1
|
Hirota K, Yamauchi R, Miyata M, Kojima M, Kako K, Fukamizu A. Dietary methionine functions in proliferative zone maintenance and egg production via sams-1 in Caenorhabditis elegans. J Biochem 2024; 176:359-367. [PMID: 39046461 DOI: 10.1093/jb/mvae054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 06/29/2024] [Accepted: 07/22/2024] [Indexed: 07/25/2024] Open
Abstract
The maintenance of germ cells is critical for the prosperity of offspring. The amount of food consumption is known to be closely related to reproduction, i.e. the number of eggs decreases under calorie-restricted conditions in various organisms. Previous studies in Caenorhabditis elegans have reported that calorie restriction reduces the number of eggs and the reduction can be rescued by methionine. However, the effect of methionine on the reproductive process has not been fully understood. In this study, to assess the gonadal function of methionine metabolism, we firstly demonstrated that a depletion in dietary methionine resulted in reduced levels of S-adenosyl-l-methionine (SAM) and S-adenosyl homocysteine in wild-type N2, but not in glp-1 mutants, which possess only a few germ cells. Second, we found no recovery in egg numbers upon methionine administration in SAM synthase (sams)-1 mutants. Furthermore, a reduced number of proliferative zone nuclei exhibited in the sams-1 mutants was not rescued via methionine. Thus, our results have shown that dietary methionine is required for the normal establishment of both the germline progenitor pool and fecundity, mediated by sams-1.
Collapse
Affiliation(s)
- Keiko Hirota
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance, University of Tsukuba, 1-1-1 Tennoudai, Tsukuba, Ibaraki, 305-8577, Japan
- Department of Hygiene and Public Health, School of Medicine, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku, Tokyo,162-8666, Japan
| | - Rieko Yamauchi
- Graduate School of Life and Environmental Sciences, University of Tsukuba, 1-1-1 Tennoudai, Tsukuba, Ibaraki, 305-8577, Japan
| | - Mai Miyata
- Graduate School of Life and Environmental Sciences, University of Tsukuba, 1-1-1 Tennoudai, Tsukuba, Ibaraki, 305-8577, Japan
| | - Mariko Kojima
- Graduate School of Life and Environmental Sciences, University of Tsukuba, 1-1-1 Tennoudai, Tsukuba, Ibaraki, 305-8577, Japan
| | - Koichiro Kako
- Institute of Life and Environmental Sciences, University of Tsukuba, 1-1-1 Tennoudai, Tsukuba, Ibaraki, 305-8577, Japan
| | - Akiyoshi Fukamizu
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance, University of Tsukuba, 1-1-1 Tennoudai, Tsukuba, Ibaraki, 305-8577, Japan
| |
Collapse
|
2
|
Liu Q, Wang M, Hou Y, Chen R, Liu H, Han T, Liu D. Deciphering the multifaceted effects of artificial sweeteners on body health and metabolic functions: a comprehensive review and future perspectives. Crit Rev Food Sci Nutr 2024:1-23. [PMID: 39368060 DOI: 10.1080/10408398.2024.2411410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/07/2024]
Abstract
As the rates of chronic diseases such as obesity and diabetes rise worldwide, there is a growing demand for low-calorie or no-calorie sweeteners to reduce sugar intake without sacrificing the sweetness of foods and beverages. Artificial sweeteners have become indispensable as substitutes for sugar due to their high sweetening power and low impact on blood sugar levels and are used in a variety of low-calorie foods and beverages. Although artificial sweeteners offer an alternative for reducing sugar intake while maintaining sweetness, research into their long-term health effects, particularly at high doses, is ongoing, further scientific research and regulatory review are needed to clarify these potential health risks. This article reviews the latest research on the health effects of artificial sweeteners, based on recent studies, introduces the classification, performance, and safety standards for artificial sweeteners, analyses their potential harms to the nervous, immune, and circulatory systems, reproductive system, as well as their effects on gut microbiota, liver function, cancer, diabetes, and obesity. In addition, consumer perceptions of artificial sweeteners and future research directions are discussed, providing insights into current research controversies and knowledge gaps, as well as the health research and market application of artificial sweeteners.
Collapse
Affiliation(s)
- Qiang Liu
- College of Food Science and Engineering, Bohai University, Jinzhou, China
| | - Min Wang
- College of Food Science and Engineering, Bohai University, Jinzhou, China
| | - Yuting Hou
- College of Food Science and Engineering, Bohai University, Jinzhou, China
- Meat Innovation Center of Liaoning Province, Jinzhou, China
- Liaoning Kazuo Hybrid Wild Boar Science and Technology Backyard, Chaoyang, China
| | - Rui Chen
- College of Food Science and Engineering, Bohai University, Jinzhou, China
| | - Haixia Liu
- College of Food Science and Engineering, Bohai University, Jinzhou, China
| | - Tianlong Han
- College of Food Science and Engineering, Bohai University, Jinzhou, China
- Liaoning Kazuo Hybrid Wild Boar Science and Technology Backyard, Chaoyang, China
| | - Dengyong Liu
- College of Food Science and Engineering, Bohai University, Jinzhou, China
- Meat Innovation Center of Liaoning Province, Jinzhou, China
| |
Collapse
|
3
|
Martínez-Arranz I, Alonso C, Mayo R, Mincholé I, Mato JM, Lee DJ. Genetic algorithms applied to translational strategy in metabolic-dysfunction associated steatohepatitis (MASH). Learning from mouse models. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2024; 255:108346. [PMID: 39089186 DOI: 10.1016/j.cmpb.2024.108346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 02/05/2024] [Accepted: 07/20/2024] [Indexed: 08/03/2024]
Abstract
BACKGROUND & AIMS We previously identified subsets of patients with metabolic (dysfunction)-associated steatotic liver disease (MASLD) with different metabolic phenotypes. Here, we aimed to refine this classification based on genetic algorithms implemented in a Python package. The use of these genetic algorithms can help scientists to solve problems which cannot be solved with other methods. We present this package and its capabilities with specific problems. The name, PyGenMet, comes from its main goal, solving problems in Python with Genetic Algorithms and Metabolomics data. METHODS We collected serum from methionine adenosyltransferase 1a knockout (Mat1a-KO) mice, which have chronically low level of hepatic S-adenosylmethionine (SAMe) and the metabolomes of all samples were determined. We also analyzed serum metabolomes of 541 patients with biopsy proven MASLD (182 with simple steatosis and 359 with metabolic (dysfunction)-associated steatohepatitis or MASH) and compared them with the serum metabolomes of this specific MASLD mouse model using Genetic Algorithms in order to select patients with a specific phenotype. RESULTS By applying genetic algorithms, we have found a subgroup of patients with a lipid profile similar to that observed in the mouse model. When analyzing the two groups of patients, we have seen that patients with a lipid profile reflecting the mouse model characteristics show significant differences in lipoproteins, especially in LDL-4, LDL-5, and LDL-6 associated with atherogenic risk. CONCLUSION The results show that the application of genetic algorithms to subclassify patients with MASLD (or other metabolic disease) give consistent results and are a good approximation for the treatment of large volumes of data such as those from omics sciences and patient classification.
Collapse
Affiliation(s)
- Ibon Martínez-Arranz
- OWL Metabolomics (Rubió Metabolomics), Derio, Bizkaia, Spain; Department of Mathematics, University of the Basque Country UPV/EHU, Bilbao, Spain.
| | - Cristina Alonso
- OWL Metabolomics (Rubió Metabolomics), Derio, Bizkaia, Spain
| | - Rebeca Mayo
- OWL Metabolomics (Rubió Metabolomics), Derio, Bizkaia, Spain
| | - Itziar Mincholé
- OWL Metabolomics (Rubió Metabolomics), Derio, Bizkaia, Spain
| | - José M Mato
- CIC bioGUNE, BRTA, CIBERehd, Derio, Bizkaia, Spain
| | - Dae-Jin Lee
- IE University - School of Science and Technology, Madrid, Spain
| |
Collapse
|
4
|
Wusiman M, Huang SY, Liu ZY, He TT, Fang AP, Li MC, Yang MT, Wang C, Zhang YJ, Zhu HL. Serum S-adenosylhomocysteine, rather than homocysteine, is associated with hepatocellular carcinoma survival: a prospective cohort study. Am J Clin Nutr 2024; 120:481-490. [PMID: 39025328 DOI: 10.1016/j.ajcnut.2024.07.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 06/29/2024] [Accepted: 07/15/2024] [Indexed: 07/20/2024] Open
Abstract
BACKGROUND Emerging evidence suggested that S-adenosylhomocysteine (SAH) may be a better serum biomarker for cardiovascular disease than homocysteine (Hcy). However, the role of SAH in hepatocellular carcinoma (HCC) prognosis remains unclear. OBJECTIVES We aimed to prospectively explore the relationships between serum SAH and related metabolites [Hcy, S-adenosylmethionine (SAM)] with HCC survival, and to evaluate the effect modifications by gene polymorphisms in one-carbon metabolism key enzymes. METHODS We included 1080 newly diagnosed patients with HCC from the Guangdong Liver Cancer Cohort. Serum SAH, Hcy, and SAM were measured utilizing high-performance liquid chromatography-tandem mass spectrometry. Gene polymorphisms in one-carbon metabolism key enzymes were identified using kompetitive allele-specific polymerase chain reaction. Primary outcomes were liver cancer-specific survival (LCSS) and overall survival (OS). Hazard ratios (HRs) and 95% confidence intervals (CIs) were computed using multivariate Cox proportional hazards models. RESULTS After a median follow-up of 3.6 y, 601 deaths occurred, with 552 (92%) attributed to HCC. Multivariable analysis revealed that patients in the highest quartile of serum SAH concentrations were significantly associated with worse survival compared with those in the lowest quartile, with HRs of 1.58 (95% CI: 1.19, 2.10; P-trend = 0.002) for LCSS and 1.54 (95% CI: 1.18, 2.02; P-trend = 0.001) for OS. There were no significant interactions between serum SAH concentrations and genetic variants of one-carbon metabolism key enzymes. No significant associations were found between serum Hcy, SAM concentrations, and SAM/SAH ratio with LCSS or OS. CONCLUSIONS Higher serum SAH concentrations, rather than Hcy, were independently associated with worse survival in patients with HCC, regardless of the genetic variants of one-carbon metabolism key enzymes. These findings suggest that SAH may be a novel metabolism-related prognostic biomarker for HCC.
Collapse
Affiliation(s)
- Maierhaba Wusiman
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Si-Yu Huang
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Zhao-Yan Liu
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Food, Nutrition and Health, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Tong-Tong He
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Ai-Ping Fang
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Food, Nutrition and Health, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Meng-Chu Li
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Meng-Tao Yang
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Chen Wang
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Yao-Jun Zhang
- Department of Hepatobiliary Surgery, Sun Yat-sen University Cancer Center, Guangzhou, China; State Key Laboratory of Oncology in South China, Guangzhou, China.
| | - Hui-Lian Zhu
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Food, Nutrition and Health, School of Public Health, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
5
|
Fogal V, Michopoulos F, Jarnuczak AF, Hamza GM, Harlfinger S, Davey P, Hulme H, Atkinson SJ, Gabrowski P, Cheung T, Grondine M, Hoover C, Rose J, Bray C, Foster AJ, Askin S, Majumder MM, Fitzpatrick P, Miele E, Macdonald R, Keun HC, Coen M. Mechanistic safety assessment via multi-omic characterisation of systemic pathway perturbations following in vivo MAT2A inhibition. Arch Toxicol 2024; 98:2589-2603. [PMID: 38755480 PMCID: PMC11272821 DOI: 10.1007/s00204-024-03771-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 04/24/2024] [Indexed: 05/18/2024]
Abstract
The tumour suppressor p16/CDKN2A and the metabolic gene, methyl-thio-adenosine phosphorylase (MTAP), are frequently co-deleted in some of the most aggressive and currently untreatable cancers. Cells with MTAP deletion are vulnerable to inhibition of the metabolic enzyme, methionine-adenosyl transferase 2A (MAT2A), and the protein arginine methyl transferase (PRMT5). This synthetic lethality has paved the way for the rapid development of drugs targeting the MAT2A/PRMT5 axis. MAT2A and its liver- and pancreas-specific isoform, MAT1A, generate the universal methyl donor S-adenosylmethionine (SAM) from ATP and methionine. Given the pleiotropic role SAM plays in methylation of diverse substrates, characterising the extent of SAM depletion and downstream perturbations following MAT2A/MAT1A inhibition (MATi) is critical for safety assessment. We have assessed in vivo target engagement and the resultant systemic phenotype using multi-omic tools to characterise response to a MAT2A inhibitor (AZ'9567). We observed significant SAM depletion and extensive methionine accumulation in the plasma, liver, brain and heart of treated rats, providing the first assessment of both global SAM depletion and evidence of hepatic MAT1A target engagement. An integrative analysis of multi-omic data from liver tissue identified broad perturbations in pathways covering one-carbon metabolism, trans-sulfuration and lipid metabolism. We infer that these pathway-wide perturbations represent adaptive responses to SAM depletion and confer a risk of oxidative stress, hepatic steatosis and an associated disturbance in plasma and cellular lipid homeostasis. The alterations also explain the dramatic increase in plasma and tissue methionine, which could be used as a safety and PD biomarker going forward to the clinic.
Collapse
Affiliation(s)
- Valentina Fogal
- Oncology Safety, Safety Sciences, Clinical Pharmacology & Safety Sciences, R&D, AstraZeneca, Cambridge, UK
- Cancer Metabolism & Systems Toxicology Group, Division of Cancer, Department of Surgery and Cancer, Imperial College London, London, UK
| | - Filippos Michopoulos
- Bioscience, Research and Early Development, Oncology R&D, AstraZeneca, Cambridge, UK
| | - Andrew F Jarnuczak
- Data Sciences & Quantitative Biology, Discovery Sciences, R&D, AstraZeneca, Cambridge, UK
| | - Ghaith M Hamza
- Discovery Biology, Discovery Sciences, R&D, AstraZeneca, R&D Boston, Waltham, USA
| | | | - Paul Davey
- Chemistry, Oncology R&D AstraZeneca, Cambridge, UK
| | - Heather Hulme
- Imaging and Data Analytics, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Cambridge, UK
| | | | - Piotr Gabrowski
- Biological Insights Knowledge Graph, R&D IT, AstraZeneca, Barcelona, Spain
| | - Tony Cheung
- Oncology R&D, AstraZeneca, R&D Boston, Waltham, USA
| | | | - Clare Hoover
- Oncology Safety Pathology, Clinical Pharmacology & Safety Sciences, R&D, AstraZeneca, R&D Boston, Waltham, USA
| | - Jonathan Rose
- Animal Science & Technologies, R&D, AstraZeneca, Cambridge, UK
| | - Chandler Bray
- Cancer Metabolism & Systems Toxicology Group, Division of Cancer, Department of Surgery and Cancer, Imperial College London, London, UK
| | - Alison J Foster
- Regulatory Toxicology and Safety Pharmacology, Clinical Pharmacology & Safety Sciences, R&D, AstraZeneca, Cambridge, UK
| | - Sean Askin
- Advanced Drug Delivery, Pharmaceutical Sci, R&D, AstraZeneca, Cambridge, UK
| | - Muntasir Mamun Majumder
- Safety Sciences, Clinical Pharmacology & Safety Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - Paul Fitzpatrick
- Safety Sciences, Clinical Pharmacology & Safety Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - Eric Miele
- Discovery Biology, Discovery Sciences, R&D, AstraZeneca, R&D Boston, Waltham, USA
| | - Ruth Macdonald
- Animal Science & Technologies, R&D, AstraZeneca, Cambridge, UK
| | - Hector C Keun
- Cancer Metabolism & Systems Toxicology Group, Division of Cancer, Department of Surgery and Cancer, Imperial College London, London, UK
| | - Muireann Coen
- Oncology Safety, Safety Sciences, Clinical Pharmacology & Safety Sciences, R&D, AstraZeneca, Cambridge, UK.
| |
Collapse
|
6
|
Marigorta UM, Millet O, Lu SC, Mato JM. Dysfunctional VLDL metabolism in MASLD. NPJ METABOLIC HEALTH AND DISEASE 2024; 2:16. [PMID: 39049993 PMCID: PMC11263124 DOI: 10.1038/s44324-024-00018-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Accepted: 06/22/2024] [Indexed: 07/27/2024]
Abstract
Lipidomics has unveiled the intricate human lipidome, emphasizing the extensive diversity within lipid classes in mammalian tissues critical for cellular functions. This diversity poses a challenge in maintaining a delicate balance between adaptability to recurring physiological changes and overall stability. Metabolic Dysfunction-Associated Steatotic Liver Disease (MASLD), linked to factors such as obesity and diabetes, stems from a compromise in the structural and functional stability of the liver within the complexities of lipid metabolism. This compromise inaccurately senses an increase in energy status, such as during fasting-feeding cycles or an upsurge in lipogenesis. Serum lipidomic studies have delineated three distinct metabolic phenotypes, or "metabotypes" in MASLD. MASLD-A is characterized by lower very low-density lipoprotein (VLDL) secretion and triglyceride (TG) levels, associated with a reduced risk of cardiovascular disease (CVD). In contrast, MASLD-C exhibits increased VLDL secretion and TG levels, correlating with elevated CVD risk. An intermediate subtype, with a blend of features, is designated as the MASLD-B metabotype. In this perspective, we examine into recent findings that show the multifaceted regulation of VLDL secretion by S-adenosylmethionine, the primary cellular methyl donor. Furthermore, we explore the differential CVD and hepatic cancer risk across MASLD metabotypes and discuss the context and potential paths forward to gear the findings from genetic studies towards a better understanding of the observed heterogeneity in MASLD.
Collapse
Affiliation(s)
- Urko M. Marigorta
- Integrative Genomics Lab, CIC bioGUNE, Basque Research and Technology Alliance (BRTA), 48160 Derio, Spain
- Ikerbasque, Basque Foundation for Science, 48013 Bilbao, Spain
| | - Oscar Millet
- Precision Medicine and Metabolism Lab, CIC bioGUNE, Basque Research and Technology Alliance (BRTA), CIBERehd, 48160 Derio, Spain
| | - Shelly C. Lu
- Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, Los Angeles, CA 90048 USA
| | - José M. Mato
- Precision Medicine and Metabolism Lab, CIC bioGUNE, Basque Research and Technology Alliance (BRTA), CIBERehd, 48160 Derio, Spain
| |
Collapse
|
7
|
Hlady RA, Zhao X, El Khoury LY, Wagner RT, Luna A, Pham K, Pyrosopoulos NT, Jain D, Wang L, Liu C, Robertson KD. Epigenetic heterogeneity hotspots in human liver disease progression. Hepatology 2024:01515467-990000000-00966. [PMID: 39028883 DOI: 10.1097/hep.0000000000001023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 06/30/2024] [Indexed: 07/21/2024]
Abstract
BACKGROUND AND AIMS Disruption of the epigenome is a hallmark of human disease, including liver cirrhosis and HCC. While genetic heterogeneity is an established effector of pathologic phenotypes, epigenetic heterogeneity is less well understood. Environmental exposures alter the liver-specific DNA methylation landscape and influence the onset of liver cancer. Given that currently available treatments are unable to target frequently mutated genes in HCC, there is an unmet need for novel therapeutics to prevent or reverse liver damage leading to hepatic tumorigenesis, which the epigenome may provide. APPROACH AND RESULTS We performed genome-wide profiling of DNA methylation, copy number, and gene expression from multiple liver regions from 31 patients with liver disease to examine their crosstalk and define the individual and combinatorial contributions of these processes to liver disease progression. We identified epigenetic heterogeneity hotspots that are conserved across patients. Elevated epigenetic heterogeneity is associated with increased gene expression heterogeneity. Cirrhotic regions comprise 2 distinct cohorts-one exclusively epigenetic, and the other where epigenetic and copy number variations collaborate. Epigenetic heterogeneity hotspots are enriched for genes central to liver function (eg, HNF1A ) and known tumor suppressors (eg, RASSF1A ). These hotspots encompass genes including ACSL1 , ACSL5 , MAT1A , and ELFN1 , which have phenotypic effects in functional screens, supporting their relevance to hepatocarcinogenesis. Moreover, epigenetic heterogeneity hotspots are linked to clinical measures of outcome. CONCLUSIONS Substantial epigenetic heterogeneity arises early in liver disease development, targeting key pathways in the progression and initiation of both cirrhosis and HCC. Integration of epigenetic and transcriptional heterogeneity unveils putative epigenetic regulators of hepatocarcinogenesis.
