1
|
Guo D, Meng Y, Zhao G, Wu Q, Lu Z. Moonlighting functions of glucose metabolic enzymes and metabolites in cancer. Nat Rev Cancer 2025:10.1038/s41568-025-00800-3. [PMID: 40175621 DOI: 10.1038/s41568-025-00800-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/13/2025] [Indexed: 04/04/2025]
Abstract
Glucose metabolic enzymes and their metabolites not only provide energy and building blocks for synthesizing macromolecules but also possess non-canonical or moonlighting functions in response to extracellular and intracellular signalling. These moonlighting functions modulate various cellular activities, including gene expression, cell cycle progression, DNA repair, autophagy, senescence and apoptosis, cell proliferation, remodelling of the tumour microenvironment and immune responses. These functions integrate glucose metabolism with other essential cellular activities, driving cancer progression. Targeting these moonlighting functions could open new therapeutic avenues and lead to cancer-specific treatments.
Collapse
Affiliation(s)
- Dong Guo
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, China
| | - Ying Meng
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, China
| | - Gaoxiang Zhao
- Department of Oncology, Cancer Institute of The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, China
| | - Qingang Wu
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, China
| | - Zhimin Lu
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China.
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, China.
| |
Collapse
|
2
|
Luque-Urbano MR, Fernández-Ramos D, Lopitz-Otsoa F, Gutiérrez de Juan V, Bizkarguenaga M, Castro-Espadas L, Hermoso-Martínez U, Barbier-Torres L, Lu SC, Millet O, Mato JM. S-Adenosylmethionine Deficit Disrupts Very Low-Density Lipoprotein Metabolism Promoting Liver Lipid Accumulation in Mice. J Lipid Res 2025:100794. [PMID: 40180215 DOI: 10.1016/j.jlr.2025.100794] [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: 11/05/2024] [Revised: 03/20/2025] [Accepted: 03/29/2025] [Indexed: 04/05/2025] Open
Abstract
Hepatic deletion of methionine adenosyltransferase-1a (Mat1a) in mice reduces S-adenosylmethionine (SAMe), a key methyl donor essential for many biological processes, which promotes the development and progression of metabolic dysfunction-associated steatotic liver disease (MASLD). Hyperglycemia and reduced MAT1A expression, along with low SAMe levels, are common in MASLD patients. This study explores how Mat1a-knockout (KO) hepatocytes respond to prolonged high glucose conditions, focusing on glucose metabolism and lipid accumulation. Hepatocytes from Mat1a-KO mice were incubated in high glucose conditions overnight, allowing for analysis of key metabolic intermediates and gene expression related to glycolysis, gluconeogenesis, glyceroneogenesis, phospholipid synthesis, and very low-density lipoprotein (VLDL) secretion. SAMe deficiency in Mat1a-KO hepatocytes led to reduced protein methyltransferase-1 activity, resulting in increased expression of glycolytic enzymes (glucokinase, phosphofructokinase, and pyruvate kinase) and decreased expression of gluconeogenic enzymes (phosphoenolpyruvate carboxykinase, fructose-1,6-bisphosphatase, and glucose-6-phosphatase). These alterations led to a reduction in dihydroxyacetone phosphate (DHAP), which subsequently inhibited mammalian target of rapamycine complex 1 (mTORC1) activity. This inhibition resulted in decreased phosphatidylcholine synthesis via the CDP-choline pathway and impaired VLDL secretion, ultimately causing lipid accumulation. Thus, under high glucose conditions, SAMe deficiency in hepatocytes depletes DHAP, inhibits mTORC1 activity, and promotes lipid buildup.
Collapse
Affiliation(s)
- María R Luque-Urbano
- Atlas Molecular Pharma, 48160 Derio, Spain,; Precision Medicine and Metabolism Lab, CIC bioGUNE, Basque Research and Technology Alliance (BRTA), 48160 Derio, Spain
| | - David Fernández-Ramos
- Precision Medicine and Metabolism Lab, CIC bioGUNE, Basque Research and Technology Alliance (BRTA), 48160 Derio, Spain,; CIBERehd, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Fernando Lopitz-Otsoa
- Precision Medicine and Metabolism Lab, CIC bioGUNE, Basque Research and Technology Alliance (BRTA), 48160 Derio, Spain
| | - Virginia Gutiérrez de Juan
- Precision Medicine and Metabolism Lab, CIC bioGUNE, Basque Research and Technology Alliance (BRTA), 48160 Derio, Spain
| | - Maider Bizkarguenaga
- Precision Medicine and Metabolism Lab, CIC bioGUNE, Basque Research and Technology Alliance (BRTA), 48160 Derio, Spain
| | - Lia Castro-Espadas
- Precision Medicine and Metabolism Lab, CIC bioGUNE, Basque Research and Technology Alliance (BRTA), 48160 Derio, Spain
| | - Uxue Hermoso-Martínez
- Precision Medicine and Metabolism Lab, CIC bioGUNE, Basque Research and Technology Alliance (BRTA), 48160 Derio, Spain
| | - Lucía Barbier-Torres
- Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Shelly C Lu
- Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Oscar Millet
- Atlas Molecular Pharma, 48160 Derio, Spain,; Precision Medicine and Metabolism Lab, CIC bioGUNE, Basque Research and Technology Alliance (BRTA), 48160 Derio, Spain,; CIBERehd, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - José M Mato
- Precision Medicine and Metabolism Lab, CIC bioGUNE, Basque Research and Technology Alliance (BRTA), 48160 Derio, Spain,; CIBERehd, Instituto de Salud Carlos III, 28029 Madrid, Spain,.
| |
Collapse
|
3
|
Zhu Y, Wei D, Karin M, Gu L. The FBP1-TP53-NRF2 metabolic switch in metabolic dysfunction-associated steatohepatitis-hepatocellular carcinoma progression and senescence reversal. Clin Transl Med 2025; 15:e70293. [PMID: 40159462 PMCID: PMC11955276 DOI: 10.1002/ctm2.70293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2025] [Accepted: 03/22/2025] [Indexed: 04/02/2025] Open
Affiliation(s)
- Yahui Zhu
- School of MedicineChongqing UniversityChongqingChina
| | - Donglin Wei
- School of MedicineChongqing UniversityChongqingChina
| | - Michael Karin
- Departments of Pharmacology and PathologyLaboratory of Gene Regulation and Signal TransductionSchool of Medicine, University of California San Diego (UCSD)La JollaCaliforniaUSA
| | - Li Gu
- Department of Laboratory MedicineWest China Hospital, Sichuan UniversityChengduChina
- Clinical Laboratory Medicine Research CenterWest China Hospital, Sichuan UniversityChengduChina
| |
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
|
Lai P, Miao G, Zhao Y, Han Y, Li Y, Liu Y, Guo J, Zhang W, Guo X, Xu Y, Zhang L, Chen G, Zhou Z, Mei S, Chen J, Chen J, Xu L, Zhang C, Ding Y, Dou X, Wen S, Lam SM, Shui G, Wang Y, Huang W, Zhao D, Xian X. SR-A3 suppresses AKT activation to protect against MAFLD by inhibiting XIAP-mediated PTEN degradation. Nat Commun 2025; 16:2430. [PMID: 40069146 PMCID: PMC11897346 DOI: 10.1038/s41467-025-57585-0] [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: 04/22/2024] [Accepted: 02/26/2025] [Indexed: 03/15/2025] Open
Abstract
Scavenger receptor class A member 3 (SR-A3) is implicated in metabolic diseases; however, the relationship between SR-A3 and metabolic dysfunction-associated fatty liver disease (MAFLD) has not been documented. Here, we show that hepatic SR-A3 expression is significantly reduced in human and animal models in the context of MAFLD. Genetic inhibition of SR-A3 in hamsters elicits hyperlipidemia, hyperglycemia, insulin resistance, and hepatic steatosis under chow-diet condition, yet escalates in diet-induced MAFLD. Mechanistically, SR-A3 ablation enhances E3 ligase XIAP-mediated proteasomal ubiquitination of PTEN, leading to AKT hyperactivation. By contrast, hepatic overexpression of human SR-A3 is sufficient to attenuate metabolic disorders in WT hamsters fed a high-fat-high-cholesterol diet and ob/ob mice via suppressing the XIAP/PTEN/AKT axis. In parallel, pharmacological intervention by PTEN agonist oroxin B or lipid lowering agent ezetimibe differentially corrects MAFLD in hamsters.
Collapse
Affiliation(s)
- Pingping Lai
- Institute of Cardiovascular Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Guolin Miao
- Institute of Cardiovascular Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, School of Basic Medical Sciences, Peking University, Beijing, China
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, China
| | - Yinqi Zhao
- Institute of Cardiovascular Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Yufei Han
- Institute of Cardiovascular Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Yanwei Li
- Department of Infectious Diseases, Shengjing Hospital, China Medical University, Shenyang, China
| | - Yiran Liu
- Department of Biomedical Informatics, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Jiabao Guo
- Institute of Cardiovascular Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Wenxi Zhang
- Institute of Cardiovascular Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Xin Guo
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, China
| | - Yitong Xu
- Institute of Cardiovascular Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Lianxin Zhang
- Institute of Cardiovascular Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Gonglie Chen
- Institute of Cardiovascular Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Zihao Zhou
- Institute of Cardiovascular Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Si Mei
- Institute of Cardiovascular Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Jingxuan Chen
- Institute of Cardiovascular Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Jinxuan Chen
- Institute of Cardiovascular Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Luzheng Xu
- Peking University Medical and Health Analysis Center, Peking University, Beijing, China
| | - Chong Zhang
- Department of Infectious Diseases, Shengjing Hospital, China Medical University, Shenyang, China
| | - Yang Ding
- Department of Infectious Diseases, Shengjing Hospital, China Medical University, Shenyang, China
| | - Xiaoguang Dou
- Department of Infectious Diseases, Shengjing Hospital, China Medical University, Shenyang, China
| | - Shengmei Wen
- NGGT (Suzhou) Biotechnology Co. Ltd, Suzhou, China
| | - Sin Man Lam
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
- Lipidall Technologies Company Limited, Changzhou, 213022, Jiangsu Province, China
| | - Guanghou Shui
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
| | - Yuhui Wang
- Institute of Cardiovascular Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Wei Huang
- Institute of Cardiovascular Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Dongyu Zhao
- Department of Biomedical Informatics, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Xunde Xian
- Institute of Cardiovascular Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, School of Basic Medical Sciences, Peking University, Beijing, China.
- Beijing Key Laboratory of Cardiovascular Receptors Research, Peking University Third Hospital, Beijing, China.
| |
Collapse
|
6
|
Zhang F, Xia C, Yang G, Shang B, Huang G, Yuan K, Wang H, Gong X, Jiang Q. Multiomics analysis of human serum and animal experiments reveals the protective mechanism of Qingre Huoxue Decoction against rheumatoid arthritis. Front Immunol 2025; 16:1526110. [PMID: 40124380 PMCID: PMC11926152 DOI: 10.3389/fimmu.2025.1526110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Accepted: 02/17/2025] [Indexed: 03/25/2025] Open
Abstract
Objective Qingre Huoxue Decoction (QRHXD) is a traditional Chinese herbal prescription widely used in clinical practice with significant therapeutic effects on RA; however, its mechanism of action remains unclear. This study aimed to investigate the efficacy and underlying mechanisms of QRHXD in treating RA through clinical research, multiomics approaches, and animal experiments. Methods We conducted a 24-week clinical study in which QRHXD was the primary treatment, collecting serum samples from patients before and after treatment for integrated proteomic and metabolomic analysis to identify potential therapeutic targets. Bioinformatics analysis of differentially expressed proteins (DEPs) and differential metabolites (DMs) was performed using hierarchical clustering, volcano plots, heat maps, Gene Ontology (GO), and Kyoto Encyclopaedia of Genes and Genomes (KEGG) analysis. To validate the identified therapeutic targets, we constructed a collagen-induced arthritis (CIA) mouse model. Results Clinical research has shown that QRHXD can improve clinical symptoms and relevant indicators in RA patients, including the disease activity score-28 (DAS28), C-reactive protein (CRP), erythrocyte sedimentation rate (ESR), tender joint count (TJC), swollen joint count (SJC), visual analogue scale (VAS), patient-reported outcome (PRO), and health assessment questionnaire (HAQ). Proteomics and metabolomics analysis identified 83 DEPs and 54 DMs, including 46 upregulated and 37 downregulated proteins, as well as 11 upregulated and 43 downregulated metabolites. KEGG enrichment analysis revealed that DEPs are primarily associated with fatty acid degradation, ferroptosis, glycerolipid metabolism, and related pathways. The identified DMs are primarily associated with the AMPK signalling pathway, FoxO signalling pathway, glycolysis/gluconeogenesis, MTOR signalling pathway, and so on. GO enrichment analysis indicated that the DEPs were mainly associated with apoptotic mitochondrial changes, protein modification processes, fatty-acyl-CoA binding, and so on. Integrated proteomics and metabolomics analyses revealed a significant increase in fructose-1,6-biphosphatase 1 (FBP1) levels and a reduction in AMP-activated protein kinase (AMPK) levels in patients with RA. QRHXD inhibited FBP1 and activated AMPK signalling. Animal experiments validated the findings from proteomics and metabolomics analyses, demonstrating that QRHXD could also delay bone destruction and reduce inflammatory factor levels in CIA mice. Conclusion QRHXD may reduce the disease activity of RA, attenuate the inflammatory response, and delay bone destruction by inhibiting FBP1 and activating the AMPK signalling pathway.
