51
|
Gu J, Jing L, Ma X, Zhang Z, Guo Q, Li Y. GC-TOF-MS-based serum metabolomic investigations of naked oat bran supplementation in high-fat-diet-induced dyslipidemic rats. J Nutr Biochem 2015; 26:1509-19. [PMID: 26388495 DOI: 10.1016/j.jnutbio.2015.07.019] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Revised: 06/19/2015] [Accepted: 07/20/2015] [Indexed: 01/15/2023]
Abstract
The present study aimed to explore the metabolic response of oat bran consumption in dyslipidemic rats by a high-throughput metabolomics approach. Four groups of Sprague-Dawley rats were used: N group (normal chow diet), M group (dyslipidemia induced by 4-week high-fat feeding, then normal chow diet), OL group and OH group (dyslipidemia induced, then normal chow diet supplemented with 10.8% or 43.4% naked oat bran). Intervention lasted for 12weeks. Gas chromatography quadrupole time-of-flight mass spectrometry was used to identify serum metabolite profiles. Results confirmed the effects of oat bran on improving lipidemic variables and showed distinct metabolomic profiles associated with diet intervention. A number of endogenous molecules were changed by high-fat diet and normalized following supplementation of naked oat bran. Elevated levels of serum unsaturated fatty acids including arachidonic acid (Log2Fold of change=0.70, P=.02 OH vs. M group), palmitoleic acid (Log2Fold of change=1.24, P=.02 OH vs. M group) and oleic acid (Log2Fold of change=0.66, P=.04 OH vs. M group) were detected after oat bran consumption. Furthermore, consumption of oat bran was also characterized by higher levels of methionine and S-adenosylmethionine. Pathway exploration found that most of the discriminant metabolites were involved in fatty acid biosynthesis, biosynthesis and metabolism of amino acids, microbial metabolism in diverse environments and biosynthesis of plant secondary metabolites. These results point to potential biomarkers and underlying benefit of naked oat bran in the context of diet-induced dyslipidemia and offer some insights into the mechanism exploration.
Collapse
Affiliation(s)
- Jiaojiao Gu
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Beijing, 100191, PR China
| | - Lulu Jing
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Beijing, 100191, PR China
| | - Xiaotao Ma
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Beijing, 100191, PR China; Department of Nutrition, China-Japan Friendship Hospital, Beijing, 100029, PR China
| | - Zhaofeng Zhang
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Beijing, 100191, PR China
| | - Qianying Guo
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Beijing, 100191, PR China
| | - Yong Li
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Beijing, 100191, PR China.
| |
Collapse
|
52
|
Deciphering non-alcoholic fatty liver disease through metabolomics. Biochem Soc Trans 2015; 42:1447-52. [PMID: 25233430 DOI: 10.1042/bst20140138] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is one of the most common liver disorders in industrialized countries. NAFLD develops in the absence of alcohol abuse and encompasses a wide spectrum of disorders ranging from benign fatty liver to non-alcoholic steatohepatitis (NASH). NASH often leads to fibrosis, cirrhosis and, finally, hepatocellular carcinoma (HCC). Therefore the earlier NAFLD is diagnosed, the better the patient's outlook. A tightly connected basic and applied research is essential to find the molecular mechanisms that accompany illness and to translate them into the clinic. From the simple starting point for triacylglycerol (TG) accumulation in the liver to the more complex implications of phospholipids in membrane biophysics, the influence of lipids may be the clue to understand NAFLD pathophysiology. Nowadays, it is achievable to diagnose non-invasively the initial symptoms to stop, revert or even prevent disease development. In this context, merging metabolomics with other techniques and the interpretation of the huge information obtained resembles the 'Rosetta stone' to decipher the pathological metabolic fluxes that must be targeted to find a cure. In the present review, we have tackled the application of metabolomics to find out the metabolic fluxes that underlie membrane integrity in NAFLD.
Collapse
|
53
|
Noureddin M, Mato JM, Lu SC. Nonalcoholic fatty liver disease: update on pathogenesis, diagnosis, treatment and the role of S-adenosylmethionine. Exp Biol Med (Maywood) 2015; 240:809-20. [PMID: 25873078 PMCID: PMC4818965 DOI: 10.1177/1535370215579161] [Citation(s) in RCA: 82] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is currently the most common liver disease worldwide affecting over one-third of the population in the U.S. It has been associated with obesity, type 2 diabetes, hyperlipidemia, and insulin resistance and is initiated by the accumulation of triglycerides in hepatocytes. Isolated hepatic steatosis (IHS) remains a benign process, while a subset develops superimposed inflammatory activity and progression to nonalcoholic steatohepatitis (NASH) with or without fibrosis. However, the molecular mechanisms underlying NAFLD progression are not completely understood. Liver biopsy is still required to differentiate IHS from NASH as easily accessible noninvasive biomarkers are lacking. In terms of treatments for NASH, pioglitazone, vitamin E, and obeticholic acid have shown some benefit. All of these agents have potential complications associated with long-term use. Nowadays, a complex hypothesis suggests that multiple parallel hits are involved in NASH development. However, the 'key switch' between IHS and NASH remains to be discovered. We have recently shown that knocking out enzymes involved in S-adenosylmethionine (SAMe) metabolism, the main biological methyl donor in humans that is abundant in the liver, will lead to NASH development in mice. This could be due to the fact that a normal SAMe level is required to establish the proper ratio of phosphatidylethanolamine to phosphatidylcholine that has been found to be important in NAFLD progression. New data from humans have also suggested that these enzymes play a role in the pathogenesis of NAFLD and that some of SAMe cycle metabolites may serve as noninvasive biomarkers of NASH. In this review, we discuss the evidence of the role of SAMe in animal models and humans with NAFLD and how studying this area may lead to the discovery of new noninvasive biomarkers and possibly personalized treatment for NASH.
Collapse
Affiliation(s)
| | - José M Mato
- Ibaizabal Kalea, 101, 48170 Zamudio, Bizkaia, Spain
| | - Shelly C Lu
- Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| |
Collapse
|
54
|
Hepatocyte-Specific Depletion of UBXD8 Induces Periportal Steatosis in Mice Fed a High-Fat Diet. PLoS One 2015; 10:e0127114. [PMID: 25970332 PMCID: PMC4430229 DOI: 10.1371/journal.pone.0127114] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2014] [Accepted: 04/10/2015] [Indexed: 12/19/2022] Open
Abstract
We showed previously that UBXD8 plays a key role in proteasomal degradation of lipidated ApoB in hepatocarcinoma cell lines. In the present study, we aimed to investigate the functions of UBXD8 in liver in vivo. For this purpose, hepatocyte-specific UBXD8 knockout (UBXD8-LKO) mice were generated. They were fed with a normal or high-fat diet, and the phenotypes were compared with those of littermate control mice. Hepatocytes obtained from UBXD8-LKO and control mice were analyzed in culture. After 26 wk of a high-fat diet, UBXD8-LKO mice exhibited macrovesicular steatosis in the periportal area and microvesicular steatosis in the perivenular area, whereas control mice exhibited steatosis only in the perivenular area. Furthermore, UBXD8-LKO mice on a high-fat diet had significantly lower concentrations of serum triglyceride and VLDL than control mice. A Triton WR-1339 injection study revealed that VLDL secretion from hepatocytes was reduced in UBXD8-LKO mice. The decrease of ApoB secretion upon UBXD8 depletion was recapitulated in cultured primary hepatocytes. Accumulation of lipidated ApoB in lipid droplets was observed only in UBXD8-null hepatocytes. The results showed that depletion of UBXD8 in hepatocytes suppresses VLDL secretion, and could lead to periportal steatosis when mice are fed a high-fat diet. This is the first demonstration that an abnormality in the intracellular ApoB degradation mechanism can cause steatosis, and provides a useful model for periportal steatosis, which occurs in several human diseases.
Collapse
|
55
|
Arisqueta L, Navarro-Imaz H, Rueda Y, Fresnedo O. Cholesterol mobilization from hepatic lipid droplets during endotoxemia is altered in obese ob/ob mice. J Biochem 2015; 158:321-9. [DOI: 10.1093/jb/mvv047] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Accepted: 03/31/2015] [Indexed: 11/12/2022] Open
|
56
|
Benet M, Guzmán C, Pisonero-Vaquero S, García-Mediavilla MV, Sánchez-Campos S, Martínez-Chantar ML, Donato MT, Castell JV, Jover R. Repression of the nuclear receptor small heterodimer partner by steatotic drugs and in advanced nonalcoholic fatty liver disease. Mol Pharmacol 2015; 87:582-94. [PMID: 25576488 DOI: 10.1124/mol.114.096313] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The small heterodimer partner (SHP) (NR0B2) is an atypical nuclear receptor that lacks a DNA-binding domain. It interacts with and inhibits many transcription factors, affecting key metabolic processes, including bile acid, cholesterol, fatty acid, and drug metabolism. Our aim was to determine the influence of steatotic drugs and nonalcoholic fatty liver disease (NAFLD) on SHP expression and investigate the potential mechanisms. SHP was found to be repressed by steatotic drugs (valproate, doxycycline, tetracycline, and cyclosporin A) in cultured hepatic cells and the livers of different animal models of NAFLD: iatrogenic (tetracycline-treated rats), genetic (glycine N-methyltransferase-deficient mice), and nutritional (mice fed a methionine- and choline-deficient diet). Among the different transcription factors investigated, CCAAT-enhancer-binding protein α (C/EBPα) showed the strongest dominant-repressive effect on SHP expression in HepG2 and human hepatocytes. Reporter assays revealed that the inhibitory effect of C/EBPα and steatotic drugs colocalize between -340 and -509 base pair of the SHP promoter, and mutation of a predicted C/EBPα response element at -473 base pair abolished SHP repression by both C/EBPα and drugs. Moreover, inhibition of major stress signaling pathways demonstrated that the mitogen-activated protein kinase kinase 1/2 pathway activates, while the phosphatidylinositol 3 kinase pathway represses SHP in a C/EBP-dependent manner. We conclude that SHP is downregulated by several steatotic drugs and in advanced NAFLD. These conditions can activate signals that target C/EBPα and consequently repress SHP, thus favoring the progression and severity of NAFLD.
Collapse
Affiliation(s)
- Marta Benet
- Experimental Hepatology Unit, IIS Hospital La Fe, Valencia (M.B., C.G., M.T.D., J.V.C., R.J.); CIBERehd, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas, Barcelona (M.B., M.V.G.-M., S.S.-C., M.L.M.-C., M.T.D., J.V.C., R.J.); Institute of Biomedicine, University of León, León (S.P.-V., M.V.G.-M., S.S.-C.); CIC bioGUNE, Technology Park of Bizkaia, Derio (M.L.M.-C.); and Department of Biochemistry and Molecular Biology, University of Valencia, Valencia, Spain (M.T.D., J.V.C., R.J.)
| | - Carla Guzmán
- Experimental Hepatology Unit, IIS Hospital La Fe, Valencia (M.B., C.G., M.T.D., J.V.C., R.J.); CIBERehd, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas, Barcelona (M.B., M.V.G.-M., S.S.-C., M.L.M.-C., M.T.D., J.V.C., R.J.); Institute of Biomedicine, University of León, León (S.P.-V., M.V.G.-M., S.S.-C.); CIC bioGUNE, Technology Park of Bizkaia, Derio (M.L.M.-C.); and Department of Biochemistry and Molecular Biology, University of Valencia, Valencia, Spain (M.T.D., J.V.C., R.J.)
| | - Sandra Pisonero-Vaquero
- Experimental Hepatology Unit, IIS Hospital La Fe, Valencia (M.B., C.G., M.T.D., J.V.C., R.J.); CIBERehd, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas, Barcelona (M.B., M.V.G.-M., S.S.-C., M.L.M.-C., M.T.D., J.V.C., R.J.); Institute of Biomedicine, University of León, León (S.P.-V., M.V.G.-M., S.S.-C.); CIC bioGUNE, Technology Park of Bizkaia, Derio (M.L.M.-C.); and Department of Biochemistry and Molecular Biology, University of Valencia, Valencia, Spain (M.T.D., J.V.C., R.J.)
| | - M Victoria García-Mediavilla
- Experimental Hepatology Unit, IIS Hospital La Fe, Valencia (M.B., C.G., M.T.D., J.V.C., R.J.); CIBERehd, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas, Barcelona (M.B., M.V.G.-M., S.S.-C., M.L.M.-C., M.T.D., J.V.C., R.J.); Institute of Biomedicine, University of León, León (S.P.-V., M.V.G.-M., S.S.-C.); CIC bioGUNE, Technology Park of Bizkaia, Derio (M.L.M.-C.); and Department of Biochemistry and Molecular Biology, University of Valencia, Valencia, Spain (M.T.D., J.V.C., R.J.)
| | - Sonia Sánchez-Campos
- Experimental Hepatology Unit, IIS Hospital La Fe, Valencia (M.B., C.G., M.T.D., J.V.C., R.J.); CIBERehd, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas, Barcelona (M.B., M.V.G.-M., S.S.-C., M.L.M.-C., M.T.D., J.V.C., R.J.); Institute of Biomedicine, University of León, León (S.P.-V., M.V.G.-M., S.S.-C.); CIC bioGUNE, Technology Park of Bizkaia, Derio (M.L.M.-C.); and Department of Biochemistry and Molecular Biology, University of Valencia, Valencia, Spain (M.T.D., J.V.C., R.J.)
| | - M Luz Martínez-Chantar
- Experimental Hepatology Unit, IIS Hospital La Fe, Valencia (M.B., C.G., M.T.D., J.V.C., R.J.); CIBERehd, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas, Barcelona (M.B., M.V.G.-M., S.S.-C., M.L.M.-C., M.T.D., J.V.C., R.J.); Institute of Biomedicine, University of León, León (S.P.-V., M.V.G.-M., S.S.-C.); CIC bioGUNE, Technology Park of Bizkaia, Derio (M.L.M.-C.); and Department of Biochemistry and Molecular Biology, University of Valencia, Valencia, Spain (M.T.D., J.V.C., R.J.)
| | - M Teresa Donato
- Experimental Hepatology Unit, IIS Hospital La Fe, Valencia (M.B., C.G., M.T.D., J.V.C., R.J.); CIBERehd, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas, Barcelona (M.B., M.V.G.-M., S.S.-C., M.L.M.-C., M.T.D., J.V.C., R.J.); Institute of Biomedicine, University of León, León (S.P.-V., M.V.G.-M., S.S.-C.); CIC bioGUNE, Technology Park of Bizkaia, Derio (M.L.M.-C.); and Department of Biochemistry and Molecular Biology, University of Valencia, Valencia, Spain (M.T.D., J.V.C., R.J.)
| | - José Vicente Castell
- Experimental Hepatology Unit, IIS Hospital La Fe, Valencia (M.B., C.G., M.T.D., J.V.C., R.J.); CIBERehd, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas, Barcelona (M.B., M.V.G.-M., S.S.-C., M.L.M.-C., M.T.D., J.V.C., R.J.); Institute of Biomedicine, University of León, León (S.P.-V., M.V.G.-M., S.S.-C.); CIC bioGUNE, Technology Park of Bizkaia, Derio (M.L.M.-C.); and Department of Biochemistry and Molecular Biology, University of Valencia, Valencia, Spain (M.T.D., J.V.C., R.J.)
| | - Ramiro Jover
- Experimental Hepatology Unit, IIS Hospital La Fe, Valencia (M.B., C.G., M.T.D., J.V.C., R.J.); CIBERehd, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas, Barcelona (M.B., M.V.G.-M., S.S.-C., M.L.M.-C., M.T.D., J.V.C., R.J.); Institute of Biomedicine, University of León, León (S.P.-V., M.V.G.-M., S.S.-C.); CIC bioGUNE, Technology Park of Bizkaia, Derio (M.L.M.-C.); and Department of Biochemistry and Molecular Biology, University of Valencia, Valencia, Spain (M.T.D., J.V.C., R.J.)
| |
Collapse
|
57
|
Martínez-Uña M, Varela-Rey M, Mestre D, Fernández-Ares L, Fresnedo O, Fernandez-Ramos D, Gutiérrez-de Juan V, Martin-Guerrero I, García-Orad A, Luka Z, Wagner C, Lu SC, García-Monzón C, Finnell RH, Aurrekoetxea I, Buqué X, Martínez-Chantar ML, Mato JM, Aspichueta P. S-Adenosylmethionine increases circulating very-low density lipoprotein clearance in non-alcoholic fatty liver disease. J Hepatol 2015; 62:673-81. [PMID: 25457203 PMCID: PMC4336596 DOI: 10.1016/j.jhep.2014.10.019] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2014] [Revised: 09/05/2014] [Accepted: 10/09/2014] [Indexed: 12/12/2022]
Abstract
BACKGROUND & AIMS Very-low-density lipoproteins (VLDLs) export lipids from the liver to peripheral tissues and are the precursors of low-density-lipoproteins. Low levels of hepatic S-adenosylmethionine (SAMe) decrease triglyceride (TG) secretion in VLDLs, contributing to hepatosteatosis in methionine adenosyltransferase 1A knockout mice but nothing is known about the effect of SAMe on the circulating VLDL metabolism. We wanted to investigate whether excess SAMe could disrupt VLDL plasma metabolism and unravel the mechanisms involved. METHODS Glycine N-methyltransferase (GNMT) knockout (KO) mice, GNMT and perilipin-2 (PLIN2) double KO (GNMT-PLIN2-KO) and their respective wild type (WT) controls were used. A high fat diet (HFD) or a methionine deficient diet (MDD) was administrated to exacerbate or recover VLDL metabolism, respectively. Finally, 33 patients with non-alcoholic fatty-liver disease (NAFLD); 11 with hypertriglyceridemia and 22 with normal lipidemia were used in this study. RESULTS We found that excess SAMe increases the turnover of hepatic TG stores for secretion in VLDL in GNMT-KO mice, a model of NAFLD with high SAMe levels. The disrupted VLDL assembly resulted in the secretion of enlarged, phosphatidylethanolamine-poor, TG- and apoE-enriched VLDL-particles; special features that lead to increased VLDL clearance and decreased serum TG levels. Re-establishing normal SAMe levels restored VLDL secretion, features and metabolism. In NAFLD patients, serum TG levels were lower when hepatic GNMT-protein expression was decreased. CONCLUSIONS Excess hepatic SAMe levels disrupt VLDL assembly and features and increase circulating VLDL clearance, which will cause increased VLDL-lipid supply to tissues and might contribute to the extrahepatic complications of NAFLD.
Collapse
Affiliation(s)
- Maite Martínez-Uña
- Department of Physiology, University of the Basque Country UPV/EHU, Spain; Biocruces Research Institute, Spain
| | - Marta Varela-Rey
- CIC bioGUNE, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Technology Park of Bizkaia, Spain
| | - Daniela Mestre
- Department of Physiology, University of the Basque Country UPV/EHU, Spain; Biocruces Research Institute, Spain
| | - Larraitz Fernández-Ares
- Department of Physiology, University of the Basque Country UPV/EHU, Spain; Biocruces Research Institute, Spain
| | - Olatz Fresnedo
- Department of Physiology, University of the Basque Country UPV/EHU, Spain
| | - David Fernandez-Ramos
- CIC bioGUNE, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Technology Park of Bizkaia, Spain
| | - Virginia Gutiérrez-de Juan
- CIC bioGUNE, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Technology Park of Bizkaia, Spain
| | - Idoia Martin-Guerrero
- Department of Genetics, Physical Anthropology and Animal Physiology, University of the Basque Country UPV/EHU, Spain
| | - Africa García-Orad
- Department of Genetics, Physical Anthropology and Animal Physiology, University of the Basque Country UPV/EHU, Spain
| | - Zigmund Luka
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Conrad Wagner
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Shelly C Lu
- Division of Gastroenterology and Liver Diseases, University of Southern California Research Center for Liver Diseases, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Carmelo García-Monzón
- Liver Research Unit, University Hospital Santa Cristina, Instituto de Investigación Sanitaria Princesa, Madrid, Spain, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Spain
| | - Richard H Finnell
- Department of Nutritional Sciences, Dell Pediatric Institute, The University of Texas at Austin, Austin, TX, USA
| | - Igor Aurrekoetxea
- Department of Physiology, University of the Basque Country UPV/EHU, Spain; Biocruces Research Institute, Spain
| | - Xabier Buqué
- Department of Physiology, University of the Basque Country UPV/EHU, Spain; Biocruces Research Institute, Spain
| | - M Luz Martínez-Chantar
- CIC bioGUNE, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Technology Park of Bizkaia, Spain; Department of Biochemistry and Molecular Biology, University of the Basque Country UPV/EHU, Spain
| | - José M Mato
- CIC bioGUNE, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Technology Park of Bizkaia, Spain
| | - Patricia Aspichueta
- Department of Physiology, University of the Basque Country UPV/EHU, Spain; Biocruces Research Institute, Spain.
| |
Collapse
|
58
|
Christensen KE, Mikael LG, Leung KY, Lévesque N, Deng L, Wu Q, Malysheva OV, Best A, Caudill MA, Greene NDE, Rozen R. High folic acid consumption leads to pseudo-MTHFR deficiency, altered lipid metabolism, and liver injury in mice. Am J Clin Nutr 2015; 101:646-58. [PMID: 25733650 PMCID: PMC4340065 DOI: 10.3945/ajcn.114.086603] [Citation(s) in RCA: 110] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2014] [Accepted: 12/05/2014] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND Increased consumption of folic acid is prevalent, leading to concerns about negative consequences. The effects of folic acid on the liver, the primary organ for folate metabolism, are largely unknown. Methylenetetrahydrofolate reductase (MTHFR) provides methyl donors for S-adenosylmethionine (SAM) synthesis and methylation reactions. OBJECTIVE Our goal was to investigate the impact of high folic acid intake on liver disease and methyl metabolism. DESIGN Folic acid-supplemented diet (FASD, 10-fold higher than recommended) and control diet were fed to male Mthfr(+/+) and Mthfr(+/-) mice for 6 mo to assess gene-nutrient interactions. Liver pathology, folate and choline metabolites, and gene expression in folate and lipid pathways were examined. RESULTS Liver and spleen weights were higher and hematologic profiles were altered in FASD-fed mice. Liver histology revealed unusually large, degenerating cells in FASD Mthfr(+/-) mice, consistent with nonalcoholic fatty liver disease. High folic acid inhibited MTHFR activity in vitro, and MTHFR protein was reduced in FASD-fed mice. 5-Methyltetrahydrofolate, SAM, and SAM/S-adenosylhomocysteine ratios were lower in FASD and Mthfr(+/-) livers. Choline metabolites, including phosphatidylcholine, were reduced due to genotype and/or diet in an attempt to restore methylation capacity through choline/betaine-dependent SAM synthesis. Expression changes in genes of one-carbon and lipid metabolism were particularly significant in FASD Mthfr(+/-) mice. The latter changes, which included higher nuclear sterol regulatory element-binding protein 1, higher Srepb2 messenger RNA (mRNA), lower farnesoid X receptor (Nr1h4) mRNA, and lower Cyp7a1 mRNA, would lead to greater lipogenesis and reduced cholesterol catabolism into bile. CONCLUSIONS We suggest that high folic acid consumption reduces MTHFR protein and activity levels, creating a pseudo-MTHFR deficiency. This deficiency results in hepatocyte degeneration, suggesting a 2-hit mechanism whereby mutant hepatocytes cannot accommodate the lipid disturbances and altered membrane integrity arising from changes in phospholipid/lipid metabolism. These preliminary findings may have clinical implications for individuals consuming high-dose folic acid supplements, particularly those who are MTHFR deficient.
Collapse
Affiliation(s)
- Karen E Christensen
- From the Departments of Human Genetics and Pediatrics, McGill University, and the Montreal Children's Hospital site of the McGill University Health Centre Research Institute, Montreal, Quebec, Canada (KEC, LGM, NL, LD, QW, and RR); Developmental Biology and Cancer Programme, Institute of Child Health, University College London, London, United Kingdom (K-YL and NDEG); the Division of Nutritional Sciences and Genomics, Cornell University, Ithaca, NY (OVM and MAC); and the Department of Mathematics and Statistics, McGill University, Montreal, Quebec, Canada (AB)
| | - Leonie G Mikael
- From the Departments of Human Genetics and Pediatrics, McGill University, and the Montreal Children's Hospital site of the McGill University Health Centre Research Institute, Montreal, Quebec, Canada (KEC, LGM, NL, LD, QW, and RR); Developmental Biology and Cancer Programme, Institute of Child Health, University College London, London, United Kingdom (K-YL and NDEG); the Division of Nutritional Sciences and Genomics, Cornell University, Ithaca, NY (OVM and MAC); and the Department of Mathematics and Statistics, McGill University, Montreal, Quebec, Canada (AB)
| | - Kit-Yi Leung
- From the Departments of Human Genetics and Pediatrics, McGill University, and the Montreal Children's Hospital site of the McGill University Health Centre Research Institute, Montreal, Quebec, Canada (KEC, LGM, NL, LD, QW, and RR); Developmental Biology and Cancer Programme, Institute of Child Health, University College London, London, United Kingdom (K-YL and NDEG); the Division of Nutritional Sciences and Genomics, Cornell University, Ithaca, NY (OVM and MAC); and the Department of Mathematics and Statistics, McGill University, Montreal, Quebec, Canada (AB)
| | - Nancy Lévesque
- From the Departments of Human Genetics and Pediatrics, McGill University, and the Montreal Children's Hospital site of the McGill University Health Centre Research Institute, Montreal, Quebec, Canada (KEC, LGM, NL, LD, QW, and RR); Developmental Biology and Cancer Programme, Institute of Child Health, University College London, London, United Kingdom (K-YL and NDEG); the Division of Nutritional Sciences and Genomics, Cornell University, Ithaca, NY (OVM and MAC); and the Department of Mathematics and Statistics, McGill University, Montreal, Quebec, Canada (AB)
| | - Liyuan Deng
- From the Departments of Human Genetics and Pediatrics, McGill University, and the Montreal Children's Hospital site of the McGill University Health Centre Research Institute, Montreal, Quebec, Canada (KEC, LGM, NL, LD, QW, and RR); Developmental Biology and Cancer Programme, Institute of Child Health, University College London, London, United Kingdom (K-YL and NDEG); the Division of Nutritional Sciences and Genomics, Cornell University, Ithaca, NY (OVM and MAC); and the Department of Mathematics and Statistics, McGill University, Montreal, Quebec, Canada (AB)
| | - Qing Wu
- From the Departments of Human Genetics and Pediatrics, McGill University, and the Montreal Children's Hospital site of the McGill University Health Centre Research Institute, Montreal, Quebec, Canada (KEC, LGM, NL, LD, QW, and RR); Developmental Biology and Cancer Programme, Institute of Child Health, University College London, London, United Kingdom (K-YL and NDEG); the Division of Nutritional Sciences and Genomics, Cornell University, Ithaca, NY (OVM and MAC); and the Department of Mathematics and Statistics, McGill University, Montreal, Quebec, Canada (AB)
| | - Olga V Malysheva
- From the Departments of Human Genetics and Pediatrics, McGill University, and the Montreal Children's Hospital site of the McGill University Health Centre Research Institute, Montreal, Quebec, Canada (KEC, LGM, NL, LD, QW, and RR); Developmental Biology and Cancer Programme, Institute of Child Health, University College London, London, United Kingdom (K-YL and NDEG); the Division of Nutritional Sciences and Genomics, Cornell University, Ithaca, NY (OVM and MAC); and the Department of Mathematics and Statistics, McGill University, Montreal, Quebec, Canada (AB)
| | - Ana Best
- From the Departments of Human Genetics and Pediatrics, McGill University, and the Montreal Children's Hospital site of the McGill University Health Centre Research Institute, Montreal, Quebec, Canada (KEC, LGM, NL, LD, QW, and RR); Developmental Biology and Cancer Programme, Institute of Child Health, University College London, London, United Kingdom (K-YL and NDEG); the Division of Nutritional Sciences and Genomics, Cornell University, Ithaca, NY (OVM and MAC); and the Department of Mathematics and Statistics, McGill University, Montreal, Quebec, Canada (AB)
| | - Marie A Caudill
- From the Departments of Human Genetics and Pediatrics, McGill University, and the Montreal Children's Hospital site of the McGill University Health Centre Research Institute, Montreal, Quebec, Canada (KEC, LGM, NL, LD, QW, and RR); Developmental Biology and Cancer Programme, Institute of Child Health, University College London, London, United Kingdom (K-YL and NDEG); the Division of Nutritional Sciences and Genomics, Cornell University, Ithaca, NY (OVM and MAC); and the Department of Mathematics and Statistics, McGill University, Montreal, Quebec, Canada (AB)
| | - Nicholas D E Greene
- From the Departments of Human Genetics and Pediatrics, McGill University, and the Montreal Children's Hospital site of the McGill University Health Centre Research Institute, Montreal, Quebec, Canada (KEC, LGM, NL, LD, QW, and RR); Developmental Biology and Cancer Programme, Institute of Child Health, University College London, London, United Kingdom (K-YL and NDEG); the Division of Nutritional Sciences and Genomics, Cornell University, Ithaca, NY (OVM and MAC); and the Department of Mathematics and Statistics, McGill University, Montreal, Quebec, Canada (AB)
| | - Rima Rozen
- From the Departments of Human Genetics and Pediatrics, McGill University, and the Montreal Children's Hospital site of the McGill University Health Centre Research Institute, Montreal, Quebec, Canada (KEC, LGM, NL, LD, QW, and RR); Developmental Biology and Cancer Programme, Institute of Child Health, University College London, London, United Kingdom (K-YL and NDEG); the Division of Nutritional Sciences and Genomics, Cornell University, Ithaca, NY (OVM and MAC); and the Department of Mathematics and Statistics, McGill University, Montreal, Quebec, Canada (AB)
| |
Collapse
|
59
|
Pandey V, Sultan M, Kashofer K, Ralser M, Amstislavskiy V, Starmann J, Osprian I, Grimm C, Hache H, Yaspo ML, Sültmann H, Trauner M, Denk H, Zatloukal K, Lehrach H, Wierling C. Comparative analysis and modeling of the severity of steatohepatitis in DDC-treated mouse strains. PLoS One 2014; 9:e111006. [PMID: 25347188 PMCID: PMC4210132 DOI: 10.1371/journal.pone.0111006] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2014] [Accepted: 09/20/2014] [Indexed: 01/11/2023] Open
Abstract
Background Non-alcoholic fatty liver disease (NAFLD) has a broad spectrum of disease states ranging from mild steatosis characterized by an abnormal retention of lipids within liver cells to steatohepatitis (NASH) showing fat accumulation, inflammation, ballooning and degradation of hepatocytes, and fibrosis. Ultimately, steatohepatitis can result in liver cirrhosis and hepatocellular carcinoma. Methodology and Results In this study we have analyzed three different mouse strains, A/J, C57BL/6J, and PWD/PhJ, that show different degrees of steatohepatitis when administered a 3,5-diethoxycarbonyl-1,4-dihydrocollidine (DDC) containing diet. RNA-Seq gene expression analysis, protein analysis and metabolic profiling were applied to identify differentially expressed genes/proteins and perturbed metabolite levels of mouse liver samples upon DDC-treatment. Pathway analysis revealed alteration of arachidonic acid (AA) and S-adenosylmethionine (SAMe) metabolism upon other pathways. To understand metabolic changes of arachidonic acid metabolism in the light of disease expression profiles a kinetic model of this pathway was developed and optimized according to metabolite levels. Subsequently, the model was used to study in silico effects of potential drug targets for steatohepatitis. Conclusions We identified AA/eicosanoid metabolism as highly perturbed in DDC-induced mice using a combination of an experimental and in silico approach. Our analysis of the AA/eicosanoid metabolic pathway suggests that 5-hydroxyeicosatetraenoic acid (5-HETE), 15-hydroxyeicosatetraenoic acid (15-HETE) and prostaglandin D2 (PGD2) are perturbed in DDC mice. We further demonstrate that a dynamic model can be used for qualitative prediction of metabolic changes based on transcriptomics data in a disease-related context. Furthermore, SAMe metabolism was identified as being perturbed due to DDC treatment. Several genes as well as some metabolites of this module show differences between A/J and C57BL/6J on the one hand and PWD/PhJ on the other.
Collapse
Affiliation(s)
- Vikash Pandey
- Max Planck Institute for Molecular Genetics, Department Vertebrate Genomics, Berlin, Germany
| | - Marc Sultan
- Max Planck Institute for Molecular Genetics, Department Vertebrate Genomics, Berlin, Germany
| | - Karl Kashofer
- Institute of Pathology, Medical University of Graz, Graz, Austria
| | - Meryem Ralser
- Max Planck Institute for Molecular Genetics, Department Vertebrate Genomics, Berlin, Germany
| | | | | | - Ingrid Osprian
- BIOCRATES Life Sciences AG, Innsbruck, Austria
- LKH Wagna, Department of Internal Medicine, Wagna, Austria
| | - Christina Grimm
- Rheumatology and Clinical Immunology, Charité-University Medicine, Berlin, Germany
| | - Hendrik Hache
- Max Planck Institute for Molecular Genetics, Department Vertebrate Genomics, Berlin, Germany
| | - Marie-Laure Yaspo
- Max Planck Institute for Molecular Genetics, Department Vertebrate Genomics, Berlin, Germany
- Dahlem Centre for Genome Research and Medical Systems Biology, Berlin, Germany
| | | | - Michael Trauner
- Hans Popper Laboratory of Molecular Hepatology, Division of Gastroenterology and Hepatology, Department of Medicine, Medical University of Vienna, Vienna, Austria
| | - Helmut Denk
- Institute of Pathology, Medical University of Graz, Graz, Austria
| | - Kurt Zatloukal
- Institute of Pathology, Medical University of Graz, Graz, Austria
| | - Hans Lehrach
- Max Planck Institute for Molecular Genetics, Department Vertebrate Genomics, Berlin, Germany
- Dahlem Centre for Genome Research and Medical Systems Biology, Berlin, Germany
| | - Christoph Wierling
- Max Planck Institute for Molecular Genetics, Department Vertebrate Genomics, Berlin, Germany
- * E-mail:
| |
Collapse
|
60
|
Abstract
Non-alcoholic fatty liver disease (NAFLD) and non-alcoholic steatohepatitis (NASH) are an increasingly common chronic liver disease closely associated with diabetes and obesity that have reached epidemic proportions. Reports on the prevalence of NAFLD have suggested that 27-34% of the general population in the USA and 40-90% of the obese population worldwide have this disease. Increasing urbanisation rate and associated inappropriate lifestyle changes are not only the risk factors of diabetes, but also unmask genetic predisposition in various populations for the metabolic syndrome and its manifestations including NAFLD and NASH. Lifestyle modifications and balanced nutrition are among the foremost management strategies along with ursodeoxycholic acid, metformin, vitamin E and pentoxifylline. Although weight reduction associated with current therapeutic strategies has shown some promise, maintaining it in the long run is largely unsuccessful. With the safety of pharmacotherapy still being uncertain and can be started only after confirmation, other reasonable interventions such as nutrition hold promise in preventing disease progression. The role of dietary components including branched-chain amino acids, methionine, choline and folic acid is currently being evaluated in various clinical trials. Nutritional approaches sought to overcome the limitations of pharmacotherapy also include evaluating the effects of natural ingredients, such as silymarin and spirulina, on liver disease. Understanding the specific interaction between nutrients and dietary needs in NAFLD and maintaining this balance through either a diet or a nutritional product thus becomes extremely important in providing a more realistic and feasible alternative to treat NAFLD. A planned complete nutritional combination addressing specific needs and helping to prevent the progression of NAFLD is the need of the hour to avert people from ending up with complications.
Collapse
|
61
|
Elshorbagy AK, Nijpels G, Valdivia-Garcia M, Stehouwer CDA, Ocke M, Refsum H, Dekker JM. S-adenosylmethionine is associated with fat mass and truncal adiposity in older adults. J Nutr 2013; 143:1982-8. [PMID: 24068793 DOI: 10.3945/jn.113.179192] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
S-adenosylmethionine (SAM) is synthesized from methionine, which is abundant in animal-derived protein, in an energy-consuming reaction. SAM and S-adenosylhomocysteine (SAH) correlate with body mass index (BMI). Plasma total concentration of the SAM-associated product cysteine (tCys) correlates with fat mass in humans and cysteine promotes adiposity in animals. In a cross-sectional study of 610 participants, we investigated whether SAM and SAH are associated with BMI via lean mass or fat mass and dietary protein sources as determinants of SAM and tCys concentrations. Plasma SAM was not associated with lean mass, but mean adjusted fat mass increased from 24 kg (95% CI: 22.6, 25.1) to 30 kg (95% CI: 28.7, 31.3) across SAM quartiles (P < 0.001) and trunk fat:total fat ratio increased from 0.48 to 0.52 (P < 0.001). Erythrocyte SAM was also positively associated with fat mass and trunk fat:total fat ratio. The association of SAM with fat mass was not weakened by adjustment for serum tCys, lipids, creatinine, or dietary or lifestyle confounders. Concentrations of the SAM precursor, methionine, and the SAM product, SAH, were not independently associated with adiposity. Intake of animal-derived protein was not related to serum methionine but was positively associated with plasma SAM (partial r = 0.11) and serum tCys (partial r = 0.13; P < 0.05 for both after adjustment for age, gender, and total energy intake). In conclusion, plasma SAM, but not methionine, is independently associated with fat mass and truncal adiposity, suggesting increased conversion of methionine to SAM in obese individuals. Prospective studies are needed to investigate the interactions among dietary energy and animal protein content, SAM concentrations, and change in body weight and cardiometabolic risk.
Collapse
|
62
|
Murphy SK, Yang H, Moylan CA, Pang H, Dellinger A, Abdelmalek MF, Garrett ME, Ashley-Koch A, Suzuki A, Tillmann HL, Hauser MA, Mae Diehl A. Relationship between methylome and transcriptome in patients with nonalcoholic fatty liver disease. Gastroenterology 2013; 145:1076-87. [PMID: 23916847 PMCID: PMC3805742 DOI: 10.1053/j.gastro.2013.07.047] [Citation(s) in RCA: 309] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2013] [Revised: 07/25/2013] [Accepted: 07/26/2013] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS Cirrhosis and liver cancer are potential outcomes of advanced nonalcoholic fatty liver disease (NAFLD). It is not clear what factors determine whether patients will develop advanced or mild NAFLD, limiting noninvasive diagnosis and treatment before clinical sequelae emerge. We investigated whether DNA methylation profiles can distinguish patients with mild disease from those with advanced NAFLD, and how these patterns are functionally related to hepatic gene expression. METHODS We collected frozen liver biopsies and clinical data from patients with biopsy-proven NAFLD (56 in the discovery cohort and 34 in the replication cohort). Samples were divided into groups based on histologic severity of fibrosis: F0-1 (mild) and F3-4 (advanced). DNA methylation profiles were determined and coupled with gene expression data from the same biopsies; differential methylation was validated in subsets of the discovery and replication cohorts. We then analyzed interactions between the methylome and transcriptome. RESULTS Clinical features did not differ between patients known to have mild or advanced fibrosis based on biopsy analysis. There were 69,247 differentially methylated CpG sites (76% hypomethylated, 24% hypermethylated) in patients with advanced vs mild NAFLD (P < .05). Methylation at fibroblast growth factor receptor 2, methionine adenosyl methyltransferase 1A, and caspase 1 was validated by bisulfite pyrosequencing and the findings were reproduced in the replication cohort. Methylation correlated with gene transcript levels for 7% of differentially methylated CpG sites, indicating that differential methylation contributes to differences in expression. In samples with advanced NAFLD, many tissue repair genes were hypomethylated and overexpressed, and genes in certain metabolic pathways, including 1-carbon metabolism, were hypermethylated and underexpressed. CONCLUSIONS Functionally relevant differences in methylation can distinguish patients with advanced vs mild NAFLD. Altered methylation of genes that regulate processes such as steatohepatitis, fibrosis, and carcinogenesis indicate the role of DNA methylation in progression of NAFLD.
Collapse
Affiliation(s)
- Susan K. Murphy
- Department of Obstetrics and Gynecology, Duke University, Durham, NC
| | - Hyuna Yang
- Department of Bioinformatics and Biostatistics, Duke University, Durham, NC
| | - Cynthia A. Moylan
- Division of Gastroenterology and Hepatology, Duke University, Durham, NC
| | - Herbert Pang
- Department of Bioinformatics and Biostatistics, Duke University, Durham, NC
| | - Andrew Dellinger
- