxygenase (CYP2C50) pathway of arachidonic acid metabolism as a novel target of ChREBP⍺. Over-expression of CYP2C50 partially restores hepatic FAO in primary hepatocytes with Chrebp⍺ deficiency and attenuates preexisting MASH in the livers of hepatocyte-specific Chrebp⍺-deleted mice. CONCLUSIONS Our findings support the protective role of hepatocyte ChREBPa against diet-induced MASLD/MASH in mouse models in part via promoting CYP2C50-driven FAO.
Collapse
Affiliation(s)
- Deqiang Zhang
- Department of Molecular & Integrative Physiology, USA; Caswell Diabetes Institute, University of Michigan Medical School, NCRC Building 20-3843, 2800 Plymouth Road, Ann Arbor, MI 48105, USA
| | - Yuee Zhao
- Department of Molecular & Integrative Physiology, USA; Caswell Diabetes Institute, University of Michigan Medical School, NCRC Building 20-3843, 2800 Plymouth Road, Ann Arbor, MI 48105, USA; Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, The Second Xiangya Hospital, Central South University, 139 Renmin Middle Rd, Furong District, Changsha, Hunan Province 410011, PR China
| | - Gary Zhang
- Department of Molecular & Integrative Physiology, USA; Caswell Diabetes Institute, University of Michigan Medical School, NCRC Building 20-3843, 2800 Plymouth Road, Ann Arbor, MI 48105, USA
| | - Daniel Lank
- Department of Pharmacology, University of Virginia, 1340 Jefferson Park Avenue, Charlottesville, VA 22908, USA
| | - Sarah Cooke
- Neurosciences Graduate Program, Case Western Reserve University School of Medicine, Cleveland, OH 44016, USA
| | - Sujuan Wang
- Department of Infectious Diseases, The Second Xiangya Hospital, Central South University, 139 Renmin Middle Rd, Furong District, Changsha, Hunan Province 410011, PR China
| | - Alli Nuotio-Antar
- Children Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Xin Tong
- Department of Molecular & Integrative Physiology, USA; Caswell Diabetes Institute, University of Michigan Medical School, NCRC Building 20-3843, 2800 Plymouth Road, Ann Arbor, MI 48105, USA
| | - Lei Yin
- Department of Molecular & Integrative Physiology, USA; Caswell Diabetes Institute, University of Michigan Medical School, NCRC Building 20-3843, 2800 Plymouth Road, Ann Arbor, MI 48105, USA.
| |
Collapse
|
33
|
Khan F, Elsori D, Verma M, Pandey S, Obaidur Rab S, Siddiqui S, Alabdallah NM, Saeed M, Pandey P. Unraveling the intricate relationship between lipid metabolism and oncogenic signaling pathways. Front Cell Dev Biol 2024; 12:1399065. [PMID: 38933330 PMCID: PMC11199418 DOI: 10.3389/fcell.2024.1399065] [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: 03/11/2024] [Accepted: 05/28/2024] [Indexed: 06/28/2024] Open
Abstract
Lipids, the primary constituents of the cell membrane, play essential roles in nearly all cellular functions, such as cell-cell recognition, signaling transduction, and energy provision. Lipid metabolism is necessary for the maintenance of life since it regulates the balance between the processes of synthesis and breakdown. Increasing evidence suggests that cancer cells exhibit abnormal lipid metabolism, significantly affecting their malignant characteristics, including self-renewal, differentiation, invasion, metastasis, and drug sensitivity and resistance. Prominent oncogenic signaling pathways that modulate metabolic gene expression and elevate metabolic enzyme activity include phosphoinositide 3-kinase (PI3K)/AKT, MAPK, NF-kB, Wnt, Notch, and Hippo pathway. Conversely, when metabolic processes are not regulated, they can lead to malfunctions in cellular signal transduction pathways. This, in turn, enables uncontrolled cancer cell growth by providing the necessary energy, building blocks, and redox potentials. Therefore, targeting lipid metabolism-associated oncogenic signaling pathways could be an effective therapeutic approach to decrease cancer incidence and promote survival. This review sheds light on the interactions between lipid reprogramming and signaling pathways in cancer. Exploring lipid metabolism as a target could provide a promising approach for creating anticancer treatments by identifying metabolic inhibitors. Additionally, we have also provided an overview of the drugs targeting lipid metabolism in cancer in this review.
Collapse
Affiliation(s)
- Fahad Khan
- Center for Global Health Research, Saveetha Medical College and Hospital, Saveetha Institute of Medical and Technical Sciences, Chennai, Tamil Nadu, India
| | - Deena Elsori
- Faculty of Resilience, Rabdan Academy, Abu Dhabi, United Arab Emirates
| | - Meenakshi Verma
- University Centre for Research and Development, Chandigarh University, Mohali, Punjab, India
| | - Shivam Pandey
- School of Applied and Life Sciences, Uttaranchal University, Dehradun, Uttarakhand, India
| | - Safia Obaidur Rab
- Department of Clinical Laboratory Sciences, College of Applied Medical Science, King Khalid University, Abha, Saudi Arabia
| | - Samra Siddiqui
- Department of Health Service Management, College of Public Health and Health Informatics, University of Hail, Haʼil, Saudi Arabia
| | - Nadiyah M. Alabdallah
- Department of Biology, College of Science, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
- Basic and Applied Scientific Research Centre, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Mohd Saeed
- Department of Biology, College of Science, University of Hail, Haʼil, Saudi Arabia
| | - Pratibha Pandey
- Chitkara Centre for Research and Development, Chitkara University, Himachal Pradesh, India
- Centre of Research Impact and Outcome, Chitkara University, Rajpura, Punjab, India
| |
Collapse
|
34
|
Bo T, Gao L, Yao Z, Shao S, Wang X, Proud CG, Zhao J. Hepatic selective insulin resistance at the intersection of insulin signaling and metabolic dysfunction-associated steatotic liver disease. Cell Metab 2024; 36:947-968. [PMID: 38718757 DOI: 10.1016/j.cmet.2024.04.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 01/22/2024] [Accepted: 04/09/2024] [Indexed: 06/26/2024]
Abstract
Insulin resistance (IR) is a major pathogenic factor in the progression of MASLD. In the liver, insulin suppresses gluconeogenesis and enhances de novo lipogenesis (DNL). During IR, there is a defect in insulin-mediated suppression of gluconeogenesis, but an unrestrained increase in hepatic lipogenesis persists. The mechanism of increased hepatic steatosis in IR is unclear and remains controversial. The key discrepancy is whether insulin retains its ability to directly regulate hepatic lipogenesis. Blocking insulin/IRS/AKT signaling reduces liver lipid deposition in IR, suggesting insulin can still regulate lipid metabolism; hepatic glucose metabolism that bypasses insulin's action may contribute to lipogenesis; and due to peripheral IR, other tissues are likely to impact liver lipid deposition. We here review the current understanding of insulin's action in governing different aspects of hepatic lipid metabolism under normal and IR states, with the purpose of highlighting the essential issues that remain unsettled.