Collapse
Affiliation(s)
- Fuyuan Zhang
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Congmin Xia
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Guang Yang
- Department of Chinese and Western Medicine, Peking University First Hospital, Beijing, China
| | - Biyue Shang
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Guangrui Huang
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Kai Yuan
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Hesong Wang
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Xun Gong
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Quan Jiang
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
7
|
Cai D, Zhong G, Dai X, Zhao Z, Chen M, Hu J, Wu Z, Cheng L, Li S, Gong J. Targeting FDFT1 Reduces Cholesterol and Bile Acid Production and Delays Hepatocellular Carcinoma Progression Through the HNF4A/ALDOB/AKT1 Axis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2411719. [PMID: 39899681 PMCID: PMC11948044 DOI: 10.1002/advs.202411719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 12/17/2024] [Indexed: 02/05/2025]
Abstract
Targeting cholesterol metabolism is a novel direction for tumor therapy. Unfortunately, the current use of statins for hepatocellular carcinoma (HCC) is controversial. Herein, farnesyl-diphosphate farnesyltransferase 1 (FDFT1) is identified as a novel target for treating HCC and a potential alternative to statins. Twenty-three key genes in cholesterol biosynthesis are screened, and FDFT1 is identified via public databases (The Cancer Genome Atlas, International Cancer Genome Consortium and Gene Expression Omnibus). Clinical samples reveal that FDFT1 is highly expressed in HCC tissues, and this phenotype is strongly associated with a poor prognosis. Functionally, FDFT1 knockdown inhibits the proliferation and metastasis of HCC cells and suppresses hepatocarcinogenesis in vitro and in vivo, whereas FDFT1 overexpression promotes HCC cell proliferation and metastasis. Mechanistically, FDFT1 downregulation decreases cholesterol and bile acid levels and then increases hepatocyte nuclear factor 4 alpha (HNF4A) transcriptional activity. Experiments indicate that HNF4A combines with the promoter of aldolase B (ALDOB) and promotes the ALDOB transcription and that ALDOB combines with AKT serine/threonine kinase 1 (AKT1) and inhibits AKT1 phosphorylation. Moreover, FDFT1 knockdown combined with AKT inhibitor (AZD5363) treatment shows remarkable therapeutic potential. FDFT1 inhibition reduces cholesterol and bile acid levels to delay HCC progression through the HNF4A/ALDOB/AKT1 axis. Thus, targeting FDFT1 may be a novel potential strategy for treating HCC.
Collapse
Affiliation(s)
- Dong Cai
- Department of Hepatobiliary SurgeryThe Second Affiliated Hospital of Chongqing Medical UniversityChongqing400010China
| | - Guo‐Chao Zhong
- Department of Hepatobiliary SurgeryThe Second Affiliated Hospital of Chongqing Medical UniversityChongqing400010China
| | - Xin Dai
- Department of Hepatobiliary SurgeryThe Second Affiliated Hospital of Chongqing Medical UniversityChongqing400010China
| | - Zhibo Zhao
- Department of Hepatobiliary SurgeryThe Second Affiliated Hospital of Chongqing Medical UniversityChongqing400010China
| | - Menglin Chen
- Institute of Clinical PathologyKey Laboratory of Transplant Engineering and ImmunologyNHCWest China HospitalSichuan UniversityChengduSichuan610041China
| | - Jiejun Hu
- Department of Hepatobiliary SurgeryThe Second Affiliated Hospital of Chongqing Medical UniversityChongqing400010China
| | - Zhenru Wu
- Institute of Clinical PathologyKey Laboratory of Transplant Engineering and ImmunologyNHCWest China HospitalSichuan UniversityChengduSichuan610041China
| | - Lve Cheng
- Department of Hepatobiliary SurgeryThe Second Affiliated Hospital of Chongqing Medical UniversityChongqing400010China
| | - Shengwei Li
- Department of Hepatobiliary SurgeryThe Second Affiliated Hospital of Chongqing Medical UniversityChongqing400010China
| | - Jianping Gong
- Department of Hepatobiliary SurgeryThe Second Affiliated Hospital of Chongqing Medical UniversityChongqing400010China
| |
Collapse
|
8
|
Li F, Gao T, Li Z, Dou H, Ba Y, Jia S, Luo D, Xiao M. Triglyceride-glucose index and triglyceride-glucose-body mass index as prognostic factors for early stage breast cancer patients receiving neoadjuvant chemotherapy. Transl Oncol 2025; 53:102292. [PMID: 39884219 PMCID: PMC11814653 DOI: 10.1016/j.tranon.2025.102292] [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: 08/07/2024] [Revised: 01/13/2025] [Accepted: 01/21/2025] [Indexed: 02/01/2025] Open
Abstract
BACKGROUND Insulin resistance (IR) is closely associated with the risk of breast cancer. The triglyceride-glucose (TyG) index and the triglyceride-glucose-body mass index (TyG-BMI) are considered surrogate indicators of IR; however, their prognostic value in breast cancer patients has not been discussed. The purpose of this study is not only to explore whether the TyG index and the TyG-BMI can predict the chemotherapy response and long-term prognosis of breast cancer patients receiving neoadjuvant chemotherapy (NACT) but also to investigate the possible mediating mechanism and to analyze the relationship between TyG-related enzyme expression and drug resistance and prognosis. METHODS From November 2011 to December 2018, a total of 335 breast cancer patients referred to Harbin Medical University Cancer Hospital who received NACT and surgery were registered in this retrospective study. The TyG index and TyG-BMI before the first chemotherapy were retrospectively calculated. Tissue samples of breast cancer patients were obtained from the Cancer Genome Atlas database, and the associations between the expression levels of the FBP1 and G6PD enzymes and the clinicopathological features and prognosis of breast cancer were analyzed. RESULTS In receiver operating characteristic analyses, the optimal cutoff values for the TyG and TyG-BMI were determined at 8.01 and 194.91, respectively. Low levels of the TyG and TyG-BMI were not associated with pathological complete response. In multivariate analysis, high TyG was an independent prognostic factor for shorter disease-free survival (DFS; HR = 2.402, P = 0.008) and overall survival (OS; HR = 3.206, P = 0.010). After adjustments for the age group, cT stage group, and cN stage group, the dose-response relationships between TyG, TyG-BMI, and survival outcomes showed a linear correlation by restricted cubic spline analyses. Lg-transformed BMI did not significantly (P > 0.05) mediate the recurrence, metastasis, and deaths associated with TyG. The expressions of two enzymes related to TyG, FBP1 and G6PD, were higher in breast cancer tissues than in the adjacent normal tissues and were associated with the TNM stage. Survival analysis shows that patients with high expressions of FBP1 and G6PD have a shorter OS. CONCLUSION This study suggests that the TyG index level before NACT is an independent prognostic factor for DFS and OS and can serve as a promising biomarker to predict the long-term prognosis of breast cancer patients undergoing NACT. Moreover, the TyG index and TyG-BMI show a linear correlation with DFS and OS. The effect of the TyG index on DFS and OS is not significantly mediated by lg-transformed BMI. Besides, FBP1 and G6PD are prognostic indicators for breast cancer patients and may serve as biomarkers for the clinical diagnosis and treatment of breast cancer.
