1
|
Huang C, Luo Y, Liu Y, Liu J, Chen Y, Zeng B, Liao X, Liu Y, Wang X. DNA hypermethylation-induced suppression of ALKBH5 is required for folic acid to alleviate hepatic lipid deposition by enhancing autophagy in an ATG12-dependent manner. J Nutr Biochem 2025; 140:109870. [PMID: 39993647 DOI: 10.1016/j.jnutbio.2025.109870] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 12/08/2024] [Accepted: 02/13/2025] [Indexed: 02/26/2025]
Abstract
Nonalcoholic fatty liver disease (NAFLD) occurs when too much fat builds up in the liver. As a growing worldwide epidemic, NAFLD is strongly linked with multiple metabolic diseases including obesity, insulin resistance, and dyslipidemia. However, very few effective treatments are currently available. Folate, an essential B-group vitamin with important biological functions including DNA and RNA methylation regulation, has been shown to have a protective effect against NAFLD with its underlying mechanism remains largely unclear. Here, we show that administration of folic acid significantly improves glucose tolerance, insulin sensitivity, and dyslipidemia in high-fat diet (HFD) fed mice. Moreover, folic acid treatment significantly inhibits lipid deposition in hepatocytes both in vivo and in vitro. Mechanically, folic acid reduces the expression of m6A demethylase AlkB homolog 5 (ALKHB5) via promoter DNA hypermethylation. Decreased ALKBH5 causes increased m6A modification and increased expression of ATG12 in a demethylase activity-dependent manner, thereby promoting autophagy and preventing hepatic steatosis. Inhibition of ATG12 induced by overexpression of ALKBH5 could impair autophagy and the inhibitory effect of folic acid on lipid accumulation in hepatocytes. Together, these findings provide novel insights into understanding the protective role of folic acid in the treatment of NAFLD and suggest that folic acid may be a potential agent for combating NAFLD.
Collapse
Affiliation(s)
- Chaoqun Huang
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, PR China; Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, Zhejiang, PR China; Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou, Zhejiang, PR China; Zhejiang Key Laboratory of Nutrition and Breeding for High-quality Animal Products, Zhejiang, PR China
| | - Yaojun Luo
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, PR China; Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, Zhejiang, PR China; Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou, Zhejiang, PR China; Zhejiang Key Laboratory of Nutrition and Breeding for High-quality Animal Products, Zhejiang, PR China
| | - Youhua Liu
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, PR China; Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, Zhejiang, PR China; Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou, Zhejiang, PR China; Zhejiang Key Laboratory of Nutrition and Breeding for High-quality Animal Products, Zhejiang, PR China
| | - Jiaqi Liu
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, PR China; Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, Zhejiang, PR China; Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou, Zhejiang, PR China; Zhejiang Key Laboratory of Nutrition and Breeding for High-quality Animal Products, Zhejiang, PR China
| | - Yushi Chen
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, PR China; Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, Zhejiang, PR China; Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou, Zhejiang, PR China; Zhejiang Key Laboratory of Nutrition and Breeding for High-quality Animal Products, Zhejiang, PR China
| | - Botao Zeng
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, PR China; Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, Zhejiang, PR China; Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou, Zhejiang, PR China; Zhejiang Key Laboratory of Nutrition and Breeding for High-quality Animal Products, Zhejiang, PR China
| | - Xing Liao
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, PR China; Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, Zhejiang, PR China; Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou, Zhejiang, PR China; Zhejiang Key Laboratory of Nutrition and Breeding for High-quality Animal Products, Zhejiang, PR China
| | - Yuxi Liu
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, PR China; Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, Zhejiang, PR China; Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou, Zhejiang, PR China; Zhejiang Key Laboratory of Nutrition and Breeding for High-quality Animal Products, Zhejiang, PR China
| | - Xinxia Wang
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, PR China; Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, Zhejiang, PR China; Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou, Zhejiang, PR China; Zhejiang Key Laboratory of Nutrition and Breeding for High-quality Animal Products, Zhejiang, PR China.
| |
Collapse
|
2
|
Ding Y, Wang S, Qi G, Hu Y, Huang J, Zhang Y, Lian K, Li Z, Li X, Kong W, Liu Y, Duan P, Yu H. Toxic effects and mechanisms of monometallic bismuth nanoparticles on early stages of zebrafish (Danio rerio). THE SCIENCE OF THE TOTAL ENVIRONMENT 2025; 983:179702. [PMID: 40398161 DOI: 10.1016/j.scitotenv.2025.179702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 04/25/2025] [Accepted: 05/16/2025] [Indexed: 05/23/2025]
Abstract
Bismuth nanoparticles (Bi NPs) have shown great potential in the fields of biomedical applications due to their favourable properties. However, the impacts and toxic mechanisms of monometallic Bi NPs on vertebrates remain poorly explored. In the present study, we utilised zebrafish (Danio rerio) as a model to evaluate the toxic effects of acute exposure to Bi NPs on early life stages. Bi NPs-treated embryos exhibited an increase in the number of malformations and decreased hatching. Exposure to 5 mg/L Bi NPs induced embryonic developmental abnormalities, including lipid transport, behavioural, and craniofacial deformities. Moreover, exposure to 25 mg/L Bi NPs caused embryonic developmental defects, including behavioural dysfunction, skeletal deformity, cardiac teratogenesis, and ocular abnormalities. Based on transcriptomics, we identified a total of 859 differentially expressed genes. Of these, 338 upregulated genes are mainly related to skeletal system development, circadian rhythm, and oxidative stress, while 519 downregulated genes are associated with visual perception and heart contraction. Furthermore, the carbon metabolism and peroxisome proliferator-activated receptor (PPAR) signalling pathways, which are essential for embryonic development, were significantly dysregulated by exposure to Bi NPs. This study innovatively focused on evaluating the embryonic developmental toxicities of monometallic Bi NPs on zebrafish and unveiled the underlying toxic mechanisms, highlighting the potential hazard and risk posed by Bi NPs and raising concerns regarding adverse impacts on aquatic organisms.
Collapse
Affiliation(s)
- Yiwei Ding
- Key Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases of Xiangyang City, Xiangyang No. 1 People's Hospital, Hubei University of Medicine, Xiangyang 441000, China; School of Public Health, Hubei University of Medicine, Shiyan 442000, China
| | - Songtao Wang
- Key Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases of Xiangyang City, Xiangyang No. 1 People's Hospital, Hubei University of Medicine, Xiangyang 441000, China; Hubei Provincial Clinical Research Center for Accurate Fetus Malformation Diagnosis, Department of Obstetrics and Gynecology, Xiangyang No. 1 People's Hospital, Hubei University of Medicine, Xiangyang 441000, China
| | - Guilin Qi
- Key Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases of Xiangyang City, Xiangyang No. 1 People's Hospital, Hubei University of Medicine, Xiangyang 441000, China; Hubei Provincial Clinical Research Center for Accurate Fetus Malformation Diagnosis, Department of Obstetrics and Gynecology, Xiangyang No. 1 People's Hospital, Hubei University of Medicine, Xiangyang 441000, China
| | - Yangyang Hu
- Key Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases of Xiangyang City, Xiangyang No. 1 People's Hospital, Hubei University of Medicine, Xiangyang 441000, China; School of Public Health, Hubei University of Medicine, Shiyan 442000, China
| | - Jiaolong Huang
- Key Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases of Xiangyang City, Xiangyang No. 1 People's Hospital, Hubei University of Medicine, Xiangyang 441000, China; Hubei Provincial Clinical Research Center for Accurate Fetus Malformation Diagnosis, Department of Obstetrics and Gynecology, Xiangyang No. 1 People's Hospital, Hubei University of Medicine, Xiangyang 441000, China
| | - Ying Zhang
- Key Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases of Xiangyang City, Xiangyang No. 1 People's Hospital, Hubei University of Medicine, Xiangyang 441000, China
| | - Kai Lian
- Key Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases of Xiangyang City, Xiangyang No. 1 People's Hospital, Hubei University of Medicine, Xiangyang 441000, China
| | - Zhiyong Li
- Key Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases of Xiangyang City, Xiangyang No. 1 People's Hospital, Hubei University of Medicine, Xiangyang 441000, China
| | - Xinyu Li
- Key Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases of Xiangyang City, Xiangyang No. 1 People's Hospital, Hubei University of Medicine, Xiangyang 441000, China
| | - Weimeng Kong
- Key Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases of Xiangyang City, Xiangyang No. 1 People's Hospital, Hubei University of Medicine, Xiangyang 441000, China; Hubei Provincial Clinical Research Center for Accurate Fetus Malformation Diagnosis, Department of Obstetrics and Gynecology, Xiangyang No. 1 People's Hospital, Hubei University of Medicine, Xiangyang 441000, China
| | - Ying Liu
- Key Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases of Xiangyang City, Xiangyang No. 1 People's Hospital, Hubei University of Medicine, Xiangyang 441000, China
| | - Peng Duan
- Key Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases of Xiangyang City, Xiangyang No. 1 People's Hospital, Hubei University of Medicine, Xiangyang 441000, China; Hubei Provincial Clinical Research Center for Accurate Fetus Malformation Diagnosis, Department of Obstetrics and Gynecology, Xiangyang No. 1 People's Hospital, Hubei University of Medicine, Xiangyang 441000, China.
| | - Hui Yu
- Key Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases of Xiangyang City, Xiangyang No. 1 People's Hospital, Hubei University of Medicine, Xiangyang 441000, China; School of Public Health, Hubei University of Medicine, Shiyan 442000, China; Hubei Provincial Clinical Research Center for Accurate Fetus Malformation Diagnosis, Department of Obstetrics and Gynecology, Xiangyang No. 1 People's Hospital, Hubei University of Medicine, Xiangyang 441000, China.
| |
Collapse
|
3
|
Dong S, Deng R, Zeng H, Xue P, Lin S, Zhou D, Mao L. Trophic transfer of carbon-14 from algae to zebrafish leads to its blending in biomolecules and the dysregulation of metabolism via isotope effect. Natl Sci Rev 2025; 12:nwae346. [PMID: 39777205 PMCID: PMC11706001 DOI: 10.1093/nsr/nwae346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 07/26/2024] [Accepted: 09/14/2024] [Indexed: 01/11/2025] Open
Abstract
Carbon-14 (C-14) has been a major contributor to the human radioactive exposure dose, as it is released into the environment from the nuclear industry in larger quantities compared to other radionuclides. This most abundant nuclide enters the biosphere as organically bound C-14 (OBC-14), posing a potential threat to public health. Yet, it remains unknown how this relatively low radiotoxic nuclide induces health risks via chemical effects, such as isotope effect. By establishing a trophic transfer model involving algae (Scenedesmus obliquus), daphnia (Daphnia magna) and zebrafish (Danio rerio), we demonstrate that rapid incorporation and transformation of inorganic C-14 by algae into OBC-14 facilitates the blending of C-14 into the biomolecules of zebrafish. We find that internalized C-14 is persistently retained in the brain of zebrafish, affecting DNA methylation and causing alterations in neuropathology. Global isotope tracing metabolomics with C-14 exposure further reveals the involvement of C-14 in various critical metabolic pathways, including one-carbon metabolism and nucleotide metabolism. We thus characterize the kinetic isotope effects for 12C/14C in the key reactions of these metabolic pathways through kinetic experiments and density functional theory computations, showing that the isotopic substitution of carbon in biochemicals regulates metabolism by disrupting reaction ratios via isotope effects. Our results suggest that inorganic C-14 discharged by the nuclear industry can be biotransformed into OBC-14 to impact metabolism via isotope effects, providing new insights into understanding the health risk of C-14, which is traditionally considered as a low radiotoxic nuclide.