Collapse
Affiliation(s)
- Ryan A Hlady
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota, USA
| | - Xia Zhao
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota, USA
| | - Louis Y El Khoury
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota, USA
| | - Ryan T Wagner
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota, USA
| | - Aesis Luna
- Department of Pathology, Yale School of Medicine, Yale University, New Haven, Connecticut, USA
| | - Kien Pham
- Department of Pathology, Yale School of Medicine, Yale University, New Haven, Connecticut, USA
| | | | - Dhanpat Jain
- Department of Pathology, Yale School of Medicine, Yale University, New Haven, Connecticut, USA
| | - Liguo Wang
- Division of Computational Biology, Mayo Clinic, Department of Quantitative Health Sciences, Rochester, Minnesota, USA
| | - Chen Liu
- Department of Pathology, Yale School of Medicine, Yale University, New Haven, Connecticut, USA
| | - Keith D Robertson
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
8
|
Wang C, Hei Y, Liu Y, Bajpai AK, Li Y, Guan Y, Xu F, Yao C. Systems genetics identifies methionine as a high risk factor for Alzheimer's disease. Front Neurosci 2024; 18:1381889. [PMID: 39081851 PMCID: PMC11286400 DOI: 10.3389/fnins.2024.1381889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Accepted: 06/25/2024] [Indexed: 08/02/2024] Open
Abstract
As a dietary strategy, methionine restriction has been reported to promote longevity and regulate metabolic disorders. However, the role and possible regulatory mechanisms underlying methionine in neurodegenerative diseases such as Alzheimer's disease (AD), remain unexplored. This study utilized the data from BXD recombinant inbred (RI) mice to establish a correlation between the AD phenotype in mice and methionine level. Gene enrichment analysis indicated that the genes associated with the concentration of methionine in the midbrain are involved in the dopaminergic synaptic signaling pathway. Protein interaction network analysis revealed that glycogen synthase kinase 3 beta (GSK-3β) was a key regulator of the dopaminergic synaptic pathway and its expression level was significantly correlated with the AD phenotype. Finally, in vitro experiments demonstrated that methionine deprivation could reduce the expression of Aβ and phosphorylated Tau, suggesting that lowering methionine levels in humans may be a preventive or therapeutic strategy for AD. In conclusion, our findings support that methionine is a high risk factor for AD. These findings predict potential regulatory network, theoretically supporting methionine restriction to prevent AD.
Collapse
Affiliation(s)
- Congmin Wang
- School of Pharmacy, Binzhou Medical University, Yantai, China
| | - Yu Hei
- School of Pharmacy, Binzhou Medical University, Yantai, China
| | - Yu Liu
- School of Pharmacy, Binzhou Medical University, Yantai, China
- Shandong Technology Innovation Center of Molecular Targeting and Intelligent Diagnosis and Treatment, Binzhou Medical University, Yantai, China
| | - Akhilesh Kumar Bajpai
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Yuhe Li
- School of Pharmacy, Binzhou Medical University, Yantai, China
| | - Yawen Guan
- School of Pharmacy, Binzhou Medical University, Yantai, China
| | - Fuyi Xu
- School of Pharmacy, Binzhou Medical University, Yantai, China
- Shandong Technology Innovation Center of Molecular Targeting and Intelligent Diagnosis and Treatment, Binzhou Medical University, Yantai, China
| | - Cuifang Yao
- School of Pharmacy, Binzhou Medical University, Yantai, China
- Shandong Technology Innovation Center of Molecular Targeting and Intelligent Diagnosis and Treatment, Binzhou Medical University, Yantai, China
| |
Collapse
|
9
|
Jing T, Wei D, Xu X, Wu C, Yuan L, Huang Y, Liu Y, Jiang Y, Wang B. Transposable elements-mediated recruitment of KDM1A epigenetically silences HNF4A expression to promote hepatocellular carcinoma. Nat Commun 2024; 15:5631. [PMID: 38965210 PMCID: PMC11224304 DOI: 10.1038/s41467-024-49926-2] [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: 09/12/2023] [Accepted: 06/25/2024] [Indexed: 07/06/2024] Open
Abstract
Transposable elements (TEs) contribute to gene expression regulation by acting as cis-regulatory elements that attract transcription factors and epigenetic regulators. This research aims to explore the functional and clinical implications of transposable element-related molecular events in hepatocellular carcinoma, focusing on the mechanism through which liver-specific accessible TEs (liver-TEs) regulate adjacent gene expression. Our findings reveal that the expression of HNF4A is inversely regulated by proximate liver-TEs, which facilitates liver cancer cell proliferation. Mechanistically, liver-TEs are predominantly occupied by the histone demethylase, KDM1A. KDM1A negatively influences the methylation of histone H3 Lys4 (H3K4) of liver-TEs, resulting in the epigenetic silencing of HNF4A expression. The suppression of HNF4A mediated by KDM1A promotes liver cancer cell proliferation. In conclusion, this study uncovers a liver-TE/KDM1A/HNF4A regulatory axis that promotes liver cancer growth and highlights KDM1A as a promising therapeutic target. Our findings provide insight into the transposable element-related molecular mechanisms underlying liver cancer progression.
Collapse
Affiliation(s)
- Tiantian Jing
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200032, China
| | - Dianhui Wei
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200032, China
| | - Xiaoli Xu
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200032, China
| | - Chengsi Wu
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200032, China
| | - Lili Yuan
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200032, China
| | - Yiwen Huang
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200032, China
| | - Yizhen Liu
- Department of Medical Oncology, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| | - Yanyi Jiang
- Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China.
- University of Science and Technology of China, Hefei, 230026, China.
| | - Boshi Wang
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200032, China.
| |
Collapse
|
10
|
Palit S, Shrestha AK, Thapa S, L. Grimm S, Coarfa C, Theis F, Simon LM, Shivanna B. Leveraging Integrated RNA Sequencing to Decipher Adrenomedullin's Protective Mechanisms in Experimental Bronchopulmonary Dysplasia. Genes (Basel) 2024; 15:806. [PMID: 38927741 PMCID: PMC11202456 DOI: 10.3390/genes15060806] [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: 05/06/2024] [Revised: 06/12/2024] [Accepted: 06/15/2024] [Indexed: 06/28/2024] Open
Abstract
Bronchopulmonary dysplasia (BPD) is a chronic lung disease commonly affecting premature infants, with limited therapeutic options and increased long-term consequences. Adrenomedullin (Adm), a proangiogenic peptide hormone, has been found to protect rodents against experimental BPD. This study aims to elucidate the molecular and cellular mechanisms through which Adm influences BPD pathogenesis using a lipopolysaccharide (LPS)-induced model of experimental BPD in mice. Bulk RNA sequencing of Adm-sufficient (wild-type or Adm+/+) and Adm-haplodeficient (Adm+/-) mice lungs, integrated with single-cell RNA sequencing data, revealed distinct gene expression patterns and cell type alterations associated with Adm deficiency and LPS exposure. Notably, computational integration with cell atlas data revealed that Adm-haplodeficient mouse lungs exhibited gene expression signatures characteristic of increased inflammation, natural killer (NK) cell frequency, and decreased endothelial cell and type II pneumocyte frequency. Furthermore, in silico human BPD patient data analysis supported our cell type frequency finding, highlighting elevated NK cells in BPD infants. These results underscore the protective role of Adm in experimental BPD and emphasize that it is a potential therapeutic target for BPD infants with an inflammatory phenotype.
Collapse
Affiliation(s)
- Subarna Palit
- TUM School of Life Sciences Weihenstephan, Technical University of Munich, 85354 Freising, Germany
| | - Amrit Kumar Shrestha
- Division of Neonatology, Department of Pediatrics, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX 77030, USA (B.S.)
| | - Shyam Thapa
- Division of Neonatology, Department of Pediatrics, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX 77030, USA (B.S.)
| | - Sandra L. Grimm
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Cristian Coarfa
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Fabian Theis
- TUM School of Life Sciences Weihenstephan, Technical University of Munich, 85354 Freising, Germany
- Institute of Computational Biology, Helmholtz Zentrum München, 85764 Neuherberg, Germany
- Department of Mathematics, Technical University of Munich, 85748 Garching, Germany
| | - Lukas M. Simon
- Therapeutic Innovation Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Binoy Shivanna
- Division of Neonatology, Department of Pediatrics, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX 77030, USA (B.S.)
| |
Collapse
|
11
|
Karin M, Kim JY. MASH as an emerging cause of hepatocellular carcinoma: current knowledge and future perspectives. Mol Oncol 2024. [PMID: 38874196 DOI: 10.1002/1878-0261.13685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 04/15/2024] [Accepted: 06/04/2024] [Indexed: 06/15/2024] Open
Abstract
Hepatocellular carcinoma is one of the deadliest and fastest-growing cancers. Among HCC etiologies, metabolic dysfunction-associated fatty liver disease (MAFLD) has served as a major HCC driver due to its great potential for increasing cirrhosis. The obesogenic environment fosters a positive energy balance and results in a continuous rise of obesity and metabolic syndrome. However, it is difficult to understand how metabolic complications lead to the poor prognosis of liver diseases and which molecular mechanisms are underpinning MAFLD-driven HCC development. Thus, suitable preclinical models that recapitulate human etiologies are essentially required. Numerous preclinical models have been created but not many mimicked anthropometric measures and the course of disease progression shown in the patients. Here we review the literature on adipose tissues, liver-related HCC etiologies and recently discovered genetic mutation signatures found in MAFLD-driven HCC patients. We also critically review current rodent models suggested for MAFLD-driven HCC study.
Collapse
Affiliation(s)
- Michael Karin
- Laboratory of Gene Regulation and Signal Transduction, Departments of Pharmacology and Pathology, School of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Ju Youn Kim
- Department of Molecular and Life Science, Hanyang University ERICA, Ansan, Korea
| |
Collapse
|
12
|
Xiao Y, Han C, Li X, Zhu X, Li S, Jiang N, Yu C, Liu Y, Liu F. S-Adenosylmethionine (SAM) diet promotes innate immunity via histone H3K4me3 complex. Int Immunopharmacol 2024; 131:111837. [PMID: 38471365 DOI: 10.1016/j.intimp.2024.111837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 02/27/2024] [Accepted: 03/08/2024] [Indexed: 03/14/2024]
Abstract
S-adenosylmethionine (SAM) was a methyl donor for modifying histones, which had crucial roles in lipid accumulation, tissue injury, and immune responses. SAM fluctuation might be linked to variations in histone methylation. However, the underlying molecular mechanisms of whether the SAM diet influenced the immune response via histone modification remained obscure. In this study, we utilized the Caenorhabditis elegans as a model to investigate the role of SAM diet in innate immunity. We found that 50 μM SAM increased resistance to Gram-negative pathogen Pseudomonas aeruginosa PA14 by reducing the bacterial burden in the intestine. Furthermore, through the genetic screening in C. elegans, we found that SAM functioned in germline to enhance innate immunity via an H3K4 methyltransferase complex to upregulate the immune response genes, including irg-1 and T24B8.5. Intriguingly, SAM also protected mice from P. aeruginosa PA14 infection by reducing the bacterial burden in lung. These findings provided insight into the mechanisms of molecular connections among SAM diet, histone modifications and innate immunity.
Collapse
Affiliation(s)
- Yi Xiao
- Guizhou Provincial College-based Key Lab for Tumor Prevention and Treatment with Distinctive Medicines, Zunyi Medical University, Zunyi, Guizhou 563000, China; College of Basic Medicine, Zunyi Medical University, Zunyi, Guizhou 563000, China; Institute of Life Sciences, Zunyi Medical University, Zunyi, Guizhou 563000, China.
| | - Chao Han
- Guizhou Provincial College-based Key Lab for Tumor Prevention and Treatment with Distinctive Medicines, Zunyi Medical University, Zunyi, Guizhou 563000, China; College of Basic Medicine, Zunyi Medical University, Zunyi, Guizhou 563000, China; Institute of Life Sciences, Zunyi Medical University, Zunyi, Guizhou 563000, China
| | - Xiaocong Li
- Guizhou Provincial College-based Key Lab for Tumor Prevention and Treatment with Distinctive Medicines, Zunyi Medical University, Zunyi, Guizhou 563000, China; College of Basic Medicine, Zunyi Medical University, Zunyi, Guizhou 563000, China; Institute of Life Sciences, Zunyi Medical University, Zunyi, Guizhou 563000, China
| | - Xinting Zhu
- Guizhou Provincial College-based Key Lab for Tumor Prevention and Treatment with Distinctive Medicines, Zunyi Medical University, Zunyi, Guizhou 563000, China; College of Basic Medicine, Zunyi Medical University, Zunyi, Guizhou 563000, China
| | - Sanhua Li
- Guizhou Provincial College-based Key Lab for Tumor Prevention and Treatment with Distinctive Medicines, Zunyi Medical University, Zunyi, Guizhou 563000, China; College of Basic Medicine, Zunyi Medical University, Zunyi, Guizhou 563000, China; Institute of Life Sciences, Zunyi Medical University, Zunyi, Guizhou 563000, China
| | - Nian Jiang
- Guizhou Provincial College-based Key Lab for Tumor Prevention and Treatment with Distinctive Medicines, Zunyi Medical University, Zunyi, Guizhou 563000, China; College of Basic Medicine, Zunyi Medical University, Zunyi, Guizhou 563000, China; Institute of Life Sciences, Zunyi Medical University, Zunyi, Guizhou 563000, China
| | - Changyan Yu
- Guizhou Provincial College-based Key Lab for Tumor Prevention and Treatment with Distinctive Medicines, Zunyi Medical University, Zunyi, Guizhou 563000, China; College of Basic Medicine, Zunyi Medical University, Zunyi, Guizhou 563000, China; Institute of Life Sciences, Zunyi Medical University, Zunyi, Guizhou 563000, China
| | - Yun Liu
- Guizhou Provincial College-based Key Lab for Tumor Prevention and Treatment with Distinctive Medicines, Zunyi Medical University, Zunyi, Guizhou 563000, China; College of Basic Medicine, Zunyi Medical University, Zunyi, Guizhou 563000, China; Institute of Life Sciences, Zunyi Medical University, Zunyi, Guizhou 563000, China.
| | - Fang Liu
- Guizhou Provincial College-based Key Lab for Tumor Prevention and Treatment with Distinctive Medicines, Zunyi Medical University, Zunyi, Guizhou 563000, China; College of Basic Medicine, Zunyi Medical University, Zunyi, Guizhou 563000, China.
| |
Collapse
|
13
|
Liu Y, Tian C, Yang Z, Huang C, Jiao K, Yang L, Duan C, Zhang Z, Li G. Effects of Chronic Heat Stress on Growth, Apoptosis, Antioxidant Enzymes, Transcriptomic Profiles, and Immune-Related Genes of Hong Kong Catfish ( Clarias fuscus). Animals (Basel) 2024; 14:1006. [PMID: 38612245 PMCID: PMC11010891 DOI: 10.3390/ani14071006] [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: 01/19/2024] [Revised: 03/14/2024] [Accepted: 03/19/2024] [Indexed: 04/14/2024] Open
Abstract
Chronic heat stress can have detrimental effects on the survival of fish. This study aimed to investigate the impact of prolonged high temperatures on the growth, antioxidant capacity, apoptosis, and transcriptome analysis of Hong Kong catfish (Clarias fuscus). By analyzing the morphological statistics of C. fuscus subjected to chronic high-temperature stress for 30, 60, and 90 days, it was observed that the growth of C. fuscus was inhibited compared to the control group. The experimental group showed a significant decrease in body weight and body length compared to the control group after 60 and 90 days of high-temperature stress (p < 0.05, p < 0.01). A biochemical analysis revealed significant alterations in the activities of three antioxidant enzymes superoxide dismutase activity (SOD); catalase activity (CAT); glutathione peroxidase activity (GPx), the malondialdehyde content (MDA), and the concentrations of serum alkaline phosphatase (ALP); Aspartate aminotransferase (AST); and alanine transaminase (ALT) in the liver. TUNEL staining indicated stronger apoptotic signals in the high-temperature-stress group compared to the control group, suggesting that chronic high-temperature-induced oxidative stress, leading to liver tissue injury and apoptosis. Transcriptome analysis identified a total of 1330 DEGs, with 835 genes being upregulated and 495 genes being downregulated compared to the control group. These genes may be associated with oxidative stress, apoptosis, and immune response. The findings elucidate the growth changes in C. fuscus under chronic high temperature and provide insights into the underlying response mechanisms to a high-temperature environment.