Collapse
Affiliation(s)
- Fucheng Li
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, No. 150, Haping Road, Nangang District, Harbin, Heilongjiang 150081, China
| | - Tian Gao
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, No. 150, Haping Road, Nangang District, Harbin, Heilongjiang 150081, China
| | - Zhaoting Li
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, No. 150, Haping Road, Nangang District, Harbin, Heilongjiang 150081, China
| | - He Dou
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, No. 150, Haping Road, Nangang District, Harbin, Heilongjiang 150081, China
| | - Yuling Ba
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, No. 150, Haping Road, Nangang District, Harbin, Heilongjiang 150081, China
| | - Siyuan Jia
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, No. 150, Haping Road, Nangang District, Harbin, Heilongjiang 150081, China
| | - Danli Luo
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, No. 150, Haping Road, Nangang District, Harbin, Heilongjiang 150081, China
| | - Min Xiao
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, No. 150, Haping Road, Nangang District, Harbin, Heilongjiang 150081, China.
| |
Collapse
|
9
|
Han W, Cheng W, Fan M, Liu D, Cao Y, Mei X, Wan J, Hu G, Gao H, Ji N. Effects of Alexandrium pacificum Exposure on Exopalaemon carinicauda: Hepatopancreas Histology, Antioxidant Enzyme Activity, and Transcriptome Analysis. Int J Mol Sci 2025; 26:1605. [PMID: 40004076 PMCID: PMC11855214 DOI: 10.3390/ijms26041605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Revised: 02/08/2025] [Accepted: 02/11/2025] [Indexed: 02/27/2025] Open
Abstract
Alexandrium pacificum, a dinoflagellate known for causing harmful algal blooms (HABs), has garnered significant attention due to its potential toxicity to marine ecosystems, fisheries, and human health. However, the effects of this toxin-producing alga on shrimp are not yet comprehensively understood. This study aimed to assess the hepatopancreas damage induced by A. pacificum in the economically important shrimp species E. carinicauda and to elucidate the underlying molecular mechanisms through histology, antioxidant enzyme activity, and transcriptome analysis. The shrimp were assigned to either a control group or an exposed group, with the latter involving exposure to A. pacificum at a concentration of 1.0 × 104 cells/mL for 7 days. A histological analysis subsequently revealed pathological changes in the hepatopancreas tissue of the exposed group, including lumen expansion and the separation of the basement membrane from epithelial cells, while antioxidant enzyme activity assays demonstrated that exposure to A. pacificum weakened the antioxidant defense system, as evidenced by the reduced activities of catalase, superoxide dismutase, and glutathione, along with increased malondialdehyde levels. Transcriptome analysis further identified 663 significantly upregulated genes and 1735 significantly downregulated ones in the exposed group, with these differentially expressed genes being primarily associated with pathways such as protein processing in the endoplasmic reticulum, mitophagy, glycolysis/gluconeogenesis, sphingolipid metabolism, and glycerophospholipid metabolism. This study provides novel insights into the toxicological effects of A. pacificum on aquatic organisms and enhances the current understanding of the ecotoxicological risks posed by HABs.
Collapse
Affiliation(s)
- Wanyu Han
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Jiangsu Ocean University, Lianyungang 222005, China; (W.H.); (W.C.); (M.F.); (D.L.); (Y.C.); (X.M.); (J.W.); (H.G.)
| | - Weitao Cheng
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Jiangsu Ocean University, Lianyungang 222005, China; (W.H.); (W.C.); (M.F.); (D.L.); (Y.C.); (X.M.); (J.W.); (H.G.)
| | - Menghao Fan
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Jiangsu Ocean University, Lianyungang 222005, China; (W.H.); (W.C.); (M.F.); (D.L.); (Y.C.); (X.M.); (J.W.); (H.G.)
| | - Dexue Liu
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Jiangsu Ocean University, Lianyungang 222005, China; (W.H.); (W.C.); (M.F.); (D.L.); (Y.C.); (X.M.); (J.W.); (H.G.)
| | - Yanrong Cao
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Jiangsu Ocean University, Lianyungang 222005, China; (W.H.); (W.C.); (M.F.); (D.L.); (Y.C.); (X.M.); (J.W.); (H.G.)
| | - Xuao Mei
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Jiangsu Ocean University, Lianyungang 222005, China; (W.H.); (W.C.); (M.F.); (D.L.); (Y.C.); (X.M.); (J.W.); (H.G.)
| | - Jiaxuan Wan
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Jiangsu Ocean University, Lianyungang 222005, China; (W.H.); (W.C.); (M.F.); (D.L.); (Y.C.); (X.M.); (J.W.); (H.G.)
| | - Guangwei Hu
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Jiangsu Ocean University, Lianyungang 222005, China; (W.H.); (W.C.); (M.F.); (D.L.); (Y.C.); (X.M.); (J.W.); (H.G.)