Collapse
Affiliation(s)
- Shipeng Dong
- State Key Laboratory of Pollution Control and Resource Reuse, School of the Environment, Chemistry and Biomedicine Innovation Center, Nanjing University, Nanjing 210023, China
| | - Renquan Deng
- State Key Laboratory of Pollution Control and Resource Reuse, School of the Environment, Chemistry and Biomedicine Innovation Center, Nanjing University, Nanjing 210023, China
| | - Hang Zeng
- State Key Laboratory of Pollution Control and Resource Reuse, School of the Environment, Chemistry and Biomedicine Innovation Center, Nanjing University, Nanjing 210023, China
| | - Pengfei Xue
- State Key Laboratory of Pollution Control and Resource Reuse, School of the Environment, Chemistry and Biomedicine Innovation Center, Nanjing University, Nanjing 210023, China
| | - Sijie Lin
- College of Environmental Science & Engineering, State Key Laboratory of Pollution Control and Resource Reuse, Tongji University, Shanghai 200092, China
| | - Dongmei Zhou
- State Key Laboratory of Pollution Control and Resource Reuse, School of the Environment, Chemistry and Biomedicine Innovation Center, Nanjing University, Nanjing 210023, China
| | - Liang Mao
- State Key Laboratory of Pollution Control and Resource Reuse, School of the Environment, Chemistry and Biomedicine Innovation Center, Nanjing University, Nanjing 210023, China
| |
Collapse
|
4
|
He J, Wang A, Zhao Q, Zou Y, Zhang Z, Sha N, Hou G, Zhou B, Yang Y, Chen T, Zhao Y, Jiang Y. RNAi screens identify HES4 as a regulator of redox balance supporting pyrimidine synthesis and tumor growth. Nat Struct Mol Biol 2024; 31:1413-1425. [PMID: 38769389 DOI: 10.1038/s41594-024-01309-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 04/08/2024] [Indexed: 05/22/2024]
Abstract
NADH/NAD+ redox balance is pivotal for cellular metabolism. Systematic identification of NAD(H) redox regulators, although currently lacking, would help uncover unknown effectors critically implicated in the coordination of growth metabolism. In this study, we performed a genome-scale RNA interference (RNAi) screen to globally survey the genes involved in redox modulation and identified the HES family bHLH transcription factor HES4 as a negative regulator of NADH/NAD+ ratio. Functionally, HES4 is shown to be crucial for maintaining mitochondrial electron transport chain (ETC) activity and pyrimidine synthesis. More specifically, HES4 directly represses transcription of SLC44A2 and SDS, thereby inhibiting mitochondrial choline oxidation and cytosolic serine deamination, respectively, which, in turn, ensures coenzyme Q reduction capacity for DHODH-mediated UMP synthesis and serine-derived dTMP production. Accordingly, inhibition of choline oxidation preserves mitochondrial serine catabolism and ETC-coupled redox balance. Furthermore, HES4 protein stability is enhanced under EGFR activation, and increased HES4 levels facilitate EGFR-driven tumor growth and predict poor prognosis of lung adenocarcinoma. These findings illustrate an unidentified mechanism, underlying pyrimidine biosynthesis in the intersection between serine and choline catabolism, and underscore the physiological importance of HES4 in tumor metabolism.
Collapse
Affiliation(s)
- Jing He
- Department of Liver Surgery and Shanghai Cancer Institute, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Aoxue Wang
- Optogenetics & Synthetic Biology Interdisciplinary Research Center, Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, State Key Laboratory of Bioreactor Engineering, School of Pharmacy, East China University of Science and Technology, Shanghai, China
- Research Unit of New Techniques for Live-Cell Metabolic Imaging, Chinese Academy of Medical Sciences, Beijing, China
| | - Qin Zhao
- Department of Liver Surgery and Shanghai Cancer Institute, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yejun Zou
- Optogenetics & Synthetic Biology Interdisciplinary Research Center, Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, State Key Laboratory of Bioreactor Engineering, School of Pharmacy, East China University of Science and Technology, Shanghai, China
- Research Unit of New Techniques for Live-Cell Metabolic Imaging, Chinese Academy of Medical Sciences, Beijing, China
- Department of Gynecology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Zhuo Zhang
- Optogenetics & Synthetic Biology Interdisciplinary Research Center, Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, State Key Laboratory of Bioreactor Engineering, School of Pharmacy, East China University of Science and Technology, Shanghai, China
- Research Unit of New Techniques for Live-Cell Metabolic Imaging, Chinese Academy of Medical Sciences, Beijing, China
| | - Nannan Sha
- Department of Liver Surgery and Shanghai Cancer Institute, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Guofang Hou
- Department of Liver Surgery and Shanghai Cancer Institute, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bei Zhou
- Department of Liver Surgery and Shanghai Cancer Institute, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yi Yang
- Optogenetics & Synthetic Biology Interdisciplinary Research Center, Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, State Key Laboratory of Bioreactor Engineering, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Tao Chen
- Endoscopy Center, Department of Gastroenterology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yuzheng Zhao
- Optogenetics & Synthetic Biology Interdisciplinary Research Center, Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, State Key Laboratory of Bioreactor Engineering, School of Pharmacy, East China University of Science and Technology, Shanghai, China.
- Research Unit of New Techniques for Live-Cell Metabolic Imaging, Chinese Academy of Medical Sciences, Beijing, China.
| | - Yuhui Jiang
- Department of Liver Surgery and Shanghai Cancer Institute, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
5
|
Kadam I, Trasino SE, Korsmo H, Lucas J, Pinkas M, Jiang X. Prenatal Choline Supplementation Improves Glucose Tolerance and Reduces Liver Fat Accumulation in Mouse Offspring Exposed to Ethanol during the Prenatal and Postnatal Periods. Nutrients 2024; 16:1264. [PMID: 38732511 PMCID: PMC11085373 DOI: 10.3390/nu16091264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 04/18/2024] [Accepted: 04/21/2024] [Indexed: 05/13/2024] Open
Abstract
Prenatal alcohol exposure (AE) affects cognitive development. However, it is unclear whether prenatal AE influences the metabolic health of offspring and whether postnatal AE exacerbates metabolic deterioration resulting from prenatal AE. Choline is a semi-essential nutrient that has been demonstrated to mitigate the cognitive impairment of prenatal AE. This study investigated how maternal choline supplementation (CS) may modify the metabolic health of offspring with prenatal and postnatal AE (AE/AE). C57BL/6J female mice were fed either a Lieber-DeCarli diet with 1.4% ethanol between embryonic day (E) 9.5 and E17.5 or a control diet. Choline was supplemented with 4 × concentrations versus the control throughout pregnancy. At postnatal week 7, offspring mice were exposed to 1.4% ethanol for females and 3.9% ethanol for males for 4 weeks. AE/AE increased hepatic triglyceride accumulation in male offspring only, which was normalized by prenatal CS. Prenatal CS also improved glucose tolerance compared to AE/AE animals. AE/AE suppressed hepatic gene expression of peroxisome proliferator activated receptor alpha (Ppara) and low-density lipoprotein receptor (Ldlr), which regulate fatty acid catabolism and cholesterol reuptake, respectively, in male offspring. However, these changes were not rectified by prenatal CS. In conclusion, AE/AE led to an increased risk of steatosis and was partially prevented by prenatal CS in male mice.
Collapse
Affiliation(s)
- Isma’il Kadam
- PhD Program in Biochemistry, Graduate Center of the City University of New York, New York, NY 10016, USA; (I.K.); (H.K.)
- Department of Health and Nutrition Sciences, Brooklyn College of the City University of New York, Brooklyn, NY 11210, USA; (J.L.); (M.P.)
| | - Steven E. Trasino
- Nutrition Program, School of Urban Public Health, Hunter College, City University of New York, New York, NY 10065, USA
| | - Hunter Korsmo
- PhD Program in Biochemistry, Graduate Center of the City University of New York, New York, NY 10016, USA; (I.K.); (H.K.)
- Department of Health and Nutrition Sciences, Brooklyn College of the City University of New York, Brooklyn, NY 11210, USA; (J.L.); (M.P.)
| | - Jessica Lucas
- Department of Health and Nutrition Sciences, Brooklyn College of the City University of New York, Brooklyn, NY 11210, USA; (J.L.); (M.P.)
| | - Myriam Pinkas
- Department of Health and Nutrition Sciences, Brooklyn College of the City University of New York, Brooklyn, NY 11210, USA; (J.L.); (M.P.)
| | - Xinyin Jiang
- PhD Program in Biochemistry, Graduate Center of the City University of New York, New York, NY 10016, USA; (I.K.); (H.K.)
- Department of Health and Nutrition Sciences, Brooklyn College of the City University of New York, Brooklyn, NY 11210, USA; (J.L.); (M.P.)
| |
Collapse
|
6
|
Deng Y, Hu M, Huang S, Fu N. Molecular mechanism and therapeutic significance of essential amino acids in metabolically associated fatty liver disease. J Nutr Biochem 2024; 126:109581. [PMID: 38219809 DOI: 10.1016/j.jnutbio.2024.109581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 01/01/2024] [Accepted: 01/06/2024] [Indexed: 01/16/2024]
Abstract
Non-alcoholic fatty liver disease (NAFLD), also known as metabolically associated fatty liver disease (MAFLD), is a systemic metabolic disease characterized by lipid accumulation in the liver, lipid toxicity, insulin resistance, intestinal dysbiosis, and inflammation that can progress from simple steatosis to nonalcoholic steatohepatitis (NASH) and even cirrhosis or cancer. It is the most prevalent illness threatening world health. Currently, there are almost no approved drug interventions for MAFLD, mainly dietary changes and exercise to control weight and regulate metabolic disorders. Meanwhile, the metabolic pathway involved in amino acid metabolism also influences the onset and development of MAFLD in the body, and most amino acid metabolism takes place in the liver. Essential amino acids are those amino acids that must be supplemented from outside the diet and that cannot be synthesized in the body or cannot be synthesized at a rate sufficient to meet the body's needs, including leucine, isoleucine, valine (collectively known as branched-chain amino acids), tryptophan, phenylalanine (which are aromatic amino acids), histidine, methionine, threonine and lysine. The metabolic balance of the body is closely linked to these essential amino acids, and essential amino acids are closely linked to the pathophysiological process of MAFLD. In this paper, we will focus on the metabolism of essential amino acids in the body and further explore the therapeutic strategies for MAFLD based on the studies conducted in recent years.
Collapse
Affiliation(s)
- Yuting Deng
- The Affiliated Nanhua Hospital, Department of Gastroenterology, Hunan Provincial Clinical Research Center of Metabolic Associated Fatty Liver Disease, Hengyang Medical School, University of South China, Hengyang, Hunan, 421002, China
| | - Mengsi Hu
- The Affiliated Nanhua Hospital, Department of Gastroenterology, Hunan Provincial Clinical Research Center of Metabolic Associated Fatty Liver Disease, Hengyang Medical School, University of South China, Hengyang, Hunan, 421002, China
| | - Shufang Huang
- The Affiliated Nanhua Hospital, Hunan Provincial Clinical Research Center of Metabolic Associated Fatty Liver Disease, Hengyang Medical School, University of South China, Hengyang, Hunan, 421002, China.
| | - Nian Fu
- The Affiliated Nanhua Hospital, Department of Gastroenterology, Hunan Provincial Clinical Research Center of Metabolic Associated Fatty Liver Disease, Hengyang Medical School, University of South China, Hengyang, Hunan, 421002, China; The Affiliated Nanhua Hospital, Institute of Clinical Research, Hengyang Medical School, University of South China, Hengyang, Hunan, 421002, China.
| |
Collapse
|
7
|
Ghrayeb A, Finney AC, Agranovich B, Peled D, Anand SK, McKinney MP, Sarji M, Yang D, Weissman N, Drucker S, Fernandes SI, Fernández-García J, Mahan K, Abassi Z, Tan L, Lorenzi PL, Traylor J, Zhang J, Abramovich I, Chen YE, Rom O, Mor I, Gottlieb E. Serine synthesis via reversed SHMT2 activity drives glycine depletion and acetaminophen hepatotoxicity in MASLD. Cell Metab 2024; 36:116-129.e7. [PMID: 38171331 PMCID: PMC10777734 DOI: 10.1016/j.cmet.2023.12.013] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 10/27/2023] [Accepted: 12/08/2023] [Indexed: 01/05/2024]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) affects one-third of the global population. Understanding the metabolic pathways involved can provide insights into disease progression and treatment. Untargeted metabolomics of livers from mice with early-stage steatosis uncovered decreased methylated metabolites, suggesting altered one-carbon metabolism. The levels of glycine, a central component of one-carbon metabolism, were lower in mice with hepatic steatosis, consistent with clinical evidence. Stable-isotope tracing demonstrated that increased serine synthesis from glycine via reverse serine hydroxymethyltransferase (SHMT) is the underlying cause for decreased glycine in steatotic livers. Consequently, limited glycine availability in steatotic livers impaired glutathione synthesis under acetaminophen-induced oxidative stress, enhancing acute hepatotoxicity. Glycine supplementation or hepatocyte-specific ablation of the mitochondrial SHMT2 isoform in mice with hepatic steatosis mitigated acetaminophen-induced hepatotoxicity by supporting de novo glutathione synthesis. Thus, early metabolic changes in MASLD that limit glycine availability sensitize mice to xenobiotics even at the reversible stage of this disease.