Collapse
Affiliation(s)
- Yong Liu
- Guangdong Research Center on Reproductive Control and Breeding Technology of Indigenous Valuable Fish Species, Guangdong Provincial Engineering Laboratory for Mariculture Organism Breeding, Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, Fisheries College, Guangdong Ocean University, Zhanjiang 524088, China; (Y.L.); (C.T.); (Z.Y.); (K.J.); (L.Y.); (C.D.)
| | - Changxu Tian
- Guangdong Research Center on Reproductive Control and Breeding Technology of Indigenous Valuable Fish Species, Guangdong Provincial Engineering Laboratory for Mariculture Organism Breeding, Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, Fisheries College, Guangdong Ocean University, Zhanjiang 524088, China; (Y.L.); (C.T.); (Z.Y.); (K.J.); (L.Y.); (C.D.)
| | - Zhihua Yang
- Guangdong Research Center on Reproductive Control and Breeding Technology of Indigenous Valuable Fish Species, Guangdong Provincial Engineering Laboratory for Mariculture Organism Breeding, Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, Fisheries College, Guangdong Ocean University, Zhanjiang 524088, China; (Y.L.); (C.T.); (Z.Y.); (K.J.); (L.Y.); (C.D.)
| | - Cailin Huang
- Guangxi Introduction and Breeding Center of Aquaculture, Nanning 530001, China; (C.H.); (Z.Z.)
| | - Kaizhi Jiao
- Guangdong Research Center on Reproductive Control and Breeding Technology of Indigenous Valuable Fish Species, Guangdong Provincial Engineering Laboratory for Mariculture Organism Breeding, Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, Fisheries College, Guangdong Ocean University, Zhanjiang 524088, China; (Y.L.); (C.T.); (Z.Y.); (K.J.); (L.Y.); (C.D.)
| | - Lei Yang
- Guangdong Research Center on Reproductive Control and Breeding Technology of Indigenous Valuable Fish Species, Guangdong Provincial Engineering Laboratory for Mariculture Organism Breeding, Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, Fisheries College, Guangdong Ocean University, Zhanjiang 524088, China; (Y.L.); (C.T.); (Z.Y.); (K.J.); (L.Y.); (C.D.)
| | - Cunyu Duan
- Guangdong Research Center on Reproductive Control and Breeding Technology of Indigenous Valuable Fish Species, Guangdong Provincial Engineering Laboratory for Mariculture Organism Breeding, Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, Fisheries College, Guangdong Ocean University, Zhanjiang 524088, China; (Y.L.); (C.T.); (Z.Y.); (K.J.); (L.Y.); (C.D.)
| | - Zhixin Zhang
- Guangxi Introduction and Breeding Center of Aquaculture, Nanning 530001, China; (C.H.); (Z.Z.)
| | - Guangli Li
- Guangdong Research Center on Reproductive Control and Breeding Technology of Indigenous Valuable Fish Species, Guangdong Provincial Engineering Laboratory for Mariculture Organism Breeding, Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, Fisheries College, Guangdong Ocean University, Zhanjiang 524088, China; (Y.L.); (C.T.); (Z.Y.); (K.J.); (L.Y.); (C.D.)
| |
Collapse
|
14
|
Fan W, Cao D, Yang B, Wang J, Li X, Kitka D, Li TWH, You S, Shiao S, Gangi A, Posadas E, Di Vizio D, Tomasi ML, Seki E, Mato JM, Yang H, Lu SC. Hepatic prohibitin 1 and methionine adenosyltransferase α1 defend against primary and secondary liver cancer metastasis. J Hepatol 2024; 80:443-453. [PMID: 38086446 PMCID: PMC10922446 DOI: 10.1016/j.jhep.2023.11.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 11/06/2023] [Accepted: 11/13/2023] [Indexed: 12/24/2023]
Abstract
BACKGROUND & AIMS The liver is a common site of cancer metastasis, most commonly from colorectal cancer, and primary liver cancers that have metastasized are associated with poor outcomes. The underlying mechanisms by which the liver defends against these processes are largely unknown. Prohibitin 1 (PHB1) and methionine adenosyltransferase 1A (MAT1A) are highly expressed in the liver. They positively regulate each other and their deletion results in primary liver cancer. Here we investigated their roles in primary and secondary liver cancer metastasis. METHODS We identified common target genes of PHB1 and MAT1A using a metastasis array, and measured promoter activity and transcription factor binding using luciferase reporter assays and chromatin immunoprecipitation, respectively. We examined how PHB1 or MAT1A loss promotes liver cancer metastasis and whether their loss sensitizes to colorectal liver metastasis (CRLM). RESULTS Matrix metalloproteinase-7 (MMP-7) is a common target of MAT1A and PHB1 and its induction is responsible for increased migration and invasion when MAT1A or PHB1 is silenced. Mechanistically, PHB1 and MAT1A negatively regulate MMP7 promoter activity via an AP-1 site by repressing the MAFG-FOSB complex. Loss of MAT1A or PHB1 also increased MMP-7 in extracellular vesicles, which were internalized by colon and pancreatic cancer cells to enhance their oncogenicity. Low hepatic MAT1A or PHB1 expression sensitized to CRLM, but not if endogenous hepatic MMP-7 was knocked down first, which lowered CD4+ T cells while increasing CD8+ T cells in the tumor microenvironment. Hepatocytes co-cultured with colorectal cancer cells express less MAT1A/PHB1 but more MMP-7. Consistently, CRLM raised distant hepatocytes' MMP-7 expression in mice and humans. CONCLUSION We have identified a PHB1/MAT1A-MAFG/FOSB-MMP-7 axis that controls primary liver cancer metastasis and sensitization to CRLM. IMPACT AND IMPLICATIONS Primary and secondary liver cancer metastasis is associated with poor outcomes but whether the liver has underlying defense mechanism(s) against metastasis is unknown. Here we examined the hypothesis that hepatic prohibitin 1 (PHB1) and methionine adenosyltransferase 1A (MAT1A) cooperate to defend the liver against metastasis. Our studies found PHB1 and MAT1A form a complex that suppresses matrix metalloproteinase-7 (MMP-7) at the transcriptional level and loss of either PHB1 or MAT1A sensitizes the liver to metastasis via MMP-7 induction. Strategies that target the PHB1/MAT1A-MMP-7 axis may be a promising approach for the treatment of primary and secondary liver cancer metastasis.
Collapse
Affiliation(s)
- Wei Fan
- Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center (CSMC), Los Angeles, CA 90048, USA
| | - DuoYao Cao
- Department of Biomedical Sciences, CSMC, Los Angeles, CA 90048, USA
| | - Bing Yang
- Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center (CSMC), Los Angeles, CA 90048, USA; Department of Geriatric Endocrinology and Metabolism, Guangxi Key Laboratory of Precision Medicine in Cardio-cerebrovascular Diseases Control and Prevention, Guangxi Clinical Research Center for Cardio-cerebrovascular Diseases, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Jiaohong Wang
- Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center (CSMC), Los Angeles, CA 90048, USA
| | - Xiaomo Li
- Department of Pathology, CSMC, Los Angeles CA 90048, USA
| | - Diana Kitka
- Department of Biomedical Sciences, CSMC, Los Angeles, CA 90048, USA; Department of Surgery, Cedars-Sinai Cancer, CSMC, Los Angeles, CA, 90048, USA
| | - Tony W H Li
- Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center (CSMC), Los Angeles, CA 90048, USA
| | - Sungyong You
- Department of Biomedical Sciences, CSMC, Los Angeles, CA 90048, USA; Department of Surgery, Cedars-Sinai Cancer, CSMC, Los Angeles, CA, 90048, USA
| | - Stephen Shiao
- Department of Radiation Oncology, CSMC, LA, CA 90048, USA
| | | | | | - Dolores Di Vizio
- Department of Biomedical Sciences, CSMC, Los Angeles, CA 90048, USA; Department of Surgery, Cedars-Sinai Cancer, CSMC, Los Angeles, CA, 90048, USA
| | - Maria Lauda Tomasi
- Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center (CSMC), Los Angeles, CA 90048, USA
| | - Ekihiro Seki
- Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center (CSMC), Los Angeles, CA 90048, USA
| | - José M Mato
- CIC bioGUNE, Centro de Investigación Biomédica en Red de Enfermedades Hepáticasy Digestivas (Ciberehd), Basque Research and Technology Alliance (BRTA), Technology, Park of Bizkaia, 48160 Derio, Bizkaia, Spain
| | - Heping Yang
- Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center (CSMC), Los Angeles, CA 90048, USA
| | - Shelly C Lu
- Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center (CSMC), Los Angeles, CA 90048, USA.
| |
Collapse
|
15
|
Barbier-Torres L, Chhimwal J, Kim SY, Ramani K, Robinson A, Yang H, Van Eyk J, Liangpunsakul S, Seki E, Mato JM, Lu SC. S-Adenosylmethionine Negatively Regulates the Mitochondrial Respiratory Chain Repressor MCJ in the Liver. Int J Biol Sci 2024; 20:1218-1237. [PMID: 38385082 PMCID: PMC10878152 DOI: 10.7150/ijbs.90104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 12/29/2023] [Indexed: 02/23/2024] Open
Abstract
MCJ (Methylation-Controlled J protein), an endogenous repressor of the mitochondrial respiratory chain, is upregulated in multiple liver diseases but little is known about how it is regulated. S-adenosylmethionine (SAMe), the biological methyl donor, is frequently depleted in chronic liver diseases. Here, we show that SAMe negatively regulates MCJ in the liver. While deficiency in methionine adenosyltransferase alpha 1 (MATα1), enzyme that catalyzes SAMe biosynthesis, leads to hepatic MCJ upregulation, MAT1A overexpression and SAMe treatment reduced MCJ expression. We found that MCJ is methylated at lysine residues and that it interacts with MATα1 in liver mitochondria, likely to facilitate its methylation. Lastly, we observed that MCJ is upregulated in alcohol-associated liver disease, a condition characterized by reduced MAT1A expression and SAMe levels along with mitochondrial injury. MCJ silencing protected against alcohol-induced mitochondrial dysfunction and lipid accumulation. Our study demonstrates a new role of MATα1 and SAMe in reducing hepatic MCJ expression.
Collapse
Affiliation(s)
- Lucía Barbier-Torres
- Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Jyoti Chhimwal
- Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - So Yeon Kim
- Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Komal Ramani
- Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Aaron Robinson
- Smidt Heart Institute and Advanced Clinical Biosystems Research Institute, Los Angeles, CA, USA
| | - Heping Yang
- Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Jenny Van Eyk
- Smidt Heart Institute and Advanced Clinical Biosystems Research Institute, Los Angeles, CA, USA
| | - Suthat Liangpunsakul
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, USA
- Roudebush Veterans Administration Medical Center, Indianapolis, IN, USA
| | - Ekihiro Seki
- Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - José M Mato
- bioGUNE, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (Ciberehd), Basque Research and Technology Assembly (BRTA), Technology Park of Bizkaia, 48160 Derio, Bizkaia, Spain
| | - Shelly C Lu
- Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| |
Collapse
|
16
|
Jiang C, Liu J, He S, Xu W, Huang R, Pan W, Li X, Dai X, Guo J, Zhang T, Inuzuka H, Wang P, Asara JM, Xiao J, Wei W. PRMT1 orchestrates with SAMTOR to govern mTORC1 methionine sensing via Arg-methylation of NPRL2. Cell Metab 2023; 35:2183-2199.e7. [PMID: 38006878 PMCID: PMC11192564 DOI: 10.1016/j.cmet.2023.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 09/22/2023] [Accepted: 11/01/2023] [Indexed: 11/27/2023]
Abstract
Methionine is an essential branch of diverse nutrient inputs that dictate mTORC1 activation. In the absence of methionine, SAMTOR binds to GATOR1 and inhibits mTORC1 signaling. However, how mTORC1 is activated upon methionine stimulation remains largely elusive. Here, we report that PRMT1 senses methionine/SAM by utilizing SAM as a cofactor for an enzymatic activity-based regulation of mTORC1 signaling. Under methionine-sufficient conditions, elevated cytosolic SAM releases SAMTOR from GATOR1, which confers the association of PRMT1 with GATOR1. Subsequently, SAM-loaded PRMT1 methylates NPRL2, the catalytic subunit of GATOR1, thereby suppressing its GAP activity and leading to mTORC1 activation. Notably, genetic or pharmacological inhibition of PRMT1 impedes hepatic methionine sensing by mTORC1 and improves insulin sensitivity in aged mice, establishing the role of PRMT1-mediated methionine sensing at physiological levels. Thus, PRMT1 coordinates with SAMTOR to form the methionine-sensing apparatus of mTORC1 signaling.
Collapse
Affiliation(s)
- Cong Jiang
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA; Joint Research Center for Musculoskeletal Tumor of Shanghai Changzheng Hospital and University of Shanghai for Science and Technology, Spinal Tumor Center, Department of Orthopedic Oncology, Shanghai Changzheng Hospital, Shanghai 200003, China; Tongji University Cancer Center, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| | - Jing Liu
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | - Shaohui He
- Joint Research Center for Musculoskeletal Tumor of Shanghai Changzheng Hospital and University of Shanghai for Science and Technology, Spinal Tumor Center, Department of Orthopedic Oncology, Shanghai Changzheng Hospital, Shanghai 200003, China
| | - Wei Xu
- Joint Research Center for Musculoskeletal Tumor of Shanghai Changzheng Hospital and University of Shanghai for Science and Technology, Spinal Tumor Center, Department of Orthopedic Oncology, Shanghai Changzheng Hospital, Shanghai 200003, China
| | - Runzhi Huang
- Tongji University Cancer Center, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| | - Weijuan Pan
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Xiaolong Li
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | - Xiaoming Dai
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | - Jianping Guo
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | - Tao Zhang
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | - Hiroyuki Inuzuka
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | - Ping Wang
- Tongji University Cancer Center, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| | - John M Asara
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | - Jianru Xiao
- Joint Research Center for Musculoskeletal Tumor of Shanghai Changzheng Hospital and University of Shanghai for Science and Technology, Spinal Tumor Center, Department of Orthopedic Oncology, Shanghai Changzheng Hospital, Shanghai 200003, China.
| | - Wenyi Wei
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
17
|
Wang S, Friedman SL. Found in translation-Fibrosis in metabolic dysfunction-associated steatohepatitis (MASH). Sci Transl Med 2023; 15:eadi0759. [PMID: 37792957 PMCID: PMC10671253 DOI: 10.1126/scitranslmed.adi0759] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Accepted: 09/15/2023] [Indexed: 10/06/2023]
Abstract
Metabolic dysfunction-associated steatohepatitis (MASH) is a severe form of liver disease that poses a global health threat because of its potential to progress to advanced fibrosis, leading to cirrhosis and liver cancer. Recent advances in single-cell methodologies, refined disease models, and genetic and epigenetic insights have provided a nuanced understanding of MASH fibrogenesis, with substantial cellular heterogeneity in MASH livers providing potentially targetable cell-cell interactions and behavior. Unlike fibrogenesis, mechanisms underlying fibrosis regression in MASH are still inadequately understood, although antifibrotic targets have been recently identified. A refined antifibrotic treatment framework could lead to noninvasive assessment and targeted therapies that preserve hepatocellular function and restore the liver's architectural integrity.
Collapse
Affiliation(s)
- Shuang Wang
- Division of Liver Diseases, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Scott L. Friedman
- Division of Liver Diseases, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| |
Collapse
|
18
|
Jiachen Z, Paul Kwong Hang T, Kenneth Kak Yuen W, Vincent Chi Hang L. Pathological role of methionine in the initiation and progression of biliary atresia. Front Pediatr 2023; 11:1263836. [PMID: 37772039 PMCID: PMC10522914 DOI: 10.3389/fped.2023.1263836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 08/21/2023] [Indexed: 09/30/2023] Open
Abstract
Methionine (Met) is an essential amino acid, and its excessive dietary intake and/or its metabolism disturbance could lead to accumulation/depletion of hepatic Met and some of the key intermediates of these pathways, which would interfere normal liver function and would be associated with liver diseases. Biliary atresia (BA) is a life-threatening disease characterized by inflammatory fibrosclerosing changes of the intrahepatic and extrahepatic biliary systems and is the primary cause of obstructive neonatal cholestasis with a rapid course of liver failure. However, its pathogenesis remains unknown. Previous studies reported elevated Met level in patients with obstructive cholestasis, suggesting a potential link between Met and BA. This paper reviews the Met metabolism in normal conditions and its dysregulation under abnormal conditions, the possible causes of hypermethioninemia, and its connection to BA pathogenesis: Abnormal hepatic level of Met could lead to a perturbation of redox homeostasis and mitochondrial functions of hepatocytes, enhancement of viral infectivity, and dysregulation of innate and adaptative immune cells in response to infection/damage of the liver contributing to the initiation/progression of BA.
Collapse
Affiliation(s)
- Zheng Jiachen
- Department of Surgery, School of Clinical Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Tam Paul Kwong Hang
- Faculty of Medicine, Macau University of Science and Technology, Macau, Macau SAR, China
| | - Wong Kenneth Kak Yuen
- Department of Surgery, School of Clinical Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Department of Surgery, University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Lui Vincent Chi Hang
- Department of Surgery, School of Clinical Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Dr. Li Dak-Sum Research Centre, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| |
Collapse
|
19
|
Di Pietrantonio N, Cappellacci I, Mandatori D, Baldassarre MPA, Pandolfi A, Pipino C. Role of Epigenetics and Metabolomics in Predicting Endothelial Dysfunction in Type 2 Diabetes. Adv Biol (Weinh) 2023; 7:e2300172. [PMID: 37616517 DOI: 10.1002/adbi.202300172] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 06/15/2023] [Indexed: 08/26/2023]
Abstract
Type 2 diabetes (T2D) is a worldwide health problem and cardiovascular disease (CVD) is a leading cause of morbidity and mortality in T2D patients, making the prevention of CVD onset a major priority. It is therefore crucial to optimize diagnosis and treatment to reduce this burden. Endothelial dysfunction is one of the most important prognostic factors for CVD progression, thus novel approaches to identify the early phase of endothelial dysfunction may lead to specific preventive measures to reduce the occurrence of CVD. Nowadays, multiomics approaches have provided unprecedented opportunities to stratify T2D patients into endotypes, improve therapeutic treatment and outcome and amend the survival prediction. Among omics strategies, epigenetics and metabolomics are gaining increasing interest. Recently, a dynamic correlation between metabolic pathways and gene expression through chromatin remodeling, such as DNA methylation, has emerged, indicating new perspectives on the regulatory networks impacting cellular processes. Thus, a better understanding of epigenetic-metabolite relationships can provide insight into the physiological processes altered early in the endothelium that ultimately head to disease development. Here, recent studies on epigenetics and metabolomics related to CVD prevention potentially useful to identify disease biomarkers, as well as new therapies hopefully targeting the early phase of endothelial dysfunction are highlighted.
Collapse
Affiliation(s)
- Nadia Di Pietrantonio
- Department of Medical, Oral and Biotechnological Sciences, "G. d'Annunzio" University of Chieti-Pescara, Chieti, 66100, Italy
- Center for Advanced Studies and Technology-CAST, "G. d'Annunzio" University of Chieti-Pescara, Chieti, 66100, Italy
| | - Ilaria Cappellacci
- Department of Medical, Oral and Biotechnological Sciences, "G. d'Annunzio" University of Chieti-Pescara, Chieti, 66100, Italy
- Center for Advanced Studies and Technology-CAST, "G. d'Annunzio" University of Chieti-Pescara, Chieti, 66100, Italy
| | - Domitilla Mandatori
- Department of Medical, Oral and Biotechnological Sciences, "G. d'Annunzio" University of Chieti-Pescara, Chieti, 66100, Italy
- Center for Advanced Studies and Technology-CAST, "G. d'Annunzio" University of Chieti-Pescara, Chieti, 66100, Italy
| | - Maria Pompea Antonia Baldassarre
- Center for Advanced Studies and Technology-CAST, "G. d'Annunzio" University of Chieti-Pescara, Chieti, 66100, Italy
- Department of Medicine and Aging Sciences, "G. d'Annunzio" University Chieti-Pescara, Chieti, 66100, Italy
| | - Assunta Pandolfi
- Department of Medical, Oral and Biotechnological Sciences, "G. d'Annunzio" University of Chieti-Pescara, Chieti, 66100, Italy
- Center for Advanced Studies and Technology-CAST, "G. d'Annunzio" University of Chieti-Pescara, Chieti, 66100, Italy
| | - Caterina Pipino
- Department of Medical, Oral and Biotechnological Sciences, "G. d'Annunzio" University of Chieti-Pescara, Chieti, 66100, Italy
- Center for Advanced Studies and Technology-CAST, "G. d'Annunzio" University of Chieti-Pescara, Chieti, 66100, Italy
| |
Collapse
|
20
|
Aguree S, Zolnoori M, Atwood TP, Owora A. Association between choline supplementation and Alzheimer's disease risk: a systematic review protocol. Front Aging Neurosci 2023; 15:1242853. [PMID: 37700814 PMCID: PMC10493398 DOI: 10.3389/fnagi.2023.1242853] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 08/15/2023] [Indexed: 09/14/2023] Open
Abstract
Background and aims There is growing evidence suggesting choline intake might have beneficial effects on cognitive function in the elderly. However, some studies report no relationship between choline intake and cognitive function or improvement in Alzheimer's disease patients. This protocol is for a systematic review of choline intake and Alzheimer's disease that aims to assess the comparative clinical effectiveness of choline supplementation on Alzheimer's disease risk. Methods and analysis literature search will be performed in PubMed, MEDLINE, EMBASE, CINAHL, Scopus, Cochrane, and the Web of Science electronic databases from inception until October 2023. We will follow the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines. Studies will be included if they compared two different time points of choline biomarkers measures in men or women (65+) with Alzheimer's Disease. The risk of bias in the included studies will be assessed within the Covidence data-management software. Results This review will summarize the clinical trial and quasi-experimental evidence of choline intake on Alzheimer's disease risk for adults aged 65+. The results from all eligible studies included in the analysis will be presented in tables, text, and figures. A descriptive synthesis will present the characteristics of included studies (e.g., age, sex of participants, type, length of intervention and comparator, and outcome measures), critical appraisal results, and descriptions of the main findings. Discussion This systematic review will summarize the existing evidence on the association between Choline intake and AD and to make recommendations if appropriate. The results of this review will be considered with respect to whether there is enough evidence of benefit to merit a more definitive randomized controlled trial. The results will be disseminated through peer-reviewed journals population. Conclusion This protocol outlines the methodology for a systematic review of choline intake and AD. The resulting systematic review from this protocol will form an evidence-based foundation to advance nutrition care for individuals with AD or poor cognitive function. Systematic review registration http://www.crd.york.ac.uk/PROSPERO, identifier CRD42023395004.
Collapse
Affiliation(s)
- Sixtus Aguree
- Department of Applied Health Science, School of Public Health, Indiana University Bloomington, Bloomington, IN, United States
| | - Maryam Zolnoori
- Columbia University Irving Medical Center, New York, NY, United States
| | - Thea Patricia Atwood
- Department of Chemistry, Indiana University Bloomington, Bloomington, IN, United States
| | - Arthur Owora
- Department of Pediatrics, School of Medicine, Indiana University, Indianapolis, IN, United States
| |
Collapse
|
21
|
Capelo-Diz A, Lachiondo-Ortega S, Fernández-Ramos D, Cañas-Martín J, Goikoetxea-Usandizaga N, Serrano-Maciá M, González-Rellan MJ, Mosca L, Blazquez-Vicens J, Tinahones-Ruano A, Fondevila MF, Buyan M, Delgado TC, Gutierrez de Juan V, Ayuso-García P, Sánchez-Rueda A, Velasco-Avilés S, Fernández-Susavila H, Riobello-Suárez C, Dziechciarz B, Montiel-Duarte C, Lopitz-Otsoa F, Bizkarguenaga M, Bilbao-García J, Bernardo-Seisdedos G, Senra A, Soriano-Navarro M, Millet O, Díaz-Lagares Á, Crujeiras AB, Bao-Caamano A, Cabrera D, van Liempd S, Tamayo-Carro M, Borzacchiello L, Gomez-Santos B, Buqué X, Sáenz de Urturi D, González-Romero F, Simon J, Rodríguez-Agudo R, Ruiz A, Matute C, Beiroa D, Falcon-Perez JM, Aspichueta P, Rodríguez-Cuesta J, Porcelli M, Pajares MA, Ameneiro C, Fidalgo M, Aransay AM, Lama-Díaz T, Blanco MG, López M, Villa-Bellosta R, Müller TD, Nogueiras R, Woodhoo A, Martínez-Chantar ML, Varela-Rey M. Hepatic levels of S-adenosylmethionine regulate the adaptive response to fasting. Cell Metab 2023; 35:1373-1389.e8. [PMID: 37527658 PMCID: PMC10432853 DOI: 10.1016/j.cmet.2023.07.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 03/30/2023] [Accepted: 07/06/2023] [Indexed: 08/03/2023]
Abstract
There has been an intense focus to uncover the molecular mechanisms by which fasting triggers the adaptive cellular responses in the major organs of the body. Here, we show that in mice, hepatic S-adenosylmethionine (SAMe)-the principal methyl donor-acts as a metabolic sensor of nutrition to fine-tune the catabolic-fasting response by modulating phosphatidylethanolamine N-methyltransferase (PEMT) activity, endoplasmic reticulum-mitochondria contacts, β-oxidation, and ATP production in the liver, together with FGF21-mediated lipolysis and thermogenesis in adipose tissues. Notably, we show that glucagon induces the expression of the hepatic SAMe-synthesizing enzyme methionine adenosyltransferase α1 (MAT1A), which translocates to mitochondria-associated membranes. This leads to the production of this metabolite at these sites, which acts as a brake to prevent excessive β-oxidation and mitochondrial ATP synthesis and thereby endoplasmic reticulum stress and liver injury. This work provides important insights into the previously undescribed function of SAMe as a new arm of the metabolic adaptation to fasting.
Collapse
Affiliation(s)
- Alba Capelo-Diz
- Gene Regulatory Control in Disease Laboratory, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), University of Santiago de Compostela, Santiago de Compostela, A Coruña 15706, Spain
| | - Sofía Lachiondo-Ortega
- Liver Disease Laboratory, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, 48160 Derio, Spain
| | - David Fernández-Ramos
- Precision Medicine and Metabolism Laboratory, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, 48160 Derio, Spain; Centro de investigación Biomedica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de salud Carlos III, 28029 Madrid, Spain
| | - Jorge Cañas-Martín
- Gene Regulatory Control in Disease Laboratory, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), University of Santiago de Compostela, Santiago de Compostela, A Coruña 15706, Spain
| | - Naroa Goikoetxea-Usandizaga
- Liver Disease Laboratory, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, 48160 Derio, Spain
| | - Marina Serrano-Maciá
- Liver Disease Laboratory, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, 48160 Derio, Spain
| | - Maria J González-Rellan
- Department of Physiology, CIMUS, University of Santiago de Compostela, Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), Santiago de Compostela, A Coruña 15706, Spain
| | - Laura Mosca
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Via Luigi De Crecchio 7, 80138 Naples, Italy
| | - Joan Blazquez-Vicens
- Gene Regulatory Control in Disease Laboratory, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), University of Santiago de Compostela, Santiago de Compostela, A Coruña 15706, Spain
| | - Alberto Tinahones-Ruano
- Gene Regulatory Control in Disease Laboratory, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), University of Santiago de Compostela, Santiago de Compostela, A Coruña 15706, Spain
| | - Marcos F Fondevila
- Department of Physiology, CIMUS, University of Santiago de Compostela, Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), Santiago de Compostela, A Coruña 15706, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela, A Coruña 15706, Spain
| | - Mason Buyan
- Liver Disease Laboratory, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, 48160 Derio, Spain
| | - Teresa C Delgado
- Liver Disease Laboratory, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, 48160 Derio, Spain
| | - Virginia Gutierrez de Juan
- Precision Medicine and Metabolism Laboratory, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, 48160 Derio, Spain
| | - Paula Ayuso-García
- Gene Regulatory Control in Disease Laboratory, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), University of Santiago de Compostela, Santiago de Compostela, A Coruña 15706, Spain
| | - Alejandro Sánchez-Rueda
- Gene Regulatory Control in Disease Laboratory, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), University of Santiago de Compostela, Santiago de Compostela, A Coruña 15706, Spain
| | - Sergio Velasco-Avilés
- Gene Regulatory Control in Disease Laboratory, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), University of Santiago de Compostela, Santiago de Compostela, A Coruña 15706, Spain
| | - Héctor Fernández-Susavila
- Gene Regulatory Control in Disease Laboratory, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), University of Santiago de Compostela, Santiago de Compostela, A Coruña 15706, Spain
| | - Cristina Riobello-Suárez
- Gene Regulatory Control in Disease Laboratory, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), University of Santiago de Compostela, Santiago de Compostela, A Coruña 15706, Spain
| | - Bartlomiej Dziechciarz
- Gene Regulatory Control in Disease Laboratory, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), University of Santiago de Compostela, Santiago de Compostela, A Coruña 15706, Spain
| | - Cristina Montiel-Duarte
- The John van Geest Cancer Research Centre, School of Science and Technology, Nottingham Trent University, Nottingham NG11 8NS, UK
| | - Fernando Lopitz-Otsoa
- Precision Medicine and Metabolism Laboratory, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, 48160 Derio, Spain
| | - Maider Bizkarguenaga
- Precision Medicine and Metabolism Laboratory, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, 48160 Derio, Spain
| | - Jon Bilbao-García
- Precision Medicine and Metabolism Laboratory, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, 48160 Derio, Spain
| | - Ganeko Bernardo-Seisdedos
- Precision Medicine and Metabolism Laboratory, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, 48160 Derio, Spain
| | - Ana Senra
- CIMUS, University of Santiago de Compostela, Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), Santiago de Compostela, A Coruña 15706, Spain
| | - Mario Soriano-Navarro
- Electron Microscopy Core Facility, Centro de Investigación Príncipe Felipe (CIPF), Valencia 46012, Spain
| | - Oscar Millet
- Precision Medicine and Metabolism Laboratory, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, 48160 Derio, Spain
| | - Ángel Díaz-Lagares
- Epigenomics Unit, Cancer Epigenomics, Translational Medical Oncology Group (ONCOMET), Health Research Institute of Santiago de Compostela (IDIS), University Clinical Hospital of Santiago (CHUS/SERGAS), Santiago de Compostela, A Coruña 15706, Spain
| | - Ana B Crujeiras
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela, A Coruña 15706, Spain; Epigenomics in Endocrinology and Nutrition Group, Epigenomics Unit, Instituto de Investigacion Sanitaria de Santiago de Compostela (IDIS), Complejo Hospitalario Universitario de Santiago de Compostela (CHUS/SERGAS), 15706 Santiago de Compostela, Spain
| | - Aida Bao-Caamano
- Epigenomics in Endocrinology and Nutrition Group, Epigenomics Unit, Instituto de Investigacion Sanitaria de Santiago de Compostela (IDIS), Complejo Hospitalario Universitario de Santiago de Compostela (CHUS/SERGAS), 15706 Santiago de Compostela, Spain
| | - Diana Cabrera
- Metabolomics Platform, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, 48160 Derio, Spain
| | - Sebastiaan van Liempd
- Metabolomics Platform, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, 48160 Derio, Spain
| | - Miguel Tamayo-Carro
- Nerve Disorders Laboratory, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, 48160 Derio, Spain
| | - Luigi Borzacchiello
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Via Luigi De Crecchio 7, 80138 Naples, Italy
| | - Beatriz Gomez-Santos
- Department of Physiology, Faculty of Medicine and Nursing, University of the Basque Country UPV/EHU, Leioa, Spain
| | - Xabier Buqué
- Department of Physiology, Faculty of Medicine and Nursing, University of the Basque Country UPV/EHU, Leioa, Spain
| | - Diego Sáenz de Urturi
- Department of Physiology, Faculty of Medicine and Nursing, University of the Basque Country UPV/EHU, Leioa, Spain
| | - Francisco González-Romero
- Department of Physiology, Faculty of Medicine and Nursing, University of the Basque Country UPV/EHU, Leioa, Spain
| | - Jorge Simon
- Liver Disease Laboratory, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, 48160 Derio, Spain
| | - Rubén Rodríguez-Agudo
- Liver Disease Laboratory, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, 48160 Derio, Spain
| | - Asier Ruiz
- Laboratory of Neurobiology, Achucarro Basque Center for Neuroscience, Department of Neurosciences, University of Basque Country (UPV/EHU), Centro de investigación Biomedica en Red de Enfermedades Neurodegenerativas (CIBERNED), 48940 Leioa, Spain
| | - Carlos Matute
- Laboratory of Neurobiology, Achucarro Basque Center for Neuroscience, Department of Neurosciences, University of Basque Country (UPV/EHU), Centro de investigación Biomedica en Red de Enfermedades Neurodegenerativas (CIBERNED), 48940 Leioa, Spain
| | - Daniel Beiroa
- Experimental Biomedicine Center (CEBEGA), University of Santiago de Compostela, A Coruña 15706, Spain
| | - Juan M Falcon-Perez
- Centro de investigación Biomedica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de salud Carlos III, 28029 Madrid, Spain; Metabolomics Platform, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, 48160 Derio, Spain; IKERBASQUE, Basque Foundation for Science, Bilbao, Bizkaia 48009, Spain
| | - Patricia Aspichueta
- Centro de investigación Biomedica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de salud Carlos III, 28029 Madrid, Spain; Department of Physiology, Faculty of Medicine and Nursing, University of the Basque Country UPV/EHU, Leioa, Spain; Biocruces Bizkaia Health Research Institute, Barakaldo, Spain
| | - Juan Rodríguez-Cuesta
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, 48160 Derio, Spain
| | - Marina Porcelli
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Via Luigi De Crecchio 7, 80138 Naples, Italy
| | - María A Pajares
- Centro de Investigaciones Biológicas Margarita Salas (CSIC), Ramiro de Maeztu 9, 28040 Madrid, Spain
| | - Cristina Ameneiro
- Stem Cells and Human Diseases, CIMUS, University of Santiago de Compostela, Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), Santiago de Compostela, A Coruña 15706, Spain
| | - Miguel Fidalgo
- Stem Cells and Human Diseases, CIMUS, University of Santiago de Compostela, Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), Santiago de Compostela, A Coruña 15706, Spain
| | - Ana M Aransay
- Genome Analysis Plataform, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, 48160 Derio, Spain
| | - Tomas Lama-Díaz
- DNA Repair and Genome Integrity Laboratory, CIMUS, University of Santiago de Compostela, Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), Santiago de Compostela, A Coruña 15706, Spain
| | - Miguel G Blanco
- DNA Repair and Genome Integrity Laboratory, CIMUS, University of Santiago de Compostela, Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), Santiago de Compostela, A Coruña 15706, Spain; Department of Biochemistry and Molecular Biology, University of Santiago de Compostela, Plaza do Obradoiro s/n, Santiago de Compostela, Spain
| | - Miguel López
- Department of Physiology, CIMUS, University of Santiago de Compostela, Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), Santiago de Compostela, A Coruña 15706, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela, A Coruña 15706, Spain
| | - Ricardo Villa-Bellosta
- Department of Biochemistry and Molecular Biology, University of Santiago de Compostela, Plaza do Obradoiro s/n, Santiago de Compostela, Spain; Metabolic Homeostasis and Vascular Calcification Laboratory, Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), Universidade de Santiago de Compostela, Santiago de Compostela, A Coruña 15706, Spain
| | - Timo D Müller
- Institute for Diabetes and Obesity, Helmholtz Zentrum Munich, and German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany
| | - Rubén Nogueiras
- Department of Physiology, CIMUS, University of Santiago de Compostela, Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), Santiago de Compostela, A Coruña 15706, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela, A Coruña 15706, Spain; Oportunius Program, Galician Agency of Innovation (GAIN), Xunta de Galicia, Santiago de Compostela, A Coruña, Spain
| | - Ashwin Woodhoo
- Gene Regulatory Control in Disease Laboratory, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), University of Santiago de Compostela, Santiago de Compostela, A Coruña 15706, Spain; Nerve Disorders Laboratory, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, 48160 Derio, Spain; IKERBASQUE, Basque Foundation for Science, Bilbao, Bizkaia 48009, Spain; Oportunius Program, Galician Agency of Innovation (GAIN), Xunta de Galicia, Santiago de Compostela, A Coruña, Spain; Department of Functional Biology, University of Santiago de Compostela, Plaza do Obradoiro s/n, Santiago de Compostela, Spain
| | - María Luz Martínez-Chantar
- Liver Disease Laboratory, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, 48160 Derio, Spain; Centro de investigación Biomedica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de salud Carlos III, 28029 Madrid, Spain.
| | - Marta Varela-Rey
- Gene Regulatory Control in Disease Laboratory, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), University of Santiago de Compostela, Santiago de Compostela, A Coruña 15706, Spain; Liver Disease Laboratory, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, 48160 Derio, Spain; Centro de investigación Biomedica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de salud Carlos III, 28029 Madrid, Spain; Department of Biochemistry and Molecular Biology, University of Santiago de Compostela, Plaza do Obradoiro s/n, Santiago de Compostela, Spain.
| |
Collapse
|
22
|
Valdés S, Paredes SD, García Carreras C, Zuluaga P, Rancan L, Linillos-Pradillo B, Arias-Díaz J, Vara E. S-Adenosylmethionine Decreases Bacterial Translocation, Proinflammatory Cytokines, Oxidative Stress and Apoptosis Markers in Hepatic Ischemia-Reperfusion Injury in Wistar Rats. Antioxidants (Basel) 2023; 12:1539. [PMID: 37627534 PMCID: PMC10451188 DOI: 10.3390/antiox12081539] [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: 06/01/2023] [Revised: 07/22/2023] [Accepted: 07/27/2023] [Indexed: 08/27/2023] Open
Abstract
Hepatic ischemia/reperfusion injury (IRI) can seriously impair liver function. It is initiated by oxidative stress, resulting in inflammation and apoptosis-induced cellular damage. Glutathione (GSH) prevents oxidative stress. S-Adenosylmethionine (SAMet) is a GSH synthesis precursor that avoids the deficit in SAMet-synthetase activity and contributes to intracellular ATP repletion. It also acts as a methyl group donor, stabilizing hepatocyte membranes, among other functions. This study investigated the effect of SAMet on bacterial translocation and levels of proinflammatory cytokines, oxidative stress and apoptosis markers in male Wistar rats subjected to hepatic IRI. Animals were randomly divided into six groups: (1) sham operation, (3) animals undergoing 60 min of ischemia of the right lateral lobe for temporary occlusion of the portal vein and hepatic artery plus 10 min of reperfusion, and (5) the same as (3) but with a reperfusion period of 120 min. Groups 2, 4 and 6, respectively, are the same as (1), (3) and (5), except that animals received SAMet (20 mg/kg) 15 min before ischemia. GSH, ATP, lipid peroxidation (LPO), TNF-α, IL-1β, IL-6, total caspase-1 and caspase-9, total and cleaved caspase-3, and phosphatidylcholine were determined in the liver. Endotoxin, TNF-α, IL-1β, IL-6, IL-10 and LPO in vena cava and portal vein blood samples were also measured. Endotoxin and LPO levels as well as proinflammatory cytokines and apoptotic markers increased significantly in animals undergoing IRI, both after 10 and 120 min of reperfusion. IRI produced a significant decrease in GSH, ATP, portal IL-10 and phosphatidylcholine. SAMet treatment prevented these effects significantly and increased survival rate. The study suggests that SAMet exerts protective effects in hepatic IRI.
Collapse
Affiliation(s)
- Sergio Valdés
- Department of Biochemistry and Molecular Biology, School of Medicine, Complutense University of Madrid, Avda. Complutense, s/n, 28040 Madrid, Spain; (S.V.); (C.G.C.); (L.R.); (B.L.-P.); (E.V.)
| | - Sergio D. Paredes
- Department of Physiology, School of Medicine, Complutense University of Madrid, Avda. Complutense, s/n, 28040 Madrid, Spain
| | - Carmen García Carreras
- Department of Biochemistry and Molecular Biology, School of Medicine, Complutense University of Madrid, Avda. Complutense, s/n, 28040 Madrid, Spain; (S.V.); (C.G.C.); (L.R.); (B.L.-P.); (E.V.)
| | - Pilar Zuluaga
- Departmental Unit of Biostatistics—Department of Statistics and Operations Research, School of Medicine, Complutense University of Madrid, Avda. Complutense, s/n, 28040 Madrid, Spain;
| | - Lisa Rancan
- Department of Biochemistry and Molecular Biology, School of Medicine, Complutense University of Madrid, Avda. Complutense, s/n, 28040 Madrid, Spain; (S.V.); (C.G.C.); (L.R.); (B.L.-P.); (E.V.)
| | - Beatriz Linillos-Pradillo
- Department of Biochemistry and Molecular Biology, School of Medicine, Complutense University of Madrid, Avda. Complutense, s/n, 28040 Madrid, Spain; (S.V.); (C.G.C.); (L.R.); (B.L.-P.); (E.V.)
| | - Javier Arias-Díaz
- Department of Surgery, School of Medicine, Complutense University of Madrid, Avda. Complutense, s/n, 28040 Madrid, Spain;
| | - Elena Vara
- Department of Biochemistry and Molecular Biology, School of Medicine, Complutense University of Madrid, Avda. Complutense, s/n, 28040 Madrid, Spain; (S.V.); (C.G.C.); (L.R.); (B.L.-P.); (E.V.)
| |
Collapse
|
23
|
Basha A, May SC, Anderson RM, Samala N, Mirmira RG. Non-Alcoholic Fatty Liver Disease: Translating Disease Mechanisms into Therapeutics Using Animal Models. Int J Mol Sci 2023; 24:9996. [PMID: 37373143 PMCID: PMC10298283 DOI: 10.3390/ijms24129996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 06/06/2023] [Accepted: 06/09/2023] [Indexed: 06/29/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a range of pathologies arising from fat accumulation in the liver in the absence of excess alcohol use or other causes of liver disease. Its complications include cirrhosis and liver failure, hepatocellular carcinoma, and eventual death. NAFLD is the most common cause of liver disease globally and is estimated to affect nearly one-third of individuals in the United States. Despite knowledge that the incidence and prevalence of NAFLD are increasing, the pathophysiology of the disease and its progression to cirrhosis remain insufficiently understood. The molecular pathogenesis of NAFLD involves insulin resistance, inflammation, oxidative stress, and endoplasmic reticulum stress. Better insight into these molecular pathways would allow for therapies that target specific stages of NAFLD. Preclinical animal models have aided in defining these mechanisms and have served as platforms for screening and testing of potential therapeutic approaches. In this review, we will discuss the cellular and molecular mechanisms thought to contribute to NAFLD, with a focus on the role of animal models in elucidating these mechanisms and in developing therapies.
Collapse
Affiliation(s)
- Amina Basha
- Kovler Diabetes Center, Section of Adult and Pediatric Endocrinology, Diabetes and Metabolism, Department of Medicine, The University of Chicago, Chicago, IL 60637, USA
| | - Sarah C. May
- Kovler Diabetes Center, Section of Adult and Pediatric Endocrinology, Diabetes and Metabolism, Department of Medicine, The University of Chicago, Chicago, IL 60637, USA
| | - Ryan M. Anderson
- Kovler Diabetes Center, Section of Adult and Pediatric Endocrinology, Diabetes and Metabolism, Department of Medicine, The University of Chicago, Chicago, IL 60637, USA
| | - Niharika Samala
- Department of Medicine, Division of Gastroenterology and Hepatology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Raghavendra G. Mirmira
- Kovler Diabetes Center, Section of Adult and Pediatric Endocrinology, Diabetes and Metabolism, Department of Medicine, The University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|
24
|
Harada S, Taketomi Y, Aiba T, Kawaguchi M, Hirabayashi T, Uranbileg B, Kurano M, Yatomi Y, Murakami M. The Lysophospholipase PNPLA7 Controls Hepatic Choline and Methionine Metabolism. Biomolecules 2023; 13:biom13030471. [PMID: 36979406 PMCID: PMC10046082 DOI: 10.3390/biom13030471] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 02/28/2023] [Accepted: 02/28/2023] [Indexed: 03/08/2023] Open
Abstract
The in vivo roles of lysophospholipase, which cleaves a fatty acyl ester of lysophospholipid, remained unclear. Recently, we have unraveled a previously unrecognized physiological role of the lysophospholipase PNPLA7, a member of the Ca2+-independent phospholipase A2 (iPLA2) family, as a key regulator of the production of glycerophosphocholine (GPC), a precursor of endogenous choline, whose methyl groups are preferentially fluxed into the methionine cycle in the liver. PNPLA7 deficiency in mice markedly decreases hepatic GPC, choline, and several metabolites related to choline/methionine metabolism, leading to various symptoms reminiscent of methionine shortage. Overall metabolic alterations in the liver of Pnpla7-null mice in vivo largely recapitulate those in methionine-deprived hepatocytes in vitro. Reduction of the methyl donor S-adenosylmethionine (SAM) after methionine deprivation decreases the methylation of the PNPLA7 gene promoter, relieves PNPLA7 expression, and thereby increases GPC and choline levels, likely as a compensatory adaptation. In line with the view that SAM prevents the development of liver cancer, the expression of PNPLA7, as well as several enzymes in the choline/methionine metabolism, is reduced in human hepatocellular carcinoma. These findings uncover an unexplored role of a lysophospholipase in hepatic phospholipid catabolism coupled with choline/methionine metabolism.
Collapse
Affiliation(s)
- Sayaka Harada
- Laboratory of Microenvironmental and Metabolic Health Sciences, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan
| | - Yoshitaka Taketomi
- Laboratory of Microenvironmental and Metabolic Health Sciences, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan
| | - Toshiki Aiba
- Department of Radiation Effects Research, National Institutes for Quantum and Radiological Science and Technology, Chiba 263-8555, Japan
| | - Mai Kawaguchi
- Laboratory of Microenvironmental and Metabolic Health Sciences, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan
- Laboratory of Biomembrane, Department of Basic Medical Sciences, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan
| | - Tetsuya Hirabayashi
- Laboratory of Biomembrane, Department of Basic Medical Sciences, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan
| | - Baasanjav Uranbileg
- Department of Clinical Laboratory Medicine, The University of Tokyo, Tokyo 113-8655, Japan
| | - Makoto Kurano
- Department of Clinical Laboratory Medicine, The University of Tokyo, Tokyo 113-8655, Japan
| | - Yutaka Yatomi
- Department of Clinical Laboratory Medicine, The University of Tokyo, Tokyo 113-8655, Japan
| | - Makoto Murakami
- Laboratory of Microenvironmental and Metabolic Health Sciences, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan
- Correspondence: ; Tel.: +81-3-5841-1431
| |
Collapse
|
25
|
Godbole AA, Gopalan S, Nguyen TK, Munden AL, Lui DS, Fanelli MJ, Vo P, Lewis CA, Spinelli JB, Fazzio TG, Walker AK. S-adenosylmethionine synthases specify distinct H3K4me3 populations and gene expression patterns during heat stress. eLife 2023; 12:e79511. [PMID: 36756948 PMCID: PMC9984191 DOI: 10.7554/elife.79511] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 02/07/2023] [Indexed: 02/10/2023] Open
Abstract
Methylation is a widely occurring modification that requires the methyl donor S-adenosylmethionine (SAM) and acts in regulation of gene expression and other processes. SAM is synthesized from methionine, which is imported or generated through the 1-carbon cycle (1 CC). Alterations in 1 CC function have clear effects on lifespan and stress responses, but the wide distribution of this modification has made identification of specific mechanistic links difficult. Exploiting a dynamic stress-induced transcription model, we find that two SAM synthases in Caenorhabditis elegans, SAMS-1 and SAMS-4, contribute differently to modification of H3K4me3, gene expression and survival. We find that sams-4 enhances H3K4me3 in heat shocked animals lacking sams-1, however, sams-1 cannot compensate for sams-4, which is required to survive heat stress. This suggests that the regulatory functions of SAM depend on its enzymatic source and that provisioning of SAM may be an important regulatory step linking 1 CC function to phenotypes in aging and stress.
Collapse
Affiliation(s)
- Adwait A Godbole
- Program in Molecular Medicine, UMASS Chan Medical SchoolWorcesterUnited States
| | - Sneha Gopalan
- Cancer Center, UMASS Chan Medical SchoolWorcesterUnited States
- Department of Molecular, Cell, and Cancer Biology, UMASS Chan Medical SchoolWorcesterUnited States
| | - Thien-Kim Nguyen
- Program in Molecular Medicine, UMASS Chan Medical SchoolWorcesterUnited States
| | - Alexander L Munden
- Program in Molecular Medicine, UMASS Chan Medical SchoolWorcesterUnited States
| | - Dominique S Lui
- Program in Molecular Medicine, UMASS Chan Medical SchoolWorcesterUnited States
| | - Matthew J Fanelli
- Program in Molecular Medicine, UMASS Chan Medical SchoolWorcesterUnited States
| | - Paula Vo
- Program in Molecular Medicine, UMASS Chan Medical SchoolWorcesterUnited States
| | - Caroline A Lewis
- Program in Molecular Medicine, UMASS Chan Medical SchoolWorcesterUnited States
| | - Jessica B Spinelli
- Program in Molecular Medicine, UMASS Chan Medical SchoolWorcesterUnited States
- Cancer Center, UMASS Chan Medical SchoolWorcesterUnited States
| | - Thomas G Fazzio
- Cancer Center, UMASS Chan Medical SchoolWorcesterUnited States
- Department of Molecular, Cell, and Cancer Biology, UMASS Chan Medical SchoolWorcesterUnited States
| | - Amy K Walker
- Program in Molecular Medicine, UMASS Chan Medical SchoolWorcesterUnited States
- Department of Molecular, Cell, and Cancer Biology, UMASS Chan Medical SchoolWorcesterUnited States
| |
Collapse
|
26
|
Kitamura H. Ubiquitin-Specific Proteases (USPs) and Metabolic Disorders. Int J Mol Sci 2023; 24:3219. [PMID: 36834633 PMCID: PMC9966627 DOI: 10.3390/ijms24043219] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 02/02/2023] [Accepted: 02/03/2023] [Indexed: 02/10/2023] Open
Abstract
Ubiquitination and deubiquitination are reversible processes that modify the characteristics of target proteins, including stability, intracellular localization, and enzymatic activity. Ubiquitin-specific proteases (USPs) constitute the largest deubiquitinating enzyme family. To date, accumulating evidence indicates that several USPs positively and negatively affect metabolic diseases. USP22 in pancreatic β-cells, USP2 in adipose tissue macrophages, USP9X, 20, and 33 in myocytes, USP4, 7, 10, and 18 in hepatocytes, and USP2 in hypothalamus improve hyperglycemia, whereas USP19 in adipocytes, USP21 in myocytes, and USP2, 14, and 20 in hepatocytes promote hyperglycemia. In contrast, USP1, 5, 9X, 14, 15, 22, 36, and 48 modulate the progression of diabetic nephropathy, neuropathy, and/or retinopathy. USP4, 10, and 18 in hepatocytes ameliorates non-alcoholic fatty liver disease (NAFLD), while hepatic USP2, 11, 14, 19, and 20 exacerbate it. The roles of USP7 and 22 in hepatic disorders are controversial. USP9X, 14, 17, and 20 in vascular cells are postulated to be determinants of atherosclerosis. Moreover, mutations in the Usp8 and Usp48 loci in pituitary tumors cause Cushing syndrome. This review summarizes the current knowledge about the modulatory roles of USPs in energy metabolic disorders.
Collapse
Affiliation(s)
- Hiroshi Kitamura
- Laboratory of Comparative Medicine, School of Veterinary Medicine, Rakuno Gakuen University, Ebetsu 069-8501, Japan
| |
Collapse
|
27
|
Robinson AE, Binek A, Ramani K, Sundararaman N, Barbier-Torres L, Murray B, Venkatraman V, Kreimer S, Ardle AM, Noureddin M, Fernández-Ramos D, Lopitz-Otsoa F, Gutiérrez de Juan V, Millet O, Mato JM, Lu SC, Van Eyk JE. Hyperphosphorylation of hepatic proteome characterizes nonalcoholic fatty liver disease in S-adenosylmethionine deficiency. iScience 2023; 26:105987. [PMID: 36756374 PMCID: PMC9900401 DOI: 10.1016/j.isci.2023.105987] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 09/15/2022] [Accepted: 01/11/2023] [Indexed: 01/15/2023] Open
Abstract
Methionine adenosyltransferase 1a (MAT1A) is responsible for hepatic S-adenosyl-L-methionine (SAMe) biosynthesis. Mat1a -/- mice have hepatic SAMe depletion, develop nonalcoholic steatohepatitis (NASH) which is reversed with SAMe administration. We examined temporal alterations in the proteome/phosphoproteome in pre-disease and NASH Mat1a -/- mice, effects of SAMe administration, and compared to human nonalcoholic fatty liver disease (NAFLD). Mitochondrial and peroxisomal lipid metabolism proteins were altered in pre-disease mice and persisted in NASH Mat1a -/- mice, which exhibited more progressive alterations in cytoplasmic ribosomes, ER, and nuclear proteins. A common mechanism found in both pre-disease and NASH livers was a hyperphosphorylation signature consistent with casein kinase 2α (CK2α) and AKT1 activation, which was normalized by SAMe administration. This was mimicked in human NAFLD with a metabolomic signature (M-subtype) resembling Mat1a -/- mice. In conclusion, we have identified a common proteome/phosphoproteome signature between Mat1a -/- mice and human NAFLD M-subtype that may have pathophysiological and therapeutic implications.
Collapse
Affiliation(s)
- Aaron E. Robinson
- Advanced Clinical Biosystems Research Institute, The Smidt Heart Institute, Cedars Sinai Medical Center, Advanced Health Sciences Pavilion, 127 S. San Vicente Blvd, Room 9302, Los Angeles, CA 90048, USA
| | - Aleksandra Binek
- Advanced Clinical Biosystems Research Institute, The Smidt Heart Institute, Cedars Sinai Medical Center, Advanced Health Sciences Pavilion, 127 S. San Vicente Blvd, Room 9302, Los Angeles, CA 90048, USA
| | - Komal Ramani
- Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Davis Building, Room 2097, Los Angeles, CA 90048, USA
| | - Niveda Sundararaman
- Advanced Clinical Biosystems Research Institute, The Smidt Heart Institute, Cedars Sinai Medical Center, Advanced Health Sciences Pavilion, 127 S. San Vicente Blvd, Room 9302, Los Angeles, CA 90048, USA
| | - Lucía Barbier-Torres
- Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Davis Building, Room 2097, Los Angeles, CA 90048, USA
| | - Ben Murray
- Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Davis Building, Room 2097, Los Angeles, CA 90048, USA
| | - Vidya Venkatraman
- Advanced Clinical Biosystems Research Institute, The Smidt Heart Institute, Cedars Sinai Medical Center, Advanced Health Sciences Pavilion, 127 S. San Vicente Blvd, Room 9302, Los Angeles, CA 90048, USA
| | - Simion Kreimer
- Advanced Clinical Biosystems Research Institute, The Smidt Heart Institute, Cedars Sinai Medical Center, Advanced Health Sciences Pavilion, 127 S. San Vicente Blvd, Room 9302, Los Angeles, CA 90048, USA
| | - Angela Mc Ardle
- Advanced Clinical Biosystems Research Institute, The Smidt Heart Institute, Cedars Sinai Medical Center, Advanced Health Sciences Pavilion, 127 S. San Vicente Blvd, Room 9302, Los Angeles, CA 90048, USA
| | - Mazen Noureddin
- Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Davis Building, Room 2097, Los Angeles, CA 90048, USA
| | - David Fernández-Ramos
- CIC bioGUNE, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (Ciberehd), Technology Park of Bizkaia, 48160 Derio, Bizkaia, Spain
| | - Fernando Lopitz-Otsoa
- CIC bioGUNE, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (Ciberehd), Technology Park of Bizkaia, 48160 Derio, Bizkaia, Spain
| | - Virginia Gutiérrez de Juan
- CIC bioGUNE, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (Ciberehd), Technology Park of Bizkaia, 48160 Derio, Bizkaia, Spain
| | - Oscar Millet
- CIC bioGUNE, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (Ciberehd), Technology Park of Bizkaia, 48160 Derio, Bizkaia, Spain
| | - José M. Mato
- CIC bioGUNE, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (Ciberehd), Technology Park of Bizkaia, 48160 Derio, Bizkaia, Spain
| | - Shelly C. Lu
- Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Davis Building, Room 2097, Los Angeles, CA 90048, USA
- Corresponding author
| | - Jennifer E. Van Eyk
- Advanced Clinical Biosystems Research Institute, The Smidt Heart Institute, Cedars Sinai Medical Center, Advanced Health Sciences Pavilion, 127 S. San Vicente Blvd, Room 9302, Los Angeles, CA 90048, USA
- Corresponding author
| |
Collapse
|
28
|
Alarcón-Vila C, Insausti-Urkia N, Torres S, Segalés-Rovira P, Conde de la Rosa L, Nuñez S, Fucho R, Fernández-Checa JC, García-Ruiz C. Dietary and genetic disruption of hepatic methionine metabolism induce acid sphingomyelinase to promote steatohepatitis. Redox Biol 2023; 59:102596. [PMID: 36610223 PMCID: PMC9827379 DOI: 10.1016/j.redox.2022.102596] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 12/28/2022] [Indexed: 01/03/2023] Open
Abstract
Alcoholic (ASH) and nonalcoholic. (NASH).steatohepatitis are advanced.stages.of.fatty.liver.disease.Methionine adenosyltransferase 1A (MAT1A) plays a key role in hepatic methionine metabolism and germline Mat1a deletion in mice promotes NASH. Acid sphingomyelinase (ASMase) triggers hepatocellular apoptosis and liver fibrosis and has been shown to downregulate MAT1A expression in the context of fulminant liver failure. Given the role of ASMase in steatohepatitis development, we investigated the status of ASMase in Mat1a-/- mice and the regulation of ASMase by SAM/SAH. Consistent with its role in NASH, Mat1a-/- mice fed a choline-deficient (CD) diet exhibited macrosteatosis, inflammation, fibrosis and liver injury as well as reduced total and mitochondrial GSH levels. Our data uncovered an increased basal expression and activity of ASMase but not neutral SMase in Mat1a-/- mice, which further increased upon CD feeding. Interestingly, adenovirus-mediated shRNA expression targeting ASMase reduced ASMase activity and protected Mat1a-/- mice against CD diet-induced NASH. Similar results were observed in CD fed Mat1a-/- mice by pharmacological inhibition of ASMase with amitriptyline. Moreover, Mat1a/ASMase double knockout mice were resistant to CD-induced NASH. ASMase knockdown protected wild type mice against NASH induced by feeding a diet deficient in methionine and choline. Furthermore, Mat1a-/- mice developed acute-on-chronic ASH and this outcome was ameliorated by amitriptyline treatment. In vitro data in primary mouse hepatocytes revealed that decreased SAM/SAH ratio increased ASMase mRNA level and activity. MAT1A and ASMase mRNA levels exhibited an inverse correlation in liver samples from patients with ASH and NASH. Thus, disruption of methionine metabolism sensitizes to steatohepatitis by ASMase activation via decreased SAM/SAH. These findings imply that MAT1A deletion and ASMase activation engage in a self-sustained loop of relevance for steatohepatitis.