- Co-Innovation Center of Jiangsu Marine Bio-industry Technology, Jiangsu Ocean University, Lianyungang 222005, China
- The Jiangsu Provincial Infrastructure for Conservation and Utilization of Agricultural Germplasm, Nanjing 210014, China
| | - Huan Gao
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Jiangsu Ocean University, Lianyungang 222005, China; (W.H.); (W.C.); (M.F.); (D.L.); (Y.C.); (X.M.); (J.W.); (H.G.)
- Co-Innovation Center of Jiangsu Marine Bio-industry Technology, Jiangsu Ocean University, Lianyungang 222005, China
- The Jiangsu Provincial Infrastructure for Conservation and Utilization of Agricultural Germplasm, Nanjing 210014, China
| | - Nanjing Ji
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Jiangsu Ocean University, Lianyungang 222005, China; (W.H.); (W.C.); (M.F.); (D.L.); (Y.C.); (X.M.); (J.W.); (H.G.)
- Co-Innovation Center of Jiangsu Marine Bio-industry Technology, Jiangsu Ocean University, Lianyungang 222005, China
| |
Collapse
|
10
|
Gu L, Zhu Y, Nandi SP, Lee M, Watari K, Bareng B, Ohira M, Liu Y, Sakane S, Carlessi R, Sauceda C, Dhar D, Ganguly S, Hosseini M, Teneche MG, Adams PD, Gonzalez DJ, Kisseleva T, Tirnitz-Parker JEE, Simon MC, Alexandrov LB, Karin M. FBP1 controls liver cancer evolution from senescent MASH hepatocytes. Nature 2025; 637:461-469. [PMID: 39743585 DOI: 10.1038/s41586-024-08317-9] [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: 07/22/2023] [Accepted: 10/30/2024] [Indexed: 01/04/2025]
Abstract
Hepatocellular carcinoma (HCC) originates from differentiated hepatocytes undergoing compensatory proliferation in livers damaged by viruses or metabolic-dysfunction-associated steatohepatitis (MASH)1. While increasing HCC risk2, MASH triggers p53-dependent hepatocyte senescence3, which we found to parallel hypernutrition-induced DNA breaks. How this tumour-suppressive response is bypassed to license oncogenic mutagenesis and enable HCC evolution was previously unclear. Here we identified the gluconeogenic enzyme fructose-1,6-bisphosphatase 1 (FBP1) as a p53 target that is elevated in senescent-like MASH hepatocytes but suppressed through promoter hypermethylation and proteasomal degradation in most human HCCs. FBP1 first declines in metabolically stressed premalignant disease-associated hepatocytes and HCC progenitor cells4,5, paralleling the protumorigenic activation of AKT and NRF2. By accelerating FBP1 and p53 degradation, AKT and NRF2 enhance the proliferation and metabolic activity of previously senescent HCC progenitors. The senescence-reversing and proliferation-supportive NRF2-FBP1-AKT-p53 metabolic switch, operative in mice and humans, also enhances the accumulation of DNA-damage-induced somatic mutations needed for MASH-to-HCC progression.
Collapse
Affiliation(s)
- Li Gu
- Laboratory of Gene Regulation and Signal Transduction, Departments of Pharmacology and Pathology, School of Medicine, University of California San Diego (UCSD), La Jolla, CA, USA.
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China.
- Clinical Laboratory Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China.
| | - Yahui Zhu
- School of Medicine, Chongqing University, Chongqing, China.
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.
| | - Shuvro P Nandi
- Department of Cellular and Molecular Medicine, UCSD, La Jolla, CA, USA
- Department of Bioengineering, UCSD, La Jolla, CA, USA
- Moores Cancer Center, UCSD, La Jolla, CA, USA
| | - Maiya Lee
- Laboratory of Gene Regulation and Signal Transduction, Departments of Pharmacology and Pathology, School of Medicine, University of California San Diego (UCSD), La Jolla, CA, USA
| | - Kosuke Watari
- Laboratory of Gene Regulation and Signal Transduction, Departments of Pharmacology and Pathology, School of Medicine, University of California San Diego (UCSD), La Jolla, CA, USA
| | - Breanna Bareng
- Laboratory of Gene Regulation and Signal Transduction, Departments of Pharmacology and Pathology, School of Medicine, University of California San Diego (UCSD), La Jolla, CA, USA
| | - Masafumi Ohira
- Laboratory of Gene Regulation and Signal Transduction, Departments of Pharmacology and Pathology, School of Medicine, University of California San Diego (UCSD), La Jolla, CA, USA
| | - Yuxiao Liu
- Laboratory of Gene Regulation and Signal Transduction, Departments of Pharmacology and Pathology, School of Medicine, University of California San Diego (UCSD), La Jolla, CA, USA
| | | | - Rodrigo Carlessi
- Curtin Medical School, Curtin Health Innovation Research Institute, Curtin University, Bentley, Western Australia, Australia
- Harry Perkins Institute of Medical Research, QEII Medical Centre and Centre for Medical Research, The University of Western Australia, Nedlands, Western Australia, Australia
| | - Consuelo Sauceda
- Department of Pharmacology, UCSD, La Jolla, CA, USA
- Skaggs School of Pharmacy and Pharmaceutical Sciences, UCSD, La Jolla, CA, USA
| | | | | | | | - Marcos G Teneche
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Peter D Adams
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - David J Gonzalez
- Department of Pharmacology, UCSD, La Jolla, CA, USA
- Skaggs School of Pharmacy and Pharmaceutical Sciences, UCSD, La Jolla, CA, USA
| | | | - Janina E E Tirnitz-Parker
- Curtin Medical School, Curtin Health Innovation Research Institute, Curtin University, Bentley, Western Australia, Australia
- Harry Perkins Institute of Medical Research, QEII Medical Centre and Centre for Medical Research, The University of Western Australia, Nedlands, Western Australia, Australia
| | - M Celeste Simon
- Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ludmil B Alexandrov
- Department of Cellular and Molecular Medicine, UCSD, La Jolla, CA, USA
- Department of Bioengineering, UCSD, La Jolla, CA, USA
- Moores Cancer Center, UCSD, La Jolla, CA, USA
| | - Michael Karin
- Laboratory of Gene Regulation and Signal Transduction, Departments of Pharmacology and Pathology, School of Medicine, University of California San Diego (UCSD), La Jolla, CA, USA.
