1
|
Su W, Wang X, Bu W, Fan H, Chen J, Guo S, Zhang S, Weng X, Zheng B, Xiang X. Recirculating aquaculture-fasting strategy (RASF) to modulate gut microbiota and enhance the fish meat quality of bighead carp (Aristichthys nobilis). Food Chem 2025; 481:143946. [PMID: 40154061 DOI: 10.1016/j.foodchem.2025.143946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 02/25/2025] [Accepted: 03/17/2025] [Indexed: 04/01/2025]
Abstract
Intensive aquaculture often leads to a decline in fish quality, and requires a strategy to restore the post-culture quality of fish. This study presents a novel recirculating aquaculture-fasting strategy (RASF) and assesses its impact on meat quality and gut microbiota of bighead carp. Results revealed RAS-F notably reduced body lipid and protein, achieving weight loss and body reshaping. RAS-F decreased the hardness, increased the springiness, and effectively improved the texture. Off-flavor compounds, especially aldehydes and ketones, were diminished, while umami amino acids (UAA) increased significantly. Noticeably, fatty acid profiles shifted, with reduced saturated fatty acids (SFA) and significantly increased polyunsaturated fatty acid (PUFA). Gut microbiota diversity and richness improved over fasting, with key microorganisms (Cetobacterium, Bacteroides, Akkermansia) contributing to PUFA and UAA synthesis as well as the off-flavor compounds degradation. These findings highlight the potential of RAS-F to enhance fish quality and reveal microbial mechanisms, offering insights for post-culture improvement.
Collapse
Affiliation(s)
- Weifa Su
- College of Food Science and Technology, Zhejiang University of Technology, Hangzhou 310014, China
| | - Xini Wang
- College of Food Science and Technology, Zhejiang University of Technology, Hangzhou 310014, China
| | - Weichao Bu
- College of Food Science and Technology, Zhejiang University of Technology, Hangzhou 310014, China
| | - Hongjie Fan
- College of Food Science and Technology, Zhejiang University of Technology, Hangzhou 310014, China
| | - Jinyu Chen
- Eco-Industrial Innovation Institute ZJUT, Quzhou 324400, China
| | - Saiqin Guo
- Eco-Industrial Innovation Institute ZJUT, Quzhou 324400, China
| | - Shuangqi Zhang
- Zhejiang Longhe Aquaculture Development Co., Ltd, Quzhou 324400, China
| | - Xudong Weng
- Zhejiang Fish Boss Agricultural Technology Co., Quzhou 324400, China
| | - Bin Zheng
- College of Food Science and Technology, Zhejiang University of Technology, Hangzhou 310014, China.
| | - Xingwei Xiang
- College of Food Science and Technology, Zhejiang University of Technology, Hangzhou 310014, China.
| |
Collapse
|
2
|
Han Z, Han S, Fang X, Lu M, Mao Y, Shi L, Song J, Wang T, Xiao J, Xiang L, Yang C, Zhu Z, Wang Y, Feng J. Acetyl-CoA carboxylase activation disrupts iron homeostasis to drive ferroptosis. Free Radic Biol Med 2025:S0891-5849(25)00720-8. [PMID: 40449808 DOI: 10.1016/j.freeradbiomed.2025.05.421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2025] [Revised: 05/20/2025] [Accepted: 05/27/2025] [Indexed: 06/03/2025]
Abstract
Acetyl-CoA carboxylase (ACC) is a rate-limiting enzyme in de novo lipogenesis. Here, we show a unique function of ACC in disrupting cellular iron homeostasis to drive ferroptosis, an iron-dependent, lipid peroxidation-induced form of cell death. We observed neuronal lipid accumulation and elevated labile iron pool associated with ACC dephosphorylation in mouse models of obstructive sleep apnea (OSA), a highly prevalent neurodegenerative disorder. ACC gene (Acaca) knockout (KO) or inhibition of its enzymatic activity rescued cellular iron metabolism through restoring lysosomal integrity and function, suppressing neuronal ferroptosis. ACC inactivation-driven lysosomal iron homeostasis requires the NFE2L2/NRF2-TFEB axis. Empagliflozin mitigates cellular iron overload via the ACC-NRF2-TFEB-lysosome pathway to alleviate neuronal ferroptosis, cognitive impairment, and mood dysfunction in OSA mice. Furthermore, inhibiting neuronal ACC reduces microglial activation, characterized by elevated complement proteins and pro-inflammatory cytokines, while microglia-specific C1qa KO prevents neuronal injury in OSA mice. Our findings identify a unique coupling between iron homeostasis and lipogenic signaling, suggesting ACC as a potential therapeutic target for neuronal ferroptosis and the resultant microgliosis in neurodegenerative diseases.
Collapse
Affiliation(s)
- Ziqi Han
- Department of Respiratory and Critical Care Medicine, Tianjin Medical University General Hospital
| | - Shuangyu Han
- Department of Respiratory and Critical Care Medicine, Tianjin Medical University General Hospital
| | - Xiaoyan Fang
- Department of Respiratory and Critical Care Medicine, Tianjin Medical University General Hospital
| | - Mengyu Lu
- Department of Respiratory and Critical Care Medicine, Tianjin Medical University General Hospital
| | - Yuanling Mao
- Department of Respiratory and Critical Care Medicine, Tianjin Medical University General Hospital
| | - Leilei Shi
- Department of Respiratory and Critical Care Medicine, Tianjin Medical University General Hospital
| | - Junxiu Song
- Department of Respiratory and Critical Care Medicine, Tianjin Medical University General Hospital
| | - Tian Wang
- Department of Respiratory and Critical Care Medicine, Tianjin Medical University General Hospital
| | - Jichen Xiao
- Department of Respiratory and Critical Care Medicine, Tianjin Medical University General Hospital
| | - Li Xiang
- Department of Respiratory and Critical Care Medicine, Tianjin Medical University General Hospital
| | - Changqing Yang
- Department of Respiratory and Critical Care Medicine, Tianjin Medical University General Hospital
| | - Zhigang Zhu
- Department of Respiratory and Critical Care Medicine, Tianjin Medical University General Hospital
| | - Yubao Wang
- Department of Respiratory and Critical Care Medicine, Tianjin Medical University General Hospital.
| | - Jing Feng
- Department of Respiratory and Critical Care Medicine, Tianjin Medical University General Hospital.
| |
Collapse
|
3
|
Wang X, Shi SR, Sun MM, Zhang XY, Zhang XH, Song SL, Yin F, Guo SD. Mechanisms of action of Fucus vesiculosus-derived fucoidan on improving dyslipidemia in New Zealand rabbits fed a high-fat diet. Int J Biol Macromol 2025; 314:144148. [PMID: 40368205 DOI: 10.1016/j.ijbiomac.2025.144148] [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: 01/15/2025] [Revised: 03/30/2025] [Accepted: 05/10/2025] [Indexed: 05/16/2025]
Abstract
Dyslipidemia is a major contributor to various diseases, including atherosclerotic cardiovascular disease and obesity. Treatment strategies for dyslipidemia continue to evolve as our understanding of this metabolic disorder and potential therapeutic candidates advance. Notably, fucoidan demonstrates promising effects in ameliorating dyslipidemia in rodents, although their lipid metabolism differs significantly from humans. This study, investigates the lipid-regulatory effects of Fucus vesiculosus-derived fucoidan (FvF) and elucidates the underlying mechanisms of action using New Zealand rabbits fed a high-fat diet, whose lipid profiles closely resemble those of patients with dyslipidemia. The results demonstrate that FvF intervention ameliorates dyslipidemia and lipid deposition in a dose-dependent manner. Mechanistically, FvF intervention modulates the expression levels of multiple molecules involved in lipid transport, fatty acid synthesis and beta-oxidation, and redox balance, as revealed by quantitative reverse transcription polymerase chain reaction, western blotting, and proteomic analysis. This study is the first to report that FvF, consisting of alternating [→4)-α-L-Fucp(1 → 3)-α-L-Fucp(1→] glycosyls ameliorates dyslipidemia by directly modulating lipid metabolism and indirectly attenuating oxidative stress. These findings suggest that FvF holds significant potential as a candidate for the treatment of lipid disorder-related diseases.
Collapse
Affiliation(s)
- Xue Wang
- Institute of Lipid Metabolism and Atherosclerosis, School of Pharmacy, Shandong Second Medical University, Weifang 261053, China
| | - Shan-Rui Shi
- Institute of Lipid Metabolism and Atherosclerosis, School of Pharmacy, Shandong Second Medical University, Weifang 261053, China
| | - Min-Min Sun
- School of Stomatology, Shandong Second Medical University, Weifang 261053, China
| | - Xue-Ying Zhang
- Institute of Lipid Metabolism and Atherosclerosis, School of Pharmacy, Shandong Second Medical University, Weifang 261053, China
| | - Xu-Hang Zhang
- Institute of Lipid Metabolism and Atherosclerosis, School of Pharmacy, Shandong Second Medical University, Weifang 261053, China
| | - Shi-Lin Song
- Institute of Lipid Metabolism and Atherosclerosis, School of Pharmacy, Shandong Second Medical University, Weifang 261053, China
| | - Fan Yin
- Institute of Lipid Metabolism and Atherosclerosis, School of Pharmacy, Shandong Second Medical University, Weifang 261053, China
| | - Shou-Dong Guo
- Institute of Lipid Metabolism and Atherosclerosis, School of Pharmacy, Shandong Second Medical University, Weifang 261053, China.