Collapse
Affiliation(s)
- Tao Bo
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China; Central Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China; Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Ling Gao
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China; Central Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China; Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Jinan, Shandong, China
| | - Zhenyu Yao
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China; Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Jinan, Shandong, China
| | - Shanshan Shao
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China; Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Jinan, Shandong, China
| | - Xuemin Wang
- Lifelong Health, South Australian Health & Medical Research Institute, North Terrace, Adelaide, SA, Australia
| | - Christopher G Proud
- Lifelong Health, South Australian Health & Medical Research Institute, North Terrace, Adelaide, SA, Australia.
| | - Jiajun Zhao
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China; Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Jinan, Shandong, China.
| |
Collapse
|
35
|
Taylor BC, Steinthal LH, Dias M, Yalamanchili HK, Ochsner SA, Zapata GE, Mehta NR, McKenna NJ, Young NL, Nuotio-Antar AM. Histone proteoform analysis reveals epigenetic changes in adult mouse brown adipose tissue in response to cold stress. Epigenetics Chromatin 2024; 17:12. [PMID: 38678237 PMCID: PMC11055387 DOI: 10.1186/s13072-024-00536-8] [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: 02/09/2024] [Accepted: 04/09/2024] [Indexed: 04/29/2024] Open
Abstract
BACKGROUND Regulation of the thermogenic response by brown adipose tissue (BAT) is an important component of energy homeostasis with implications for the treatment of obesity and diabetes. Our preliminary analyses of RNA-Seq data uncovered many nodes representing epigenetic modifiers that are altered in BAT in response to chronic thermogenic activation. Thus, we hypothesized that chronic thermogenic activation broadly alters epigenetic modifications of DNA and histones in BAT. RESULTS Motivated to understand how BAT function is regulated epigenetically, we developed a novel method for the first-ever unbiased top-down proteomic quantitation of histone modifications in BAT and validated our results with a multi-omic approach. To test our hypothesis, wildtype male C57BL/6J mice were housed under chronic conditions of thermoneutral temperature (TN, 28°C), mild cold/room temperature (RT, 22°C), or severe cold (SC, 8°C) and BAT was analyzed for DNA methylation and histone modifications. Methylation of promoters and intragenic regions in genomic DNA decrease in response to chronic cold exposure. Integration of DNA methylation and RNA expression datasets suggest a role for epigenetic modification of DNA in regulation of gene expression in response to cold. In response to cold housing, we observe increased bulk acetylation of histones H3.2 and H4, increased histone H3.2 proteoforms with di- and trimethylation of lysine 9 (K9me2 and K9me3), and increased histone H4 proteoforms with acetylation of lysine 16 (K16ac) in BAT. CONCLUSIONS Our results reveal global epigenetically-regulated transcriptional "on" and "off" signals in murine BAT in response to varying degrees of chronic cold stimuli and establish a novel methodology to quantitatively study histones in BAT, allowing for direct comparisons to decipher mechanistic changes during the thermogenic response. Additionally, we make histone PTM and proteoform quantitation, RNA splicing, RRBS, and transcriptional footprint datasets available as a resource for future research.
Collapse
Affiliation(s)
- Bethany C Taylor
- Verna and Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, TX, USA
| | - Loic H Steinthal
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Division of Nutrition, Baylor College of Medicine, Houston, TX, USA
| | - Michelle Dias
- Department of Pediatrics, Division of Neurology, Baylor College of Medicine, Houston, TX, USA
| | - Hari Krishna Yalamanchili
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Division of Nutrition, Baylor College of Medicine, Houston, TX, USA
- Department of Pediatrics, Division of Neurology, Baylor College of Medicine, Houston, TX, USA
- Jan and Dan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA
| | - Scott A Ochsner
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Gladys E Zapata
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Division of Nutrition, Baylor College of Medicine, Houston, TX, USA
| | - Nitesh R Mehta
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Division of Nutrition, Baylor College of Medicine, Houston, TX, USA
| | - Neil J McKenna
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Nicolas L Young
- Verna and Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, TX, USA.
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA.
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, TX, USA.
| | - Alli M Nuotio-Antar
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Division of Nutrition, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
36
|
Aging Biomarker Consortium, Jiang M, Zheng Z, Wang X, Chen Y, Qu J, Ding Q, Zhang W, Liu YS, Yang J, Tang W, Hou Y, He J, Wang L, Huang P, Li LC, He Z, Gao Q, Lu Q, Wei L, Wang YJ, Ju Z, Fan JG, Ruan XZ, Guan Y, Liu GH, Pei G, Li J, Wang Y. A biomarker framework for liver aging: the Aging Biomarker Consortium consensus statement. LIFE MEDICINE 2024; 3:lnae004. [PMID: 39872390 PMCID: PMC11749002 DOI: 10.1093/lifemedi/lnae004] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 01/29/2024] [Indexed: 01/11/2025]
Abstract
In human aging, liver aging per se not only increases susceptibility to liver diseases but also increases vulnerability of other organs given its central role in regulating metabolism. Total liver function tends to be well maintained in the healthy elderly, so liver aging is generally difficult to identify early. In response to this critical challenge, the Aging Biomarker Consortium of China has formulated an expert consensus on biomarkers of liver aging by synthesizing the latest scientific literature, comprising insights from both scientists and clinicians. This consensus provides a comprehensive assessment of biomarkers associated with liver aging and presents a systematic framework to characterize these into three dimensions: functional, imaging, and humoral. For the functional domain, we highlight biomarkers associated with cholesterol metabolism and liver-related coagulation function. For the imaging domain, we note that hepatic steatosis and liver blood flow can serve as measurable biomarkers for liver aging. Finally, in the humoral domain, we pinpoint hepatokines and enzymatic alterations worthy of attention. The aim of this expert consensus is to establish a foundation for assessing the extent of liver aging and identify early signs of liver aging-related diseases, thereby improving liver health and the healthy life expectancy of the elderly population.
Collapse
Affiliation(s)
| | - Mengmeng Jiang
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China
| | - Zhuozhao Zheng
- Department of Radiology, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing 102218, China
| | - Xuan Wang
- Hepatopancreatobiliary Center, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing 102218, China
| | - Yanhao Chen
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Jing Qu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Qiurong Ding
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Weiqi Zhang
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing 100101, China
| | - You-Shuo Liu
- Department of Geriatrics, the Second Xiangya Hospital, and the Institute of Aging and Geriatrics, Central South University, Changsha 410011, China
| | - Jichun Yang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Center for Non-coding RNA Medicine, Peking University Health Science Center, Beijing 100191, China
| | - Weiqing Tang
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing 100730, China
| | - Yunlong Hou
- Yiling Pharmaceutical Academician Workstation, Shijiazhuang 050035, China
| | - Jinhan He
- Department of Pharmacy, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Lin Wang
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an 710032, China
| | - Pengyu Huang
- State Key Laboratory of Advanced Medical Materials and Devices, Engineering Research Center of Pulmonary and Critical Care Medicine Technology and Device (Ministry of Education), Institute of Biomedical Engineering, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin 300192, China
| | - Lin-Chen Li
- Clinical Translational Science Center, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing 102218, China
| | - Zhiying He
- Institute for Regenerative Medicine, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai Engineering Research Center of Stem Cells Translational Medicine, Shanghai 200092, China
| | - Qiang Gao
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Qian Lu
- Hepatopancreatobiliary Center, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing 102218, China
- Key Laboratory of Digital Intelligence Hepatology (Ministry of Education), School of Clinical Medicine, Tsinghua University, Beijing 102218, China
| | - Lai Wei
- Hepatopancreatobiliary Center, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing 102218, China
| | - Yan-Jiang Wang
- Department of Neurology, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - Zhenyu Ju
- Key Laboratory of Regenerative Medicine of Ministry of Education, Institute of Aging and Regenerative Medicine, Jinan University, Guangzhou 510632, China
| | - Jian-Gao Fan
- Department of Gastroenterology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Xiong Zhong Ruan
- Centre for Lipid Research & Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, the Second Affiliated Hospital, Chongqing Medical University, Chongqing 400016, China
| | - Youfei Guan
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian 116044, China
| | - Guang-Hui Liu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Gang Pei
- Collaborative Innovation Center for Brain Science, School of Life Science and Technology, Tongji University, Shanghai 200092, China
| | - Jian Li
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing 100730, China
| | - Yunfang Wang
- Hepatopancreatobiliary Center, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing 102218, China
- Clinical Translational Science Center, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing 102218, China
- Key Laboratory of Digital Intelligence Hepatology (Ministry of Education), School of Clinical Medicine, Tsinghua University, Beijing 102218, China
- Research Unit of Precision Hepatobiliary Surgery Paradigm, Chinese Academy of Medical Sciences, Beijing 102218, China
| |
Collapse
|
37
|
Kuriyama T, Ishibashi C, Kozawa J, Baden MY, Horii T, Niki A, Ozawa H, Hosokawa Y, Fujita Y, Sadahiro K, Satoh T, Hamaguchi T, Shimomura I. Effects of liraglutide on intrapancreatic fat deposition in patients with type 2 diabetes. Clin Nutr ESPEN 2024; 59:208-213. [PMID: 38220377 DOI: 10.1016/j.clnesp.2023.12.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 12/01/2023] [Accepted: 12/05/2023] [Indexed: 01/16/2024]
Abstract
BACKGROUND & AIMS Ectopic fat deposition is associated with worsening of glycemic control. This study was conducted to determine whether liraglutide reduces ectopic fat deposition, especially in pancreas, in patients with type 2 diabetes (T2D). METHODS We retrospectively recruited T2D patients who underwent abdominal unenhanced CT scans both before and after administration of liraglutide (N = 13) or glimepiride (N = 29). Using CT values of pancreas (P), liver (L) and spleen (S), we defined the indices of intrapancreatic and liver fat as P-S value and L-S value, respectively. Increase of each value suggests the reduction of each fat deposition. RESULTS The values of HbA1c (p = 0.0017) and body weight (p = 0.0081) decreased, and L-S (p = 0.0024) increased significantly after administration of liraglutide compared with those at baseline. Similarly, P-S tended to increase in the liraglutide group (p = 0.0547) and increased significantly in the liraglutide subgroup with fatty pancreas (p = 0.0303), defined as having baseline P-S less than -5. In the glimepiride group, P-S did not increase regardless of baseline P-S. Among patients with fatty pancreas, administration of liraglutide tended to be a significant factor for the change in P-S after adjustment for the change in HbA1c (p = 0.1090) and the change in visceral fat area (p = 0.1030). CONCLUSIONS Intrapancreatic fat deposition was decreased after treatment with liraglutide, but not glimepiride, in T2D patients with fatty pancreas. Liraglutide might reduce intrapancreatic fat deposition independently of decreases in HbA1c and visceral fat volume.
Collapse
Affiliation(s)
- Tsukasa Kuriyama
- Division of Diabetes, Department of Internal Medicine, Itami City Hospital, Itami, Hyogo, Japan; Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Chisaki Ishibashi
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Junji Kozawa
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Suita, Japan; Department of Diabetes Care Medicine, Graduate School of Medicine, Osaka University, Suita, Japan.
| | - Megu Y Baden
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Suita, Japan; Department of Lifestyle Medicine, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Tomomi Horii
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Akiko Niki
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Harutoshi Ozawa
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Suita, Japan; Department of Diabetes Care Medicine, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Yoshiya Hosokawa
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Yukari Fujita
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Katsuhiko Sadahiro
- Division of Diabetes, Department of Internal Medicine, Itami City Hospital, Itami, Hyogo, Japan
| | - Tomomi Satoh
- Division of Diabetes, Department of Internal Medicine, Itami City Hospital, Itami, Hyogo, Japan
| | - Tomoya Hamaguchi
- Division of Diabetes, Department of Internal Medicine, Itami City Hospital, Itami, Hyogo, Japan
| | - Iichiro Shimomura
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Suita, Japan
| |
Collapse
|
38
|
Taylor BC, Steinthal LH, Dias M, Yalamanchili HK, Ochsner SA, Zapata GE, Mehta NR, McKenna NJ, Young NL, Nuotio-Antar AM. Histone proteoform analysis reveals epigenetic changes in adult mouse brown adipose tissue in response to cold stress. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.07.30.551059. [PMID: 38328142 PMCID: PMC10849524 DOI: 10.1101/2023.07.30.551059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2024]
Abstract
Regulation of the thermogenic response by brown adipose tissue (BAT) is an important component of energy homeostasis with implications for the treatment of obesity and diabetes. Our preliminary analyses uncovered many nodes representing epigenetic modifiers that are altered in BAT in response to chronic thermogenic activation. Thus, we hypothesized that chronic thermogenic activation broadly alters epigenetic modifications of DNA and histones in BAT. Motivated to understand how BAT function is regulated epigenetically, we developed a novel method for the first-ever unbiased top-down proteomic quantitation of histone modifications in BAT and validated our results with a multi-omic approach. To test our hypothesis, wildtype male C57BL/6J mice were housed under chronic conditions of thermoneutral temperature (TN, 28.8°C), mild cold/room temperature (RT, 22°C), or severe cold (SC, 8°C) and BAT was analyzed for DNA methylation and histone modifications. Methylation of promoters and intragenic regions in genomic DNA decrease in response to chronic cold exposure. Integration of DNA methylation and RNA expression data suggest a role for epigenetic modification of DNA in gene regulation in response to cold. In response to cold housing, we observe increased bulk acetylation of histones H3.2 and H4, increased histone H3.2 proteoforms with di- and trimethylation of lysine 9 (K9me2 and K9me3), and increased histone H4 proteoforms with acetylation of lysine 16 (K16ac) in BAT. Taken together, our results reveal global epigenetically-regulated transcriptional "on" and "off" signals in murine BAT in response to varying degrees of chronic cold stimuli and establish a novel methodology to quantitatively study histones in BAT, allowing for direct comparisons to decipher mechanistic changes during the thermogenic response. Additionally, we make histone PTM and proteoform quantitation, RNA splicing, RRBS, and transcriptional footprint datasets available as a resource for future research.
Collapse
Affiliation(s)
- Bethany C. Taylor
- Verna and Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, TX
| | - Loic H. Steinthal
- Children’s Nutrition Research Center, Baylor College of Medicine, Houston, TX
| | - Michelle Dias
- Jan and Dan Duncan Neurological Research Institute, Baylor College of Medicine, Houston, TX
| | - Hari K. Yalamanchili
- Children’s Nutrition Research Center, Baylor College of Medicine, Houston, TX
- Jan and Dan Duncan Neurological Research Institute, Baylor College of Medicine, Houston, TX
| | - Scott A. Ochsner
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX
| | - Gladys E. Zapata
- Children’s Nutrition Research Center, Baylor College of Medicine, Houston, TX
| | - Nitesh R. Mehta
- Children’s Nutrition Research Center, Baylor College of Medicine, Houston, TX
| | - Neil J. McKenna
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX
| | - Nicolas L. Young
- Verna and Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, TX
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, TX
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX
| | | |
Collapse
|
39
|
Rabbani N, Thornalley PJ. Hexokinase-linked glycolytic overload and unscheduled glycolysis in hyperglycemia-induced pathogenesis of insulin resistance, beta-cell glucotoxicity, and diabetic vascular complications. Front Endocrinol (Lausanne) 2024; 14:1268308. [PMID: 38292764 PMCID: PMC10824962 DOI: 10.3389/fendo.2023.1268308] [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: 07/27/2023] [Accepted: 12/12/2023] [Indexed: 02/01/2024] Open
Abstract
Hyperglycemia is a risk factor for the development of insulin resistance, beta-cell glucotoxicity, and vascular complications of diabetes. We propose the hypothesis, hexokinase-linked glycolytic overload and unscheduled glycolysis, in explanation. Hexokinases (HKs) catalyze the first step of glucose metabolism. Increased flux of glucose metabolism through glycolysis gated by HKs, when occurring without concomitant increased activity of glycolytic enzymes-unscheduled glycolysis-produces increased levels of glycolytic intermediates with overspill into effector pathways of cell dysfunction and pathogenesis. HK1 is saturated with glucose in euglycemia and, where it is the major HK, provides for basal glycolytic flux without glycolytic overload. HK2 has similar saturation characteristics, except that, in persistent hyperglycemia, it is stabilized to proteolysis by high intracellular glucose concentration, increasing HK activity and initiating glycolytic overload and unscheduled glycolysis. This drives the development of vascular complications of diabetes. Similar HK2-linked unscheduled glycolysis in skeletal muscle and adipose tissue in impaired fasting glucose drives the development of peripheral insulin resistance. Glucokinase (GCK or HK4)-linked glycolytic overload and unscheduled glycolysis occurs in persistent hyperglycemia in hepatocytes and beta-cells, contributing to hepatic insulin resistance and beta-cell glucotoxicity, leading to the development of type 2 diabetes. Downstream effector pathways of HK-linked unscheduled glycolysis are mitochondrial dysfunction and increased reactive oxygen species (ROS) formation; activation of hexosamine, protein kinase c, and dicarbonyl stress pathways; and increased Mlx/Mondo A signaling. Mitochondrial dysfunction and increased ROS was proposed as the initiator of metabolic dysfunction in hyperglycemia, but it is rather one of the multiple downstream effector pathways. Correction of HK2 dysregulation is proposed as a novel therapeutic target. Pharmacotherapy addressing it corrected insulin resistance in overweight and obese subjects in clinical trial. Overall, the damaging effects of hyperglycemia are a consequence of HK-gated increased flux of glucose metabolism without increased glycolytic enzyme activities to accommodate it.
Collapse
Affiliation(s)
| | - Paul J. Thornalley
- College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar
| |
Collapse
|
40
|
Mostofinejad Z, Cremonini E, Kang J, Oteiza PI. Effects of (-)-epicatechin on hepatic triglyceride metabolism. Food Funct 2024; 15:326-337. [PMID: 38086683 DOI: 10.1039/d3fo03666a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2024]
Abstract
(-)-Epicatechin (EC) consumption is associated with an improvement of hyperlipemia and other metabolic changes linked to obesity and western-style diets. This work investigated the effects of EC on triglyceride (TG) metabolism both in vivo, where mice were supplemented with EC (2 and 20 mg EC per kg body weight), and in vitro, when human HepG2 hepatocytes were incubated in the presence of EC and the main EC metabolites found in human plasma. Increased hepatic TG levels were only observed after 24 weeks supplementation with EC (20 mg per kg body weight), with a preserved liver structure and absence of inflammation or oxidative stress. EC caused increased expression of diacylglycerol acyltransferases (DGAT2), key enzymes in TG synthesis, and the upregulation of PPARα, which promotes free fatty acid (FFA) oxidation. On the other hand, incubation of HepG2 cells in the presence of high concentrations of EC (1-10 μM) did not affect TG deposition nor DGAT2 expression. In summary, in mouse liver, EC upregulated mechanisms that can neutralize the potential toxicity of FFA, i.e. TG synthesis and FFA β-oxidation. Results in mouse liver and HepG2 cells stress the safety of EC in terms of TG metabolism and development of hepatopathies in doses within the limits given by a rational time and dose for human consumption.
Collapse
Affiliation(s)
- Zahra Mostofinejad
- Department of Nutrition, University of California, One Shields Avenue, Davis, CA 95616, USA.
| | - Eleonora Cremonini
- Department of Nutrition, University of California, One Shields Avenue, Davis, CA 95616, USA.
| | - Jiye Kang
- Department of Nutrition, University of California, One Shields Avenue, Davis, CA 95616, USA.
| | - Patricia I Oteiza
- Department of Nutrition, University of California, One Shields Avenue, Davis, CA 95616, USA.
- Department of Environmental Toxicology, University of California, Davis, USA
| |
Collapse
|
41
|
Singh C, Jin B, Shrestha N, Markhard AL, Panda A, Calvo SE, Deik A, Pan X, Zuckerman AL, Ben Saad A, Corey KE, Sjoquist J, Osganian S, AminiTabrizi R, Rhee EP, Shah H, Goldberger O, Mullen AC, Cracan V, Clish CB, Mootha VK, Goodman RP. ChREBP is activated by reductive stress and mediates GCKR-associated metabolic traits. Cell Metab 2024; 36:144-158.e7. [PMID: 38101397 PMCID: PMC10842884 DOI: 10.1016/j.cmet.2023.11.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 07/24/2023] [Accepted: 11/21/2023] [Indexed: 12/17/2023]
Abstract
Common genetic variants in glucokinase regulator (GCKR), which encodes GKRP, a regulator of hepatic glucokinase (GCK), influence multiple metabolic traits in genome-wide association studies (GWASs), making GCKR one of the most pleiotropic GWAS loci in the genome. It is unclear why. Prior work has demonstrated that GCKR influences the hepatic cytosolic NADH/NAD+ ratio, also referred to as reductive stress. Here, we demonstrate that reductive stress is sufficient to activate the transcription factor ChREBP and necessary for its activation by the GKRP-GCK interaction, glucose, and ethanol. We show that hepatic reductive stress induces GCKR GWAS traits such as increased hepatic fat, circulating FGF21, and circulating acylglycerol species, which are also influenced by ChREBP. We define the transcriptional signature of hepatic reductive stress and show its upregulation in fatty liver disease and downregulation after bariatric surgery in humans. These findings highlight how a GCKR-reductive stress-ChREBP axis influences multiple human metabolic traits.
Collapse
Affiliation(s)
- Charandeep Singh
- Liver Center, Division of Gastroenterology, Massachusetts General Hospital, Boston, MA 02114, USA; Endocrine Unit, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Byungchang Jin
- Liver Center, Division of Gastroenterology, Massachusetts General Hospital, Boston, MA 02114, USA; Endocrine Unit, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Nirajan Shrestha
- Liver Center, Division of Gastroenterology, Massachusetts General Hospital, Boston, MA 02114, USA; Endocrine Unit, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Andrew L Markhard
- Howard Hughes Medical Institute and Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Apekshya Panda
- Howard Hughes Medical Institute and Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Sarah E Calvo
- Howard Hughes Medical Institute and Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Amy Deik
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Xingxiu Pan
- The Scintillon Institute, San Diego, CA 92121, USA
| | - Austin L Zuckerman
- The Scintillon Institute, San Diego, CA 92121, USA; Program in Mathematics and Science Education, University of California, San Diego, La Jolla, CA 92093; Program in Mathematics and Science Education, San Diego State University, San Diego, CA 92120
| | - Amel Ben Saad
- Division of Gastroenterology, University of Massachusetts Chan Medical School, Worcester, MA 01655, USA
| | - Kathleen E Corey
- Liver Center, Division of Gastroenterology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Julia Sjoquist
- Liver Center, Division of Gastroenterology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Stephanie Osganian
- Liver Center, Division of Gastroenterology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Roya AminiTabrizi
- Metabolomics Platform, Comprehensive Cancer Center, the University of Chicago, Chicago, IL 60637, USA
| | - Eugene P Rhee
- Endocrine Unit, Massachusetts General Hospital, Boston, MA 02114, USA; Nephrology Division, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Hardik Shah
- Metabolomics Platform, Comprehensive Cancer Center, the University of Chicago, Chicago, IL 60637, USA
| | - Olga Goldberger
- Howard Hughes Medical Institute and Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Alan C Mullen
- Division of Gastroenterology, University of Massachusetts Chan Medical School, Worcester, MA 01655, USA
| | - Valentin Cracan
- The Scintillon Institute, San Diego, CA 92121, USA; Department of Chemistry, the Scripps Research Institute, La Jolla, CA 92037, USA
| | - Clary B Clish
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Vamsi K Mootha
- Howard Hughes Medical Institute and Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Russell P Goodman
- Liver Center, Division of Gastroenterology, Massachusetts General Hospital, Boston, MA 02114, USA; Endocrine Unit, Massachusetts General Hospital, Boston, MA 02114, USA.
| |
Collapse
|
42
|
Krishnamurthy KA, Rutten MGS, Hoogerland JA, van Dijk TH, Bos T, Koehorst M, de Vries MP, Kloosterhuis NJ, Havinga H, Schomakers BV, van Weeghel M, Wolters JC, Bakker BM, Oosterveer MH. Hepatic ChREBP orchestrates intrahepatic carbohydrate metabolism to limit hepatic glucose 6-phosphate and glycogen accumulation in a mouse model for acute Glycogen Storage Disease type Ib. Mol Metab 2024; 79:101838. [PMID: 37995884 PMCID: PMC10716006 DOI: 10.1016/j.molmet.2023.101838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 11/13/2023] [Indexed: 11/25/2023] Open
Abstract
OBJECTIVE Carbohydrate Response Element Binding Protein (ChREBP) is a glucose 6-phosphate (G6P)-sensitive transcription factor that acts as a metabolic switch to maintain intracellular glucose and phosphate homeostasis. Hepatic ChREBP is well-known for its regulatory role in glycolysis, the pentose phosphate pathway, and de novo lipogenesis. The physiological role of ChREBP in hepatic glycogen metabolism and blood glucose regulation has not been assessed in detail, and ChREBP's contribution to carbohydrate flux adaptations in hepatic Glycogen Storage Disease type 1 (GSD I) requires further investigation. METHODS The current study aimed to investigate the role of ChREBP as a regulator of glycogen metabolism in response to hepatic G6P accumulation, using a model for acute hepatic GSD type Ib. The immediate biochemical and regulatory responses to hepatic G6P accumulation were evaluated upon G6P transporter inhibition by the chlorogenic acid S4048 in mice that were either treated with a short hairpin RNA (shRNA) directed against ChREBP (shChREBP) or a scrambled shRNA (shSCR). Complementary stable isotope experiments were performed to quantify hepatic carbohydrate fluxes in vivo. RESULTS ShChREBP treatment normalized the S4048-mediated induction of hepatic ChREBP target genes to levels observed in vehicle- and shSCR-treated controls. In parallel, hepatic shChREBP treatment in S4048-infused mice resulted in a more pronounced accumulation of hepatic glycogen and further reduction of blood glucose levels compared to shSCR treatment. Hepatic ChREBP knockdown modestly increased glucokinase (GCK) flux in S4048-treated mice while it enhanced UDP-glucose turnover as well as glycogen synthase and phosphorylase fluxes. Hepatic GCK mRNA and protein levels were induced by shChREBP treatment in both vehicle- and S4048-treated mice, while glycogen synthase 2 (GYS2) and glycogen phosphorylase (PYGL) mRNA and protein levels were reduced. Finally, knockdown of hepatic ChREBP expression reduced starch domain binding protein 1 (STBD1) mRNA and protein levels while it inhibited acid alpha-glucosidase (GAA) activity, suggesting reduced capacity for lysosomal glycogen breakdown. CONCLUSIONS Our data show that ChREBP activation controls hepatic glycogen and blood glucose levels in acute hepatic GSD Ib through concomitant regulation of glucose phosphorylation, glycogenesis, and glycogenolysis. ChREBP-mediated control of GCK enzyme levels aligns with corresponding adaptations in GCK flux. In contrast, ChREBP activation in response to acute hepatic GSD Ib exerts opposite effects on GYS2/PYGL enzyme levels and their corresponding fluxes, indicating that GYS2/PYGL expression levels are not limiting to their respective fluxes under these conditions.
Collapse
Affiliation(s)
- K A Krishnamurthy
- Laboratory of Pediatrics, University of Groningen, University Medical Center Groningen, The Netherlands
| | - M G S Rutten
- Laboratory of Pediatrics, University of Groningen, University Medical Center Groningen, The Netherlands
| | - J A Hoogerland
- Laboratory of Pediatrics, University of Groningen, University Medical Center Groningen, The Netherlands
| | - T H van Dijk
- Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, The Netherlands
| | - T Bos
- Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, The Netherlands
| | - M Koehorst
- Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, The Netherlands
| | - M P de Vries
- Laboratory of Pediatrics, University of Groningen, University Medical Center Groningen, The Netherlands; Interfaculty Mass Spectrometry Center, University of Groningen, University Medical Center Groningen, The Netherlands
| | - N J Kloosterhuis
- Laboratory of Pediatrics, University of Groningen, University Medical Center Groningen, The Netherlands
| | - H Havinga
- Laboratory of Pediatrics, University of Groningen, University Medical Center Groningen, The Netherlands
| | - B V Schomakers
- Laboratory Genetic Metabolic Diseases, UMC Amsterdam, The Netherlands; Core Facility Metabolomics, UMC Amsterdam, The Netherlands
| | - M van Weeghel
- Laboratory Genetic Metabolic Diseases, UMC Amsterdam, The Netherlands; Core Facility Metabolomics, UMC Amsterdam, The Netherlands
| | - J C Wolters
- Laboratory of Pediatrics, University of Groningen, University Medical Center Groningen, The Netherlands; Interfaculty Mass Spectrometry Center, University of Groningen, University Medical Center Groningen, The Netherlands
| | - B M Bakker
- Laboratory of Pediatrics, University of Groningen, University Medical Center Groningen, The Netherlands
| | - M H Oosterveer
- Laboratory of Pediatrics, University of Groningen, University Medical Center Groningen, The Netherlands; Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, The Netherlands.
| |
Collapse
|
43
|
Sun Q, Xing X, Wang H, Wan K, Fan R, Liu C, Wang Y, Wu W, Wang Y, Wang R. SCD1 is the critical signaling hub to mediate metabolic diseases: Mechanism and the development of its inhibitors. Biomed Pharmacother 2024; 170:115586. [PMID: 38042113 DOI: 10.1016/j.biopha.2023.115586] [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: 07/23/2023] [Revised: 09/23/2023] [Accepted: 09/25/2023] [Indexed: 12/04/2023] Open
Abstract
Metabolic diseases, featured with dysregulated energy homeostasis, have become major global health challenges. Patients with metabolic diseases have high probability to manifest multiple complications in lipid metabolism, e.g. obesity, insulin resistance and fatty liver. Therefore, targeting the hub genes in lipid metabolism may systemically ameliorate the metabolic diseases, along with the complications. Stearoyl-CoA desaturase 1(SCD1) is a key enzyme that desaturates the saturated fatty acids (SFAs) derived from de novo lipogenesis or diet to generate monounsaturated fatty acids (MUFAs). SCD1 maintains the metabolic and tissue homeostasis by responding to, and integrating the multiple layers of endogenous stimuli, which is mediated by the synthesized MUFAs. It critically regulates a myriad of physiological processes, including energy homeostasis, development, autophagy, tumorigenesis and inflammation. Aberrant transcriptional and epigenetic activation of SCD1 regulates AMPK/ACC, SIRT1/PGC1α, NcDase/Wnt, etc, and causes aberrant lipid accumulation, thereby promoting the progression of obesity, non-alcoholic fatty liver, diabetes and cancer. This review critically assesses the integrative mechanisms of the (patho)physiological functions of SCD1 in metabolic homeostasis, inflammation and autophagy. For translational perspective, potent SCD1 inhibitors have been developed to treat various types of cancer. We thus discuss the multidisciplinary advances that greatly accelerate the development of SCD1 new inhibitors. In conclusion, besides cancer treatment, SCD1 may serve as the promising target to combat multiple metabolic complications simultaneously.
Collapse
Affiliation(s)
- Qin Sun
- School of Kinesiology, Shanghai University of Sport, Shanghai 200438, China
| | - Xiaorui Xing
- School of Kinesiology, Shanghai University of Sport, Shanghai 200438, China
| | - Huanyu Wang
- School of Kinesiology, Shanghai University of Sport, Shanghai 200438, China
| | - Kang Wan
- School of Kinesiology, Shanghai University of Sport, Shanghai 200438, China
| | - Ruobing Fan
- School of Kinesiology, Shanghai University of Sport, Shanghai 200438, China
| | - Cheng Liu
- School of Kinesiology, Shanghai University of Sport, Shanghai 200438, China
| | - Yongjian Wang
- School of Kinesiology, Shanghai University of Sport, Shanghai 200438, China
| | - Wenyi Wu
- School of Kinesiology, Shanghai University of Sport, Shanghai 200438, China
| | - Yibing Wang
- School of Kinesiology, Shanghai University of Sport, Shanghai 200438, China.
| | - Ru Wang
- School of Kinesiology, Shanghai University of Sport, Shanghai 200438, China.
| |
Collapse
|
44
|
Wang B, Pu R. Association between glycolysis markers and prognosis of liver cancer: a systematic review and meta-analysis. World J Surg Oncol 2023; 21:390. [PMID: 38114977 PMCID: PMC10731852 DOI: 10.1186/s12957-023-03275-4] [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: 07/13/2023] [Accepted: 12/08/2023] [Indexed: 12/21/2023] Open
Abstract
BACKGROUND In recent years, the capacity of tumor cells to maintain high levels of glycolysis, even in the presence of oxygen, has emerged as one of the main metabolic traits and garnered considerable attention. The purpose of this meta-analysis is to investigate the prognostic value of glycolysis markers in liver cancer. METHODS PubMed, Embase, and Cochrane Library databases were searched for articles on glycolytic marker expression levels associated with the prognosis of liver cancer until April 2023. Stata SE14.0 was used to calculate the aggregate hazard ratios and 95% confidence intervals. RESULTS Thirty-five studies were included. The worse overall survival (OS) (P < 0.001), disease-free survival (DFS) (P = 0.001), recurrence-free survival (RFS) (P = 0.004), and time to recurrence (TTR) (P < 0.001) were significantly associated with elevated expression of glycolysis markers. Higher expression of PKM2 (P < 0.001), STMN1 (P = 0.002), MCT4 (P < 0.001), GLUT1 (P = 0.025), HK-2 (P < 0.001), and CA9 (P < 0.001) were significantly related to shorter OS. Increased levels of PKM2 (P < 0.001), CA9 (P = 0.005), and MCT4 (P < 0.001) were associated with worse DFS. Elevated PKM2 expression (P = 0.002) was also associated with poorer RFS in hepatocellular carcinoma patients. GLUT2 expression was not correlated with the prognosis of liver cancer (P = 0.134). CONCLUSIONS Elevated expression of glycolysis markers was associated with worse OS, DFS, RFS, and TTR in patients with liver cancer. Therefore, these glycolysis markers could serve as potential prognostic markers and therapeutic targets in liver cancer. TRIAL REGISTRATION PROSPERO registration: CRD42023469645.
Collapse
Affiliation(s)
- Boqin Wang
- Department of Clinical Laboratory, SSL Central Hospital of Dongguan City, Dongguan, Guangdong, China
| | - Rong Pu
- Department of Clinical Laboratory, SSL Central Hospital of Dongguan City, Dongguan, Guangdong, China.
| |
Collapse
|
45
|
Park YJ, Kim HY, Shin S, Lee J, Heo I, Cha YY, An HJ. Anti-obesity effect of Lythri herba water extracts in vitro and in vivo. JOURNAL OF ETHNOPHARMACOLOGY 2023; 317:116789. [PMID: 37328083 DOI: 10.1016/j.jep.2023.116789] [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: 02/16/2023] [Revised: 06/07/2023] [Accepted: 06/13/2023] [Indexed: 06/18/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Lythrum salicaria L., also called purple loosestrife, has traditionally been used as a medicinal plant to treat internal dysfunction, such as gastrointestinal disorders or hemorrhages. It contains numerous phytochemical compounds, including orientin, and has been reported to have anti-diarrheal, anti-inflammatory, antioxidant, and antimicrobial properties. AIM OF THE STUDY The effects of Lythrum salicaria L. on obesity have not been explored. Therefore, we investigated the anti-obesity effects of Lythri Herba, the aerial part of this plant, in vitro and in vivo. MATERIALS AND METHODS Using distilled water, Lythri Herba water extracts (LHWE) were prepared by extracting Lythri Herba at 100°Ϲ. The contents of orientin in LHWE were identified using High Performance Liquid Chromatography (HPLC) analysis. To evaluate the anti-obesity effect of LHWE, 3T3-L1 adipocytes and a high-fat diet (HFD)-fed mice were used. Oil-red O staining was performed to examine the anti-adipogenic effects of LHWE in vitro. The histological changes in epididymal white adipose tissue (epiWAT) by LHWE were examined using hematoxylin and eosin staining. Serum leptin levels were measured by enzyme-linked immunosorbent assay. Specific quantification kits measured total cholesterol and triglyceride levels in the serum. The relative fold induction of protein and mRNA was determined using western blot and Quantitative real-time Polymerase Chain Reaction analysis, respectively. RESULTS HPLC analysis demonstrated the presence of orientin in LHWE. LHWE treatment markedly reduced lipid accumulation in differentiated 3T3-L1 adipocytes. LHWE administration also conferred resistance to HFD-induced weight gain in mice and reduced epiWAT mass. Mechanistically, LHWE significantly decreased lipogenesis by downregulating lipoprotein lipase (LPL), glucose-6-phosphate dehydrogenase, ATP-citrate lyase, fatty acid synthase, stearoyl-CoA desaturase 1, sterol regulatory element binding transcription factor 1, and carbohydrate response element binding protein expression and increased the expression of genes involved in fatty acid oxidation (FAO), peroxisome proliferator-activated receptor α and carnitine palmitoyltransferase 1 in 3T3-L1 adipocytes and epiWAT. Furthermore, LHWE significantly up-regulated the phosphorylation of AMP-activated protein kinase in 3T3-L1 adipocytes and epiWAT. CONCLUSION LHWE decreases white adipogenesis in vitro and HFD-induced weight gain in vivo, which is associated with reduced lipogenesis and enhanced FAO.
Collapse
Affiliation(s)
- Yea-Jin Park
- Department of Rehabilitative Medicine of Korean Medicine and Neuropsychiatry, College of Korean Medicine, Sangji University, Wonju, Gangwon-do, 26339, Republic of Korea; Department of Oriental Pharmaceutical Science, College of Pharmacy, Kyung Hee University, Seoul, 02447, Republic of Korea.
| | - Hee-Young Kim
- Department of Oriental Pharmaceutical Science, College of Pharmacy, Kyung Hee University, Seoul, 02447, Republic of Korea; Department of Integrated Drug Development and Natural Products, Graduate School, Kyung Hee University, Seoul, 02447, Republic of Korea.
| | - Su Shin
- Research Institute, BIO PORT KOREA INC., 36, Ballyongsandan 1-ro, Jangan-eup, Gijang-gun, Busan, 46034, Republic of Korea.
| | - JungHyun Lee
- Research Institute, BIO PORT KOREA INC., 36, Ballyongsandan 1-ro, Jangan-eup, Gijang-gun, Busan, 46034, Republic of Korea.
| | - In Heo
- School of Korean Medicine, Pusan National University, Yangsan, Gyeongsangnam-do, Republic of Korea; Department of Rehabilitation Medicine of Korean Medicine, Pusan National University Korean Medicine Hospital, Yangsan, Gyeongsangnam-do, Republic of Korea.
| | - Yun-Yeop Cha
- Department of Rehabilitative Medicine of Korean Medicine and Neuropsychiatry, College of Korean Medicine, Sangji University, Wonju, Gangwon-do, 26339, Republic of Korea.
| | - Hyo-Jin An
- Department of Oriental Pharmaceutical Science, College of Pharmacy, Kyung Hee University, Seoul, 02447, Republic of Korea; Department of Integrated Drug Development and Natural Products, Graduate School, Kyung Hee University, Seoul, 02447, Republic of Korea.
| |
Collapse
|
46
|
Shi JH, Chen YX, Feng Y, Yang X, Lin J, Wang T, Wei CC, Ma XH, Yang R, Cao D, Zhang H, Xie X, Xie Z, Zhang WJ. Fructose overconsumption impairs hepatic manganese homeostasis and ammonia disposal. Nat Commun 2023; 14:7934. [PMID: 38040719 PMCID: PMC10692208 DOI: 10.1038/s41467-023-43609-0] [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: 03/28/2023] [Accepted: 11/15/2023] [Indexed: 12/03/2023] Open
Abstract
Arginase, a manganese (Mn)-dependent enzyme, is indispensable for urea generation and ammonia disposal in the liver. The potential role of fructose in Mn and ammonia metabolism is undefined. Here we demonstrate that fructose overconsumption impairs hepatic Mn homeostasis and ammonia disposal in male mice. Fructose overexposure reduces liver Mn content as well as its activity of arginase and Mn-SOD, and impairs the clearance of blood ammonia under liver dysfunction. Mechanistically, fructose activates the Mn exporter Slc30a10 gene transcription in the liver in a ChREBP-dependent manner. Hepatic overexpression of Slc30a10 can mimic the effect of fructose on liver Mn content and ammonia disposal. Hepatocyte-specific deletion of Slc30a10 or ChREBP increases liver Mn contents and arginase activity, and abolishes their responsiveness to fructose. Collectively, our data establish a role of fructose in hepatic Mn and ammonia metabolism through ChREBP/Slc30a10 pathway, and postulate fructose dietary restriction for the prevention and treatment of hyperammonemia.
Collapse
Affiliation(s)
- Jian-Hui Shi
- National Key Laboratory of Immunity & Inflammation and Department of Pathophysiology, Naval Medical University, Shanghai, China
| | - Yu-Xia Chen
- National Key Laboratory of Immunity & Inflammation and Department of Pathophysiology, Naval Medical University, Shanghai, China
| | - Yingying Feng
- National Key Laboratory of Immunity & Inflammation and Department of Pathophysiology, Naval Medical University, Shanghai, China
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, China
| | - Xiaohang Yang
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, China
| | - Jie Lin
- National Key Laboratory of Immunity & Inflammation and Department of Pathophysiology, Naval Medical University, Shanghai, China
| | - Ting Wang
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, China
| | - Chun-Chun Wei
- National Key Laboratory of Immunity & Inflammation and Department of Pathophysiology, Naval Medical University, Shanghai, China
| | - Xian-Hua Ma
- National Key Laboratory of Immunity & Inflammation and Department of Pathophysiology, Naval Medical University, Shanghai, China
| | - Rui Yang
- National Key Laboratory of Immunity & Inflammation and Department of Pathophysiology, Naval Medical University, Shanghai, China
| | - Dongmei Cao
- National Key Laboratory of Immunity & Inflammation and Department of Pathophysiology, Naval Medical University, Shanghai, China
| | - Hai Zhang
- National Key Laboratory of Immunity & Inflammation and Department of Pathophysiology, Naval Medical University, Shanghai, China
| | - Xiangyang Xie
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, China
| | - Zhifang Xie
- National Key Laboratory of Immunity & Inflammation and Department of Pathophysiology, Naval Medical University, Shanghai, China
| | - Weiping J Zhang
- National Key Laboratory of Immunity & Inflammation and Department of Pathophysiology, Naval Medical University, Shanghai, China.
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, China.
| |
Collapse
|
47
|
Rabadán-Chávez G, Díaz de la Garza RI, Jacobo-Velázquez DA. White adipose tissue: Distribution, molecular insights of impaired expandability, and its implication in fatty liver disease. Biochim Biophys Acta Mol Basis Dis 2023; 1869:166853. [PMID: 37611674 DOI: 10.1016/j.bbadis.2023.166853] [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: 04/15/2023] [Revised: 07/17/2023] [Accepted: 08/18/2023] [Indexed: 08/25/2023]
Abstract
We are far behind the 2025 World Health Organization (WHO) goal of a zero increase in obesity. Close to 360 million people in Latin America and the Caribbean are overweight, with the highest rates observed in the Bahamas, Mexico, and Chile. To achieve relevant progress against the obesity epidemic, scientific research is essential to establish uniform practices in the study of obesity pathophysiology (using pre-clinical and clinical models) that ensure accuracy, reproducibility, and transcendent outcomes. The present review focuses on relevant aspects of white adipose tissue (WAT) expansion, underlying mechanisms of inefficient expandability, and its repercussion in ectopic lipid accumulation in the liver during nutritional abundance. In addition, we highlight the potential role of disrupted circadian rhythm in WAT metabolism. Since genetic factors also play a key role in determining an individual's predisposition to weight gain, we describe the most relevant genes associated with obesity in the Mexican population, underlining that most of them are related to appetite control.
Collapse
Affiliation(s)
- Griselda Rabadán-Chávez
- Tecnologico de Monterrey, Institute for Obesity Research, Av. Eugenio Garza Sada 2501 Sur, 64849 Monterrey, NL, Mexico
| | - Rocío I Díaz de la Garza
- Tecnologico de Monterrey, Institute for Obesity Research, Av. Eugenio Garza Sada 2501 Sur, 64849 Monterrey, NL, Mexico; Tecnologico de Monterrey, Escuela de Ingeniería y Ciencias, Campus Monterrey, Av. Eugenio Garza Sada 2501 Sur, 64849 Monterrey, NL, Mexico.
| | - Daniel A Jacobo-Velázquez
- Tecnologico de Monterrey, Institute for Obesity Research, Av. Eugenio Garza Sada 2501 Sur, 64849 Monterrey, NL, Mexico; Tecnologico de Monterrey, Escuela de Ingeniería y Ciencias, Campus Guadalajara, Av. General Ramon Corona 2514, C.P. 45201 Zapopan, Jalisco, Mexico.
| |
Collapse
|
48
|
Gong Y, Lu Q, Xi L, Liu Y, Yang B, Su J, Liu H, Jin J, Zhang Z, Yang Y, Zhu X, Xie S, Han D. F6P/G6P-mediated ChREBP activation promotes the insulin resistance-driven hepatic lipid deposition in zebrafish. J Nutr Biochem 2023; 122:109452. [PMID: 37748621 DOI: 10.1016/j.jnutbio.2023.109452] [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: 10/25/2022] [Revised: 08/15/2023] [Accepted: 09/21/2023] [Indexed: 09/27/2023]
Abstract
Insulin-sensitive lipogenesis dominates the body lipid deposition; however, nonalcoholic fatty liver disease (NAFLD) develops in the insulin-resistant state. The regulation mechanism of insulin resistance-driven NAFLD remains elusive. Using zebrafish model of insulin resistance (ZIR, insrb-/-) and mouse hepatocytes (NCTC 1469), we explored the regulation mechanism of insulin resistance-driven hepatic lipid deposition under the stimulation of carbohydrate diet (CHD). In ZIR model, insulin resistance induced hyperlipidemia and elevated hepatic lipid deposition via elevating the gene/protein expressions of lipogenic enzymes, that was activated by carbohydrate response element binding protein (ChREBP), rather than sterol regulatory element binding proteins 1c (SREBP-1c). The metabolomic analysis in zebrafish and silencing of chrebp in mouse hepatocytes revealed that the increased hepatic frucotose-6-phosphate (F6P) and glucose-6-phosphate (G6P) promoted the ChREBP-mediated lipid deposition. We further identified that F6P alone was sufficient to activate ChREBP-mediated lipid deposition by a SREBP-1c-independent manner. Moreover, we clarified the suppressed hepatic phosphofructokinase/glucose-6-phosphatase functions and the normal glucokinase function preserved by glucose transporter 2 (GLUT2) manipulated the increased F6P/G6P content in ZIR. In conclusion, the present study revealed that insulin resistance promoted hepatic lipid deposition via the F6P/G6P-mediated ChREBP activation. Our findings deciphered the main regulation pathway for the liver lipid deposition in the insulin-resistant state and identified F6P as a new potential regulator for ChREBP.
Collapse
Affiliation(s)
- Yulong Gong
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
| | - Qisheng Lu
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China; College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Longwei Xi
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China; College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Yulong Liu
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China; College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Bingyuan Yang
- Department of Molecular Physiology & Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Jingzhi Su
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China; College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Haokun Liu
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
| | - Junyan Jin
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
| | - Zhimin Zhang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
| | - Yunxia Yang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
| | - Xiaoming Zhu
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
| | - Shouqi Xie
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China; College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, China; The Innovative Academy of Seed Design, Chinese Academy of Sciences, Wuhan, China
| | - Dong Han
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China; College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, China; Hubei Hongshan Laboratory, Wuhan, China.
| |
Collapse
|
49
|
Rao G, Peng X, Li X, An K, He H, Fu X, Li S, An Z. Unmasking the enigma of lipid metabolism in metabolic dysfunction-associated steatotic liver disease: from mechanism to the clinic. Front Med (Lausanne) 2023; 10:1294267. [PMID: 38089874 PMCID: PMC10711211 DOI: 10.3389/fmed.2023.1294267] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 10/26/2023] [Indexed: 06/21/2024] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD), formerly defined as non-alcoholic fatty liver disease (NAFLD), is a disorder marked by the excessive deposition of lipids in the liver, giving rise to a spectrum of liver pathologies encompassing steatohepatitis, fibrosis/cirrhosis, and hepatocellular carcinoma. Despite the alarming increase in its prevalence, the US Food and Drug Administration has yet to approve effective pharmacological therapeutics for clinical use. MASLD is characterized by the accretion of lipids within the hepatic system, arising from a disarray in lipid provision (whether through the absorption of circulating lipids or de novo lipogenesis) and lipid elimination (via free fatty acid oxidation or the secretion of triglyceride-rich lipoproteins). This disarray leads to the accumulation of lipotoxic substances, cellular pressure, damage, and fibrosis. Indeed, the regulation of the lipid metabolism pathway is intricate and multifaceted, involving a myriad of factors, such as membrane transport proteins, metabolic enzymes, and transcription factors. Here, we will review the existing literature on the key process of lipid metabolism in MASLD to understand the latest progress in this molecular mechanism. Notably, de novo lipogenesis and the roles of its two main transcription factors and other key metabolic enzymes are highlighted. Furthermore, we will delve into the realm of drug research, examining the recent progress made in understanding lipid metabolism in MASLD. Additionally, we will outline prospective avenues for future drug research on MASLD based on our unique perspectives.
Collapse
Affiliation(s)
- Guocheng Rao
- Department of Endocrinology and Metabolism, West China Hospital, Sichuan University, Chengdu, China
| | - Xi Peng
- Department of Endocrinology and Metabolism, West China Hospital, Sichuan University, Chengdu, China
- Department of Endocrinology and Metabolism, Affiliated Hospital of North Sichuan Medical College, North Sichuan Medical College, Nanchong, China
| | - Xinqiong Li
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, China
| | - Kang An
- General Practice Ward/International Medical Center Ward, General Practice Medical Center, National Clinical Research Center for Geriatrics, Multimorbidity Laboratory, West China Hospital, Sichuan University, Chengdu, China
| | - He He
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Xianghui Fu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, China
| | - Shuangqing Li
- General Practice Ward/International Medical Center Ward, General Practice Medical Center, National Clinical Research Center for Geriatrics, Multimorbidity Laboratory, West China Hospital, Sichuan University, Chengdu, China
| | - Zhenmei An
- Department of Endocrinology and Metabolism, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
50
|
Wang H, Stevens T, Lu J, Roberts A, Land CV, Muzumdar R, Gong Z, Vockley J, Prochownik EV. The Myc-Like Mlx Network Impacts Aging and Metabolism. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.26.568749. [PMID: 38076995 PMCID: PMC10705233 DOI: 10.1101/2023.11.26.568749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/23/2023]
Abstract
The "Mlx" and "Myc" Networks share many common gene targets. Just as Myc's activity depends upon its heterodimerization with Max, the Mlx Network requires that the Max-like factor Mlx associate with the Myc-like factors MondoA or ChREBP. We show here that body-wide Mlx inactivation, like that of Myc, accelerates numerous aging-related phenotypes pertaining to body habitus and metabolism. The deregulation of numerous aging-related Myc target gene sets is also accelerated. Among other functions, these gene sets often regulate ribosomal and mitochondrial structure and function, genomic stability and aging. Whereas "MycKO" mice have an extended lifespan because of a lower cancer incidence, "MlxKO" mice have normal lifespans and a somewhat higher cancer incidence. Like Myc, Mlx, MondoA and ChREBP expression and that of their target genes, deteriorate with age in both mice and humans, underscoring the importance of life-long and balanced cross-talk between the two Networks to maintain normal aging.
Collapse
Affiliation(s)
- Huabo Wang
- Division of Hematology/Oncology, UPMC Children’s Hospital of Pittsburgh
| | - Taylor Stevens
- Division of Hematology/Oncology, UPMC Children’s Hospital of Pittsburgh
| | - Jie Lu
- Division of Hematology/Oncology, UPMC Children’s Hospital of Pittsburgh
| | - Alexander Roberts
- Division of Hematology/Oncology, UPMC Children’s Hospital of Pittsburgh
| | | | - Radhika Muzumdar
- Division of Endocrinology, UPMC Children’s Hospital of Pittsburgh
| | - Zhenwei Gong
- Division of Endocrinology, UPMC Children’s Hospital of Pittsburgh
| | - Jerry Vockley
- Division of Medical Genetics, UPMC Children’s Hospital of Pittsburgh
| | - Edward V. Prochownik
- Division of Hematology/Oncology, UPMC Children’s Hospital of Pittsburgh
- The Department of Microbiology and Molecular Genetics, UPMC
- The Hillman Cancer Center of UPMC
- The Pittsburgh Liver Research Center, UPMC, Pittsburgh, PA. 15224
| |
Collapse
|