Collapse
Affiliation(s)
- Cristina Alarcón-Vila
- Cell Death and Proliferation, Instituto de Investigaciones Biomédicas de Barcelona, CSIC, Barcelona, Spain; Liver Unit, Hospital Clínic I Provincial, IDIBAPS, Barcelona, Spain; CIBERehd, University of Barcelona, Spain
| | - Naroa Insausti-Urkia
- Cell Death and Proliferation, Instituto de Investigaciones Biomédicas de Barcelona, CSIC, Barcelona, Spain; Liver Unit, Hospital Clínic I Provincial, IDIBAPS, Barcelona, Spain; CIBERehd, University of Barcelona, Spain
| | - Sandra Torres
- Cell Death and Proliferation, Instituto de Investigaciones Biomédicas de Barcelona, CSIC, Barcelona, Spain; Liver Unit, Hospital Clínic I Provincial, IDIBAPS, Barcelona, Spain; CIBERehd, University of Barcelona, Spain
| | - Paula Segalés-Rovira
- Cell Death and Proliferation, Instituto de Investigaciones Biomédicas de Barcelona, CSIC, Barcelona, Spain; Liver Unit, Hospital Clínic I Provincial, IDIBAPS, Barcelona, Spain; CIBERehd, University of Barcelona, Spain
| | - Laura Conde de la Rosa
- Cell Death and Proliferation, Instituto de Investigaciones Biomédicas de Barcelona, CSIC, Barcelona, Spain; Liver Unit, Hospital Clínic I Provincial, IDIBAPS, Barcelona, Spain; CIBERehd, University of Barcelona, Spain
| | - Susana Nuñez
- Cell Death and Proliferation, Instituto de Investigaciones Biomédicas de Barcelona, CSIC, Barcelona, Spain; Liver Unit, Hospital Clínic I Provincial, IDIBAPS, Barcelona, Spain; CIBERehd, University of Barcelona, Spain
| | - Raquel Fucho
- Cell Death and Proliferation, Instituto de Investigaciones Biomédicas de Barcelona, CSIC, Barcelona, Spain; Liver Unit, Hospital Clínic I Provincial, IDIBAPS, Barcelona, Spain; CIBERehd, University of Barcelona, Spain
| | - Jose C Fernández-Checa
- Cell Death and Proliferation, Instituto de Investigaciones Biomédicas de Barcelona, CSIC, Barcelona, Spain; Liver Unit, Hospital Clínic I Provincial, IDIBAPS, Barcelona, Spain; CIBERehd, University of Barcelona, Spain; University of Southern California Research Center for Liver Diseases, Keck School of Medicine, USC, Los Angeles, CA, USA.
| | - Carmen García-Ruiz
- Cell Death and Proliferation, Instituto de Investigaciones Biomédicas de Barcelona, CSIC, Barcelona, Spain; Liver Unit, Hospital Clínic I Provincial, IDIBAPS, Barcelona, Spain; CIBERehd, University of Barcelona, Spain; University of Southern California Research Center for Liver Diseases, Keck School of Medicine, USC, Los Angeles, CA, USA.
| |
Collapse
|
29
|
de Moraes Meine B, Franceschi TS, Bona NP, Spohr L, Pedra NS, Spanevello RM, de Aguiar MSS, Stefanello FM. Chemical hypermethioninemia in young mice: oxidative damage and reduction of antioxidant enzyme activity in brain, kidney, and liver. Metab Brain Dis 2023; 38:223-232. [PMID: 36308587 DOI: 10.1007/s11011-022-01107-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 10/13/2022] [Indexed: 02/03/2023]
Abstract
High levels of methionine (Met) and its metabolites, such as methionine sulfoxide (MetO), found in hypermethioninemia, can be detrimental to the body; however, the underlying mechanisms are still uncertain. Using a recently standardized protocol, the aim of this study was to investigate the effects of chronic administration of Met and/or MetO on parameters of oxidative damage in the total brain, liver, and kidney of young mice. Swiss male mice were subcutaneously injected with Met and MetO at concentrations of 0.35-1.2 g/kg body weight and 0.09-0.3 g/kg body weight, respectively, from the 10th-38th day post-birth, while the control group was treated with saline solution. Results showed that Met and/or MetO caused an increase in reactive oxygen species (ROS) and lipoperoxidation, along with a reduction of superoxide dismutase (SOD) and catalase (CAT) activities in the brain. In the liver, Met and/or MetO enhanced ROS and nitrite levels, and reduced SOD, CAT, and delta aminolevulinic dehydratase activities. The effects on the kidney were an increase in ROS production and SOD activity, and a reduction in thiol content and CAT activity. These data demonstrated the contribution of redox imbalance to the systemic changes found in patients with hypermethioninemia. In conclusion, our findings may help future studies to better understand the pathophysiological mechanisms of hypermethioninemia as well as contribute to the search for new therapeutic agents for this pathology.
Collapse
Affiliation(s)
- Bernardo de Moraes Meine
- Programa de Pós-Graduação em Bioquímica e Bioprospecção, Laboratório de Biomarcadores, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas, Campus Universitário S/N, Pelotas, RS, Brazil
| | - Thaís Scolari Franceschi
- Programa de Pós-Graduação em Bioquímica e Bioprospecção, Laboratório de Biomarcadores, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas, Campus Universitário S/N, Pelotas, RS, Brazil
| | - Natália Pontes Bona
- Programa de Pós-Graduação em Bioquímica e Bioprospecção, Laboratório de Biomarcadores, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas, Campus Universitário S/N, Pelotas, RS, Brazil
| | - Luiza Spohr
- Programa de Pós-Graduação em Bioquímica e Bioprospecção, Laboratório de Neuroquímica, Inflamação e Câncer, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas, Campus Universitário s/n, Pelotas, RS, CEP: 96010-900, Brazil
| | - Nathalia Stark Pedra
- Programa de Pós-Graduação em Bioquímica e Bioprospecção, Laboratório de Neuroquímica, Inflamação e Câncer, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas, Campus Universitário s/n, Pelotas, RS, CEP: 96010-900, Brazil
| | - Roselia Maria Spanevello
- Programa de Pós-Graduação em Bioquímica e Bioprospecção, Laboratório de Neuroquímica, Inflamação e Câncer, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas, Campus Universitário s/n, Pelotas, RS, CEP: 96010-900, Brazil
| | - Mayara Sandrielly Soares de Aguiar
- Programa de Pós-Graduação em Bioquímica e Bioprospecção, Laboratório de Neuroquímica, Inflamação e Câncer, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas, Campus Universitário s/n, Pelotas, RS, CEP: 96010-900, Brazil.
| | - Francieli Moro Stefanello
- Programa de Pós-Graduação em Bioquímica e Bioprospecção, Laboratório de Biomarcadores, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas, Campus Universitário S/N, Pelotas, RS, Brazil
| |
Collapse
|
30
|
Flessa CM, Nasiri-Ansari N, Kyrou I, Leca BM, Lianou M, Chatzigeorgiou A, Kaltsas G, Kassi E, Randeva HS. Genetic and Diet-Induced Animal Models for Non-Alcoholic Fatty Liver Disease (NAFLD) Research. Int J Mol Sci 2022; 23:ijms232415791. [PMID: 36555433 PMCID: PMC9780957 DOI: 10.3390/ijms232415791] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 12/05/2022] [Accepted: 12/10/2022] [Indexed: 12/15/2022] Open
Abstract
A rapidly increasing incidence of non-alcoholic fatty liver disease (NAFLD) is noted worldwide due to the adoption of western-type lifestyles and eating habits. This makes the understanding of the molecular mechanisms that drive the pathogenesis of this chronic disease and the development of newly approved treatments of utmost necessity. Animal models are indispensable tools for achieving these ends. Although the ideal mouse model for human NAFLD does not exist yet, several models have arisen with the combination of dietary interventions, genetic manipulations and/or administration of chemical substances. Herein, we present the most common mouse models used in the research of NAFLD, either for the whole disease spectrum or for a particular disease stage (e.g., non-alcoholic steatohepatitis). We also discuss the advantages and disadvantages of each model, along with the challenges facing the researchers who aim to develop and use animal models for translational research in NAFLD. Based on these characteristics and the specific study aims/needs, researchers should select the most appropriate model with caution when translating results from animal to human.
Collapse
Affiliation(s)
- Christina-Maria Flessa
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism (WISDEM), University Hospitals Coventry and Warwickshire NHS Trust, Coventry CV2 2DX, UK
| | - Narjes Nasiri-Ansari
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Ioannis Kyrou
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism (WISDEM), University Hospitals Coventry and Warwickshire NHS Trust, Coventry CV2 2DX, UK
- Warwick Medical School, University of Warwick, Coventry CV4 7AL, UK
- Research Institute for Health and Wellbeing, Coventry University, Coventry CV1 5FB, UK
- Aston Medical School, College of Health and Life Sciences, Aston University, Birmingham B4 7ET, UK
- Laboratory of Dietetics and Quality of Life, Department of Food Science and Human Nutrition, School of Food and Nutritional Sciences, Agricultural University of Athens, 11855 Athens, Greece
| | - Bianca M. Leca
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism (WISDEM), University Hospitals Coventry and Warwickshire NHS Trust, Coventry CV2 2DX, UK
| | - Maria Lianou
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Antonios Chatzigeorgiou
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Gregory Kaltsas
- Endocrine Unit, 1st Department of Propaedeutic Internal Medicine, Laiko Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Eva Kassi
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
- Endocrine Unit, 1st Department of Propaedeutic Internal Medicine, Laiko Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece
- Correspondence: (E.K.); (H.S.R.)
| | - Harpal S. Randeva
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism (WISDEM), University Hospitals Coventry and Warwickshire NHS Trust, Coventry CV2 2DX, UK
- Warwick Medical School, University of Warwick, Coventry CV4 7AL, UK
- Correspondence: (E.K.); (H.S.R.)
| |
Collapse
|
31
|
Wang H, Qi S, Mu X, Yuan L, Li Y, Qiu J. Bisphenol F induces liver-gut alteration in zebrafish. THE SCIENCE OF THE TOTAL ENVIRONMENT 2022; 851:157974. [PMID: 35963407 DOI: 10.1016/j.scitotenv.2022.157974] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 07/23/2022] [Accepted: 08/07/2022] [Indexed: 06/15/2023]
Abstract
The unease of consumers with bisphenol A has led to the increased industrial usage of bisphenol F (BPF), which is a new hazard to environmental health. Here, zebrafish were exposed to three BPF concentrations (0.5, 5, and 50 μg/L) from the embryonic stage for 180 days. Results showed that zebrafish body length and weight decreased and hepatosomatic index values increased, even at environmentally relevant concentration. Histological analysis identified the occurrence of hepatic fibrosis and steatosis in 5 and 50 μg/L groups, which indicated the liver injury caused by BPF. Based on the untargeted metabolomics results, a dose-dependent variation in the effects of BPF on liver metabolism was found, and amino acids, purines and one carbon metabolism were the main affected processes in the 0.5, 5, and 50 μg/L treatments, respectively. At the same time, BPF induced a shift in intestinal microbiome composition, including decreased abundance of Erysipelotrichaceae, Rhodobacteraceae and Gemmobacter. In addition, the correlation analysis suggested an association between gut microbiome changes and affected hepatic metabolites after BPF exposure. These findings indicate that a liver-gut alteration is induced by long-term BPF exposure.
Collapse
Affiliation(s)
- Hui Wang
- Institute of Quality Standard and Testing Technology for Agro-Products, Chinese Academy of Agricultural Sciences, Beijing, China; Fishery Resource and Environment Research Center, Chinese Academy of Fishery Sciences, Beijing, China
| | - Suzhen Qi
- Institute of Apicultural Research, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Xiyan Mu
- Institute of Quality Standard and Testing Technology for Agro-Products, Chinese Academy of Agricultural Sciences, Beijing, China; Fishery Resource and Environment Research Center, Chinese Academy of Fishery Sciences, Beijing, China.
| | - Lilai Yuan
- Fishery Resource and Environment Research Center, Chinese Academy of Fishery Sciences, Beijing, China
| | - Yingren Li
- Fishery Resource and Environment Research Center, Chinese Academy of Fishery Sciences, Beijing, China
| | - Jing Qiu
- Institute of Quality Standard and Testing Technology for Agro-Products, Chinese Academy of Agricultural Sciences, Beijing, China.
| |
Collapse
|
32
|
Fernández-Ramos D, Lopitz-Otsoa F, Millet O, Alonso C, Lu SC, Mato JM. One Carbon Metabolism and S-Adenosylmethionine in Non-Alcoholic Fatty Liver Disease Pathogenesis and Subtypes. LIVERS 2022; 2:243-257. [PMID: 37123053 PMCID: PMC10137169 DOI: 10.3390/livers2040020] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
One carbon metabolism (1CM) can be defined as the transfer of a carbon unit from one metabolite to another and its replenishment by different sources of labile methyl-group nutrients: primarily choline, methionine, betaine, and serine. This flow of carbon units allows the biosynthesis of nucleotides, amino acids, formylated methionyl-tRNA, polyamines, glutathione, phospholipids, detoxification reactions, maintenance of the redox status and the concentration of NAD, and methylation reactions including epigenetic modifications. That is, 1CM functions as a nutrient sensor and integrator of cellular metabolism. A critical process in 1CM is the synthesis of S-adenosylmethionine (SAMe), the source of essentially all the hundreds of millions of daily methyl transfer reactions in a cell. This versatility of SAMe imposes a tight control in its synthesis and catabolism. Much of our knowledge concerning 1CM has been gained from studies in the production and prevention of nonalcoholic fatty liver disease (NAFLD). Here, we discuss in detail the function of the most important enzymes for their quantitative contribution to maintaining the flux of carbon units through 1CM in the liver and discuss how alterations in their enzymatic activity contribute to the development of NAFLD. Next, we discuss NAFLD subtypes based on serum lipidomic profiles with different risk of cardiovascular disease. Among the latter, we highlight the so-called subtype A for its serum lipidomic profile phenocopying that of mice deficient in SAMe synthesis and because its high frequency (about 50% of the NAFLD patients).
Collapse
Affiliation(s)
- David Fernández-Ramos
- Precision Medicine and Metabolism Laboratory, CIC bioGUNE, BRTA, CIBERehd, Technology Park of Bizkaia, 48160 Derio, Bizkaia, Spain
| | - Fernando Lopitz-Otsoa
- Precision Medicine and Metabolism Laboratory, CIC bioGUNE, BRTA, CIBERehd, Technology Park of Bizkaia, 48160 Derio, Bizkaia, Spain
| | - Oscar Millet
- Precision Medicine and Metabolism Laboratory, CIC bioGUNE, BRTA, CIBERehd, Technology Park of Bizkaia, 48160 Derio, Bizkaia, Spain
| | - Cristina Alonso
- OWL Metabolomics, Technology Park of Bizkaia, 48160 Derio, Bizkaia, Spain
| | - Shelly C. Lu
- Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - José M. Mato
- Precision Medicine and Metabolism Laboratory, CIC bioGUNE, BRTA, CIBERehd, Technology Park of Bizkaia, 48160 Derio, Bizkaia, Spain
- Correspondence: ; Tel.: +34-944-061300; Fax: +34-944-0611301
| |
Collapse
|
33
|
Martínez‐Arranz I, Bruzzone C, Noureddin M, Gil‐Redondo R, Mincholé I, Bizkarguenaga M, Arretxe E, Iruarrizaga‐Lejarreta M, Fernández‐Ramos D, Lopitz‐Otsoa F, Mayo R, Embade N, Newberry E, Mittendorf B, Izquierdo‐Sánchez L, Smid V, Arnold J, Iruzubieta P, Pérez Castaño Y, Krawczyk M, Marigorta UM, Morrison MC, Kleemann R, Martín‐Duce A, Hayardeny L, Vitek L, Bruha R, Aller de la Fuente R, Crespo J, Romero‐Gomez M, Banales JM, Arrese M, Cusi K, Bugianesi E, Klein S, Lu SC, Anstee QM, Millet O, Davidson NO, Alonso C, Mato JM. Metabolic subtypes of patients with NAFLD exhibit distinctive cardiovascular risk profiles. Hepatology 2022; 76:1121-1134. [PMID: 35220605 PMCID: PMC9790568 DOI: 10.1002/hep.32427] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 01/18/2022] [Accepted: 02/15/2022] [Indexed: 12/31/2022]
Abstract
BACKGROUND AND AIMS We previously identified subsets of patients with NAFLD with different metabolic phenotypes. Here we align metabolomic signatures with cardiovascular disease (CVD) and genetic risk factors. APPROACH AND RESULTS We analyzed serum metabolome from 1154 individuals with biopsy-proven NAFLD, and from four mouse models of NAFLD with impaired VLDL-triglyceride (TG) secretion, and one with normal VLDL-TG secretion. We identified three metabolic subtypes: A (47%), B (27%), and C (26%). Subtype A phenocopied the metabolome of mice with impaired VLDL-TG secretion; subtype C phenocopied the metabolome of mice with normal VLDL-TG; and subtype B showed an intermediate signature. The percent of patients with NASH and fibrosis was comparable among subtypes, although subtypes B and C exhibited higher liver enzymes. Serum VLDL-TG levels and secretion rate were lower among subtype A compared with subtypes B and C. Subtype A VLDL-TG and VLDL-apolipoprotein B concentrations were independent of steatosis, whereas subtypes B and C showed an association with these parameters. Serum TG, cholesterol, VLDL, small dense LDL5,6 , and remnant lipoprotein cholesterol were lower among subtype A compared with subtypes B and C. The 10-year high risk of CVD, measured with the Framingham risk score, and the frequency of patatin-like phospholipase domain-containing protein 3 NAFLD risk allele were lower in subtype A. CONCLUSIONS Metabolomic signatures identify three NAFLD subgroups, independent of histological disease severity. These signatures align with known CVD and genetic risk factors, with subtype A exhibiting a lower CVD risk profile. This may account for the variation in hepatic versus cardiovascular outcomes, offering clinically relevant risk stratification.