| |
Collapse
|
4
|
Brejchova K, Rahm M, Benova A, Domanska V, Reyes-Gutierez P, Dzubanova M, Trubacova R, Vondrackova M, Cajka T, Tencerova M, Vrabel M, Kuda O. Uncovering mechanisms of thiazolidinediones on osteogenesis and adipogenesis using spatial fluxomics. Metabolism 2025; 166:156157. [PMID: 39947516 DOI: 10.1016/j.metabol.2025.156157] [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: 11/28/2024] [Revised: 01/27/2025] [Accepted: 02/08/2025] [Indexed: 02/17/2025]
Abstract
OBJECTIVE Insulin-sensitizing drugs, despite their broad use against type 2 diabetes, can adversely affect bone health, and the mechanisms underlying these side effects remain largely unclear. Here, we investigated the different metabolic effects of a series of thiazolidinediones, including rosiglitazone, pioglitazone, and the second-generation compound MSDC-0602K, on human mesenchymal stem cells (MSCs). METHODS We developed 13C subcellular metabolomic tracer analysis measuring separate mitochondrial and cytosolic metabolite pools, lipidomic network-based isotopologue models, and bioorthogonal click chemistry, to demonstrate that MSDC-0602K differentially affected bone marrow-derived MSCs (BM-MSCs) and adipose tissue-derived MSCs (AT-MSCs). In BM-MSCs, MSDC-0602K promoted osteoblastic differentiation and suppressed adipogenesis. This effect was clearly distinct from that of the earlier drugs and that on AT-MSCs. RESULTS Fluxomic data reveal unexpected differences between this drug's effect on MSCs and provide mechanistic insight into the pharmacologic inhibition of mitochondrial pyruvate carrier 1 (MPC). Our study demonstrates that MSDC-0602K retains the capacity to inhibit MPC, akin to rosiglitazone but unlike pioglitazone, enabling the utilization of alternative metabolic pathways. Notably, MSDC-0602K exhibits a limited lipogenic potential compared to both rosiglitazone and pioglitazone, each of which employs a distinct lipogenic strategy. CONCLUSIONS These findings indicate that the new-generation drugs do not compromise bone structure, offering a safer alternative for treating insulin resistance. Moreover, these results highlight the ability of cell compartment-specific metabolite labeling by click reactions and tracer metabolomics analysis of complex lipids to discover molecular mechanisms within the intersection of carbohydrate and lipid metabolism.
Collapse
Affiliation(s)
- Kristyna Brejchova
- Laboratory of Metabolism of Bioactive Lipids, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, 14200 Prague, Czechia
| | - Michal Rahm
- Chemistry of Bioconjugates, Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo nám. 2, 166 10 Prague, Czechia
| | - Andrea Benova
- Laboratory of Molecular Physiology of Bone, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, 14200 Prague, Czechia
| | - Veronika Domanska
- Laboratory of Metabolism of Bioactive Lipids, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, 14200 Prague, Czechia
| | - Paul Reyes-Gutierez
- Chemistry of Bioconjugates, Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo nám. 2, 166 10 Prague, Czechia
| | - Martina Dzubanova
- Laboratory of Molecular Physiology of Bone, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, 14200 Prague, Czechia; Faculty of Science, Charles University, Albertov 6, 128 00 Prague, Czech Republic
| | - Radka Trubacova
- Laboratory of Metabolism of Bioactive Lipids, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, 14200 Prague, Czechia
| | - Michaela Vondrackova
- Laboratory of Metabolism of Bioactive Lipids, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, 14200 Prague, Czechia
| | - Tomas Cajka
- Laboratory of Translational Metabolism, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, 14200 Prague, Czechia
| | - Michaela Tencerova
- Laboratory of Molecular Physiology of Bone, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, 14200 Prague, Czechia
| | - Milan Vrabel
- Chemistry of Bioconjugates, Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo nám. 2, 166 10 Prague, Czechia
| | - Ondrej Kuda
- Laboratory of Metabolism of Bioactive Lipids, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, 14200 Prague, Czechia.
| |
Collapse
|
5
|
Kao YS, Lauterbach M, Lopez Krol A, Distler U, Godoy GJ, Klein M, Argüello RJ, Boukhallouk F, Vallejo Fuente S, Braband KL, Nurbekova A, Romero M, Mamareli P, Silva L, Damasceno LEA, Rampoldi F, Berod L, Lynch L, Hiller K, Sparwasser T. Metabolic reprogramming of interleukin-17-producing γδ T cells promotes ACC1-mediated de novo lipogenesis under psoriatic conditions. Nat Metab 2025; 7:966-984. [PMID: 40360755 PMCID: PMC12116387 DOI: 10.1038/s42255-025-01276-z] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 03/13/2025] [Indexed: 05/15/2025]
Abstract
Metabolic reprogramming determines γδ T cell fate during thymic development; however, the metabolic requirements of interleukin (IL)-17A-producing γδ T cells (γδT17 cells) under psoriatic conditions are unclear. Combining high-throughput techniques, including RNA sequencing, SCENITH, proteomics and stable isotope tracing, we demonstrated that psoriatic inflammation caused γδT17 cells to switch toward aerobic glycolysis. Under psoriatic conditions, γδT17 cells upregulated ATP-citrate synthase to convert citrate to acetyl-CoA, linking carbohydrate metabolism and fatty acid synthesis (FAS). Accordingly, we used a pharmacological inhibitor, Soraphen A, which blocks acetyl-CoA carboxylase (ACC), to impair FAS in γδT17 cells, reducing their intracellular lipid stores and ability to produce IL-17A under psoriatic conditions in vitro. We pinpointed the pathogenic role of ACC1 in γδT17 cells in vivo by genetic ablation, ameliorating inflammation in a psoriatic mouse model. Furthermore, ACC inhibition limited human IL-17A-producing γδT17 cells. Targeting ACC1 to attenuate pathogenic γδT17 cell function has important implications for psoriasis management.
Collapse
Affiliation(s)
- Yu-San Kao
- Institute of Medical Microbiology and Hygiene, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany.
- Ludwig Cancer Research Institute, Princeton Branch, Princeton University, Princeton, NJ, USA.
| | - Mario Lauterbach
- Department of Bioinformatics and Biochemistry, Braunschweig Integrated Centre of Systems Biology, Technical University of Braunschweig, Braunschweig, Germany
| | - Aleksandra Lopez Krol
- Institute of Medical Microbiology and Hygiene, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Ute Distler
- Research Center for Immunotherapy (FZI), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
- Institute for Immunology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Gloria Janet Godoy
- Institute of Medical Microbiology and Hygiene, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
- Institute of Molecular Medicine, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Matthias Klein
- Research Center for Immunotherapy (FZI), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
- Institute for Immunology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Rafael Jose Argüello
- Aix Marseille University, CNRS, INSERM, CIML, Centre d'Immunologie de Marseille-Luminy, Marseille, France
| | - Fatima Boukhallouk
- Institute of Medical Microbiology and Hygiene, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Sara Vallejo Fuente
- Institute of Medical Microbiology and Hygiene, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Kathrin Luise Braband
- Institute of Medical Microbiology and Hygiene, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
- Institute for Immunology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Assel Nurbekova
- Institute for Immunology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Monica Romero
- Institute of Medical Microbiology and Hygiene, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Panagiota Mamareli
- Institute of Medical Microbiology and Hygiene, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Luana Silva
- Institute of Medical Microbiology and Hygiene, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Luis Eduardo Alves Damasceno
- Institute of Medical Microbiology and Hygiene, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
- Center for Research in Inflammatory Diseases (CRID), Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, São Paulo, Brazil
| | - Francesca Rampoldi
- Institute of Medical Microbiology and Hygiene, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
- DKFZ German Cancer Research Center, Heidelberg, Germany
| | - Luciana Berod
- Research Center for Immunotherapy (FZI), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
- Institute of Molecular Medicine, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Lydia Lynch
- Molecular Biology, Princeton University, Princeton, NJ, USA
- Ludwig Cancer Research Institute, Princeton Branch, Princeton University, Princeton, NJ, USA
| | - Karsten Hiller
- Department of Bioinformatics and Biochemistry, Braunschweig Integrated Centre of Systems Biology, Technical University of Braunschweig, Braunschweig, Germany
| | - Tim Sparwasser
- Institute of Medical Microbiology and Hygiene, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany.
- Research Center for Immunotherapy (FZI), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany.
| |
Collapse
|
6
|
Zhou M, Zhang Y, Song Z, Tang S, Liu Z, Pang M, Zhang D, Wu X, Yu X, Wang P, Wei Y. Enhanced bioactivity of honeysuckle-Cassia seeds extracts through Lactobacillus acidophilus and Bacillus subtilis co-fermentation: Impact on alcoholic liver disease and gut microbiota. Food Chem 2025; 486:144463. [PMID: 40339419 DOI: 10.1016/j.foodchem.2025.144463] [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: 12/27/2024] [Revised: 04/13/2025] [Accepted: 04/20/2025] [Indexed: 05/10/2025]
Abstract
This study investigated the hepatoprotective potential of Honeysuckle-Cassia seeds extracts co-fermented with Lactobacillus acidophilus and Bacillus subtilis against alcoholic liver disease (ALD) and gut microbiota dysbiosis. Through network pharmacology analysis, 209 overlapping targets between Honeysuckle-Cassia seeds bioactive components and ALD-related targets were identified, with 39 core targets subsequently determined. Comparative analysis of aqueous extract (AE), Lactobacillus acidophilus fermentation broth (LAF), and mixed bacteria fermentation broth (MBF) revealed that MBF significantly enhanced the content of bioactive compounds: total polysaccharides (72.6 ± 3.8 mg/g), flavonoids (34.7 ± 2.5 mg/g), and saponins (15.2 ± 1.1 mg/g), representing 275 %, 72 %, and 62 % increases over AE, respectively (p < 0.05). In a murine ALD model, MBF intervention (12.5 mL/kg, 30 days) significantly reduced serum markers of liver injury (ALT: 35 %, AST: 28 %, TC: 42 %, TG: 39 %; (p < 0.05) and hepatic oxidative stress (MDA ↓52 %, GSH ↑156.55 %, SOD ↑76.71 %). Mechanistically, MBF suppressed pro-inflammatory cytokines (IL-1β, IL-6, TNF-α) by 40-50 % while elevating anti-inflammatory mediators (IL-4, IL-10, PGE2) 1.6-2.0-fold via AMPK/ACC/SREBP1c signaling modulation. Gut microbiota analysis revealed that MBF restored α-diversity indices (Shannon ↑10.06 %, ACE ↑32.34 %) and reversed alcohol-induced dysbiosis by enriching Lachnospiraceae and Blautia while suppressing Alloprevotella. Structural degradation of plant residues (SEM) confirmed microbial synergy in releasing insoluble-bound phytochemicals (100-400 m/z range). These findings validate co-fermentation as a potent strategy to amplify the hepatoprotective and microbiota-modulating activities of traditional herbs, offering a scientific foundation for developing functional foods against ALD.
Collapse
Affiliation(s)
- Mengtian Zhou
- Key Food Science and Engineering College, Beijing University of Agriculture, Beijing 102206, China
| | - Yingao Zhang
- Key Food Science and Engineering College, Beijing University of Agriculture, Beijing 102206, China
| | - Zeyi Song
- College of Bioscience and Resources Environment, Beijing University of Agriculture, Beijing 102206, China
| | - Shuping Tang
- Key Food Science and Engineering College, Beijing University of Agriculture, Beijing 102206, China
| | - Zhaoxue Liu
- Key Food Science and Engineering College, Beijing University of Agriculture, Beijing 102206, China
| | - Meixia Pang
- Key Food Science and Engineering College, Beijing University of Agriculture, Beijing 102206, China
| | - Di Zhang
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang 212013, China
| | - Xiaoyan Wu
- Ningbo Yufangtang Biotechnology Co., Ltd., Ningbo 315012, China
| | - Xindi Yu
- Zhejiang Dayidemei Biotechnology Co., Ltd., Ningbo 315012, China
| | - Pengze Wang
- Ningbo Yufangtang Biotechnology Co., Ltd., Ningbo 315012, China
| | - Ying Wei
- Key Food Science and Engineering College, Beijing University of Agriculture, Beijing 102206, China
| |
Collapse
|
7
|
Liu YQ, Yang Q, He GW. Post-translational acylation of proteins in cardiac hypertrophy. Nat Rev Cardiol 2025:10.1038/s41569-025-01150-1. [PMID: 40229510 DOI: 10.1038/s41569-025-01150-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/20/2025] [Indexed: 04/16/2025]
Abstract
Acylations are post-translational modifications in which functional groups are attached to amino acids on proteins. Most acylations (acetylation, butyrylation, crotonylation, lactylation, malonylation, propionylation and succinylation) involve lysine but cysteine (palmitoylation) and glycine (myristoylation) residues can also be altered. Acylations have important roles in physiological and pathophysiological processes, including cardiac hypertrophy and related cardiovascular diseases. These post-translational modifications influence chromatin architecture, transcriptional regulation and metabolic pathways, thereby affecting cardiomyocyte function and pathology. The dynamic interaction between these acylations and their regulatory enzymes, such as histone acetyltransferases, histone deacetylases and sirtuins, underscores the complexity of cellular homeostasis and pathological processes. Emerging evidence highlights the therapeutic potential of targeting acylations to modulate enzyme activity and metabolite levels, offering promising avenues for novel treatments. In this Review, we explore the diverse mechanisms through which acylations contribute to cardiac hypertrophy, highlighting the complexity and potential therapeutic targets in this regulatory network.
Collapse
Affiliation(s)
- Ying-Qi Liu
- Department of Cardiovascular Surgery & The Institute of Cardiovascular Diseases, TEDA International Cardiovascular Hospital, Tianjin University & Chinese Academy of Medical Sciences, Tianjin, China
- Tianjin Key Laboratory of Molecular Regulation of Cardiovascular Diseases and Translational Medicine, Tianjin, China
| | - Qin Yang
- Department of Cardiovascular Surgery & The Institute of Cardiovascular Diseases, TEDA International Cardiovascular Hospital, Tianjin University & Chinese Academy of Medical Sciences, Tianjin, China
- Tianjin Key Laboratory of Molecular Regulation of Cardiovascular Diseases and Translational Medicine, Tianjin, China
| | - Guo-Wei He
- Department of Cardiovascular Surgery & The Institute of Cardiovascular Diseases, TEDA International Cardiovascular Hospital, Tianjin University & Chinese Academy of Medical Sciences, Tianjin, China.
- Tianjin Key Laboratory of Molecular Regulation of Cardiovascular Diseases and Translational Medicine, Tianjin, China.
- Division of Cardiothoracic Surgery, Department of Surgery, Oregon Health & Science University, Portland, OR, USA.
| |
Collapse
|
8
|
He B, Copps KD, Stöhr O, Liu B, Hu S, Joshi S, Haigis MC, White MF, Zhu H, Tao R. Spatial regulation of glucose and lipid metabolism by hepatic insulin signaling. Cell Metab 2025:S1550-4131(25)00207-4. [PMID: 40245868 DOI: 10.1016/j.cmet.2025.03.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 02/18/2025] [Accepted: 03/27/2025] [Indexed: 04/19/2025]
Abstract
Hepatic insulin sensitivity is critical for systemic glucose and lipid homeostasis. The liver is spatially organized into zones in which hepatocytes express distinct metabolic enzymes; however, the functional significance of this zonation to metabolic dysregulation caused by insulin resistance is undetermined. Here, we used CreER mice to selectively disrupt insulin signaling in periportal (PP) and pericentral (PC) hepatocytes. PP-insulin resistance has been suggested to drive combined hyperglycemia and excess lipogenesis in individuals with type 2 diabetes. However, PP-insulin resistance in mice impaired lipogenesis and suppressed high-fat diet (HFD)-induced hepatosteatosis, despite elevated gluconeogenesis and insulin. In contrast, PC-insulin resistance reduced HFD-induced PC steatosis while preserving normal glucose homeostasis, in part by shifting glycolytic metabolism from the liver to the muscle. These results demonstrate distinct roles of insulin in PP versus PC hepatocytes and suggest that PC-insulin resistance might be therapeutically useful to combat hepatosteatosis without compromising glucose homeostasis.
Collapse
Affiliation(s)
- Baiyu He
- Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02215, USA
| | - Kyle D Copps
- Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02215, USA
| | - Oliver Stöhr
- Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02215, USA
| | - Beikl Liu
- Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02215, USA
| | - Songhua Hu
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Shakchhi Joshi
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Marcia C Haigis
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Morris F White
- Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02215, USA
| | - Hao Zhu
- Divisions of Hematology-Oncology, Children's Research Institute, UT Southwestern Medical Center, Dallas, TX 75235, USA
| | - Rongya Tao
- Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02215, USA.
| |
Collapse
|
9
|
Tao J, Zhang XY, Tan HB, Huang MY, Yang YQ, Guo J. FTZ alleviates lipid deposition in diabetic kidney disease by AMPK/ACC/SREBP signaling pathway. Acta Diabetol 2025:10.1007/s00592-025-02492-5. [PMID: 40167639 DOI: 10.1007/s00592-025-02492-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 03/17/2025] [Indexed: 04/02/2025]
Abstract
Fufang Zhenzhu Tiaozhi capsule (FTZ) is a patented traditional Chinese medicine preparation that has been used clinically for nearly 10 years to treat hyperglycemia, hyperlipidemia, and other glucolipid metabolic diseases. Previous studies have shown that FTZ can improve diabetic kidney disease (DKD). However, the role and mechanism of FTZ in reducing renal lipid accumulation in DKD remain unclear. Phosphorylation of Adenosine 5'-Monophosphate-Activated Protein Kinase (AMPK), a key regulator of energy homeostasis, inhibits Acetyl-CoA Carboxylase (ACC) signaling, thereby reducing fatty acid synthesis and promoting fatty acid oxidation via carnitine palmitoyltransferase-1 (CPT-1). Sterol regulatory element-binding protein 1 (SREBP-1), a transcription factor, regulates lipid metabolism through fatty acid synthesis. This study investigated the anti-lipid accumulation effect and mechanism of FTZ in vitro and in vivo. Streptozotocin (40 mg/kg/d, i.p. for 5 days, consecutively) combined with a high-fat diet (HFD) were used to induce a DKD model in C57BL/6J mice, followed by FTZ (1, 2 g/kg/d, i.g.) or Losartan (30 mg/kg/d, i.g.) treatments for 12 weeks. High glucose (HG, 30 mM) combined with palmitic-acid (PA, 250 µM) were used to induce HK-2 cells injury, followed by FTZ (25, 50, or 100 µg/ml) or Compound C (an AMPK inhibitor, 10 µM) treatments for 24 h. Results showed that FTZ reduced blood lipids and improved renal function in DKD mice. In addition, compared with the control group, DKD mice and cells exhibited significantly increased lipid deposition. However, the effect of FTZ in alleviating lipid accumulation was reversed by Compound C. Furthermore, FTZ increased p-AMPK, p-ACC and CPT-1 protein expression while decreasing SREBP-1. These results indicate that FTZ effectively protects against lipid accumulation in DKD by regulating the AMPK/ACC/SREBP pathway, inhibiting de novo lipogenesis, providing a novel therapeutic strategy for DKD.
Collapse
Affiliation(s)
- Jie Tao
- Key Laboratory of Glucolipid Metabolic Diseases of the Ministry of Education, Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, The Institute of Chinese Medicinal Sciences, Guangdong Pharmaceutical University, Guangzhou, 510006, People's Republic of China
| | - Xiao-Yu Zhang
- Key Laboratory of Glucolipid Metabolic Diseases of the Ministry of Education, Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, The Institute of Chinese Medicinal Sciences, Guangdong Pharmaceutical University, Guangzhou, 510006, People's Republic of China
| | - Hai-Bo Tan
- Key Laboratory of Glucolipid Metabolic Diseases of the Ministry of Education, Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, The Institute of Chinese Medicinal Sciences, Guangdong Pharmaceutical University, Guangzhou, 510006, People's Republic of China
| | - Min-Yi Huang
- Key Laboratory of Glucolipid Metabolic Diseases of the Ministry of Education, Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, The Institute of Chinese Medicinal Sciences, Guangdong Pharmaceutical University, Guangzhou, 510006, People's Republic of China
| | - Yi-Qi Yang
- Key Laboratory of Glucolipid Metabolic Diseases of the Ministry of Education, Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, The Institute of Chinese Medicinal Sciences, Guangdong Pharmaceutical University, Guangzhou, 510006, People's Republic of China.
| | - Jiao Guo
- Key Laboratory of Glucolipid Metabolic Diseases of the Ministry of Education, Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, The Institute of Chinese Medicinal Sciences, Guangdong Pharmaceutical University, Guangzhou, 510006, People's Republic of China.
| |
Collapse
|
10
|
Yang L, Wang X, Wang S, Shen J, Li Y, Wan S, Xiao Z, Wu Z. Targeting lipid metabolism in regulatory T cells for enhancing cancer immunotherapy. Biochim Biophys Acta Rev Cancer 2025; 1880:189259. [PMID: 39798823 DOI: 10.1016/j.bbcan.2025.189259] [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: 06/11/2024] [Revised: 12/30/2024] [Accepted: 01/02/2025] [Indexed: 01/15/2025]
Abstract
As immunosuppressive cells, Regulatory T cells (Tregs) exert their influence on tumor immune escape within the tumor microenvironment (TME) by effectively suppressing the activity of other immune cells, thereby significantly impeding the anti-tumor immune response. In recent years, the metabolic characteristics of Tregs have become a focus of research, especially the important role of lipid metabolism in maintaining the function of Tregs. Consequently, targeted interventions aimed at modulating lipid metabolism in Tregs have been recognized as an innovative and promising approach to enhance the effectiveness of tumor immunotherapy. This review presents a comprehensive overview of the pivotal role of lipid metabolism in regulating the function of Tregs, with a specific focus on targeting Tregs lipid metabolism as an innovative approach to augment anti-tumor immune responses. Furthermore, we discuss potential opportunities and challenges associated with this strategy, aiming to provide novel insights for enhancing the efficacy of cancer immunotherapy.
Collapse
Affiliation(s)
- Liu Yang
- Department of Pharmacy, Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan 646000, China; Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China; South Sichuan Institute of Translational Medicine, Luzhou, Sichuan 646000, China; Laboratory of Personalised Cell Therapy and Cell Medicine, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Xingyue Wang
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Shurong Wang
- Department of Pharmacy, Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Jing Shen
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China; South Sichuan Institute of Translational Medicine, Luzhou, Sichuan 646000, China; Laboratory of Personalised Cell Therapy and Cell Medicine, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Yaling Li
- Department of Pharmacy, Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Shengli Wan
- Department of Pharmacy, Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Zhangang Xiao
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China; South Sichuan Institute of Translational Medicine, Luzhou, Sichuan 646000, China; Laboratory of Personalised Cell Therapy and Cell Medicine, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China.
| | - Zhigui Wu
- Department of Pharmacy, Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan 646000, China; Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China; South Sichuan Institute of Translational Medicine, Luzhou, Sichuan 646000, China; Laboratory of Personalised Cell Therapy and Cell Medicine, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China.
| |
Collapse
|
11
|
Jiang H, Zhou L, Zhang H, Yu Z. Bioinformatic analysis of glycolysis and lactate metabolism genes in head and neck squamous cell carcinoma. Sci Rep 2025; 15:10781. [PMID: 40155682 PMCID: PMC11953422 DOI: 10.1038/s41598-025-94843-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Accepted: 03/17/2025] [Indexed: 04/01/2025] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) is a heterogeneous cancer with significant global incidence. This study investigates glycolysis- and lactate metabolism-related genes (GALMRGs) in HNSCC, focusing on their impact on prognosis, the tumor immune microenvironment, and their potential as therapeutic biomarkers. Analysis of data from the Cancer Genome Atlas and Gene Expression Omnibus identified 16 GALMRGs that were differentially expressed in HNSCC compared to normal tissues. Functional analysis revealed the involvement of lactate and pyruvate metabolism and HIF-1 signaling pathways. Weighted gene co-expression network analysis identified two module genes, CDKN3 and SLC2A1. Five key genes (CAV1, CDKN3, LDHA, MB, and PER2) were identified through univariate, multivariate, and LASSO regression analyses and used to construct a prognostic model. This model demonstrated strong predictive accuracy for overall survival, stratifying patients into high- and low-risk groups. Immune cell infiltration analysis showed a negative correlation between resting and activated mast cells, and low-risk patients had higher tumor mutational burden, suggesting a better response to immunotherapy. Consensus clustering classified HNSCC into two distinct molecular subtypes with differing expression of the key genes. This GALMRG-based prognostic model is a promising biomarker for predicting HNSCC outcomes and immunotherapy responses, providing valuable insights for personalized treatment strategies.
Collapse
Affiliation(s)
- Huanyu Jiang
- School of Medicine, Southeast University, 87 Dingjiaqiao, Hunan Road, Nanjing, 210009, Jiangsu, China
- Department of Otolaryngology Head and Neck Surgery, The Affiliated Benq Hospital of Nanjing Medical University, Nanjing, 210019, Jiangsu, China
| | - Lijuan Zhou
- Department of Otolaryngology Head and Neck Surgery, The Affiliated Benq Hospital of Nanjing Medical University, Nanjing, 210019, Jiangsu, China
| | - Haidong Zhang
- School of Medicine, Southeast University, 87 Dingjiaqiao, Hunan Road, Nanjing, 210009, Jiangsu, China
- Department of Otolaryngology Head and Neck Surgery, The Affiliated Benq Hospital of Nanjing Medical University, Nanjing, 210019, Jiangsu, China
| | - Zhenkun Yu
- School of Medicine, Southeast University, 87 Dingjiaqiao, Hunan Road, Nanjing, 210009, Jiangsu, China.
- Department of Otolaryngology Head and Neck Surgery, The Affiliated Benq Hospital of Nanjing Medical University, Nanjing, 210019, Jiangsu, China.
| |
Collapse
|
12
|
Luukkonen PK. Hidden metabolic effects of acetyl-CoA carboxylase inhibition. J Hepatol 2025; 82:544-545. [PMID: 39681499 DOI: 10.1016/j.jhep.2024.11.043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 11/26/2024] [Accepted: 11/26/2024] [Indexed: 12/18/2024]
Affiliation(s)
- Panu K Luukkonen
- Department of Internal Medicine, University of Helsinki and Helsinki University Hospital, Helsinki, Finland; Minerva Foundation Institute for Medical Research, Helsinki, Finland; Abdominal Center, Helsinki University Hospital, Helsinki, Finland.
| |
Collapse
|
13
|
Li M, Ding L, Cao L, Zhang Z, Li X, Li Z, Xia Q, Yin K, Song S, Wang Z, Du H, Zhao D, Li X, Wang Z. Natural products targeting AMPK signaling pathway therapy, diabetes mellitus and its complications. Front Pharmacol 2025; 16:1534634. [PMID: 39963239 PMCID: PMC11830733 DOI: 10.3389/fphar.2025.1534634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Accepted: 01/15/2025] [Indexed: 02/20/2025] Open
Abstract
Diabetes mellitus (DM) ranks among the most prevalent chronic metabolic diseases, characterized primarily by a persistent elevation in blood glucose levels. This condition typically stems from either insufficient insulin secretion or a functional defect in the insulin itself. Clinically, diabetes is primarily classified into type 1 diabetes mellitus (T1DM) and type 2 diabetes mellitus (T2DM), with T2DM comprising nearly 90% of all diagnosed cases. Notably, the global incidence of T2DM has surged dramatically over recent decades. The adenylate-activated protein kinase (AMPK) signaling pathway is crucial in regulating cellular energy metabolism, marking it as a significant therapeutic target for diabetes and related complications. Natural products, characterized by their diverse origins, multifaceted bioactivities, and relative safety, hold considerable promise in modulating the AMPK pathway. This review article explores the advances in research on natural products that target the AMPK signaling pathway, aiming to inform the development of innovative antidiabetic therapies.
Collapse
Affiliation(s)
- Min Li
- Northeast Asia Research Institute of Traditional Chinese Medicine, Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efcacy, Ministry of Education, Jilin Provincial Key Laboratory of Bio-Macromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Jilin, China
| | - Lu Ding
- Northeast Asia Research Institute of Traditional Chinese Medicine, Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efcacy, Ministry of Education, Jilin Provincial Key Laboratory of Bio-Macromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Jilin, China
- Research Center of Traditional Chinese Medicine, The Affiliated Hospital to Changchun University of Chinese Medicine, Jilin, China
| | - Liyuan Cao
- Northeast Asia Research Institute of Traditional Chinese Medicine, Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efcacy, Ministry of Education, Jilin Provincial Key Laboratory of Bio-Macromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Jilin, China
| | - Zepeng Zhang
- Research Center of Traditional Chinese Medicine, The Affiliated Hospital to Changchun University of Chinese Medicine, Jilin, China
| | - Xueyan Li
- Northeast Asia Research Institute of Traditional Chinese Medicine, Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efcacy, Ministry of Education, Jilin Provincial Key Laboratory of Bio-Macromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Jilin, China
| | - Zirui Li
- Northeast Asia Research Institute of Traditional Chinese Medicine, Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efcacy, Ministry of Education, Jilin Provincial Key Laboratory of Bio-Macromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Jilin, China
| | - Qinjing Xia
- Northeast Asia Research Institute of Traditional Chinese Medicine, Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efcacy, Ministry of Education, Jilin Provincial Key Laboratory of Bio-Macromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Jilin, China
| | - Kai Yin
- Northeast Asia Research Institute of Traditional Chinese Medicine, Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efcacy, Ministry of Education, Jilin Provincial Key Laboratory of Bio-Macromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Jilin, China
| | - Siyu Song
- Northeast Asia Research Institute of Traditional Chinese Medicine, Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efcacy, Ministry of Education, Jilin Provincial Key Laboratory of Bio-Macromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Jilin, China
| | - Zihan Wang
- Northeast Asia Research Institute of Traditional Chinese Medicine, Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efcacy, Ministry of Education, Jilin Provincial Key Laboratory of Bio-Macromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Jilin, China
| | - Haijian Du
- Northeast Asia Research Institute of Traditional Chinese Medicine, Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efcacy, Ministry of Education, Jilin Provincial Key Laboratory of Bio-Macromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Jilin, China
| | - Daqing Zhao
- Northeast Asia Research Institute of Traditional Chinese Medicine, Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efcacy, Ministry of Education, Jilin Provincial Key Laboratory of Bio-Macromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Jilin, China
| | - Xiangyan Li
- Northeast Asia Research Institute of Traditional Chinese Medicine, Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efcacy, Ministry of Education, Jilin Provincial Key Laboratory of Bio-Macromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Jilin, China
| | - Zeyu Wang
- Northeast Asia Research Institute of Traditional Chinese Medicine, Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efcacy, Ministry of Education, Jilin Provincial Key Laboratory of Bio-Macromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Jilin, China
- Research Center of Traditional Chinese Medicine, The Affiliated Hospital to Changchun University of Chinese Medicine, Jilin, China
| |
Collapse
|
14
|
Smith K, Dennis KMJH, Hodson L. The ins and outs of liver fat metabolism: The effect of phenotype and diet on risk of intrahepatic triglyceride accumulation. Exp Physiol 2025. [PMID: 39861959 DOI: 10.1113/ep092001] [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: 07/05/2024] [Accepted: 11/22/2024] [Indexed: 01/27/2025]
Abstract
In health, the liver is a metabolically flexible organ that plays a key role in regulating systemic lipid and glucose concentrations. There is a constant flux of fatty acids (FAs) to the liver from multiple sources, including adipose tissue, dietary, endogenously synthesized from non-lipid precursors, intrahepatic lipid droplets and recycling of triglyceride-rich remnants. Within the liver, FAs are used for triglyceride synthesis, which can be oxidized, stored or secreted in very low-density lipoproteins into the systemic circulation. The processes of FA uptake, FA synthesis and the intracellular partitioning of FAs into storage, oxidation or secretory pathways are tightly regulated. An imbalance in these processes causes intrahepatic triglyceride to accumulate and is associated with the development of metabolic dysfunction-associated steatotic liver disease. It is well appreciated that many factors can influence intrahepatic FA partitioning, and although there is good evidence that both phenotype (e.g., sex, ethnicity and adiposity) and dietary macronutrient composition can play a role in intrahepatic triglyceride accumulation, their interaction remains poorly understood. The aim of this review is to explore how the respective pathways of FA delivery, synthesis and disposal are altered by phenotype and understand how dietary macronutrient composition might influence the partitioning of FAs in the liver in vivo, in humans.
Collapse
Affiliation(s)
- Kieran Smith
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK
| | - Kaitlyn M J H Dennis
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK
| | - Leanne Hodson
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK
- Oxford NIHR Biomedical Research Centre, Churchill Hospital, Oxford, UK
| |
Collapse
|
15
|
Du J, Shen M, Chen J, Yan H, Xu Z, Yang X, Yang B, Luo P, Ding K, Hu Y, He Q. The impact of solute carrier proteins on disrupting substance regulation in metabolic disorders: insights and clinical applications. Front Pharmacol 2025; 15:1510080. [PMID: 39850557 PMCID: PMC11754210 DOI: 10.3389/fphar.2024.1510080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Accepted: 12/20/2024] [Indexed: 01/25/2025] Open
Abstract
Carbohydrates, lipids, bile acids, various inorganic salt ions and organic acids are the main nutrients or indispensable components of the human body. Dysregulation in the processes of absorption, transport, metabolism, and excretion of these metabolites can lead to the onset of severe metabolic disorders, such as type 2 diabetes, non-alcoholic fatty liver disease, gout and hyperbilirubinemia. As the second largest membrane receptor supergroup, several major families in the solute carrier (SLC) supergroup have been found to play key roles in the transport of substances such as carbohydrates, lipids, urate, bile acids, monocarboxylates and zinc ions. Based on common metabolic dysregulation and related metabolic substances, we explored the relationship between several major families of SLC supergroup and metabolic diseases, providing examples of drugs targeting SLC proteins that have been approved or are currently in clinical/preclinical research as well as SLC-related diagnostic techniques that are in clinical use or under investigation. By highlighting these connections, we aim to provide insights that may contribute to the development of improved treatment strategies and targeted therapies for metabolic disorders.
Collapse
Affiliation(s)
- Jiangxia Du
- Center for Medical Research and Innovation in Digestive System Tumors, Ministry of Education, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Minhui Shen
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Jiajia Chen
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Hao Yan
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Zhifei Xu
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Xiaochun Yang
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Bo Yang
- Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China
- School of Medicine, Hangzhou City University, Hangzhou, Zhejiang, China
| | - Peihua Luo
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China
- Department of Pharmaceutical and Translational Toxicology, Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Hangzhou, Zhejiang, China
| | - Kefeng Ding
- Center for Medical Research and Innovation in Digestive System Tumors, Ministry of Education, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yuhuai Hu
- Yuhong Pharmaceutical Technology Co., Ltd., Hangzhou, Zhejiang, China
| | - Qiaojun He
- Center for Medical Research and Innovation in Digestive System Tumors, Ministry of Education, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China
- School of Medicine, Hangzhou City University, Hangzhou, Zhejiang, China
- Department of Pharmaceutical and Translational Toxicology, Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Hangzhou, Zhejiang, China
| |
Collapse
|
16
|
Rauckhorst AJ, Sheldon RD, Pape DJ, Ahmed A, Falls-Hubert KC, Merrill RA, Brown RF, Deshmukh K, Vallim TA, Deja S, Burgess SC, Taylor EB. A hierarchical hepatic de novo lipogenesis substrate supply network utilizing pyruvate, acetate, and ketones. Cell Metab 2025; 37:255-273.e6. [PMID: 39471817 PMCID: PMC11856365 DOI: 10.1016/j.cmet.2024.10.013] [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: 11/03/2023] [Revised: 08/07/2024] [Accepted: 10/18/2024] [Indexed: 11/01/2024]
Abstract
Hepatic de novo lipogenesis (DNL) is a fundamental physiologic process that is often pathogenically elevated in metabolic disease. Treatment is limited by incomplete understanding of the metabolic pathways supplying cytosolic acetyl-CoA, the obligate precursor to DNL, including their interactions and proportional contributions. Here, we combined extensive 13C tracing with liver-specific knockout of key mitochondrial and cytosolic proteins mediating cytosolic acetyl-CoA production. We show that the mitochondrial pyruvate carrier (MPC) and ATP-citrate lyase (ACLY) gate the major hepatic lipogenic acetyl-CoA production pathway, operating in parallel with acetyl-CoA synthetase 2 (ACSS2). Given persistent DNL after mitochondrial citrate carrier (CiC) and ACSS2 double knockout, we tested the contribution of exogenous and leucine-derived acetoacetate to acetoacetyl-CoA synthetase (AACS)-dependent DNL. CiC knockout increased acetoacetate-supplied hepatic acetyl-CoA production and DNL, indicating that ketones function as mitochondrial-citrate reciprocal DNL precursors. By delineating a mitochondrial-cytosolic DNL substrate supply network, these findings may inform strategies to therapeutically modulate DNL.
Collapse
Affiliation(s)
- Adam J Rauckhorst
- Department of Molecular Physiology and Biophysics, University of Iowa Carver College of Medicine, Iowa City, IA 52240, USA; Fraternal Order of Eagles Diabetes Research Center (FOEDRC), University of Iowa Carver College of Medicine, Iowa City, IA 52240, USA; FOEDRC Metabolomics Core Research Facility, University of Iowa Carver College of Medicine, Iowa City, IA 52240, USA
| | - Ryan D Sheldon
- Department of Molecular Physiology and Biophysics, University of Iowa Carver College of Medicine, Iowa City, IA 52240, USA
| | - Daniel J Pape
- Department of Molecular Physiology and Biophysics, University of Iowa Carver College of Medicine, Iowa City, IA 52240, USA
| | - Adnan Ahmed
- Department of Molecular Physiology and Biophysics, University of Iowa Carver College of Medicine, Iowa City, IA 52240, USA
| | - Kelly C Falls-Hubert
- Department of Molecular Physiology and Biophysics, University of Iowa Carver College of Medicine, Iowa City, IA 52240, USA
| | - Ronald A Merrill
- Department of Molecular Physiology and Biophysics, University of Iowa Carver College of Medicine, Iowa City, IA 52240, USA
| | - Reid F Brown
- Department of Molecular Physiology and Biophysics, University of Iowa Carver College of Medicine, Iowa City, IA 52240, USA; FOEDRC Metabolomics Core Research Facility, University of Iowa Carver College of Medicine, Iowa City, IA 52240, USA
| | - Kshitij Deshmukh
- Department of Molecular Physiology and Biophysics, University of Iowa Carver College of Medicine, Iowa City, IA 52240, USA
| | - Thomas A Vallim
- Department of Biological Chemistry, University of California, Los Angeles (UCLA), David Geffen School of Medicine, Los Angeles, CA 90095, USA
| | - Stanislaw Deja
- Center for Human Nutrition, University of Texas Southwestern, Dallas, TX 75390, USA
| | - Shawn C Burgess
- Center for Human Nutrition, University of Texas Southwestern, Dallas, TX 75390, USA
| | - Eric B Taylor
- Department of Molecular Physiology and Biophysics, University of Iowa Carver College of Medicine, Iowa City, IA 52240, USA; Fraternal Order of Eagles Diabetes Research Center (FOEDRC), University of Iowa Carver College of Medicine, Iowa City, IA 52240, USA; FOEDRC Metabolomics Core Research Facility, University of Iowa Carver College of Medicine, Iowa City, IA 52240, USA; Holden Comprehensive Cancer Center, University of Iowa Carver College of Medicine, Iowa City, IA 52240, USA; Abboud Cardiovascular Research Center, University of Iowa Carver College of Medicine, Iowa City, IA 52240, USA; Pappajohn Biomedical Institute, University of Iowa Carver College of Medicine, Iowa City, IA 52240, USA.
| |
Collapse
|
17
|
Zhu S, Ruan F, Ye L, Jiang S, Yang C, Zuo Z, He C. Black phosphorus quantum dots induce lipid accumulation through PPARγ activation and mitochondrial dysfunction in adipocytes. THE SCIENCE OF THE TOTAL ENVIRONMENT 2025; 958:177972. [PMID: 39662394 DOI: 10.1016/j.scitotenv.2024.177972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 12/02/2024] [Accepted: 12/04/2024] [Indexed: 12/13/2024]
Abstract
Black phosphorus quantum dots (BPQDs) are believed to have broad prospects for application. Obesity has garnered significant attention, but the association between BPQDs and lipid metabolism has not been thoroughly investigated. Mice were orally exposed to BPQDs at doses of 0.1 and 1 mg/kg for 28 d. The exposed mice exhibited reduced insulin sensitivity, hypertrophy of white adipose tissues, and reduced thermogenic function of brown adipose tissues. In white adipocyte line (3T3-L1), exposure to 5-20 μg/mL BPQDs induced lipid accumulation, oxidative stress, and upregulated the expression of PPARγ and genes involved in de novo lipogenesis. Moreover, both a reactive oxygen species (ROS) scavenger and a PPARγ inhibitor were able to attenuate lipid accumulation and downregulate the expression of lipid-associated genes in white adipocytes. In mouse brown adipocytes, BPQDs exposure caused oxidative stress, mitochondrial dysfunction, and downregulation of thermogenic genes such as UCP1. The ROS scavenger attenuated the oxidative stress and improved the mitochondrial thermogenic function in brown adipocytes. In summary, this work demonstrates that oxidative stress induced by BPQDs mediates the lipid accumulation possibly through PPARγ activation and mitochondrial dysfunction of adipose tissues, highlighting the potential obesogenic effect of BPQDs. Our findings provide novel insights into the biosafety of BPQDs and their potential health risks to humans, offering important considerations for the sustainable application of BP materials. ENVIRONMENTAL IMPLICATION: BPQDs are a novel type of nanomaterials with unique physicochemical properties, and have broad applications in various fields, particularly in biomedicine. However, during the production and use of BPQDs as medical materials, they inevitably contact with the human body for long periods of time. Therefore, it is necessary to investigate the effects of BPQDs on organisms under long-term exposure, especially lipid metabolism. This study would be helpful decreasing the environmental health risk of BP materials and promoting their sustainable development of nanotechnology in biomedicine.
Collapse
Affiliation(s)
- Sihao Zhu
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Department of Endocrinology, Xiang'an Hospital of Xiamen University, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen 361102, China
| | - Fengkai Ruan
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Department of Endocrinology, Xiang'an Hospital of Xiamen University, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen 361102, China
| | - Lingxiao Ye
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Department of Endocrinology, Xiang'an Hospital of Xiamen University, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen 361102, China
| | - Suhua Jiang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Department of Endocrinology, Xiang'an Hospital of Xiamen University, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen 361102, China
| | - Chunyan Yang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Department of Endocrinology, Xiang'an Hospital of Xiamen University, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen 361102, China
| | - Zhenghong Zuo
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Department of Endocrinology, Xiang'an Hospital of Xiamen University, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen 361102, China
| | - Chengyong He
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Department of Endocrinology, Xiang'an Hospital of Xiamen University, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen 361102, China..
| |
Collapse
|
18
|
Vo ATT, Mouli K, Liopo AV, Lorenzi P, Tan L, Wei B, Martinez SA, McHugh EA, Tour JM, Khan U, Derry PJ, Kent TA. Pleozymes: Pleiotropic Oxidized Carbon Nanozymes Enhance Cellular Metabolic Flexibility. NANOMATERIALS (BASEL, SWITZERLAND) 2024; 14:2017. [PMID: 39728553 PMCID: PMC11728746 DOI: 10.3390/nano14242017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 12/02/2024] [Accepted: 12/06/2024] [Indexed: 12/28/2024]
Abstract
Our group has synthesized a pleiotropic synthetic nanozyme redox mediator we term a "pleozyme" that displays multiple enzymatic characteristics, including acting as a superoxide dismutase mimetic, oxidizing NADH to NAD+, and oxidizing H2S to polysulfides and thiosulfate. Benefits have been seen in acute and chronic neurological disease models. The molecule is sourced from coconut-derived activated charcoal that has undergone harsh oxidization with fuming nitric acid, which alters the structure and chemical characteristics, yielding 3-8 nm discs with broad redox potential. Prior work showed pleozymes localize to mitochondria and increase oxidative phosphorylation and glycolysis. Here, we measured cellular NAD+ and NADH levels after pleozyme treatment and observed increased total cellular NADH levels but not total NAD+ levels. A 13C-glucose metabolic flux analysis suggested pleozymes stimulate the generation of pyruvate and lactate glycolytically and from the tricarboxylic acid (TCA) cycle, pointing to malate decarboxylation. Analysis of intracellular fatty acid abundances suggests pleozymes increased fatty acid β-oxidation, with a concomitant increase in succinyl- and acetyl-CoA. Pleozymes increased total ATP, potentially via flexible enhancement of NAD+-dependent catabolic pathways such as glycolysis, fatty acid β-oxidation, and metabolic flux through the TCA cycle. These effects may be favorable for pathologies that compromise metabolism such as brain injury.
Collapse
Affiliation(s)
- Anh T. T. Vo
- Center for Genomics and Precision Medicine, Institute of Bioscience and Technology, Texas A&M Health Science Center, Houston, TX 77030, USA; (A.T.T.V.); (K.M.); (A.V.L.); (U.K.)
| | - Karthik Mouli
- Center for Genomics and Precision Medicine, Institute of Bioscience and Technology, Texas A&M Health Science Center, Houston, TX 77030, USA; (A.T.T.V.); (K.M.); (A.V.L.); (U.K.)
| | - Anton V. Liopo
- Center for Genomics and Precision Medicine, Institute of Bioscience and Technology, Texas A&M Health Science Center, Houston, TX 77030, USA; (A.T.T.V.); (K.M.); (A.V.L.); (U.K.)
- Department of Chemistry, Rice University, Houston, TX 77005, USA; (E.A.M.); (J.M.T.)
| | - Philip Lorenzi
- Metabolomics Core Facility, Department of Bioinformatics & Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (P.L.); (L.T.); (B.W.); (S.A.M.)
| | - Lin Tan
- Metabolomics Core Facility, Department of Bioinformatics & Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (P.L.); (L.T.); (B.W.); (S.A.M.)
| | - Bo Wei
- Metabolomics Core Facility, Department of Bioinformatics & Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (P.L.); (L.T.); (B.W.); (S.A.M.)
| | - Sara A. Martinez
- Metabolomics Core Facility, Department of Bioinformatics & Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (P.L.); (L.T.); (B.W.); (S.A.M.)
| | - Emily A. McHugh
- Department of Chemistry, Rice University, Houston, TX 77005, USA; (E.A.M.); (J.M.T.)
- Smalley-Curl Institute, Rice University, Houston, TX 77005, USA
| | - James M. Tour
- Department of Chemistry, Rice University, Houston, TX 77005, USA; (E.A.M.); (J.M.T.)
- Smalley-Curl Institute, Rice University, Houston, TX 77005, USA
- Rice Advanced Materials Institute, Rice University, Houston, TX 77005, USA
- The NanoCarbon Center, Rice University, Houston, TX 77005, USA
| | - Uffaf Khan
- Center for Genomics and Precision Medicine, Institute of Bioscience and Technology, Texas A&M Health Science Center, Houston, TX 77030, USA; (A.T.T.V.); (K.M.); (A.V.L.); (U.K.)
| | - Paul J. Derry
- Center for Genomics and Precision Medicine, Institute of Bioscience and Technology, Texas A&M Health Science Center, Houston, TX 77030, USA; (A.T.T.V.); (K.M.); (A.V.L.); (U.K.)
- School of Engineering Medicine, Texas A&M University, Houston, TX 77030, USA
| | - Thomas A. Kent
- Center for Genomics and Precision Medicine, Institute of Bioscience and Technology, Texas A&M Health Science Center, Houston, TX 77030, USA; (A.T.T.V.); (K.M.); (A.V.L.); (U.K.)
- Department of Chemistry, Rice University, Houston, TX 77005, USA; (E.A.M.); (J.M.T.)
- Stanley H. Appel Department of Neurology, Houston Methodist Hospital and Houston Methodist Research Institute, Houston, TX 77030, USA
| |
Collapse
|
19
|
Barroso E, Jurado-Aguilar J, Wahli W, Palomer X, Vázquez-Carrera M. Increased hepatic gluconeogenesis and type 2 diabetes mellitus. Trends Endocrinol Metab 2024; 35:1062-1077. [PMID: 38816269 DOI: 10.1016/j.tem.2024.05.006] [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: 01/28/2024] [Revised: 05/03/2024] [Accepted: 05/13/2024] [Indexed: 06/01/2024]
Abstract
Abnormally increased hepatic gluconeogenesis is a significant contributor to hyperglycemia in the fasting state in patients with type 2 diabetes mellitus (T2DM) due to insulin resistance. Metformin, the most prescribed drug for the treatment of T2DM, is believed to exert its effect mainly by reducing hepatic gluconeogenesis. Here, we discuss how increased hepatic gluconeogenesis contributes to T2DM and we review newly revealed mechanisms underlying the attenuation of gluconeogenesis by metformin. In addition, we analyze the recent findings on new determinants involved in the regulation of gluconeogenesis, which might ultimately lead to the identification of novel and targeted treatment strategies for T2DM.
Collapse
Affiliation(s)
- Emma Barroso
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, 08028 Barcelona, Spain; Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, 08028 Barcelona, Spain; Spanish Biomedical Research Center in Diabetes and Associated Metabolic Diseases (CIBERDEM)-Instituto de Salud Carlos III, 28029 Madrid, Spain; Pediatric Research Institute-Hospital Sant Joan de Déu, 08950, Esplugues de Llobregat, Barcelona, Spain
| | - Javier Jurado-Aguilar
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, 08028 Barcelona, Spain; Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, 08028 Barcelona, Spain; Spanish Biomedical Research Center in Diabetes and Associated Metabolic Diseases (CIBERDEM)-Instituto de Salud Carlos III, 28029 Madrid, Spain; Pediatric Research Institute-Hospital Sant Joan de Déu, 08950, Esplugues de Llobregat, Barcelona, Spain
| | - Walter Wahli
- Center for Integrative Genomics, University of Lausanne, CH-1015 Lausanne, Switzerland; Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Singapore 308232; ToxAlim (Research Center in Food Toxicology), INRAE, UMR1331, F-31300 Toulouse Cedex, France
| | - Xavier Palomer
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, 08028 Barcelona, Spain; Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, 08028 Barcelona, Spain; Spanish Biomedical Research Center in Diabetes and Associated Metabolic Diseases (CIBERDEM)-Instituto de Salud Carlos III, 28029 Madrid, Spain; Pediatric Research Institute-Hospital Sant Joan de Déu, 08950, Esplugues de Llobregat, Barcelona, Spain
| | - Manuel Vázquez-Carrera
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, 08028 Barcelona, Spain; Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, 08028 Barcelona, Spain; Spanish Biomedical Research Center in Diabetes and Associated Metabolic Diseases (CIBERDEM)-Instituto de Salud Carlos III, 28029 Madrid, Spain; Pediatric Research Institute-Hospital Sant Joan de Déu, 08950, Esplugues de Llobregat, Barcelona, Spain.
| |
Collapse
|
20
|
Luo M, Ma X, Ye J. Reductive stress-a common metabolic feature of obesity and cancer. Acta Pharm Sin B 2024; 14:5181-5185. [PMID: 39807313 PMCID: PMC11725146 DOI: 10.1016/j.apsb.2024.08.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 08/19/2024] [Accepted: 08/26/2024] [Indexed: 01/16/2025] Open
Abstract
Reductive stress, characterized by rising level of NADH (nicotinamide adenine dinucleotide) for a status of NADH/NAD+ ratio elevation, has been reported in obesity and cancer. However, the mechanism and significance of reductive stress remain to be established in obesity. This perspective is prepared to address the issue with new insights published recently. NADH is used in production of NADPH, glutathione, ATP and heat in the classical biochemistry. In obesity, elevation of NADH/NAD+ ratio, likely from overproduction due to substrate overloading, has been found in the liver for insulin resistance and gluconeogenesis. New evidence demonstrates that the elevation may induce lipogenesis, purine biosynthesis and gluconeogenesis through activation of transcription factors of ChREBP and NRF2. In cancer cells, NADH/NAD+ elevation under the Warburg effect is primarily derived from decreased NADH consumption in the mitochondrial respiration. Alternatively, NRF2 overactivation from gene mutation represents another mechanism of NADH/NAD+ elevation from NADH production in the cancer cells. The elevation is required for quick proliferation of cancer cells through induction of biosynthesis of the essential molecules. It appears that the causes of reductive stress are different between obesity and cancer, while its impact in anabolism is similar in the two conditions.
Collapse
Affiliation(s)
- Man Luo
- Metabolic Disease Research Center, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou 450052, China
| | - Xiwen Ma
- Metabolic Disease Research Center, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou 450052, China
- Institute of Trauma and Metabolism, Zhengzhou University, Zhengzhou 450052, China
| | - Jianping Ye
- Metabolic Disease Research Center, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou 450052, China
- Institute of Trauma and Metabolism, Zhengzhou University, Zhengzhou 450052, China
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou 450052, China
- Zhengzhou Key laboratory of Obesity Research, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou 450052, China
| |
Collapse
|
21
|
Pan S, Yin L, Liu J, Tong J, Wang Z, Zhao J, Liu X, Chen Y, Miao J, Zhou Y, Zeng S, Xu T. Metabolomics-driven approaches for identifying therapeutic targets in drug discovery. MedComm (Beijing) 2024; 5:e792. [PMID: 39534557 PMCID: PMC11555024 DOI: 10.1002/mco2.792] [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: 07/07/2024] [Revised: 09/29/2024] [Accepted: 09/30/2024] [Indexed: 11/16/2024] Open
Abstract
Identification of therapeutic targets can directly elucidate the mechanism and effect of drug therapy, which is a central step in drug development. The disconnect between protein targets and phenotypes under complex mechanisms hampers comprehensive target understanding. Metabolomics, as a systems biology tool that captures phenotypic changes induced by exogenous compounds, has emerged as a valuable approach for target identification. A comprehensive overview was provided in this review to illustrate the principles and advantages of metabolomics, delving into the application of metabolomics in target identification. This review outlines various metabolomics-based methods, such as dose-response metabolomics, stable isotope-resolved metabolomics, and multiomics, which identify key enzymes and metabolic pathways affected by exogenous substances through dose-dependent metabolite-drug interactions. Emerging techniques, including single-cell metabolomics, artificial intelligence, and mass spectrometry imaging, are also explored for their potential to enhance target discovery. The review emphasizes metabolomics' critical role in advancing our understanding of disease mechanisms and accelerating targeted drug development, while acknowledging current challenges in the field.
Collapse
Affiliation(s)
- Shanshan Pan
- Research Center for Clinical PharmacyCollege of Pharmaceutical SciencesZhejiang UniversityHangzhouZhejiangChina
- College of Pharmaceutical SciencesZhejiang UniversityHangzhouZhejiangChina
| | - Luan Yin
- College of Pharmaceutical SciencesZhejiang UniversityHangzhouZhejiangChina
| | - Jie Liu
- College of Pharmaceutical SciencesZhejiang UniversityHangzhouZhejiangChina
| | - Jie Tong
- Department of Radiology and Biomedical ImagingPET CenterYale School of MedicineNew HavenConnecticutUSA
| | - Zichuan Wang
- Research Center for Clinical PharmacyCollege of Pharmaceutical SciencesZhejiang UniversityHangzhouZhejiangChina
- College of Pharmaceutical SciencesZhejiang UniversityHangzhouZhejiangChina
| | - Jiahui Zhao
- School of Basic Medical SciencesZhejiang Chinese Medical UniversityHangzhouChina
| | - Xuesong Liu
- College of Pharmaceutical SciencesZhejiang UniversityHangzhouZhejiangChina
- Cangnan County Qiushi Innovation Research Institute of Traditional Chinese MedicineWenzhouZhejiangChina
| | - Yong Chen
- College of Pharmaceutical SciencesZhejiang UniversityHangzhouZhejiangChina
- Cangnan County Qiushi Innovation Research Institute of Traditional Chinese MedicineWenzhouZhejiangChina
| | - Jing Miao
- Research Center for Clinical PharmacyCollege of Pharmaceutical SciencesZhejiang UniversityHangzhouZhejiangChina
| | - Yuan Zhou
- School of Basic Medical SciencesZhejiang Chinese Medical UniversityHangzhouChina
| | - Su Zeng
- Research Center for Clinical PharmacyCollege of Pharmaceutical SciencesZhejiang UniversityHangzhouZhejiangChina
- College of Pharmaceutical SciencesZhejiang UniversityHangzhouZhejiangChina
| | - Tengfei Xu
- Research Center for Clinical PharmacyCollege of Pharmaceutical SciencesZhejiang UniversityHangzhouZhejiangChina
- College of Pharmaceutical SciencesZhejiang UniversityHangzhouZhejiangChina
| |
Collapse
|
22
|
Zhang X, Ge J, Wang Y, Chen M, Guo X, Zhu S, Wang H, Wang Q. Integrative Omics Reveals the Metabolic Patterns During Oocyte Growth. Mol Cell Proteomics 2024; 23:100862. [PMID: 39414232 PMCID: PMC11585809 DOI: 10.1016/j.mcpro.2024.100862] [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: 04/29/2024] [Revised: 08/01/2024] [Accepted: 10/13/2024] [Indexed: 10/18/2024] Open
Abstract
Well-controlled metabolism is associated with high-quality oocytes and optimal development of a healthy embryo. However, the metabolic framework that controls mammalian oocyte growth remains unknown. In the present study, we comprehensively depict the temporal metabolic dynamics of mouse oocytes during in vivo growth through the integrated analysis of metabolomics and proteomics. Many novel metabolic features are discovered during this process. Of note, glycolysis is enhanced, and oxidative phosphorylation capacity is reduced in the growing oocytes, presenting a Warburg-like metabolic program. For nucleotide biosynthesis, the salvage pathway is markedly activated during oocyte growth, whereas the de novo pathway is evidently suppressed. Fatty acid synthesis and channeling into phosphoinositides are specifically elevated in oocytes accompanying primordial follicle activation; nevertheless, fatty acid oxidation is reduced in these oocytes simultaneously. Our data establish the metabolic landscape during in vivo oocyte growth and serve as a broad resource for probing mammalian oocyte metabolism.
Collapse
Affiliation(s)
- Xiang Zhang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Changzhou Maternity and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, Nanjing, China
| | - Juan Ge
- State Key Laboratory of Reproductive Medicine and Offspring Health, Changzhou Maternity and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, Nanjing, China
| | - Yue Wang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Changzhou Maternity and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, Nanjing, China
| | - Minjian Chen
- Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Xuejiang Guo
- State Key Laboratory of Reproductive Medicine and Offspring Health, Changzhou Maternity and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, Nanjing, China
| | - Shuai Zhu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Changzhou Maternity and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, Nanjing, China.
| | - Hui Wang
- Department of Histology and Embryology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China.
| | - Qiang Wang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Changzhou Maternity and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, Nanjing, China; Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
23
|
Ansar Khawaja S, Alturise F, Alkhalifah T, Khan SA, Khan YD. Gluconeogenesis unraveled: A proteomic Odyssey with machine learning. Methods 2024; 232:29-42. [PMID: 39276958 DOI: 10.1016/j.ymeth.2024.09.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 08/05/2024] [Accepted: 09/01/2024] [Indexed: 09/17/2024] Open
Abstract
The metabolic pathway known as gluconeogenesis, which produces glucose from non-carbohydrate substrates, is essential for maintaining balanced blood sugar levels while fasting. It's extremely important to anticipate gluconeogenesis rates accurately to recognize metabolic disorders and create efficient treatment strategies. The implementation of deep learning and machine learning methods to forecast complex biological processes has been gaining popularity in recent years. The recognition of both the regulation of the pathway and possible therapeutic applications of proteins depends on accurate identification associated with their gluconeogenesis patterns. This article analyzes the uses of machine learning and deep learning models, to predict gluconeogenesis efficiency. The study also discusses the challenges that come with restricted data availability and model interpretability, as well as possible applications in personalized healthcare, metabolic disease treatment, and the discovery of drugs. The predictor utilizes statistics moments on the structures of gluconeogenesis and their enzymes, while Random Forest is utilized as a classifier to ensure the accuracy of this model in identifying the best outcomes. The method was validated utilizing the independent test, self-consistency, 10k fold cross-validations, and jackknife test which achieved 92.33 %, 91.87%, 87.88%, and 87.02%. An accurate prediction of gluconeogenesis has significant implications for understanding metabolic disorders and developing targeted therapies. This study contributes to the rising field of predictive biology by mixing algorithms for deep learning, and machine learning, with metabolic pathways.
Collapse
Affiliation(s)
- Seher Ansar Khawaja
- Department of Computer Science, University of Management and Technology, Lahore, Paksistan
| | - Fahad Alturise
- Department of Cybersecurity, College of Computer, Qassim University, Buraydah, Saudi Arabia.
| | - Tamim Alkhalifah
- Deparment of Computer Engineering, College of Computer, Qassim University, Buraydah, Saudi Arabia.
| | - Sher Afzal Khan
- Deparment of Computer Sciences, Abdul Wali Khan University, Mardan, Pakistan.
| | - Yaser Daanial Khan
- Department of Computer Science, University of Management and Technology, Lahore, Paksistan.
| |
Collapse
|
24
|
Pérez-Díaz AJ, Núñez-Sánchez MÁ, Ramos-Molina B. Revisiting liver metabolism through acetyl-CoA carboxylase inhibition. Trends Endocrinol Metab 2024; 35:563-565. [PMID: 38664153 DOI: 10.1016/j.tem.2024.04.010] [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: 03/26/2024] [Revised: 04/09/2024] [Accepted: 04/10/2024] [Indexed: 07/11/2024]
Abstract
Liver-targeted acetyl-coenzyme A (CoA) carboxylase (ACC) inhibitors in metabolic dysfunction-associated steatotic liver disease (MASLD) trials reveal notable secondary effects: hypertriglyceridemia and altered glucose metabolism, paradoxically with reduced hepatic steatosis. In their study, Deja et al. explored how hepatic ACC influences metabolism using different pharmacological and genetic methods, coupled with targeted metabolomics and stable isotope-based tracing techniques.
Collapse
Affiliation(s)
| | - María Ángeles Núñez-Sánchez
- Obesity, Diabetes and Metabolism Laboratory, Biomedical Research Institute of Murcia (IMIB), 30120 Murcia, Spain.
| | - Bruno Ramos-Molina
- Obesity, Diabetes and Metabolism Laboratory, Biomedical Research Institute of Murcia (IMIB), 30120 Murcia, Spain.
| |
Collapse
|
25
|
Eller MM, Zuberi AR, Fu X, Burgess SC, Lutz CM, Bailey RM. Valine and Inflammation Drive Epilepsy in a Mouse Model of ECHS1 Deficiency. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.13.598697. [PMID: 38915588 PMCID: PMC11195255 DOI: 10.1101/2024.06.13.598697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
ECHS1 Deficiency (ECHS1D) is a rare and devastating pediatric disease that currently has no defined treatments. This disorder results from missense loss-of-function mutations in the ECHS1 gene that result in severe developmental delays, encephalopathy, hypotonia, and early death. ECHS1 enzymatic activity is necessary for the beta-oxidation of fatty acids and the oxidation of branched-chain amino acids within the inner mitochondrial matrix. The pathogenesis of disease remains unknown, however it is hypothesized that disease is driven by an accumulation of toxic metabolites from impaired valine oxidation. To expand our knowledge on disease mechanisms, a novel mouse model of ECHS1D was generated that possesses a disease-associated knock-in (KI) allele and a knock-out (KO) allele. To investigate the behavioral phenotype, a battery of testing was performed at multiple time points, which included assessments of learning, motor function, endurance, sensory responses, and anxiety. Neurological abnormalities were assessed using wireless telemetry EEG recordings, pentylenetetrazol (PTZ) seizure induction, and immunohistochemistry. Metabolic perturbations were measured within the liver, serum, and brain using mass spectrometry and magnetic resonance spectroscopy. To test disease mechanisms, mice were subjected to disease pathway stressors and then survival, body weight gain, and epilepsy were assessed. Mice containing KI/KI or KI/KO alleles were viable with normal development and survival, and the presence of KI and KO alleles resulted in a significant reduction in ECHS1 protein. ECHS1D mice displayed reduced exercise capacity and pain sensation. EEG analysis revealed increased slow wave power that was associated with perturbations in sleep. ECHS1D mice had significantly increased epileptiform EEG discharges, and were sensitive to seizure induction, which resulted in death of 60% of ECHS1D mice. Under basal conditions, brain structure was grossly normal, although histological analysis revealed increased microglial activation in aged ECHS1D mice. Increased dietary valine only affected ECHS1D mice, which significantly exacerbated seizure susceptibility and resulted in death. Lastly, acute inflammatory challenge drove regression and early lethality in ECHS1D mice. In conclusion, we developed a novel model of ECHS1D that may be used to further knowledge on disease mechanisms and to develop therapeutics. Our data suggests altered metabolic signaling and inflammation may contribute to epilepsy in ECHS1D, and these alterations may be attributed to impaired valine metabolism.
Collapse
Affiliation(s)
- Meghan M. Eller
- Graduate School of Biomedical Sciences, University of Texas Southwestern Medical School, 5323 Harry Hines Blvd, Dallas, TX 75235
- Center for Alzheimer’s and Neurodegenerative Diseases, University of Texas Southwestern Medical School, 5323 Harry Hines Blvd, Dallas, TX 75235
| | - Aamir R. Zuberi
- The Jackson Laboratory Center for Precision Genetics, The Jackson Laboratory, Bar Harbor, ME 04609
| | - Xiaorong Fu
- Center for Human Nutrition, University of Texas Southwestern Medical School, 5323 Harry Hines Blvd, Dallas, TX 75235
| | - Shawn C. Burgess
- Center for Human Nutrition, University of Texas Southwestern Medical School, 5323 Harry Hines Blvd, Dallas, TX 75235
- Department of Pharmacology, University of Texas Southwestern Medical School, 5323 Harry Hines Blvd, Dallas, TX 75235
| | - Cathleen M. Lutz
- The Jackson Laboratory Center for Precision Genetics, The Jackson Laboratory, Bar Harbor, ME 04609
| | - Rachel M. Bailey
- Center for Alzheimer’s and Neurodegenerative Diseases, University of Texas Southwestern Medical School, 5323 Harry Hines Blvd, Dallas, TX 75235
- Department of Pediatrics, University of Texas Southwestern Medical School, 5323 Harry Hines Blvd, Dallas, TX 75235
| |
Collapse
|