Collapse
Affiliation(s)
- Alia Ghrayeb
- Ruth and Bruce Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa 31096, Israel
| | - Alexandra C Finney
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA 71103, USA
| | - Bella Agranovich
- Ruth and Bruce Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa 31096, Israel
| | - Daniel Peled
- Ruth and Bruce Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa 31096, Israel
| | - Sumit Kumar Anand
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA 71103, USA
| | - M Peyton McKinney
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA 71103, USA
| | - Mahasen Sarji
- Ruth and Bruce Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa 31096, Israel
| | - Dongshan Yang
- Center for Advanced Models for Translational Sciences and Therapeutics, University of Michigan Medical Center, Ann Arbor, MI 48109, USA; Department of Internal Medicine, Frankel Cardiovascular Center, University of Michigan, Ann Arbor, MI 48109, USA
| | - Natan Weissman
- Ruth and Bruce Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa 31096, Israel
| | - Shani Drucker
- Ruth and Bruce Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa 31096, Israel
| | - Sara Isabel Fernandes
- Ruth and Bruce Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa 31096, Israel
| | - Jonatan Fernández-García
- Ruth and Bruce Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa 31096, Israel
| | - Kyle Mahan
- Ruth and Bruce Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa 31096, Israel
| | - Zaid Abassi
- Ruth and Bruce Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa 31096, Israel
| | - Lin Tan
- Metabolomics Core Facility, Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Philip L Lorenzi
- Metabolomics Core Facility, Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - James Traylor
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA 71103, USA
| | - Jifeng Zhang
- Center for Advanced Models for Translational Sciences and Therapeutics, University of Michigan Medical Center, Ann Arbor, MI 48109, USA; Department of Internal Medicine, Frankel Cardiovascular Center, University of Michigan, Ann Arbor, MI 48109, USA
| | - Ifat Abramovich
- Ruth and Bruce Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa 31096, Israel
| | - Y Eugene Chen
- Center for Advanced Models for Translational Sciences and Therapeutics, University of Michigan Medical Center, Ann Arbor, MI 48109, USA; Department of Internal Medicine, Frankel Cardiovascular Center, University of Michigan, Ann Arbor, MI 48109, USA
| | - Oren Rom
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA 71103, USA; Department of Internal Medicine, Frankel Cardiovascular Center, University of Michigan, Ann Arbor, MI 48109, USA; Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA 71103, USA; Center for Cardiovascular Diseases and Sciences, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA 71103, USA.
| | - Inbal Mor
- Ruth and Bruce Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa 31096, Israel.
| | - Eyal Gottlieb
- Ruth and Bruce Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa 31096, Israel; Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA.
| |
Collapse
|
8
|
Rice PA, Kabadi SV, Doerge DR, Vanlandingham MM, Churchwell MI, Tryndyak VP, Fisher JW, Aungst J, Beland FA. Evaluating the toxicokinetics of some metabolites of a C6 polyfluorinated compound, 6:2 fluorotelomer alcohol in pregnant and nonpregnant rats after oral exposure to the parent compound. Food Chem Toxicol 2024; 183:114333. [PMID: 38061571 DOI: 10.1016/j.fct.2023.114333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 11/28/2023] [Accepted: 11/30/2023] [Indexed: 12/19/2023]
Abstract
The 6:2 fluorotelomer alcohol (6:2 FTOH) is a common impurity in per- and polyfluoroalkyl substances (PFASs) used in many applications. Our previous toxicokinetic (TK) evaluation of 6:2 FTOH calculated times to steady state (tss) of one of its metabolites, 5:3 fluorotelomer carboxylic acid (5:3A), in the plasma and tissues of up to a year after oral exposure to rats. Our current work further elucidated the TK of 5:3A and other metabolites of 6:2 FTOH in pregnant and nonpregnant rats after repeated oral exposure and examined the role of renal transporters in the biopersistence of 5:3A. The tss values for 5:3A in serum and tissues of adult nonpregnant animals ranged from 150 days to over a year. 4:3 fluorotelomer carboxylic acid (4:3A) was an additional potentially-biopersistent metabolite. 5:3A was the major metabolite of 6:2 FTOH in serum of pregnant dams and fetuses at each time interval. 5:3A was not a substrate for renal transporters in a human kidney cell line in vitro, indicating that renal reuptake of 5:3A is unlikely contribute to its biopersistence. Further research is needed to identify the underlying processes and evaluate the impact of these 6:2 FTOH metabolites on human health.
Collapse
Affiliation(s)
- Penelope A Rice
- FDA/CFSAN/OFAS/DFCN, 5001 Campus Drive, HFS 275, College Park, MD, 20740, USA.
| | - Shruti V Kabadi
- FDA/CFSAN/OFAS/DFCN, 5001 Campus Drive, HFS 275, College Park, MD, 20740, USA
| | | | | | | | | | | | - Jason Aungst
- FDA/CFSAN/OFAS/DFCN, 5001 Campus Drive, HFS 275, College Park, MD, 20740, USA
| | | |
Collapse
|
9
|
Wen J, Fei Y, Yuan L, Li K, Xu Q, Cao X, Su J, Zhu Y, Zhang Z. Analysis of the mediating role of BMI in associations of different folate forms with hepatic steatosis and liver fibrosis in adolescents in the USA: results from the NHANES 2017-2018. Front Endocrinol (Lausanne) 2023; 14:1273580. [PMID: 38116318 PMCID: PMC10728716 DOI: 10.3389/fendo.2023.1273580] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Accepted: 11/20/2023] [Indexed: 12/21/2023] Open
Abstract
Background Most studies have explored the relationship between serum total folate and nonalcoholic fatty liver disease (NAFLD) in adults, but there has been no study on the relationship between different folate forms and hepatic steatosis or liver stiffness in adolescents. Objective To investigate the association of different folate forms with hepatic steatosis or liver stiffness in adolescents, and further explore the intermediary role of BMI in this relationship. Methods The cross-sectional study included 549 participants from the 2017-2018 National Health and Nutrition Inspection Survey (NHANES) survey cycle who had complete data. Four folate data (red blood cell folate, serum total folate, 5-methyl-tetrahydrofolate and folic acid) were included in our study. Controlled attenuation parameters (CAP) and liver stiffness came from the results of liver ultrasound transient elastography. We used linear regression to analyze the relationship between different forms of folate and CAP or liver stiffness, and logistic regression to analyze the relationship between different forms of folate and NAFLD or significant fibrosis. We also used restricted cubic splines to analyze the nonlinear relationship between different forms of folate and NAFLD or significant fibrosis. Finally, we used regression-based intermediary analysis to distinguish the direct and BMI-mediated effects of folate on CAP or liver stiffness. All the analyses adjusted the relevant covariates. Results The means of CAP and liver hardness in this study were 223.02dB/m and 5.03kPa, respectively. We found that in model 2, there was a negative correlation between serum total folate (β: -18.53; 95%CI: -29.32 to -7.73) or 5-methyltetrahydrofolate (β: -14.13; 95%CI: -28.98 to -7.86) and CAP. However, when the BMI was further adjusted in model 3, this negative correlation no longer existed (serum total folate: β: -8.36; 95%CI: -17.69 to 0.97; 5-methyltetrahydrofolate: β: -8.05; 95%CI: -17.19 to 1.09). Similarly, we found a negative correlation between serum total folate or 5-Methyl-tetrahydrofolate and liver stiffness in model 2. There was no significant correlation between red blood cell folate or folic acid and CAP or liver stiffness in either model 2 or model 3. The nonlinear relationship between different folate forms and NAFLD or significant fibrosis was not significant. It is estimated that 76% of the total association between serum total folate and CAP is mediated by BMI. The mediating proportion of BMI in the total correlation between serum total folate and liver stiffness was 50%. Similarly, we found that BMI significantly mediated the relationship between 5-Methyl-tetrahydrofolate and CAP or liver stiffness, with a mediating ratio of 77% and 49%, respectively. Conclusion Our results show that serum total folate or 5-Methyl-tetrahydrofolate are negatively correlated with hepatic steatosis or liver stiffness in adolescents, and BMI plays major mediating role in this relationship. Our findings emphasize the importance of monitoring the concentration of serum folate, not just the serum total folate concentration.
Collapse
Affiliation(s)
- Jingli Wen
- Department of Infection, The Affiliated Suqian first people's Hospital of Nanjing Medical University, Suqian, JiangSu, China
| | - Yuanyuan Fei
- Department of Infection, The Affiliated Suqian first people's Hospital of Nanjing Medical University, Suqian, JiangSu, China
| | - Ling Yuan
- Department of Infection, The Affiliated Suqian first people's Hospital of Nanjing Medical University, Suqian, JiangSu, China
| | - Kai Li
- Department of Infection, The Affiliated Suqian first people's Hospital of Nanjing Medical University, Suqian, JiangSu, China
| | - Qian Xu
- Department of Infection, The Affiliated Zhangjiagang Hospital of Soochow University, Suqian, JiangSu, China
| | - Xueyan Cao
- Department of Infection, The Affiliated Lianyungang Hospital of Xuzhou Medical University, Suqian, JiangSu, China
| | - Jing Su
- Laboratory of Department of hematology, The Affiliated Suqian first people's Hospital of Nanjing Medical University, Suqian, JiangSu, China
| | - Yujing Zhu
- Clinical Research Center, The Affiliated Suqian first people's Hospital of Nanjing Medical University, Suqian, JiangSu, China
| | - Zhenjiang Zhang
- Department of Infection, The Affiliated Suqian first people's Hospital of Nanjing Medical University, Suqian, JiangSu, China
| |
Collapse
|
10
|
Herranz JM, López-Pascual A, Clavería-Cabello A, Uriarte I, Latasa MU, Irigaray-Miramon A, Adán-Villaescusa E, Castelló-Uribe B, Sangro B, Arechederra M, Berasain C, Avila MA, Fernández-Barrena MG. Comprehensive analysis of epigenetic and epitranscriptomic genes' expression in human NAFLD. J Physiol Biochem 2023; 79:901-924. [PMID: 37620598 PMCID: PMC10636027 DOI: 10.1007/s13105-023-00976-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 07/19/2023] [Indexed: 08/26/2023]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a multifactorial condition with a complex etiology. Its incidence is increasing globally in parallel with the obesity epidemic, and it is now considered the most common liver disease in Western countries. The precise mechanisms underlying the development and progression of NAFLD are complex and still poorly understood. The dysregulation of epigenetic and epitranscriptomic mechanisms is increasingly recognized to play pathogenic roles in multiple conditions, including chronic liver diseases. Here, we have performed a comprehensive analysis of the expression of epigenetic and epitranscriptomic genes in a total of 903 liver tissue samples corresponding to patients with normal liver, obese patients, and patients with non-alcoholic fatty liver (NAFL) and non-alcoholic steatohepatitis (NASH), advancing stages in NAFLD progression. We integrated ten transcriptomic datasets in an unbiased manner, enabling their robust analysis and comparison. We describe the complete landscape of epigenetic and epitranscriptomic genes' expression along the course of the disease. We identify signatures of genes significantly dysregulated in association with disease progression, particularly with liver fibrosis development. Most of these epigenetic and epitranscriptomic effectors have not been previously described in human NAFLD, and their altered expression may have pathogenic implications. We also performed a comprehensive analysis of the expression of enzymes involved in the metabolism of the substrates and cofactors of epigenetic and epitranscriptomic effectors. This study provides novel information on NAFLD pathogenesis and may also guide the identification of drug targets to treat this condition and its progression towards hepatocellular carcinoma.
Collapse
Affiliation(s)
- Jose M Herranz
- Hepatology Laboratory, Solid Tumors Program, CIMA, CCUN, University of Navarra, Pamplona, Spain
- CIBERehd, Instituto de Salud Carlos III, Madrid, Spain
| | - Amaya López-Pascual
- Hepatology Unit, CCUN, Navarra University Clinic, Pamplona, Spain
- Instituto de Investigaciones Sanitarias de Navarra IdiSNA, Pamplona, Spain
| | - Alex Clavería-Cabello
- Hepatology Laboratory, Solid Tumors Program, CIMA, CCUN, University of Navarra, Pamplona, Spain
| | - Iker Uriarte
- Hepatology Laboratory, Solid Tumors Program, CIMA, CCUN, University of Navarra, Pamplona, Spain
- CIBERehd, Instituto de Salud Carlos III, Madrid, Spain
| | - M Ujúe Latasa
- Hepatology Laboratory, Solid Tumors Program, CIMA, CCUN, University of Navarra, Pamplona, Spain
| | - Ainara Irigaray-Miramon
- Hepatology Laboratory, Solid Tumors Program, CIMA, CCUN, University of Navarra, Pamplona, Spain
| | - Elena Adán-Villaescusa
- Hepatology Laboratory, Solid Tumors Program, CIMA, CCUN, University of Navarra, Pamplona, Spain
| | - Borja Castelló-Uribe
- Hepatology Laboratory, Solid Tumors Program, CIMA, CCUN, University of Navarra, Pamplona, Spain
| | - Bruno Sangro
- CIBERehd, Instituto de Salud Carlos III, Madrid, Spain
- Hepatology Unit, CCUN, Navarra University Clinic, Pamplona, Spain
- Instituto de Investigaciones Sanitarias de Navarra IdiSNA, Pamplona, Spain
| | - María Arechederra
- Hepatology Laboratory, Solid Tumors Program, CIMA, CCUN, University of Navarra, Pamplona, Spain
- CIBERehd, Instituto de Salud Carlos III, Madrid, Spain
- Instituto de Investigaciones Sanitarias de Navarra IdiSNA, Pamplona, Spain
| | - Carmen Berasain
- Hepatology Laboratory, Solid Tumors Program, CIMA, CCUN, University of Navarra, Pamplona, Spain
- CIBERehd, Instituto de Salud Carlos III, Madrid, Spain
| | - Matías A Avila
- Hepatology Laboratory, Solid Tumors Program, CIMA, CCUN, University of Navarra, Pamplona, Spain
- CIBERehd, Instituto de Salud Carlos III, Madrid, Spain
- Instituto de Investigaciones Sanitarias de Navarra IdiSNA, Pamplona, Spain
| | - Maite G Fernández-Barrena
- Hepatology Laboratory, Solid Tumors Program, CIMA, CCUN, University of Navarra, Pamplona, Spain.
- CIBERehd, Instituto de Salud Carlos III, Madrid, Spain.
- Instituto de Investigaciones Sanitarias de Navarra IdiSNA, Pamplona, Spain.
| |
Collapse
|
11
|
Gama-Almeida MC, Pinto GDA, Teixeira L, Hottz ED, Ivens P, Ribeiro H, Garrett R, Torres AG, Carneiro TIA, Barbalho BDO, Ludwig C, Struchiner CJ, Assunção-Miranda I, Valente APC, Bozza FA, Bozza PT, Dos Santos GC, El-Bacha T. Integrated NMR and MS Analysis of the Plasma Metabolome Reveals Major Changes in One-Carbon, Lipid, and Amino Acid Metabolism in Severe and Fatal Cases of COVID-19. Metabolites 2023; 13:879. [PMID: 37512587 PMCID: PMC10384698 DOI: 10.3390/metabo13070879] [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: 06/12/2023] [Revised: 07/15/2023] [Accepted: 07/20/2023] [Indexed: 07/30/2023] Open
Abstract
Brazil has the second-highest COVID-19 death rate worldwide, and Rio de Janeiro is among the states with the highest rate in the country. Although vaccine coverage has been achieved, it is anticipated that COVID-19 will transition into an endemic disease. It is concerning that the molecular mechanisms underlying clinical evolution from mild to severe disease, as well as the mechanisms leading to long COVID-19, are not yet fully understood. NMR and MS-based metabolomics were used to identify metabolites associated with COVID-19 pathophysiology and disease outcome. Severe COVID-19 cases (n = 35) were enrolled in two reference centers in Rio de Janeiro within 72 h of ICU admission, alongside 12 non-infected control subjects. COVID-19 patients were grouped into survivors (n = 18) and non-survivors (n = 17). Choline-related metabolites, serine, glycine, and betaine, were reduced in severe COVID-19, indicating dysregulation in methyl donors. Non-survivors had higher levels of creatine/creatinine, 4-hydroxyproline, gluconic acid, and N-acetylserine, indicating liver and kidney dysfunction. Several changes were greater in women; thus, patients' sex should be considered in pandemic surveillance to achieve better disease stratification and improve outcomes. These metabolic alterations may be useful to monitor organ (dys) function and to understand the pathophysiology of acute and possibly post-acute COVID-19 syndromes.
Collapse
Affiliation(s)
- Marcos C Gama-Almeida
- LeBioME-Bioactives, Mitochondrial and Placental Metabolism Core, Institute of Nutrition Josué de Castro, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Gabriela D A Pinto
- LeBioME-Bioactives, Mitochondrial and Placental Metabolism Core, Institute of Nutrition Josué de Castro, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Lívia Teixeira
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro 21041-361, Brazil
| | - Eugenio D Hottz
- Laboratory of Immunothrombosis, Department of Biochemistry, Federal University of Juiz de Fora, Juiz de Fora 36936-900, Brazil
| | - Paula Ivens
- LabMeta, Metabolomics Laboratory, Institute of Chemistry, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-598, Brazil
| | - Hygor Ribeiro
- LabMeta, Metabolomics Laboratory, Institute of Chemistry, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-598, Brazil
- Lipid Biochemistry and Lipidomics Laboratory, Department of Chemistry, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-598, Brazil
| | - Rafael Garrett
- LabMeta, Metabolomics Laboratory, Institute of Chemistry, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-598, Brazil
| | - Alexandre G Torres
- LeBioME-Bioactives, Mitochondrial and Placental Metabolism Core, Institute of Nutrition Josué de Castro, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
- Lipid Biochemistry and Lipidomics Laboratory, Department of Chemistry, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-598, Brazil
| | - Talita I A Carneiro
- LeBioME-Bioactives, Mitochondrial and Placental Metabolism Core, Institute of Nutrition Josué de Castro, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Bianca de O Barbalho
- LeBioME-Bioactives, Mitochondrial and Placental Metabolism Core, Institute of Nutrition Josué de Castro, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Christian Ludwig
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham B15 2SQ, UK
| | - Claudio J Struchiner
- School of Applied Mathematics, Fundação Getúlio Vargas, Rio de Janeiro 22231-080, Brazil
- Institute of Social Medicine, Universidade do Estado do Rio de Janeiro, Rio de Janeiro 20550-013, Brazil
| | - Iranaia Assunção-Miranda
- LaRIV, Instituto de Microbiologia Paulo de Goes, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Ana Paula C Valente
- National Center for Nuclear Magnetic Resonance-Jiri Jonas, Institute of Medical Biochemistry, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Fernando A Bozza
- National Institute of Infectious Disease Evandro Chagas, Oswaldo Cruz Foundation, Rio de Janeiro 21040-360, Brazil
- D'Or Institute for Research and Education, Rio de Janeiro 22281-100, Brazil
| | - Patrícia T Bozza
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro 21041-361, Brazil
| | - Gilson C Dos Santos
- LabMet-Laboratory of Metabolomics, Instituto de Biologia Roberto Alcantara Gomes (IBRAG), Department of Genetics, State University of Rio de Janeiro, Rio de Janeiro 20551-030, Brazil
| | - Tatiana El-Bacha
- LeBioME-Bioactives, Mitochondrial and Placental Metabolism Core, Institute of Nutrition Josué de Castro, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
- Lipid Biochemistry and Lipidomics Laboratory, Department of Chemistry, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-598, Brazil
| |
Collapse
|
12
|
Warren D, Benedito VA, Skinner RC, Alawadi A, Vendemiatti E, Laub DJ, Showman C, Matak K, Tou JC. Low-Protein Diets Composed of Protein Recovered from Food Processing Supported Growth, but Induced Mild Hepatic Steatosis Compared with a No-Protein Diet in Young Female Rats. J Nutr 2023; 153:1668-1679. [PMID: 36990182 PMCID: PMC10447611 DOI: 10.1016/j.tjnut.2023.03.028] [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: 01/01/2023] [Revised: 03/08/2023] [Accepted: 03/20/2023] [Indexed: 03/29/2023] Open
Abstract
BACKGROUND Living in low-income countries often restricts the consumption of adequate protein and animal protein. OBJECTIVES This study aimed to investigate the effects of feeding low-protein diets on growth and liver health using proteins recovered from animal processing. METHODS Female Sprague-Dawley rats (aged 28 d) were randomly assigned (n = 8 rats/group) to be fed standard purified diets with 0% or 10% kcal protein that was comprised of either carp, whey, or casein. RESULTS Rats that were fed low-protein diets showed higher growth but developed mild hepatic steatosis compared to rats that were fed a no-protein diet, regardless of the protein source. Real-time quantitative polymerase chain reactions targeting the expression of genes involved in liver lipid homeostasis were not significantly different among groups. Global RNA-sequencing technology identified 9 differentially expressed genes linked to folate-mediated 1-carbon metabolism, endoplasmic reticulum (ER) stress, and metabolic diseases. Canonical pathway analysis revealed that mechanisms differed depending on the protein source. ER stress and dysregulated energy metabolism were implicated in hepatic steatosis in carp- and whey-fed rats. In contrast, impaired liver one-carbon methylations, lipoprotein assembly, and lipid export were implicated in casein-fed rats. CONCLUSIONS Carp sarcoplasmic protein showed comparable results to commercially available casein and whey protein. A better understanding of the molecular mechanisms in hepatic steatosis development can assist formulation of proteins recovered from food processing into a sustainable source of high-quality protein.
Collapse
Affiliation(s)
- Derek Warren
- Division of Animal and Nutritional Sciences, West Virginia University, Morgantown, WV, United States; Department of Biology, University of the Ozarks, Clarksville, AR, United States
| | - Vagner A Benedito
- Division of Plant and Soil Sciences, West Virginia University, Morgantown, WV, United States
| | - R Chris Skinner
- Food Systems Research Center, College of Agriculture and Life Sciences, University of Vermont Burlington, VT, United States
| | - Ayad Alawadi
- Division of Animal and Nutritional Sciences, West Virginia University, Morgantown, WV, United States
| | - Eloisa Vendemiatti
- Division of Plant and Soil Sciences, West Virginia University, Morgantown, WV, United States
| | - David J Laub
- Department of Biology, West Virginia University, Morgantown, WV, United States
| | - Casey Showman
- Division of Animal and Nutritional Sciences, West Virginia University, Morgantown, WV, United States
| | - Kristen Matak
- Division of Animal and Nutritional Sciences, West Virginia University, Morgantown, WV, United States
| | - Janet C Tou
- Division of Animal and Nutritional Sciences, West Virginia University, Morgantown, WV, United States.
| |
Collapse
|
13
|
Guo Z, Ma Y, Wang Y, Xiang H, Cui H, Fan Z, Zhu Y, Xing D, Chen B, Tao H, Guo Z, Wu X. Identification and validation of metabolism-related genes signature and immune infiltration landscape of rheumatoid arthritis based on machine learning. Aging (Albany NY) 2023; 15:3807-3825. [PMID: 37166429 PMCID: PMC10449312 DOI: 10.18632/aging.204714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 05/01/2023] [Indexed: 05/12/2023]
Abstract
Rheumatoid arthritis (RA) causes irreversible joint damage, but the pathogenesis is unknown. Therefore, it is crucial to identify diagnostic biomarkers of RA metabolism-related genes (MRGs). This study obtained transcriptome data from healthy individuals (HC) and RA patients from the GEO database. Weighted gene correlation network analysis (WGCNA), the least absolute shrinkage and selection operator (LASSO), and random forest (RF) algorithms were adopted to identify the diagnostic feature biomarker for RA. In addition, biomarkers were verified by qRT-PCR and Western blot analysis. We established a mouse model of collagen-induced arthritis (CIA), which was confirmed by HE staining and bone structure micro-CT analysis, and then further verified the biomarkers by immunofluorescence. In vitro NMR analysis was used to analyze and identify possible metabolites. The correlation of diagnostic feature biomarkers and immune cells was performed using the Spearman-rank correlation algorithm. In this study, a total of 434 DE-MRGs were identified. GO and KEGG enrichment analysis indicated that the DE-MRGs were significantly enriched in small molecules, catabolic process, purine metabolism, carbon metabolism, and inositol phosphate metabolism. AKR1C3, MCEE, POLE4, and PFKM were identified through WGCNA, LASSO, and RF algorithms. The nomogram result should have a significant diagnostic capacity of four biomarkers in RA. Immune infiltration landscape analysis revealed a significant difference in immune cells between HC and RA groups. Our findings suggest that AKR1C3, MCEE, POLE4, and PFKM were identified as potential diagnostic feature biomarkers associated with RA's immune cell infiltrations, providing a new perspective for future research and clinical management of RA.