| |
Collapse
|
11
|
Paradoxical link between senescent cell state and liver cancer resolved. Nature 2025:10.1038/d41586-024-04137-z. [PMID: 39743551 DOI: 10.1038/d41586-024-04137-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
12
|
Kim D, Xu H, Li O, Xue M, Bao Z, Yang F. Phenyllactic acid modulates the gut microbiota, enhances intestinal health, and alleviates physical frailty in aging mice. Eur J Pharmacol 2024; 985:177105. [PMID: 39515558 DOI: 10.1016/j.ejphar.2024.177105] [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: 06/17/2024] [Revised: 10/22/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024]
Abstract
Phenyllactic acid (PLA) is a natural antibiotic-like compound derived from certain foods and probiotics. PLA levels have been associated with age-related sarcopenia and provide benefits to metabolic health when derived from probiotics. However, the specific regulatory effects of PLA in aging remain largely unexplored. In this study, aging mice were administered PLA via gavage, followed by fecal 16S rRNA sequencing, measurements of targeted metabolites, glucose metabolism monitoring, and physical performance assessments. Our results indicate that PLA administration significantly altered gut microbiota composition, increased the abundance of short-chain fatty acids (SCFAs) and succinate producing microbiota, and enhanced gut integrity in aging mice. Furthermore, PLA treatment raised fasting blood glucose levels and improved physical activity. Mechanistically, PLA intake elevated the levels of circulating SCFAs and succinate, promoting glycogen metabolic homeostasis and maintaining skeletal muscle oxidative capacity. This study provides evidence that PLA modulates the gut microbiota in aging mice, supports intestinal health, promotes glucose homeostasis, and enhances physical activity.
Collapse
Affiliation(s)
- Dayoung Kim
- Department of Gerontology, Huadong Hospital Affiliated to Fudan University, Shanghai, China; Shanghai Key Laboratory of Clinical Geriatric Medicine, Shanghai, China
| | - Han Xu
- Department of Gerontology, Huadong Hospital Affiliated to Fudan University, Shanghai, China; Shanghai Key Laboratory of Clinical Geriatric Medicine, Shanghai, China
| | - Ouyang Li
- Department of Gerontology, Huadong Hospital Affiliated to Fudan University, Shanghai, China; Shanghai Key Laboratory of Clinical Geriatric Medicine, Shanghai, China
| | - Mengjuan Xue
- Shanghai Key Laboratory of Clinical Geriatric Medicine, Shanghai, China; Department of Endocrinology, Huadong Hospital Affiliated to Fudan University, Shanghai, China
| | - Zhijun Bao
- Department of Gerontology, Huadong Hospital Affiliated to Fudan University, Shanghai, China; Shanghai Key Laboratory of Clinical Geriatric Medicine, Shanghai, China.
| | - Fan Yang
- Department of Gerontology, Huadong Hospital Affiliated to Fudan University, Shanghai, China; Shanghai Key Laboratory of Clinical Geriatric Medicine, Shanghai, China.
| |
Collapse
|
13
|
Wu SH, Xiao MC, Liu F, Hong HY, Ding CH, Zhang X, Xie WF. Cell-permeated peptide P-T3H2 inhibits malignancy on hepatocellular carcinoma through stabilizing HNF4α protein. Discov Oncol 2024; 15:752. [PMID: 39638897 PMCID: PMC11621286 DOI: 10.1007/s12672-024-01661-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 12/02/2024] [Indexed: 12/07/2024] Open
Abstract
OBJECTIVES Hepatocyte nuclear factor 4α (HNF4α) is a key regulator of hepatocyte function and has a strong therapeutic effect on hepatocellular carcinoma (HCC) by inducing the differentiation of hepatoma cell into hepatocytes. Our previous study showed that Tribbles homolog 3 (TRIB3) directly interacts with and promotes the degradation of HNF4α in non-alcoholic fatty liver disease (NAFLD). Disrupting the TRIB3-HNF4α interaction by a cell-permeating peptide, called P-T3H2, stabilized HNF4α protein. This study aimed to assess the anti-tumor impact of P-T3H2 in HCC. METHODS The expression of TRIB3 and HNF4α was evaluated using western blot and immunohistochemistry (IHC). Hepatic functions and cellular senescence of HCC cells were evaluated through periodic acid-Schiff (PAS) staining, acetylated low-density lipoprotein (ac-LDL) uptake and senescence-associated β-galactosidase (SA-β-gal) activity staining, respectively. RNA-Seq analysis was performed to identify differentially expressed genes in Huh7 cells treated with P-T3H2. The impact of P-T3H2 on HCC malignancy was assessed in vitro and in vivo. RESULTS TRIB3 exhibited a negative correlation with HNF4α in both human and mouse HCC tissues. The administration of P-T3H2 significantly inhibited the malignancy of HCC cells. Additionally, P-T3H2 stabilized HNF4α protein and facilitated the restoration of hepatic functions and the cellular senescence in HCC cells. RNA-Seq analysis demonstrated that P-T3H2 enhanced the transcriptional activity of HNF4α in HCC. Furthermore, P-T3H2 effectively suppressed the carcinogenesis and progression of HCC in mice. CONCLUSION P-T3H2 suppressed HCC progression through the stabilization of HNF4α protein and may be a promising therapeutic candidate for clinical application in the treatment of HCC.
Collapse
Affiliation(s)
- Si-Han Wu
- Department of Gastroenterology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Meng-Chao Xiao
- Department of Gastroenterology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Fang Liu
- Department of Gastroenterology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Huan-Yu Hong
- Department of Gastroenterology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Chen-Hong Ding
- Department of Gastroenterology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Xin Zhang
- Department of Gastroenterology, Changzheng Hospital, Naval Medical University, Shanghai, China.
| | - Wei-Fen Xie
- Department of Gastroenterology, Changzheng Hospital, Naval Medical University, Shanghai, China.
| |
Collapse
|
14
|
Pakaew K, Chonpathompikunlert P, Wongmanee N, Rojanaverawong W, Sitdhipol J, Thaveethaptaikul P, Charoenphon N, Hanchang W. Lactobacillus reuteri TISTR 2736 alleviates type 2 diabetes in rats via the hepatic IRS1/PI3K/AKT signaling pathway by mitigating oxidative stress and inflammatory mediators. Eur J Nutr 2024; 64:27. [PMID: 39589518 DOI: 10.1007/s00394-024-03529-1] [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: 06/11/2024] [Accepted: 10/18/2024] [Indexed: 11/27/2024]
Abstract
PURPOSE This study investigated the beneficial effects of Lactobacillus reuteri TISTR 2736 on glucose homeostasis, carbohydrate metabolism, and the underlying mechanisms of its actions in type 2 diabetic (T2D) rats. METHODS A rat model of T2D was established by a combination of a high-fat diet and streptozotocin. The diabetic rats were treated daily with L. reuteri TISTR 2736 (2 × 108 CFU/day) for 30 days. Biochemical, histopathological, and molecular analyses were carried out to determine insulin signaling, carbohydrate metabolism, oxidative stress, and inflammation. RESULTS The results demonstrated that treatment with L. reuteri TISTR 2736 significantly ameliorated fasting blood glucose and glucose intolerance, and improved insulin sensitivity indices in the diabetic rats. The hepatic histopathology was improved with L. reuteri TISTR 2736 treatment, which was correlated with a reduction of hepatic lipid profiles. L. reuteri TISTR 2736 significantly reduced glycogen content, fructose 1,6-bisphosphatase activity, and phosphoenolpyruvate carboxykinase 1 protein expression, and enhanced hexokinase activity in the diabetic liver. The downregulation of IRS1 and phosphorylated IRS1Ser307 and upregulation of PI3K and phosphorylated AKTSer473 proteins in the liver were found in the L. reuteri TISTR 2736-treated diabetic group. Furthermore, it was able to suppress oxidative stress and inflammation in the diabetic rats, as demonstrated by decreased malondialdehyde and protein levels of NF-κB, IL-6 and TNF-α, but increased antioxidant enzyme activities of superoxide dismutase, catalase, and glutathione peroxidase. CONCLUSION By inhibiting oxidative and inflammatory stress, L. reuteri TISTR 2736 alleviated hyperglycemia and improved carbohydrate metabolism through activating IRS1/PI3K/AKT pathway in the T2D rats.
Collapse
Affiliation(s)
- Kamonthip Pakaew
- Department of Physiology, Faculty of Medical Science, Naresuan University, Phitsanulok, 65000, Thailand
| | - Pennapa Chonpathompikunlert
- Biodiversity Research Centre (BRC), Thailand Institute of Scientific and Technological Research (TISTR), Pathumthani, 12120, Thailand
| | - Navinee Wongmanee
- Department of Physiology, Faculty of Medical Science, Naresuan University, Phitsanulok, 65000, Thailand
| | - Worarat Rojanaverawong
- Department of Physiology, Faculty of Medical Science, Naresuan University, Phitsanulok, 65000, Thailand
| | - Jaruwan Sitdhipol
- Biodiversity Research Centre (BRC), Thailand Institute of Scientific and Technological Research (TISTR), Pathumthani, 12120, Thailand
| | - Punnathorn Thaveethaptaikul
- Biodiversity Research Centre (BRC), Thailand Institute of Scientific and Technological Research (TISTR), Pathumthani, 12120, Thailand
| | - Natthawut Charoenphon
- Department of Anatomy, Faculty of Medical Science, Naresuan University, Phitsanulok, 65000, Thailand
| | - Wanthanee Hanchang
- Department of Physiology, Faculty of Medical Science, Naresuan University, Phitsanulok, 65000, Thailand.
- Centre of Excellence in Medical Biotechnology, Naresuan University, Phitsanulok, 65000, Thailand.
| |
Collapse
|
15
|
Li M, Wang Z, Tao J, Jiang H, Yang H, Guo D, Zhao H, He X, Luo S, Jiang X, Yuan L, Xiao L, He H, Yu R, Fang J, Liang T, Mao Z, Xu D, Lu Z. Fructose-1,6-bisphosphatase 1 dephosphorylates and inhibits TERT for tumor suppression. Nat Chem Biol 2024; 20:1505-1513. [PMID: 38538923 DOI: 10.1038/s41589-024-01597-2] [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: 08/31/2023] [Accepted: 03/01/2024] [Indexed: 04/24/2024]
Abstract
Telomere dysfunction is intricately linked to the aging process and stands out as a prominent cancer hallmark. Here we demonstrate that telomerase activity is differentially regulated in cancer and normal cells depending on the expression status of fructose-1,6-bisphosphatase 1 (FBP1). In FBP1-expressing cells, FBP1 directly interacts with and dephosphorylates telomerase reverse transcriptase (TERT) at Ser227. Dephosphorylated TERT fails to translocate into the nucleus, leading to the inhibition of telomerase activity, reduction in telomere lengths, enhanced senescence and suppressed tumor cell proliferation and growth in mice. Lipid nanoparticle-mediated delivery of FBP1 mRNA inhibits liver tumor growth. Additionally, FBP1 expression levels inversely correlate with TERT pSer227 levels in renal and hepatocellular carcinoma specimens and with poor prognosis of the patients. These findings demonstrate that FBP1 governs cell immortality through its protein phosphatase activity and uncover a unique telomerase regulation in tumor cells attributed to the downregulation or deficiency of FBP1 expression.
Collapse
Affiliation(s)
- Min Li
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital and Institute of Translational Medicine, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Cancer Center, Zhejiang University, Hangzhou, China
| | - Zheng Wang
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital and Institute of Translational Medicine, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Cancer Center, Zhejiang University, Hangzhou, China
| | - Jingjing Tao
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital and Institute of Translational Medicine, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Cancer Center, Zhejiang University, Hangzhou, China
| | - Hongfei Jiang
- The Affiliated Hospital of Qingdao University and Qingdao Cancer Institute, Qingdao, China
| | - Huang Yang
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, China
| | - Dong Guo
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital and Institute of Translational Medicine, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
| | - Hong Zhao
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital and Institute of Translational Medicine, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
| | - Xuxiao He
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital and Institute of Translational Medicine, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
| | - Shudi Luo
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital and Institute of Translational Medicine, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
| | - Xiaoming Jiang
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital and Institute of Translational Medicine, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Cancer Center, Zhejiang University, Hangzhou, China
| | - Li Yuan
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital and Institute of Translational Medicine, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
| | - Liwei Xiao
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital and Institute of Translational Medicine, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Cancer Center, Zhejiang University, Hangzhou, China
| | - Haiyan He
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital and Institute of Translational Medicine, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
| | - Rilei Yu
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
| | - Jing Fang
- The Affiliated Hospital of Qingdao University and Qingdao Cancer Institute, Qingdao, China
| | - Tingbo Liang
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital and Institute of Translational Medicine, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
| | - Zhengwei Mao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, China
| | - Daqian Xu
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital and Institute of Translational Medicine, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China.