Collapse
Affiliation(s)
| | | | - Mazen Noureddin
- Karsh Division of Gastroenterology and HepatologyCedars‐Sinai Medical CenterLos AngelesCaliforniaUSA
| | | | | | | | | | | | | | | | | | | | - Elizabeth Newberry
- Department of MedicineWashington University School of MedicineSt. LouisMissouriUSA
| | - Bettina Mittendorf
- Center for Human NutritionWashington University School of MedicineSt. LouisMissouriUSA
| | - Laura Izquierdo‐Sánchez
- Department of Liver and Gastrointestinal DiseasesBiodonostia Research InstituteDonostia University HospitalDonostiaSpain
| | - Vaclav Smid
- First Faculty of MedicineCharles UniversityPragueCzech Republic
| | - Jorge Arnold
- Departamento de GastroenterologiaEscuela de MedicinaPontificia Universidad Católica de ChileSantiago de ChileChile
| | - Paula Iruzubieta
- Marqués de Valdecilla University HospitalCantabria UniversitySantanderSpain
| | - Ylenia Pérez Castaño
- Department of Digestive SystemOsakidetza Basque Health ServiceDonostia University HospitalSan SebastianSpain
| | - Marcin Krawczyk
- Department of Medicine IISaarland University Medical CenterHomburgGermany,Laboratory of Metabolic Liver DiseasesCenter for Preclinical ResearchDepartment of General, Transplant and Liver SurgeryMedical University of WarsawWarsawPoland
| | | | - Martine C. Morrison
- Department of Metabolic Health ResearchNetherlands Organization for Applied Scientific ResearchLeidenThe Netherlands
| | - Robert Kleemann
- Department of Metabolic Health ResearchNetherlands Organization for Applied Scientific ResearchLeidenThe Netherlands
| | - Antonio Martín‐Duce
- Alcalá University School of Medicine and Health SciencesUniversity Hospital Prıncipe de AsturiasMadridSpain
| | | | - Libor Vitek
- First Faculty of MedicineCharles UniversityPragueCzech Republic
| | - Radan Bruha
- First Faculty of MedicineCharles UniversityPragueCzech Republic
| | - Rocío Aller de la Fuente
- Department of Digestive DiseaseClinic University HospitalUniversity Hospital of ValladolidValladolidSpain
| | - Javier Crespo
- Marqués de Valdecilla University HospitalCantabria UniversitySantanderSpain
| | | | - Jesus M Banales
- Department of Liver and Gastrointestinal DiseasesBiodonostia Research InstituteDonostia University HospitalDonostiaSpain,University of the Basque CountryCIBERehdIKERBASQUEDonostiaSpain
| | - Marco Arrese
- Departamento de GastroenterologiaEscuela de MedicinaPontificia Universidad Católica de ChileSantiago de ChileChile,Centro de Envejecimiento y RegeneraciónSantiagoChile
| | - Kenneth Cusi
- Division of Endocrinology, Diabetes and MetabolismUniversity of Florida and Malcom Randall VAMCGainesvilleFloridaUSA
| | | | - Samuel Klein
- Center for Human NutritionWashington University School of MedicineSt. LouisMissouriUSA
| | - Shelly C. Lu
- Karsh Division of Gastroenterology and HepatologyCedars‐Sinai Medical CenterLos AngelesCaliforniaUSA
| | - Quentin M. Anstee
- Translational & Clinical Research InstituteFaculty of Medical SciencesNewcastle UniversityNewcastle Upon TyneUK,Newcastle NIHR Biomedical Research CenterNewcastle Upon Tyne Hospitals NHS TrustNewcastle Upon TyneUK
| | | | - Nicholas O. Davidson
- Department of MedicineWashington University School of MedicineSt. LouisMissouriUSA
| | | | | |
Collapse
|
34
|
Matye D, Gunewardena S, Chen J, Wang H, Wang Y, Hasan MN, Gu L, Clayton YD, Du Y, Chen C, Friedman JE, Lu SC, Ding WX, Li T. TFEB regulates sulfur amino acid and coenzyme A metabolism to support hepatic metabolic adaptation and redox homeostasis. Nat Commun 2022; 13:5696. [PMID: 36171419 PMCID: PMC9519740 DOI: 10.1038/s41467-022-33465-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 09/15/2022] [Indexed: 11/15/2022] Open
Abstract
Fatty liver is a highly heterogenous condition driven by various pathogenic factors in addition to the severity of steatosis. Protein insufficiency has been causally linked to fatty liver with incompletely defined mechanisms. Here we report that fatty liver is a sulfur amino acid insufficient state that promotes metabolic inflexibility via limiting coenzyme A availability. We demonstrate that the nutrient-sensing transcriptional factor EB synergistically stimulates lysosome proteolysis and methionine adenosyltransferase to increase cysteine pool that drives the production of coenzyme A and glutathione, which support metabolic adaptation and antioxidant defense during increased lipid influx. Intriguingly, mice consuming an isocaloric protein-deficient Western diet exhibit selective hepatic cysteine, coenzyme A and glutathione deficiency and acylcarnitine accumulation, which are reversed by cystine supplementation without normalizing dietary protein intake. These findings support a pathogenic link of dysregulated sulfur amino acid metabolism to metabolic inflexibility that underlies both overnutrition and protein malnutrition-associated fatty liver development.
Collapse
Affiliation(s)
- David Matye
- Harold Hamm Diabetes Center, Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Sumedha Gunewardena
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Jianglei Chen
- Harold Hamm Diabetes Center, Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Huaiwen Wang
- Laboratory for Molecular Biology and Cytometry Research, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Yifeng Wang
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Mohammad Nazmul Hasan
- Harold Hamm Diabetes Center, Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Lijie Gu
- Harold Hamm Diabetes Center, Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Yung Dai Clayton
- Harold Hamm Diabetes Center, Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Yanhong Du
- Harold Hamm Diabetes Center, Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Cheng Chen
- Harold Hamm Diabetes Center, Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Jacob E Friedman
- Harold Hamm Diabetes Center, Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Shelly C Lu
- Karsh Division of Gastroenterology and Hepatology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Wen-Xing Ding
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Tiangang Li
- Harold Hamm Diabetes Center, Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA.
| |
Collapse
|
35
|
Guerrero L, Paradela A, Corrales FJ. Targeted Proteomics for Monitoring One-Carbon Metabolism in Liver Diseases. Metabolites 2022; 12:metabo12090779. [PMID: 36144184 PMCID: PMC9501948 DOI: 10.3390/metabo12090779] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [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
- Correspondence: ; Tel.: +34-91-585-46-96
| |
Collapse
|
36
|
Wang H, Wu Y, Tang W. Methionine cycle in nonalcoholic fatty liver disease and its potential applications. Biochem Pharmacol 2022; 200:115033. [PMID: 35395242 DOI: 10.1016/j.bcp.2022.115033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 03/31/2022] [Accepted: 03/31/2022] [Indexed: 11/25/2022]
Abstract
As a chronic metabolic disease affecting epidemic proportions worldwide, the pathogenesis of Nonalcoholic Fatty Liver Disease (NAFLD) is not clear yet. There is also a lack of precise biomarkers and specific medicine for the diagnosis and treatment of NAFLD. Methionine metabolic cycle, which is critical for the maintaining of cellular methylation and redox state, is involved in the pathophysiology of NAFLD. However, the molecular basis and mechanism of methionine metabolism in NAFLD are not completely understood. Here, we mainly focus on specific enzymes that participates in methionine cycle, to reveal their interconnections with NAFLD, in order to recognize the pathogenesis of NAFLD from a new angle and at the same time, explore the clinical characteristics and therapeutic strategies.
Collapse
Affiliation(s)
- Haoyu Wang
- University of Chinese Academy of Sciences, Beijing, 100049, PR China; Laboratory of Anti-inflammation, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, PR China
| | - Yanwei Wu
- Laboratory of Anti-inflammation, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, PR China
| | - Wei Tang
- University of Chinese Academy of Sciences, Beijing, 100049, PR China; Laboratory of Anti-inflammation, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, PR China.
| |
Collapse
|
37
|
Methionine adenosyltransferase 1a antisense oligonucleotides activate the liver-brown adipose tissue axis preventing obesity and associated hepatosteatosis. Nat Commun 2022; 13:1096. [PMID: 35232994 PMCID: PMC8888704 DOI: 10.1038/s41467-022-28749-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 02/03/2022] [Indexed: 02/06/2023] Open
Abstract
Altered methionine metabolism is associated with weight gain in obesity. The methionine adenosyltransferase (MAT), catalyzing the first reaction of the methionine cycle, plays an important role regulating lipid metabolism. However, its role in obesity, when a plethora of metabolic diseases occurs, is still unknown. By using antisense oligonucleotides (ASO) and genetic depletion of Mat1a, here, we demonstrate that Mat1a deficiency in diet-induce obese or genetically obese mice prevented and reversed obesity and obesity-associated insulin resistance and hepatosteatosis by increasing energy expenditure in a hepatocyte FGF21 dependent fashion. The increased NRF2-mediated FGF21 secretion induced by targeting Mat1a, mobilized plasma lipids towards the BAT to be catabolized, induced thermogenesis and reduced body weight, inhibiting hepatic de novo lipogenesis. The beneficial effects of Mat1a ASO were abolished following FGF21 depletion in hepatocytes. Thus, targeting Mat1a activates the liver-BAT axis by increasing NRF2-mediated FGF21 secretion, which prevents obesity, insulin resistance and hepatosteatosis.
Collapse
|
38
|
Xiao Y, Liu F, Kong Q, Zhu X, Wang H, Li S, Jiang N, Yu C, Yun L. Metformin induces S-adenosylmethionine restriction to extend the Caenorhabditis elegans healthspan through H3K4me3 modifiers. Aging Cell 2022; 21:e13567. [PMID: 35146893 PMCID: PMC8920454 DOI: 10.1111/acel.13567] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Revised: 01/12/2022] [Accepted: 01/26/2022] [Indexed: 01/14/2023] Open
Abstract
Metformin, a widely prescribed first‐line drug for the treatment of type II diabetes mellitus, has been shown to extend lifespan and delay the onset of age‐related diseases. The precisely mechanisms by which these effects are realized remain elusive. We find that metformin exposure is restricted to adults, which is sufficient to extend lifespan. However, limiting metformin exposure to the larvae has no significant effect on Caenorhabditis elegans longevity. Here, we show that after metformin treatment, the level of S‐adenosylmethionine (SAM) is reduced in adults but not in the larvae. Potential mechanisms by which reduced SAM might increase lifespan include altering the histone methylation. However, the molecular connections between metformin, SAM limitation, methyltransferases, and healthspan‐associated phenotypes are unclear. Through genetic screening of C. elegans, we find that metformin promotes the healthspan through an H3K4 methyltransferase/demethylase complex to downregulate the targets, including mTOR and S6 kinase. Thus, our studies provide molecular links between meformin, SAM limitation, histone methylation, and healthspan and elucidate the mode action of metformin‐regulated healthspan extension will boost its therapeutic application in the treatment of human aging and age‐related diseases.
Collapse
Affiliation(s)
- Yi Xiao
- Institute of life sciences Zunyi Medical University Zunyi China
- Guizhou Provincial College‐based Key Lab for Tumor Prevention and Treatment with Distinctive Medicines Zunyi Medical University Zunyi China
- College of Basic Medicine Zunyi Medical University Zunyi China
| | - Fang Liu
- College of Basic Medicine Zunyi Medical University Zunyi China
| | - Qinghong Kong
- Institute of life sciences Zunyi Medical University Zunyi China
- Guizhou Provincial College‐based Key Lab for Tumor Prevention and Treatment with Distinctive Medicines Zunyi Medical University Zunyi China
| | - Xinting Zhu
- Guizhou Provincial College‐based Key Lab for Tumor Prevention and Treatment with Distinctive Medicines Zunyi Medical University Zunyi China
- College of Basic Medicine Zunyi Medical University Zunyi China
| | - Haijuan Wang
- Guizhou Provincial College‐based Key Lab for Tumor Prevention and Treatment with Distinctive Medicines Zunyi Medical University Zunyi China
- College of Basic Medicine Zunyi Medical University Zunyi China
| | - Sanhua Li
- Institute of life sciences Zunyi Medical University Zunyi China
- Guizhou Provincial College‐based Key Lab for Tumor Prevention and Treatment with Distinctive Medicines Zunyi Medical University Zunyi China
| | - Nian Jiang
- Institute of life sciences Zunyi Medical University Zunyi China
- Guizhou Provincial College‐based Key Lab for Tumor Prevention and Treatment with Distinctive Medicines Zunyi Medical University Zunyi China
| | - Changyan Yu
- Institute of life sciences Zunyi Medical University Zunyi China
- Guizhou Provincial College‐based Key Lab for Tumor Prevention and Treatment with Distinctive Medicines Zunyi Medical University Zunyi China
| | - Liu Yun
- Institute of life sciences Zunyi Medical University Zunyi China
- Guizhou Provincial College‐based Key Lab for Tumor Prevention and Treatment with Distinctive Medicines Zunyi Medical University Zunyi China
- College of Basic Medicine Zunyi Medical University Zunyi China
| |
Collapse
|
39
|
Dysregulation of S-adenosylmethionine Metabolism in Nonalcoholic Steatohepatitis Leads to Polyamine Flux and Oxidative Stress. Int J Mol Sci 2022; 23:ijms23041986. [PMID: 35216100 PMCID: PMC8878801 DOI: 10.3390/ijms23041986] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 01/28/2022] [Accepted: 02/08/2022] [Indexed: 02/01/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is the number one cause of chronic liver disease worldwide, with 25% of these patients developing nonalcoholic steatohepatitis (NASH). NASH significantly increases the risk of cirrhosis and decompensated liver failure. Past studies in rodent models have shown that glycine-N-methyltransferase (GNMT) knockout results in rapid steatosis, fibrosis, and hepatocellular carcinoma progression. However, the attenuation of GNMT in subjects with NASH and the molecular basis for its impact on the disease process is still unclear. To address this knowledge gap, we show the reduction of GNMT protein levels in the liver of NASH subjects compared to healthy controls. To gain insight into the impact of decreased GNMT in the disease process, we performed global label-free proteome studies on the livers from a murine modified amylin diet-based model of NASH. Histological and molecular characterization of the animal model demonstrate a high resemblance to human disease. We found that a reduction of GNMT leads to a significant increase in S-adenosylmethionine (AdoMet), an essential metabolite for transmethylation reactions and a substrate for polyamine synthesis. Further targeted proteomic and metabolomic studies demonstrated a decrease in GNMT transmethylation, increased flux through the polyamine pathway, and increased oxidative stress production contributing to NASH pathogenesis.
Collapse
|
40
|
Ornoy A, Weinstein-Fudim L, Becker M. SAMe, Choline, and Valproic Acid as Possible Epigenetic Drugs: Their Effects in Pregnancy with a Special Emphasis on Animal Studies. Pharmaceuticals (Basel) 2022; 15:192. [PMID: 35215304 PMCID: PMC8879727 DOI: 10.3390/ph15020192] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 01/27/2022] [Accepted: 01/28/2022] [Indexed: 11/29/2022] Open
Abstract
In this review, we discuss the functions and main effects on pregnancy outcomes of three agents that have the ability to induce epigenetic modifications: valproic acid (VPA), a well-known teratogen that is a histone deacetylase inhibitor; S-adenosylmethionine (SAMe), the most effective methyl donor; and choline, an important micronutrient involved in the one methyl group cycle and in the synthesis of SAMe. Our aim was to describe the possible effects of these compounds when administered during pregnancy on the developing embryo and fetus or, if administered postnatally, their effects on the developing child. These substances are able to modify gene expression and possibly alleviate neurobehavioral changes in disturbances that have epigenetic origins, such as autism spectrum disorder (ASD), depression, Rett syndrome, and fetal alcohol spectrum disorder (FASD). Valproic acid and SAMe are antagonistic epigenetic modulators whether administered in utero or postnatally. However, VPA is a major human teratogen and, whenever possible, should not be used by pregnant women. Most currently relevant data come from experimental animal studies that aimed to explore the possibility of using these substances as epigenetic modifiers and possible therapeutic agents. In experimental animals, each of these substances was able to alleviate the severity of several well-known diseases by inducing changes in the expression of affected genes or by other yet unknown mechanisms. We believe that additional studies are needed to further explore the possibility of using these substances, and similar compounds, for the treatment of "epigenetic human diseases".
Collapse
Affiliation(s)
- Asher Ornoy
- Adelson School of Medicine, Ariel University, Ariel 40700, Israel;
- Department of Medical Neurobiology, Hebrew University Hadassah Medical School, Jerusalem 9112102, Israel;
| | - Liza Weinstein-Fudim
- Department of Medical Neurobiology, Hebrew University Hadassah Medical School, Jerusalem 9112102, Israel;
| | - Maria Becker
- Adelson School of Medicine, Ariel University, Ariel 40700, Israel;
| |
Collapse
|
41
|
Ramani K, Robinson AE, Berlind J, Fan W, Abeynayake A, Binek A, Barbier-Torres L, Noureddin M, Nissen NN, Yildirim Z, Erbay E, Mato JM, Van Eyk JE, Lu SC. S-adenosylmethionine inhibits la ribonucleoprotein domain family member 1 in murine liver and human liver cancer cells. Hepatology 2022; 75:280-296. [PMID: 34449924 PMCID: PMC8766892 DOI: 10.1002/hep.32130] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 07/22/2021] [Accepted: 08/09/2021] [Indexed: 02/03/2023]
Abstract
BACKGROUND AND AIMS Methionine adenosyltransferase 1A (MAT1A) is responsible for S-adenosylmethionine (SAMe) biosynthesis in the liver. Mice lacking Mat1a have hepatic SAMe depletion and develop NASH and HCC spontaneously. Several kinases are activated in Mat1a knockout (KO) mice livers. However, characterizing the phospho-proteome and determining whether they contribute to liver pathology remain open for study. Our study aimed to provide this knowledge. APPROACH AND RESULTS We performed phospho-proteomics in Mat1a KO mice livers with and without SAMe treatment to identify SAMe-dependent changes that may contribute to liver pathology. Our studies used Mat1a KO mice at different ages treated with and without SAMe, cell lines, in vitro translation and kinase assays, and human liver specimens. We found that the most striking change was hyperphosphorylation and increased content of La-related protein 1 (LARP1), which, in the unphosphorylated form, negatively regulates translation of 5'-terminal oligopyrimidine (TOP)-containing mRNAs. Consistently, multiple TOP proteins are induced in KO livers. Translation of TOP mRNAs ribosomal protein S3 and ribosomal protein L18 was enhanced by LARP1 overexpression in liver cancer cells. We identified LARP1-T449 as a SAMe-sensitive phospho-site of cyclin-dependent kinase 2 (CDK2). Knocking down CDK2 lowered LARP1 phosphorylation and prevented LARP1-overexpression-mediated increase in translation. LARP1-T449 phosphorylation induced global translation, cell growth, migration, invasion, and expression of oncogenic TOP-ribosomal proteins in HCC cells. LARP1 expression is increased in human NASH and HCC. CONCLUSIONS Our results reveal a SAMe-sensitive mechanism of LARP1 phosphorylation that may be involved in the progression of NASH to HCC.
Collapse
Affiliation(s)
- Komal Ramani
- Karsh Division of Gastroenterology and Hepatology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048
| | - Aaron E. Robinson
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305
- Smidt Heart Institute and Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048
| | - Joshua Berlind
- Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, CA 90033
| | - Wei Fan
- Karsh Division of Gastroenterology and Hepatology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048
| | - Aushinie Abeynayake
- Karsh Division of Gastroenterology and Hepatology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048
| | - Aleksandra Binek
- Smidt Heart Institute and Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048
| | - Lucía Barbier-Torres
- Karsh Division of Gastroenterology and Hepatology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048
| | - Mazen Noureddin
- Karsh Division of Gastroenterology and Hepatology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048
- Comprehensive Transplant Center, Cedars-Sinai Medical Center, Los Angeles, CA 90048
| | - Nicholas N. Nissen
- Comprehensive Transplant Center, Cedars-Sinai Medical Center, Los Angeles, CA 90048
- Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, CA 90048
| | - Zehra Yildirim
- Department of Cardiology, Department of Biomedical Sciences and Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048
| | - Ebru Erbay
- Department of Cardiology, Department of Biomedical Sciences and Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048
| | - José M. Mato
- CIC bioGUNE, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (Ciberehd), Technology, Derio, Bizkaia 48160, Spain
| | - Jennifer E. Van Eyk
- Smidt Heart Institute and Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048
| | - Shelly C. Lu
- Karsh Division of Gastroenterology and Hepatology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048
| |
Collapse
|
42
|
Alshawsh MA, Alsalahi A, Alshehade SA, Saghir SAM, Ahmeda AF, Al Zarzour RH, Mahmoud AM. A Comparison of the Gene Expression Profiles of Non-Alcoholic Fatty Liver Disease between Animal Models of a High-Fat Diet and Methionine-Choline-Deficient Diet. Molecules 2022; 27:molecules27030858. [PMID: 35164140 PMCID: PMC8839835 DOI: 10.3390/molecules27030858] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 01/21/2022] [Accepted: 01/24/2022] [Indexed: 12/12/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) embraces several forms of liver disorders involving fat disposition in hepatocytes ranging from simple steatosis to the severe stage, namely, non-alcoholic steatohepatitis (NASH). Recently, several experimental in vivo animal models for NAFLD/NASH have been established. However, no reproducible experimental animal model displays the full spectrum of pathophysiological, histological, molecular, and clinical features associated with human NAFLD/NASH progression. Although methionine-choline-deficient (MCD) diet and high-fat diet (HFD) models can mimic histological and metabolic abnormalities of human disease, respectively, the molecular signaling pathways are extremely important for understanding the pathogenesis of the disease. This review aimed to assess the differences in gene expression patterns and NAFLD/NASH progression pathways among the most common dietary animal models, i.e., HFD- and MCD diet-fed animals. Studies showed that the HFD and MCD diet could induce either up- or downregulation of the expression of genes and proteins that are involved in lipid metabolism, inflammation, oxidative stress, and fibrogenesis pathways. Interestingly, the MCD diet model could spontaneously develop liver fibrosis within two to four weeks and has significant effects on the expression of genes that encode proteins and enzymes involved in the liver fibrogenesis pathway. However, such effects in the HFD model were found to occur after 24 weeks with insulin resistance but appear to cause less severe fibrosis. In conclusion, assessing the abnormal gene expression patterns caused by different diet types provides valuable information regarding the molecular mechanisms of NAFLD/NASH and predicts the clinical progression of the disease. However, expression profiling studies concerning genetic variants involved in the development and progression of NAFLD/NASH should be conducted.
Collapse
Affiliation(s)
- Mohammed Abdullah Alshawsh
- Department of Pharmacology, Faculty of Medicine, University of Malaya, Kuala Lumpur 50603, Malaysia;
- Correspondence:
| | - Abdulsamad Alsalahi
- Department of Pharmacology, Faculty of Medicine, University of Malaya, Kuala Lumpur 50603, Malaysia;
| | - Salah Abdalrazak Alshehade
- Discipline of Pharmacology, School of Pharmaceutical Sciences, Universiti Sains Malaysia (USM), Gelugor 11800, Malaysia; (S.A.A.); (R.H.A.Z.)
| | - Sultan Ayesh Mohammed Saghir
- Department of Medical Analysis, Princess Aisha Bint Al-Hussein College of Nursing and Medical Sciences, Al-Hussein Bin Talal University, Ma’an 71111, Jordan;
| | - Ahmad Faheem Ahmeda
- Department of Basic Medical Sciences, College of Medicine, Ajman University, Ajman P.O. Box 346, United Arab Emirates;
- Center of Medical and Bio-Allied Health Sciences Research, Ajman University, Ajman P.O. Box 346, United Arab Emirates
| | - Raghdaa Hamdan Al Zarzour
- Discipline of Pharmacology, School of Pharmaceutical Sciences, Universiti Sains Malaysia (USM), Gelugor 11800, Malaysia; (S.A.A.); (R.H.A.Z.)
| | - Ayman Moawad Mahmoud
- Physiology Division, Zoology Department, Faculty of Science, Beni-Suef University, Beni-Suef 62514, Egypt;
| |
Collapse
|
43
|
Pascale RM, Simile MM, Calvisi DF, Feo CF, Feo F. S-Adenosylmethionine: From the Discovery of Its Inhibition of Tumorigenesis to Its Use as a Therapeutic Agent. Cells 2022; 11:409. [PMID: 35159219 PMCID: PMC8834208 DOI: 10.3390/cells11030409] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 01/10/2022] [Accepted: 01/14/2022] [Indexed: 02/07/2023] Open
Abstract
Alterations of methionine cycle in steatohepatitis, cirrhosis, and hepatocellular carcinoma induce MAT1A decrease and MAT2A increase expressions with the consequent decrease of S-adenosyl-L-methionine (SAM). This causes non-alcoholic fatty liver disease (NAFLD). SAM administration antagonizes pathological conditions, including galactosamine, acetaminophen, and ethanol intoxications, characterized by decreased intracellular SAM. Positive therapeutic effects of SAM/vitamin E or SAM/ursodeoxycholic acid in animal models with NAFLD and intrahepatic cholestasis were not confirmed in humans. In in vitro experiments, SAM and betaine potentiate PegIFN-alpha-2a/2b plus ribavirin antiviral effects. SAM plus betaine improves early viral kinetics and increases interferon-stimulated gene expression in patients with viral hepatitis non-responders to pegIFNα/ribavirin. SAM prevents hepatic cirrhosis, induced by CCl4, inhibits experimental tumors growth and is proapoptotic for hepatocellular carcinoma and MCF-7 breast cancer cells. SAM plus Decitabine arrest cancer growth and potentiate doxorubicin effects on breast, head, and neck cancers. Furthermore, SAM enhances the antitumor effect of gemcitabine against pancreatic cancer cells, inhibits growth of human prostate cancer PC-3, colorectal cancer, and osteosarcoma LM-7 and MG-63 cell lines; increases genomic stability of SW480 cells. SAM reduces colorectal cancer progression and inhibits the proliferation of preneoplastic rat liver cells in vivo. The discrepancy between positive results of SAM treatment of experimental tumors and modest effects against human disease may depend on more advanced human disease stage at moment of diagnosis.
Collapse
Affiliation(s)
- Rosa M. Pascale
- Department of Medical, Surgical and Experimental Sciences, Division of Experimental Pathology and Oncology, University of Sassari, 07100 Sassari, Italy; (M.M.S.); (D.F.C.); (F.F.)
| | - Maria M. Simile
- Department of Medical, Surgical and Experimental Sciences, Division of Experimental Pathology and Oncology, University of Sassari, 07100 Sassari, Italy; (M.M.S.); (D.F.C.); (F.F.)
| | - Diego F. Calvisi
- Department of Medical, Surgical and Experimental Sciences, Division of Experimental Pathology and Oncology, University of Sassari, 07100 Sassari, Italy; (M.M.S.); (D.F.C.); (F.F.)
| | - Claudio F. Feo
- Department of Medical, Surgical and Experimental Sciences, Division of Surgery, University of Sassari, 07100 Sassari, Italy;
| | - Francesco Feo
- Department of Medical, Surgical and Experimental Sciences, Division of Experimental Pathology and Oncology, University of Sassari, 07100 Sassari, Italy; (M.M.S.); (D.F.C.); (F.F.)
| |
Collapse
|
44
|
Wang R, Fan H, Sun M, Lv Z, Yi W. Roles of BMI1 in the Initiation, Progression, and Treatment of Hepatocellular Carcinoma. Technol Cancer Res Treat 2022; 21:15330338211070689. [PMID: 35072573 PMCID: PMC8793120 DOI: 10.1177/15330338211070689] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Liver cancer has high rates of morbidity and mortality, and its treatment is a global health challenge. Hepatocellular carcinoma (HCC) accounts for 90% of all primary liver cancer cases. B-lymphoma Mo-MLV insertion region 1 (BMI1) has been identified as a proto-oncogene, which contributes to the initiation and progression of many malignant tumors. BMI1 expression is upregulated in HCC, and it influences the occurrence and development of HCC by various mechanisms, such as the INK4a/ARF locus, NF-κB signaling pathway, and PTEN/PI3K/AKT signaling pathway. In addition, the expression of BMI1 is related to prognosis and recurrence of HCC. Hence, there is clear evidence that BMI1 is a novel and valid therapeutic target for HCC. Accordingly, the development of therapeutic strategies targeting BMI1 has been a focus of recent research, providing new directions for HCC treatment. This review summarizes the role of BMI1 in the occurrence and treatment of HCC, which will provide a basis for using BMI1 as a potential target for the development of therapeutic strategies for HCC.
Collapse
Affiliation(s)
- Ru Wang
- 278245Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Hengwei Fan
- 535219The Eastern Hepatobiliary Surgery Hospital, Navy Medical University (Second Military Medical University), Shanghai, China
| | - Ming Sun
- 278245Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Zhongwei Lv
- 278245Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Wanwan Yi
- 278245Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
45
|
Downregulation of Methionine Cycle Genes MAT1A and GNMT Enriches Protein-Associated Translation Process and Worsens Hepatocellular Carcinoma Prognosis. Int J Mol Sci 2022; 23:ijms23010481. [PMID: 35008908 PMCID: PMC8745498 DOI: 10.3390/ijms23010481] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 12/27/2021] [Accepted: 12/28/2021] [Indexed: 01/27/2023] Open
Abstract
The major biological methyl donor, S-adenosylmethionine (adoMet) synthesis occurs mainly in the liver. Methionine adenosyltransferase 1A (MAT1A) and glycine N-methyltransferase (GNMT) are two key enzymes involved in the functional implications of that variation. We collected 42 RNA-seq data from paired hepatocellular carcinoma (HCC) and its adjacent normal liver tissue from the Cancer Genome Atlas (TCGA). There was no mutation found in MAT1A or GNMT RNA in the 42 HCC patients. The 11,799 genes were annotated in the RNA-Seq data, and their expression levels were used to investigate the phenotypes of low MAT1A and low GNMT by Gene Set Enrichment Analysis (GSEA). The REACTOME_TRANSLATION gene set was enriched and visualized in a heatmap along with corresponding differences in gene expression between low MAT1A versus high MAT1A and low GNMT versus high GNMT. We identified 43 genes of the REACTOME_TRANSLATION gene set that are powerful prognosis factors in HCC. The significantly predicted genes were referred into eukaryotic translation initiation (EIF3B, EIF3K), eukaryotic translation elongation (EEF1D), and ribosomal proteins (RPs). Cell models expressing various MAT1A and GNMT proved that simultaneous restoring the expression of MAT1A and GNMT decreased cell proliferation, invasion, as well as the REACTOME_TRANSLATION gene EEF1D, consistent with a better prognosis in human HCC. We demonstrated new findings that downregulation or defect in MAT1A and GNMT genes can enrich the protein-associated translation process that may account for poor HCC prognosis. This is the first study demonstrated that MAT1A and GNMT, the 2 key enzymes involved in methionine cycle, could attenuate the function of ribosome translation. We propose a potential novel mechanism by which the diminished GNMT and MAT1A expression may confer poor prognosis for HCC.
Collapse
|
46
|
Hyperlipidemia May Synergize with Hypomethylation in Establishing Trained Immunity and Promoting Inflammation in NASH and NAFLD. J Immunol Res 2021; 2021:3928323. [PMID: 34859106 PMCID: PMC8632388 DOI: 10.1155/2021/3928323] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 10/12/2021] [Indexed: 02/07/2023] Open
Abstract
We performed a panoramic analysis on both human nonalcoholic steatohepatitis (NASH) microarray data and microarray/RNA-seq data from various mouse models of nonalcoholic fatty liver disease NASH/NAFLD with total 4249 genes examined and made the following findings: (i) human NASH and NAFLD mouse models upregulate both cytokines and chemokines; (ii) pathway analysis indicated that human NASH can be classified into metabolic and immune NASH; methionine- and choline-deficient (MCD)+high-fat diet (HFD), glycine N-methyltransferase deficient (GNMT-KO), methionine adenosyltransferase 1A deficient (MAT1A-KO), and HFCD (high-fat-cholesterol diet) can be classified into inflammatory, SAM accumulation, cholesterol/mevalonate, and LXR/RXR-fatty acid β-oxidation NAFLD, respectively; (iii) canonical and noncanonical inflammasomes play differential roles in the pathogenesis of NASH/NAFLD; (iv) trained immunity (TI) enzymes are significantly upregulated in NASH/NAFLD; HFCD upregulates TI enzymes more than cytokines, chemokines, and inflammasome regulators; (v) the MCD+HFD is a model with the upregulation of proinflammatory cytokines and canonical and noncanonical inflammasomes; however, the HFCD is a model with upregulation of TI enzymes and lipid peroxidation enzymes; and (vi) caspase-11 and caspase-1 act as upstream master regulators, which partially upregulate the expressions of cytokines, chemokines, canonical and noncanonical inflammasome pathway regulators, TI enzymes, and lipid peroxidation enzymes. Our findings provide novel insights on the synergies between hyperlipidemia and hypomethylation in establishing TI and promoting inflammation in NASH and NAFLD progression and novel targets for future therapeutic interventions for NASH and NAFLD, metabolic diseases, transplantation, and cancers.
Collapse
|
47
|
Lee J, Song JH, Chung MY, Lee JH, Nam TG, Park JH, Hwang JT, Choi HK. 3,4-dihydroxytoluene, a metabolite of rutin, suppresses the progression of nonalcoholic fatty liver disease in mice by inhibiting p300 histone acetyltransferase activity. Acta Pharmacol Sin 2021; 42:1449-1460. [PMID: 33303988 PMCID: PMC8379200 DOI: 10.1038/s41401-020-00571-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Accepted: 11/02/2020] [Indexed: 01/07/2023] Open
Abstract
3,3',4',5,7-Pentahydroxyflavone-3-rhamnoglucoside (rutin) is a flavonoid with a wide range of pharmacological activities. Dietary rutin is hardly absorbed because the microflora in the large intestine metabolize rutin into a variety of compounds including quercetin and phenol derivatives such as 3,4-dihydroxyphenolacetic acid (DHPAA), 3,4-dihydroxytoluene (DHT), 3,4-hydroxyphenylacetic acid (HPAA) and homovanillic acid (HVA). We examined the potential of rutin and its metabolites as novel histone acetyltransferase (HAT) inhibitors. DHPAA, HPAA and DHT at the concentration of 25 μM significantly inhibited in vitro HAT activity with DHT having the strongest inhibitory activity. Furthermore, DHT was shown to be a highly efficient inhibitor of p300 HAT activity, which corresponded with its high degree of inhibition on intracellular lipid accumulation in HepG2 cells. Docking simulation revealed that DHT was bound to the p300 catalytic pocket, bromodomain. Drug affinity responsive target stability (DARTS) analysis further supported the possibility of direct binding between DHT and p300. In HepG2 cells, DHT concentration-dependently abrogated p300-histone binding and induced hypoacetylation of histone subunits H3K9, H3K36, H4K8 and H4K16, eventually leading to the downregulation of lipogenesis-related genes and attenuating lipid accumulation. In ob/ob mice, administration of DHT (10, 20 mg/kg, iv, every other day for 6 weeks) dose-dependently improved the NAFLD pathogenic features including body weight, liver mass, fat mass, lipid accumulation in the liver, and biochemical blood parameters, accompanied by the decreased mRNA expression of lipogenic genes in the liver. Our results demonstrate that DHT, a novel p300 histone acetyltransferase inhibitor, may be a potential preventive or therapeutic agent for NAFLD.
Collapse
|
48
|
Yang D, Zhang X, Yue L, Hu H, Wei X, Guo Q, Zhang B, Fan X, Xin Y, Oh Y, Gu N. Thiamethoxam induces nonalcoholic fatty liver disease in mice via methionine metabolism disturb via nicotinamide N-methyltransferase overexpression. CHEMOSPHERE 2021; 273:129727. [PMID: 33524747 DOI: 10.1016/j.chemosphere.2021.129727] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 01/10/2021] [Accepted: 01/19/2021] [Indexed: 06/12/2023]
Abstract
Thiamethoxam (TMX) is one of the major compounds of neonicotinoids, the most widely used class of insecticides worldwide. Previously, TMX was considered a non-toxic neonicotinoid insecticide to mammals. However, the genotoxicity, cytotoxicity, and hepatotoxicity of TMX in mammals were recently reported. Thus far, the effects of TMX on the mouse liver and its detailed mechanism remain unclear. NNMT, strongly expressed in the liver, plays a critical role in body energy expenditure. To confirm the potential pathogenesis of liver dysfunction induced by TMX, ICR mice were exposed to TMX at a dose of 4 mg/kg and 20 mg/kg by gavage administration for 12 weeks. The data showed that chronic TMX exposure caused dyslipidemia and nonalcoholic fatty liver disease (NAFLD) in mice. Moreover, aggravated oxidative stress, dysfunction, and disorganized structure were also observed in TMX-treated mouse livers. In addition, increases of PPARγ, fatty acid synthase, and NNMT expression, as well as decreases of PPARα and GNMT expression, S-adenosylmethionine deficiency, and methionine metabolism disorder were also observed in TMX-treated mouse livers. These results suggest that chronic TMX exposure induces dyslipidemia and NAFLD in mice. Moreover, inhibition of NNMT in hepatocytes significantly reversed the effects of TMX. The molecular mechanism of TMX-induced NAFLD is mostly through NNMT-mediated methionine metabolism and methyl donor balance, which ultimately regulates PPARα signaling pathway. Inhibition of NNMT could be a potentially novel strategy for blocking the progression of NAFLD induced by TMX.
Collapse
Affiliation(s)
- Daqian Yang
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Xiaoting Zhang
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Lei Yue
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Hailong Hu
- Department of Medicine, Renal Electrolyte and Hypertension Division, Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Xiangjuan Wei
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Qian Guo
- Department of Medical Genetics and Molecular Biochemistry, Lewis Katz School of Medicine at Temple University, Philadelphia, USA
| | - Boya Zhang
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Xingpei Fan
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Yuan Xin
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Yuri Oh
- Faculty of Education, Wakayama University, Wakayama, Japan
| | - Ning Gu
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, China; State Key Laboratory of Urban Water Resource and Environment, Harbin Institute of Technology, Harbin, China.
| |
Collapse
|
49
|
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: 89] [Impact Index Per Article: 29.7] [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
|
50
|
Sun Y, Demagny H, Schoonjans K. Emerging functions of the nuclear receptor LRH-1 in liver physiology and pathology. Biochim Biophys Acta Mol Basis Dis 2021; 1867:166145. [PMID: 33862147 DOI: 10.1016/j.bbadis.2021.166145] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 03/30/2021] [Accepted: 04/08/2021] [Indexed: 02/06/2023]
Abstract
Nuclear receptors play pleiotropic roles in cell differentiation, development, proliferation, and metabolic processes to govern liver physiology and pathology. The nuclear receptor, liver receptor homolog-1 (LRH-1, NR5A2), originally identified in the liver as a regulator of bile acid and cholesterol homeostasis, was recently recognized to coordinate a multitude of other hepatic metabolic processes, including glucose and lipid processing, methyl group sensing, and cellular stress responses. In this review, we summarize the physiological and pathophysiological functions of LRH-1 in the liver, as well as the molecular mechanisms underlying these processes. This review also focuses on the recent advances highlighting LRH-1 as an attractive target for liver-associated diseases, such as non-alcoholic fatty liver disease (NAFLD) and hepatocellular carcinoma (HCC).
Collapse
Affiliation(s)
- Yu Sun
- Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne, CH-1015 Lausanne, Switzerland
| | - Hadrien Demagny
- Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne, CH-1015 Lausanne, Switzerland
| | - Kristina Schoonjans
- Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne, CH-1015 Lausanne, Switzerland.
| |
Collapse
|