Collapse
Affiliation(s)
- Zhaoyang Guo
- Department of Orthopedics, The Affiliated Hospital of Qingdao University, Qingdao 266003, Shandong Province, China
| | - Yuanye Ma
- Department of Orthopedics, The Affiliated Hospital of Qingdao University, Qingdao 266003, Shandong Province, China
| | - Yaqing Wang
- Department of Nephrology, The First Affiliated Hospital of China Medical University, Shenyang 110001, Liaoning Province, China
| | - Hongfei Xiang
- Department of Orthopedics, The Affiliated Hospital of Qingdao University, Qingdao 266003, Shandong Province, China
| | - Huifei Cui
- Department of Orthopedics, The Affiliated Hospital of Qingdao University, Qingdao 266003, Shandong Province, China
| | - Zuoran Fan
- Department of Orthopedics, The Affiliated Hospital of Qingdao University, Qingdao 266003, Shandong Province, China
| | - Youfu Zhu
- Department of Orthopedics, The Affiliated Hospital of Qingdao University, Qingdao 266003, Shandong Province, China
| | - Dongming Xing
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao 266071, Shandong, China
- School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Bohua Chen
- Department of Orthopedics, The Affiliated Hospital of Qingdao University, Qingdao 266003, Shandong Province, China
| | - Hao Tao
- Department of Orthopedics, The Affiliated Hospital of Qingdao University, Qingdao 266003, Shandong Province, China
| | - Zhu Guo
- Department of Orthopedics, The Affiliated Hospital of Qingdao University, Qingdao 266003, Shandong Province, China
| | - Xiaolin Wu
- Department of Orthopedics, The Affiliated Hospital of Qingdao University, Qingdao 266003, Shandong Province, China
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao 266071, Shandong, China
| |
Collapse
|
14
|
Porukala M, Vinod PK. Network-level analysis of ageing and its relationship with diseases and tissue regeneration in the mouse liver. Sci Rep 2023; 13:4632. [PMID: 36944690 PMCID: PMC10030664 DOI: 10.1038/s41598-023-31315-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Accepted: 03/09/2023] [Indexed: 03/23/2023] Open
Abstract
The liver plays a vital role in maintaining whole-body metabolic homeostasis, compound detoxification and has the unique ability to regenerate itself post-injury. Ageing leads to functional impairment of the liver and predisposes the liver to non-alcoholic fatty liver disease (NAFLD) and hepatocellular carcinoma (HCC). Mapping the molecular changes of the liver with ageing may help to understand the crosstalk of ageing with different liver diseases. A systems-level analysis of the ageing-induced liver changes and its crosstalk with liver-associated conditions is lacking. In the present study, we performed network-level analyses of the ageing liver using mouse transcriptomic data and a protein-protein interaction (PPI) network. A sample-wise analysis using network entropy measure was performed, which showed an increasing trend with ageing and helped to identify ageing genes based on local entropy changes. To gain further insights, we also integrated the differentially expressed genes (DEGs) between young and different age groups with the PPI network and identified core modules and nodes associated with ageing. Finally, we computed the network proximity of the ageing network with different networks of liver diseases and regeneration to quantify the effect of ageing. Our analysis revealed the complex interplay of immune, cancer signalling, and metabolic genes in the ageing liver. We found significant network proximities between ageing and NAFLD, HCC, liver damage conditions, and the early phase of liver regeneration with common nodes including NLRP12, TRP53, GSK3B, CTNNB1, MAT1 and FASN. Overall, our study maps the network-level changes of ageing and their interconnections with the physiology and pathology of the liver.
Collapse
Affiliation(s)
- Manisri Porukala
- Centre for Computational Natural Sciences and Bioinformatics, IIIT, Hyderabad, 500032, India
| | - P K Vinod
- Centre for Computational Natural Sciences and Bioinformatics, IIIT, Hyderabad, 500032, India.
| |
Collapse
|
15
|
Gong M, Lu H, Li L, Feng M, Zou Z. Integration of transcriptomics and metabonomics revealed the protective effects of hemp seed oil against methionine-choline-deficient diet-induced non-alcoholic steatohepatitis in mice. Food Funct 2023; 14:2096-2111. [PMID: 36734470 DOI: 10.1039/d2fo03054c] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Non-alcoholic steatohepatitis (NASH) is a chronic liver disease with few therapeutic options available currently. Hemp seed oil extracted from the seeds of hemp (Cannabis sativa L.) has significant nutritional and biological properties due to the unique composition of polyunsaturated fatty acids and various antioxidant compounds. However, little is known about the beneficial effects and molecular mechanisms of hemp seed oil on NASH. Here, the hepatoprotective effects of hemp seed oil on methionine-choline-deficient (MCD) diet-induced NASH in C57BL/6 mice were explored via integration of transcriptomics and metabolomics. Hemp seed oil could improve hepatic steatosis, inflammation and fibrosis in mice with MCD diet-induced NASH. In a nuclear magnetic resonance (NMR)-based metabonomic study, the hepatic and urinary metabolic profiles of mice supplemented with hemp seed oil showed a tendency to recover to healthy controls compared to those of NASH mice. Eight potential biomarkers associated with NASH in both liver tissue and urine were restored to near normal levels by administration of hemp seed oil. The proposed pathways were mainly involved in pyrimidine metabolism, one-carbon metabolism, amino acid metabolism, glycolysis and the tricarboxylic acid (TCA) cycle. Hepatic transcriptomics based on Illumina RNA-Seq sequencing showed that hemp seed oil exerted anti-NASH activities by regulating multiple signaling pathways, e.g., downregulation of the TNF signaling pathway, the IL-17 signaling pathway, the MAPK signaling pathway and the NF-κB signaling pathway, which played a pivotal role in the pathogenesis of NASH. In particular, integration of metabonomic and transcriptomic results suggested that hemp seed oil could attenuate NASH-related liver fibrosis by inhibition of glutaminolysis. These results provided new insights into the hepatoprotective effects of hemp seed oil against MCD diet-induced NASH and hemp seed oil might have potential as an effective therapy for NASH.
Collapse
Affiliation(s)
- Mengjuan Gong
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou 510006, China.
| | - Hailong Lu
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou 510006, China.
| | - Lixi Li
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou 510006, China.
| | - Meiqi Feng
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou 510006, China.
| | - Zhongjie Zou
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou 510006, China.
| |
Collapse
|
16
|
Ghrayeb A, Agranovich B, Peled D, Finney AC, Abramovich I, Garcia JF, Traylor J, Drucker S, Fernandes SI, Weissman N, Chen YE, Rom O, Mor I, Gottlieb E. Fatty liver-mediated glycine restriction impairs glutathione synthesis and causes hypersensitization to acetaminophen. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.16.524043. [PMID: 36711913 PMCID: PMC9882121 DOI: 10.1101/2023.01.16.524043] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Non-alcoholic fatty liver disease (NAFLD) affects nearly one third of the population worldwide. Understanding metabolic pathways involved can provide insights into disease progression. Untargeted metabolomics of livers from mice with early-stage steatosis indicated a decrease in methylated metabolites suggesting altered one carbon metabolism. The levels of glycine, a central component of one carbon metabolism, were lower in steatotic mice, in line with clinical evidence. Isotope tracing studies demonstrated that increased synthesis of serine from glycine is the underlying cause for glycine limitation in fatty livers. Consequently, the low glycine availability in steatotic livers impaired glutathione (GSH) synthesis under oxidative stress induced by acetaminophen (APAP), enhancing hepatic toxicity. Glycine supplementation mitigated acute liver damage and overall toxicity caused by APAP in fatty livers by supporting de novo GSH synthesis. Thus, early metabolic changes in NAFLD that lead to glycine depletion sensitize mice to xenobiotic toxicity even at a reversible stage of NAFLD.
Collapse
|
17
|
Yang S, Ye Z, Liu M, Zhang Y, Wu Q, Zhou C, Zhang Z, He P, Zhang Y, Li H, Liu C, Qin X. Associations of different serum folate forms with indices of nonalcoholic fatty liver disease and advanced fibrosis. Obes Res Clin Pract 2023; 17:58-65. [PMID: 36746711 DOI: 10.1016/j.orcp.2023.01.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 01/24/2023] [Accepted: 01/30/2023] [Indexed: 02/07/2023]
Abstract
OBJECTIVE We aim to examine the associations of different serum folate forms (total folate, 5-methyltetrahydrofolate [5-mTHF] and unmetabolized folic acid [UMFA]), with the prevalence of nonalcoholic fatty liver disease (NAFLD) and advanced fibrosis. METHODS This cross-sectional analysis was conducted in 6610 participants aged ≥ 18 years from the 2011-2018 National Health and Nutrition Examination Survey (NHANES) database. NAFLD was defined as a United States fatty liver index (USFLI) ≥ 30. Advanced fibrosis was defined as a Fibrosis-4 score (FIB-4) > 3.25, a NAFLD Fibrosis Score (NFS) > 0.676, and a Hepamet Fibrosis Score (HFS) ≥ 0.47, respectively. RESULTS The prevalence of NAFLD was 34.5%. Overall, serum total folate and 5-mTHF were inversely associated with the prevalence of NAFLD (both P for trend across quartiles <0.001). A similar trend was found for advanced fibrosis based on NFS and HFS (both P for trend across quartiles <0.05). However, a higher concentration of UMFA was significantly related to a higher prevalence of NAFLD (P for trend across quartiles =0.004). A similar relation was found for advanced fibrosis based on NFS (P for trend across quartiles =0.024). CONCLUSIONS Higher concentrations of serum total folate and 5-mTHF were associated with a lower prevalence of NAFLD and advanced fibrosis, while a higher concentration of UMFA was related to a higher prevalence of NAFLD.