- Cancer Center, Zhejiang University, Hangzhou, China.
| | - Zhimin Lu
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital and Institute of Translational Medicine, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China.
- Cancer Center, Zhejiang University, Hangzhou, China.
| |
Collapse
|
16
|
Zhang P, Yang J, Liu X, Huang C, Tao Y, Shen P, Bai Z, Xiao C, Zhou L, Li G, Zhang L, Zhou W, Gao Y. FBP1 orchestrates keratinocyte proliferation/differentiation and suppresses psoriasis through metabolic control of histone acetylation. Cell Death Dis 2024; 15:392. [PMID: 38834617 PMCID: PMC11150480 DOI: 10.1038/s41419-024-06706-6] [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: 10/26/2023] [Revised: 04/13/2024] [Accepted: 04/24/2024] [Indexed: 06/06/2024]
Abstract
Keratinocyte proliferation and differentiation in epidermis are well-controlled and essential for reacting to stimuli such as ultraviolet light. Imbalance between proliferation and differentiation is a characteristic feature of major human skin diseases such as psoriasis and squamous cell carcinoma. However, the effect of keratinocyte metabolism on proliferation and differentiation remains largely elusive. We show here that the gluconeogenic enzyme fructose-1,6-bisphosphatase 1 (FBP1) promotes differentiation while inhibits proliferation of keratinocyte and suppresses psoriasis development. FBP1 is identified among the most upregulated genes induced by UVB using transcriptome sequencing and is elevated especially in upper epidermis. Fbp1 heterozygous mice exhibit aberrant epidermis phenotypes with local hyperplasia and dedifferentiation. Loss of FBP1 promotes proliferation and inhibits differentiation of keratinocytes in vitro. Mechanistically, FBP1 loss facilitates glycolysis-mediated acetyl-CoA production, which increases histone H3 acetylation at lysine 9, resulting in enhanced transcription of proliferation genes. We further find that the expression of FBP1 is dramatically reduced in human psoriatic lesions and in skin of mouse imiquimod psoriasis model. Fbp1 deficiency in mice facilitates psoriasis-like skin lesions development through glycolysis and acetyl-CoA production. Collectively, our findings reveal a previously unrecognized role of FBP1 in epidermal homeostasis and provide evidence for FBP1 as a metabolic psoriasis suppressor.
Collapse
Affiliation(s)
- Pengfei Zhang
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Ju Yang
- Department of Dermatology, The General Hospital of Western Theater Command PLA, Chengdu, Sichuan, 610083, China
| | - Xiong Liu
- Department of Information, The PLA Center for Disease Control and Prevention, Beijing, China
| | - Congshu Huang
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Yuandong Tao
- Department of Pediatric Urology, The Seventh Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Pan Shen
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Zhijie Bai
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Chengrong Xiao
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Lei Zhou
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Gaofu Li
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Li Zhang
- Department of Information, The PLA Center for Disease Control and Prevention, Beijing, China
| | - Wei Zhou
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, 100850, China.
| | - Yue Gao
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, 100850, China.
| |
Collapse
|
17
|
Jiang Q, Wang N, Lu S, Xiong J, Yuan Y, Liu J, Chen S. Targeting hepatic ceruloplasmin mitigates nonalcoholic steatohepatitis by modulating bile acid metabolism. J Mol Cell Biol 2024; 15:mjad060. [PMID: 37771074 PMCID: PMC10993722 DOI: 10.1093/jmcb/mjad060] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Revised: 09/19/2023] [Accepted: 09/27/2023] [Indexed: 09/30/2023] Open
Abstract
Nonalcoholic steatohepatitis (NASH) is a condition that progresses from nonalcoholic fatty liver disease (NAFLD) and is characterized by hepatic fat accumulation, inflammation, and fibrosis. It has the potential to develop into cirrhosis and liver cancer, and currently no effective pharmacological treatment is available. In this study, we investigate the therapeutic potential of targeting ceruloplasmin (Cp), a copper-containing protein predominantly secreted by hepatocytes, for treating NASH. Our result show that hepatic Cp is remarkedly upregulated in individuals with NASH and the mouse NASH model. Hepatocyte-specific Cp ablation effectively attenuates the onset of dietary-induced NASH by decreasing lipid accumulation, curbing inflammation, mitigating fibrosis, and ameliorating liver damage. By employing transcriptomics and metabolomics approaches, we have discovered that hepatic deletion of Cp brings about remarkable restoration of bile acid (BA) metabolism during NASH. Hepatic deletion of Cp effectively remodels BA metabolism by upregulating Cyp7a1 and Cyp8b1, which subsequently leads to enhanced BA synthesis and notable alterations in BA profiles. In conclusion, our studies elucidate the crucial involvement of Cp in NASH, highlighting its significance as a promising therapeutic target for the treatment of this disease.
Collapse
Affiliation(s)
- Quanxin Jiang
- Shanghai Diabetes Institute, Department of Endocrinology and Metabolism, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Ning Wang
- Shanghai Diabetes Institute, Department of Endocrinology and Metabolism, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Sijia Lu
- Shanghai Diabetes Institute, Department of Endocrinology and Metabolism, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Jie Xiong
- Shanghai Diabetes Institute, Department of Endocrinology and Metabolism, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Yanmei Yuan
- Shanghai Diabetes Institute, Department of Endocrinology and Metabolism, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Junli Liu
- Shanghai Diabetes Institute, Department of Endocrinology and Metabolism, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Suzhen Chen
- Shanghai Diabetes Institute, Department of Endocrinology and Metabolism, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| |
Collapse
|
18
|
Tournant F. Nonenzymatic prevention of insulin hyperresponsiveness by FBP1. Nat Rev Endocrinol 2023:10.1038/s41574-023-00847-y. [PMID: 37161028 DOI: 10.1038/s41574-023-00847-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
|