Collapse
Affiliation(s)
- Sisi Yang
- Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China; National Clinical Research Center for Kidney Disease, Guangzhou 510515, China; State Key Laboratory of Organ Failure Research, Guangzhou 510515, China; Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Guangdong Provincial Clinical Research Center for Kidney Disease, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou 510515, China
| | - Ziliang Ye
- Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China; National Clinical Research Center for Kidney Disease, Guangzhou 510515, China; State Key Laboratory of Organ Failure Research, Guangzhou 510515, China; Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Guangdong Provincial Clinical Research Center for Kidney Disease, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou 510515, China
| | - Mengyi Liu
- Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China; National Clinical Research Center for Kidney Disease, Guangzhou 510515, China; State Key Laboratory of Organ Failure Research, Guangzhou 510515, China; Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Guangdong Provincial Clinical Research Center for Kidney Disease, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou 510515, China
| | - Yanjun Zhang
- Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China; National Clinical Research Center for Kidney Disease, Guangzhou 510515, China; State Key Laboratory of Organ Failure Research, Guangzhou 510515, China; Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Guangdong Provincial Clinical Research Center for Kidney Disease, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou 510515, China
| | - Qimeng Wu
- Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China; National Clinical Research Center for Kidney Disease, Guangzhou 510515, China; State Key Laboratory of Organ Failure Research, Guangzhou 510515, China; Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Guangdong Provincial Clinical Research Center for Kidney Disease, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou 510515, China
| | - Chun Zhou
- Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China; National Clinical Research Center for Kidney Disease, Guangzhou 510515, China; State Key Laboratory of Organ Failure Research, Guangzhou 510515, China; Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Guangdong Provincial Clinical Research Center for Kidney Disease, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou 510515, China
| | - Zhuxian Zhang
- Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China; National Clinical Research Center for Kidney Disease, Guangzhou 510515, China; State Key Laboratory of Organ Failure Research, Guangzhou 510515, China; Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Guangdong Provincial Clinical Research Center for Kidney Disease, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou 510515, China
| | - Panpan He
- Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China; National Clinical Research Center for Kidney Disease, Guangzhou 510515, China; State Key Laboratory of Organ Failure Research, Guangzhou 510515, China; Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Guangdong Provincial Clinical Research Center for Kidney Disease, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou 510515, China
| | - Yuanyuan Zhang
- Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China; National Clinical Research Center for Kidney Disease, Guangzhou 510515, China; State Key Laboratory of Organ Failure Research, Guangzhou 510515, China; Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Guangdong Provincial Clinical Research Center for Kidney Disease, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou 510515, China
| | - Huan Li
- Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China; National Clinical Research Center for Kidney Disease, Guangzhou 510515, China; State Key Laboratory of Organ Failure Research, Guangzhou 510515, China; Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Guangdong Provincial Clinical Research Center for Kidney Disease, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou 510515, China
| | - Chengzhang Liu
- Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China; National Clinical Research Center for Kidney Disease, Guangzhou 510515, China; State Key Laboratory of Organ Failure Research, Guangzhou 510515, China; Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Guangdong Provincial Clinical Research Center for Kidney Disease, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou 510515, China; Institute of Biomedicine, Anhui Medical University, Hefei 230032, China
| | - Xianhui Qin
- Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China; National Clinical Research Center for Kidney Disease, Guangzhou 510515, China; State Key Laboratory of Organ Failure Research, Guangzhou 510515, China; Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Guangdong Provincial Clinical Research Center for Kidney Disease, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou 510515, China.
| |
Collapse
|
18
|
Creatine supplementation protects against diet-induced non-alcoholic fatty liver but exacerbates alcoholic fatty liver. Life Sci 2022; 310:121064. [PMID: 36220368 DOI: 10.1016/j.lfs.2022.121064] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 09/26/2022] [Accepted: 10/06/2022] [Indexed: 11/06/2022]
Abstract
AIMS This work investigated the effects of creatine supplementation on different pathways related to the pathogenesis of non-alcoholic fatty liver disease and alcoholic liver disease. MAIN METHODS To induce alcoholic liver disease, male Swiss mice were divided into three groups: control, ethanol and ethanol supplemented with creatine. To induce non-alcoholic fatty liver disease, mice were divided into three groups: control, high-fat diet and high-fat diet supplemented with creatine. Each group consisted of eight animals. In both cases, creatine monohydrate was added to the diets (1 %; weight/vol). KEY FINDINGS Creatine supplementation prevented high-fat diet-induced non-alcoholic fatty liver disease progression, demonstrated by attenuated liver fat accumulation and liver damage. On the other hand, when combined with ethanol, creatine supplementation up-regulated key genes related to ethanol metabolism, oxidative stress, inflammation and lipid synthesis, and exacerbated ethanol-induced liver steatosis and damage, demonstrated by increased liver fat accumulation and histopathological score, as well as elevated oxidative damage markers and inflammatory mediators. SIGNIFICANCE Our results clearly demonstrated creatine supplementation exerts different outcomes in relation to non-alcoholic fatty liver disease and alcoholic liver disease, namely it protects against high-fat diet-induced non-alcoholic fatty liver disease but exacerbates ethanol-induced alcoholic liver disease. The exacerbating effects of the creatine and ethanol combination appear to be related to oxidative stress and inflammation-mediated up-regulation of ethanol metabolism.
Collapse
|
19
|
Guerrero L, Paradela A, Corrales FJ. Targeted Proteomics for Monitoring One-Carbon Metabolism in Liver Diseases. Metabolites 2022; 12:779. [PMID: 36144184 PMCID: PMC9501948 DOI: 10.3390/metabo12090779] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 08/18/2022] [Accepted: 08/19/2022] [Indexed: 11/16/2022] Open
Abstract
Liver diseases cause approximately 2 million deaths per year worldwide and had an increasing incidence during the last decade. Risk factors for liver diseases include alcohol consumption, obesity, diabetes, the intake of hepatotoxic substances like aflatoxin, viral infection, and genetic determinants. Liver cancer is the sixth most prevalent cancer and the third in mortality (second in males). The low survival rate (less than 20% in 5 years) is partially explained by the late diagnosis, which remarks the need for new early molecular biomarkers. One-carbon metabolism integrates folate and methionine cycles and participates in essential cell processes such as redox homeostasis maintenance and the regulation of methylation reactions through the production of intermediate metabolites such as cysteine and S-Adenosylmethionine. One-carbon metabolism has a tissue specific configuration, and in the liver, the participating enzymes are abundantly expressed-a requirement to maintain hepatocyte differentiation. Targeted proteomics studies have revealed significant differences in hepatocellular carcinoma and cirrhosis, suggesting that monitoring one-carbon metabolism enzymes can be useful for stratification of liver disease patients and to develop precision medicine strategies for their clinical management. Here, reprogramming of one-carbon metabolism in liver diseases is described and the role of mass spectrometry to follow-up these alterations is discussed.
Collapse
Affiliation(s)
- Laura Guerrero
- Centro Nacional de Biotecnología (CNB), CSIC. C/Darwin 3, 28049 Madrid, Spain
| | - Alberto Paradela
- Centro Nacional de Biotecnología (CNB), CSIC. C/Darwin 3, 28049 Madrid, Spain
| | - Fernando J. Corrales
- Centro Nacional de Biotecnología (CNB), CSIC. C/Darwin 3, 28049 Madrid, Spain
- National Institute for the Study of Liver and Gastrointestinal Diseases (CIBERehd, Carlos III Health Institute), 28029 Madrid, Spain
| |
Collapse
|
20
|
Brunel M, Burkina V, Pickova J, Sampels S, Moazzami AA. Oleaginous yeast Rhodotorula toruloides biomass effect on the metabolism of Arctic char ( Salvelinus alpinus). Front Mol Biosci 2022; 9:931946. [PMID: 36052171 PMCID: PMC9425082 DOI: 10.3389/fmolb.2022.931946] [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: 04/29/2022] [Accepted: 07/11/2022] [Indexed: 12/30/2022] Open
Abstract
Sustainability issues arise when using fish oil and vegetable oils in fish feed production for aquaculture purposes. Microbial production of single cell oil is a potential alternative as a lipid ingredient in the production of fish feed. In this study, we replaced the vegetable oils with the oleaginous yeast R. toruloides biomass in the diet of Arctic char (S. alpinus) and investigated the effects on health and composition. Measurement of fish growth parameters showed a higher liver weight and hepatosomatic index in the experimental group of fish fed partly with yeast biomass compared to a control group fed a diet with vegetable oils. No significant differences in the lipid content of muscle and liver tissues were found. The fatty acid profiles in the muscle of both fish groups were similar while the experimental fish group had a higher amount of monounsaturated fatty acids in the liver. Histology of livers showed no significant difference in the number of lipid droplets. The size of hepatic lipid droplets seemed to be related to liver fat content. Quantification of metabolites in the liver revealed no differences between the fish groups while plasma metabolites involved in energy pathways such as alanine, 3-hydroxybutyrate, creatinine, serine, betaine, and choline were significantly higher in the experimental fish group.
Collapse
Affiliation(s)
- Mathilde Brunel
- Department of Molecular Sciences, Faculty of Natural Resources and Agricultural Sciences, Swedish University of Agricultural Sciences, Uppsala, Sweden,*Correspondence: Mathilde Brunel,
| | - Viktoriia Burkina
- Department of Molecular Sciences, Faculty of Natural Resources and Agricultural Sciences, Swedish University of Agricultural Sciences, Uppsala, Sweden,Faculty of Fisheries and Protection of Waters, South Bohemian Research Center of Aquaculture and Biodiversity of Hydrocenoses, Research Institute of Fish Culture and Hydrobiology, University of South Bohemia in České Budějovice, Vodňany, Czech Republic
| | - Jana Pickova
- Department of Molecular Sciences, Faculty of Natural Resources and Agricultural Sciences, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Sabine Sampels
- Department of Molecular Sciences, Faculty of Natural Resources and Agricultural Sciences, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Ali A. Moazzami
- Department of Molecular Sciences, Faculty of Natural Resources and Agricultural Sciences, Swedish University of Agricultural Sciences, Uppsala, Sweden
| |
Collapse
|
21
|
Tang Y, Chen X, Chen Q, Xiao J, Mi J, Liu Q, You Y, Chen Y, Ling W. Association of serum methionine metabolites with non-alcoholic fatty liver disease: a cross-sectional study. Nutr Metab (Lond) 2022; 19:21. [PMID: 35303918 PMCID: PMC8932073 DOI: 10.1186/s12986-022-00647-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 02/13/2022] [Indexed: 12/13/2022] Open
Abstract
Background and project Non-alcoholic fatty liver disease (NAFLD) is viewed as the hepatic manifestation of metabolic syndrome. Methionine metabolites have been linked to metabolic syndrome and its related diseases. Whether serum methionine metabolites levels are associated with NAFLD remains unclear. The study aimed to assess the association between methionine metabolites and NAFLD. Methods This cross-sectional study included a total of 2814 individuals aged 40–75 years old. All participants underwent anthropometric measurements, laboratory tests, dietary assessment and abdominal ultrasonography. Multivariable logistic regression analysis was performed to estimate the association of methionine metabolites with NAFLD. Results Overall, 1446 with and 1368 without NAFLD were enrolled in this study. Participants with NAFLD had significantly higher serum S-adenosylmethionine (SAM), S-adenosylhomocysteine (SAH) and homocysteine (Hcy) levels, and a lower S-adenosylmethionine/S-adenosylhomocysteine (SAM/SAH) ratio than those without NAFLD (all P < 0.001). After adjusting multiple confounders, odds ratios (95% confidence interval) for quartile 4 versus quartile 1 of SAH, Hcy and SAM/SAH ratio were 1.65 (1.27–2.14), 1.63 (1.26–2.12) and 0.63 (0.49–0.83), respectively (all P for trend < 0.01). In addition, serum SAH, Hcy levels and SAM/SAH ratio were significantly correlated with the degree of hepatic steatosis (all P for trend < 0.001). Conclusion Elevated serum SAH, Hcy levels and lower SAM/SAH ratio may be independently associated with the presence of NAFLD in middle-aged and elder Chinese. Supplementary Information The online version contains supplementary material available at 10.1186/s12986-022-00647-7.
Collapse
Affiliation(s)
- Yi Tang
- Department of Nutrition, School of Public Health, Sun Yat-Sen University, 74 Zhongshan Rd. 2, Guangzhou, 510080, Guangdong Province, People's Republic of China.,Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou, 510080, Guangdong Province, People's Republic of China
| | - Xu Chen
- Department of Nutrition, School of Public Health, Sun Yat-Sen University, 74 Zhongshan Rd. 2, Guangzhou, 510080, Guangdong Province, People's Republic of China.,Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou, 510080, Guangdong Province, People's Republic of China
| | - Qian Chen
- Department of Cardiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, Guangdong, People's Republic of China
| | - Jinghe Xiao
- Department of Nutrition, School of Public Health, Sun Yat-Sen University, 74 Zhongshan Rd. 2, Guangzhou, 510080, Guangdong Province, People's Republic of China.,Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou, 510080, Guangdong Province, People's Republic of China
| | - Jiaxin Mi
- Department of Nutrition, School of Public Health, Sun Yat-Sen University, 74 Zhongshan Rd. 2, Guangzhou, 510080, Guangdong Province, People's Republic of China.,Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou, 510080, Guangdong Province, People's Republic of China
| | - Qiannan Liu
- Department of Nutrition, School of Public Health, Sun Yat-Sen University, 74 Zhongshan Rd. 2, Guangzhou, 510080, Guangdong Province, People's Republic of China.,Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou, 510080, Guangdong Province, People's Republic of China
| | - Yiran You
- Department of Nutrition, School of Public Health, Sun Yat-Sen University, 74 Zhongshan Rd. 2, Guangzhou, 510080, Guangdong Province, People's Republic of China.,Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou, 510080, Guangdong Province, People's Republic of China
| | - Yuming Chen
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou, 510080, Guangdong Province, People's Republic of China. .,Department of Epidemiology, School of Public Health, Sun Yat-Sen University, 74 Zhongshan Rd. 2, Guangzhou, 510080, Guangdong Province, People's Republic of China.
| | - Wenhua Ling
- Department of Nutrition, School of Public Health, Sun Yat-Sen University, 74 Zhongshan Rd. 2, Guangzhou, 510080, Guangdong Province, People's Republic of China. .,Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou, 510080, Guangdong Province, People's Republic of China.
| |
Collapse
|
22
|
Lower plasma glutathione, choline, and betaine concentrations are associated with fatty liver in postmenopausal women. Nutr Res 2022; 101:23-30. [PMID: 35364359 DOI: 10.1016/j.nutres.2022.02.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 02/11/2022] [Accepted: 02/12/2022] [Indexed: 11/18/2022]
|
23
|
Rodríguez-Sanabria JS, Escutia-Gutiérrez R, Rosas-Campos R, Armendáriz-Borunda JS, Sandoval-Rodríguez A. An Update in Epigenetics in Metabolic-Associated Fatty Liver Disease. Front Med (Lausanne) 2022; 8:770504. [PMID: 35087844 PMCID: PMC8787199 DOI: 10.3389/fmed.2021.770504] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 12/02/2021] [Indexed: 12/17/2022] Open
Abstract
Metabolic-associated fatty liver disease (MAFLD) is characterized by hepatic steatosis accompanied by one of three features: overweight or obesity, T2DM, or lean or normal weight with evidence of metabolic dysregulation. It is distinguished by excessive fat accumulation in hepatocytes, and a decrease in the liver's ability to oxidize fats, the accumulation of ectopic fat, and the activation of proinflammatory pathways. Chronic damage will keep this pathophysiologic cycle active causing progression from hepatic steatosis to cirrhosis and eventually, hepatocarcinoma. Epigenetics affecting gene expression without altering DNA sequence allows us to study MAFLD pathophysiology from a different perspective, in which DNA methylation processes, histone modifications, and miRNAs expression have been closely associated with MAFLD progression. However, these considerations also faced us with the circumstance that modifying those epigenetics patterns might lead to MAFLD regression. Currently, epigenetics is an area of great interest because it could provide new insights in therapeutic targets and non-invasive biomarkers. This review comprises an update on the role of epigenetic patterns, as well as innovative therapeutic targets and biomarkers in MAFLD.
Collapse
Affiliation(s)
- J Samael Rodríguez-Sanabria
- Department of Molecular Biology and Genomics, Institute for Molecular Biology in Medicine and Gene Therapy, CUCS, University of Guadalajara, Guadalajara, Mexico
| | - Rebeca Escutia-Gutiérrez
- Department of Molecular Biology and Genomics, Institute for Molecular Biology in Medicine and Gene Therapy, CUCS, University of Guadalajara, Guadalajara, Mexico
| | - Rebeca Rosas-Campos
- Department of Molecular Biology and Genomics, Institute for Molecular Biology in Medicine and Gene Therapy, CUCS, University of Guadalajara, Guadalajara, Mexico
| | - Juan S Armendáriz-Borunda
- Department of Molecular Biology and Genomics, Institute for Molecular Biology in Medicine and Gene Therapy, CUCS, University of Guadalajara, Guadalajara, Mexico.,School of Medicine and Health Sciences, Tecnologico de Monterrey, Campus Guadalajara, Zapopan, Mexico
| | - Ana Sandoval-Rodríguez
- Department of Molecular Biology and Genomics, Institute for Molecular Biology in Medicine and Gene Therapy, CUCS, University of Guadalajara, Guadalajara, Mexico
| |
Collapse
|
24
|
Chang TY, Wu CH, Chang CY, Lee FJ, Wang BW, Doong JY, Lin YS, Kuo CS, Huang RFS. Optimal Dietary Intake Composition of Choline and Betaine Is Associated with Minimized Visceral Obesity-Related Hepatic Steatosis in a Case-Control Study. Nutrients 2022; 14:261. [PMID: 35057441 PMCID: PMC8779168 DOI: 10.3390/nu14020261] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 01/05/2022] [Accepted: 01/05/2022] [Indexed: 02/06/2023] Open
Abstract
Few studies on humans have comprehensively evaluated the intake composition of methyl-donor nutrients (MDNs: choline, betaine, and folate) in relation to visceral obesity (VOB)-related hepatic steatosis (HS), the hallmark of non-alcoholic fatty liver diseases. In this case-control study, we recruited 105 patients with HS and 104 without HS (controls). HS was diagnosed through ultrasound examination. VOB was measured using a whole-body analyzer. MDN intake was assessed using a validated quantitative food frequency questionnaire. After adjustment for multiple HS risk factors, total choline intake was the most significant dietary determinant of HS in patients with VOB (Beta: -0.41, p = 0.01). Low intake of choline (<6.9 mg/kg body weight), betaine (<3.1 mg/kg body weight), and folate (<8.8 μg/kg body weight) predicted increased odds ratios (ORs) of VOB-related HS (choline: OR: 22, 95% confidence interval [CI]: 6.5-80; betaine: OR: 14, 95% CI: 4.4-50; and folate: OR: 19, 95% CI: 5.2-74). Combined high intake of choline and betaine, but not folate, was associated with an 81% reduction in VOB-related HS (OR: 0.19, 95% CI: 0.05-0.69). Our data suggest that the optimal intake of choline and betaine can minimize the risk of VOB-related HS in a threshold-dependent manner.
Collapse
Affiliation(s)
- Ting-Yu Chang
- Department of Nutritional Science, Fu Jen Catholic University, New Taipei City 242062, Taiwan; (T.-Y.C.); (J.-Y.D.); (Y.-S.L.)
| | - Chien-Hsien Wu
- Ph.D. Program in Nutrition and Food Science, Fu Jen Catholic University, New Taipei City 242062, Taiwan;
- Department of Gastroenterology and Hepatology, Taipei Hospital, Ministry of Health and Welfare, New Taipei City 242, Taiwan
| | - Chi-Yang Chang
- Department of Gastroenterology and Hepatology, Fu Jen Catholic University Hospital, New Taipei City 243089, Taiwan; (C.-Y.C.); (F.-J.L.)
| | - Fu-Jen Lee
- Department of Gastroenterology and Hepatology, Fu Jen Catholic University Hospital, New Taipei City 243089, Taiwan; (C.-Y.C.); (F.-J.L.)
| | - Bei-Wen Wang
- Department of Nutrition, Fu Jen Catholic University Hospital, New Taipei City 243089, Taiwan;
| | - Jia-Yau Doong
- Department of Nutritional Science, Fu Jen Catholic University, New Taipei City 242062, Taiwan; (T.-Y.C.); (J.-Y.D.); (Y.-S.L.)
| | - Yu-Shun Lin
- Department of Nutritional Science, Fu Jen Catholic University, New Taipei City 242062, Taiwan; (T.-Y.C.); (J.-Y.D.); (Y.-S.L.)
| | - Chang-Sheng Kuo
- Department of Nutrition, Fu Jen Catholic University Hospital, New Taipei City 243089, Taiwan;
| | - Rwei-Fen S. Huang
- Department of Nutritional Science, Fu Jen Catholic University, New Taipei City 242062, Taiwan; (T.-Y.C.); (J.-Y.D.); (Y.-S.L.)
- Ph.D. Program in Nutrition and Food Science, Fu Jen Catholic University, New Taipei City 242062, Taiwan;
| |
Collapse
|
25
|
Ramírez-Contreras CY, Mehran AE, Salehzadeh M, Mussai EX, Miller JW, Smith A, Ranger M, Holsti L, Soma KK, Devlin AM. Sex-specific effects of neonatal oral sucrose treatment on growth and liver choline and glucocorticoid metabolism in adulthood. Am J Physiol Regul Integr Comp Physiol 2021; 321:R802-R811. [PMID: 34612088 PMCID: PMC11961110 DOI: 10.1152/ajpregu.00091.2021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 09/30/2021] [Accepted: 09/30/2021] [Indexed: 11/22/2022]
Abstract
Hospitalized preterm infants experience painful medical procedures. Oral sucrose is the nonpharmacological standard of care for minor procedural pain relief. Infants are treated with numerous doses of sucrose, raising concerns about potential long-term effects. The objective of this study was to determine the long-term effects of neonatal oral sucrose treatment on growth and liver metabolism in a mouse model. Neonatal female and male mice were randomly assigned to one of two oral treatments (n = 7-10 mice/group/sex): sterile water or sucrose. Pups were treated 10 times/day for the first 6 days of life with 0.2 mg/g body wt of respective treatments (24% solution; 1-4 μL/dose) to mimic what is given to preterm infants. Mice were weaned at age 3 wk onto a control diet and fed until age 16 wk. Sucrose-treated female and male mice gained less weight during the treatment period and were smaller at weaning than water-treated mice (P ≤ 0.05); no effect of sucrose treatment on body weight was observed at adulthood. However, adult sucrose-treated female mice had smaller tibias and lower serum insulin-like growth factor-1 than adult water-treated female mice (P ≤ 0.05); these effects were not observed in males. Lower liver S-adenosylmethionine, phosphocholine, and glycerophosphocholine were observed in adult sucrose-treated compared with water-treated female and male mice (P ≤ 0.05). Sucrose-treated female, but not male, mice had lower liver free choline and higher liver betaine compared with water-treated female mice (P < 0.01). Our findings suggest that repeated neonatal sucrose treatment has long-term sex-specific effects on growth and liver methionine and choline metabolism.
Collapse
Affiliation(s)
- Cynthia Y Ramírez-Contreras
- Department of Pediatrics, The University of British Columbia, Vancouver, British Columbia, Canada
- BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada
| | - Arya E Mehran
- Department of Pediatrics, The University of British Columbia, Vancouver, British Columbia, Canada
- BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada
| | - Melody Salehzadeh
- Department of Psychology, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Ei-Xia Mussai
- BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada
- Department of Obstetrics and Gynaecology, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Joshua W Miller
- Department of Nutritional Sciences, Rutgers University, The State University of New Jersey, New Brunswick, New Jersey
| | - Andre Smith
- Department of Nutritional Sciences, Rutgers University, The State University of New Jersey, New Brunswick, New Jersey
| | - Manon Ranger
- BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada
- School of Nursing, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Liisa Holsti
- BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada
- Department of Occupational Science and Occupational Therapy, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Kiran K Soma
- BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada
- Department of Psychology, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Angela M Devlin
- Department of Pediatrics, The University of British Columbia, Vancouver, British Columbia, Canada
- BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada
| |
Collapse
|
26
|
Mazi TA, Borkowski K, Newman JW, Fiehn O, Bowlus CL, Sarkar S, Matsukuma K, Ali MR, Kieffer DA, Wan YJY, Stanhope KL, Havel PJ, Medici V. Ethnicity-specific alterations of plasma and hepatic lipidomic profiles are related to high NAFLD rate and severity in Hispanic Americans, a pilot study. Free Radic Biol Med 2021; 172:490-502. [PMID: 34182070 PMCID: PMC8712226 DOI: 10.1016/j.freeradbiomed.2021.06.024] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 06/19/2021] [Accepted: 06/23/2021] [Indexed: 02/06/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a progressive condition that includes steatosis (NAFL) and nonalcoholic steatohepatitis (NASH). In the U.S., Hispanics (HIS) are afflicted with NAFLD at a higher rate and severity compared to other ethnicities. To date, the mechanisms underlying this disparity have not been elucidated. In this pilot study, we compared untargeted plasma metabolomic profiles for primary metabolism, complex lipids, choline and related compounds between a group of HIS (n = 7) and White Caucasian (CAU, n = 8) subjects with obesity and biopsy-characterized NAFL to ethnicity-matched lean healthy controls (n = 14 HIS and 8 CAU). We also compared liver and plasma metabolomic profiles in a group of HIS and CAU subjects with obesity and NASH of comparable NAFLD Activity Scores, to BMI-matched NASH-free subjects in both ethnicities. Results highlight signs of metabolic dysregulation observed in HIS, independent of obesity, including higher plasma triglycerides, acylcarnitines, and free fatty acids. With NASH progression, there were ethnicity-related differences in the hepatic profile, including higher free fatty acids and lysophospholipids seen in HIS, suggesting lipotoxicity is involved in the progression of NASH. We also observed greater hepatic triglyceride content, higher plasma triglyceride concentrations and lower hepatic phospholipids with signs of impaired hepatic mitochondrial β-oxidation. These findings provide preliminary evidence indicating ethnicity-related variations that could potentially modulate the risk for progression of NALD to NASH.
Collapse
Affiliation(s)
- Tagreed A Mazi
- Department of Nutrition, University of California Davis, 3135 Meyer Hall, One Shields Avenue, Davis, CA, 95616, USA; Department of Community Health Sciences-Clinical Nutrition, College of Applied Medical Sciences, King Saud University, P.O. Box 10219, Riyadh, 11433, Saudi Arabia.
| | - Kamil Borkowski
- West Coast Metabolomic Center, Genome Center, University of California-Davis, Davis, CA, 95616, USA
| | - John W Newman
- West Coast Metabolomic Center, Genome Center, University of California-Davis, Davis, CA, 95616, USA; United States Department of Agriculture-Agriculture Research Service-Western Human Nutrition Research Center, Davis, CA, 95616, USA; Department of Nutrition, University of California-Davis, Davis, CA, 95616, USA
| | - Oliver Fiehn
- West Coast Metabolomic Center, Genome Center, University of California-Davis, Davis, CA, 95616, USA
| | - Christopher L Bowlus
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, University of California Davis, 4150 V Street, Suite 3500 Sacramento, CA, 95817, USA
| | - Souvik Sarkar
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, University of California Davis, 4150 V Street, Suite 3500 Sacramento, CA, 95817, USA
| | - Karen Matsukuma
- Department of Pathology and Laboratory Medicine, University of California-Davis, Sacramento, CA, 95817, USA
| | - Mohamed R Ali
- Department of Surgery, University of California, Davis, 2221 Stockton Boulevard, Cypress Building, Sacramento, CA, 95817, USA
| | - Dorothy A Kieffer
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, University of California Davis, 4150 V Street, Suite 3500 Sacramento, CA, 95817, USA
| | - Yu-Jui Y Wan
- Department of Pathology and Laboratory Medicine, University of California-Davis, Sacramento, CA, 95817, USA
| | - Kimber L Stanhope
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, CA, 95616, USA
| | - Peter J Havel
- Department of Nutrition, University of California-Davis, Davis, CA, 95616, USA; Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, CA, 95616, USA
| | - Valentina Medici
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, University of California Davis, 4150 V Street, Suite 3500 Sacramento, CA, 95817, USA.
| |
Collapse
|
27
|
Korsmo HW, Jiang X. One carbon metabolism and early development: a diet-dependent destiny. Trends Endocrinol Metab 2021; 32:579-593. [PMID: 34210607 PMCID: PMC8282711 DOI: 10.1016/j.tem.2021.05.011] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 05/21/2021] [Accepted: 05/28/2021] [Indexed: 12/27/2022]
Abstract
One carbon metabolism (OCM) is critical for early development, as it provides one carbon (1C) units for the biosynthesis of DNA, proteins, and lipids and epigenetic modification of the genome. Epigenetic marks established early in life can be maintained and exert lasting impacts on gene expression and functions later in life. Animal and human studies have increasingly demonstrated that prenatal 1C nutrient deficiencies impair fetal growth, neurodevelopment, and cardiometabolic parameters in childhood, while sufficient maternal 1C nutrient intake is protective against these detrimental outcomes. However, recent studies also highlight the potential risk of maternal 1C nutrient excess or imbalance in disrupting early development. Further studies are needed to delineate the dose-response relationship among prenatal 1C nutrient exposure, epigenetic modifications, and developmental outcomes.
Collapse
Affiliation(s)
- Hunter W Korsmo
- PhD Program in Biochemistry, The Graduate Center CUNY (City University of New York), New York, NY 10016, USA; Department of Health and Nutrition Sciences, Brooklyn College of the City University of New York, Brooklyn, NY 11210, USA
| | - Xinyin Jiang
- PhD Program in Biochemistry, The Graduate Center CUNY (City University of New York), New York, NY 10016, USA; Department of Health and Nutrition Sciences, Brooklyn College of the City University of New York, Brooklyn, NY 11210, USA.
| |
Collapse
|
28
|
Arumugam MK, Paal MC, Donohue TM, Ganesan M, Osna NA, Kharbanda KK. Beneficial Effects of Betaine: A Comprehensive Review. BIOLOGY 2021; 10:456. [PMID: 34067313 PMCID: PMC8224793 DOI: 10.3390/biology10060456] [Citation(s) in RCA: 120] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 05/06/2021] [Accepted: 05/19/2021] [Indexed: 02/05/2023]
Abstract
Medicinal herbs and many food ingredients possess favorable biological properties that contribute to their therapeutic activities. One such natural product is betaine, a stable, nontoxic natural substance that is present in animals, plants, and microorganisms. Betaine is also endogenously synthesized through the metabolism of choline or exogenously consumed through dietary intake. Betaine mainly functions as (i) an osmolyte and (ii) a methyl-group donor. This review describes the major physiological effects of betaine in whole-body health and its ability to protect against both liver- as well as non-liver-related diseases and conditions. Betaine's role in preventing/attenuating both alcohol-induced and metabolic-associated liver diseases has been well studied and is extensively reviewed here. Several studies show that betaine protects against the development of alcohol-induced hepatic steatosis, apoptosis, and accumulation of damaged proteins. Additionally, it can significantly prevent/attenuate progressive liver injury by preserving gut integrity and adipose function. The protective effects are primarily associated with the regulation of methionine metabolism through removing homocysteine and maintaining cellular SAM:SAH ratios. Similarly, betaine prevents metabolic-associated fatty liver disease and its progression. In addition, betaine has a neuroprotective role, preserves myocardial function, and prevents pancreatic steatosis. Betaine also attenuates oxidant stress, endoplasmic reticulum stress, inflammation, and cancer development. To conclude, betaine exerts significant therapeutic and biological effects that are potentially beneficial for alleviating a diverse number of human diseases and conditions.
Collapse
Affiliation(s)
- Madan Kumar Arumugam
- Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE 68105, USA; (MK.A.); (M.C.P.); (T.M.D.J.); (M.G.); (N.A.O.)
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Matthew C. Paal
- Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE 68105, USA; (MK.A.); (M.C.P.); (T.M.D.J.); (M.G.); (N.A.O.)
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Terrence M. Donohue
- Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE 68105, USA; (MK.A.); (M.C.P.); (T.M.D.J.); (M.G.); (N.A.O.)
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Department of Biochemistry & Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Murali Ganesan
- Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE 68105, USA; (MK.A.); (M.C.P.); (T.M.D.J.); (M.G.); (N.A.O.)
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Natalia A. Osna
- Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE 68105, USA; (MK.A.); (M.C.P.); (T.M.D.J.); (M.G.); (N.A.O.)
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Kusum K. Kharbanda
- Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE 68105, USA; (MK.A.); (M.C.P.); (T.M.D.J.); (M.G.); (N.A.O.)
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Department of Biochemistry & Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| |
Collapse
|
29
|
Xiang L, Shao Y, Chen Y. Mitochondrial dysfunction and mitochondrion-targeted therapeutics in liver diseases. J Drug Target 2021; 29:1080-1093. [PMID: 33788656 DOI: 10.1080/1061186x.2021.1909051] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The liver is a vital metabolic and detoxifying organ and suffers diverse endogenous or exogenous damage. Hepatocyte mitochondria experience various structural and functional defects from liver injury, bearing oxidative stress, metabolic dysregulation, and the disturbance of mitochondrial quality control (MQC) mechanisms. Mitochondrial malfunction initiates the mitochondria-mediated apoptotic pathways and the release of damage signals, aggravating liver damage and disease progression via inflammation and reparative fibrogenesis. Removal of mitochondrial impairment or the improvement of MQC mechanisms restore mitochondrial homeostasis and benefit liver health. This review discusses the association of mitochondrial disorders with hepatic pathophysiological processes and the resultant potential of mitochondrion-targeting therapeutics for hepatic disorders. The recent advances in the MQC mechanisms and the mitochondrial-derived damage-associated molecular patterns (DAMPs) in the pathology and treatment of liver disease are particularly focussed.
Collapse
Affiliation(s)
- Li Xiang
- School of Chemistry and Chemical Engineering, University of South China, Hengyang, China
| | - Yaru Shao
- Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, University of South China, Hengyang, China.,Institute of Pharmacy & Pharmacology, School of Pharmaceutical Science, University of South China, Hengyang, China
| | - Yuping Chen
- Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, University of South China, Hengyang, China.,Institute of Pharmacy & Pharmacology, School of Pharmaceutical Science, University of South China, Hengyang, China
| |
Collapse
|
30
|
Gong MJ, Zhu CY, Zou ZJ, Han B, Huang P. Therapeutic potential of puerarin against methionine-choline-deficient diet-induced non-alcoholic steatohepatitis determined by combination of 1H NMR spectroscopy-based metabonomics and 16S rRNA gene sequencing. J Pharm Biomed Anal 2021; 197:113964. [PMID: 33601157 DOI: 10.1016/j.jpba.2021.113964] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 02/05/2021] [Accepted: 02/06/2021] [Indexed: 12/12/2022]
Abstract
Previously published studies have revealed the protective effect of puerarin against non-alcoholic steatohepatitis (NASH), but the definite mechanism of this effect still remains unclear. The present work was an attempt to assess the beneficial effects and the underlying mechanisms of puerarin on methionine-choline-deficient (MCD) diet-induced NASH in C57BL/6 mice by using a combination of metabonomics and 16S rRNA gene sequencing technology. Nuclear magnetic resonance (NMR)-based metabonomics showed significant hepatic and urinary metabolic phenotype changes between MCD-diet fed mice and the healthy controls. A total of eight and thirteen metabolites were identified as differential metabolites associated with NASH in liver tissue and urine of mice, respectively. The proposed pathways mainly included pyrimidine metabolism, one-carbon metabolism, amino acid metabolism, glycolysis, tricarboxylic acid (TCA) cycle and synthesis and degradation of ketone bodies. Furthermore, 16S rRNA gene sequencing analysis delineated remarkable variations in gut microbiota profiles in response to MCD diet in mice and forty differential bacterial taxa related to NASH were found between the control and model group. Puerarin could improve hepatic steatosis and inflammation in NASH mice via partially ameliorating metabolic disorders and rebalancing the gut flora. Specifically, puerarin could inhibit lipopolysaccharide (LPS)-producing genus Helicobacter, and promote butyrate-producing genus Roseburia. These findings offered novel insights into the in-depth understanding of the pathogenesis of NASH and provided further evidence for the potential use of puerarin as an anti-NASH agent.
Collapse
Affiliation(s)
- Meng-Juan Gong
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China; School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Cai-Yan Zhu
- The Sixth Affilicated Hospital, Sun Yat-sen University, Guangzhou 510655, China
| | - Zhong-Jie Zou
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Bin Han
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Ping Huang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China.
| |
Collapse
|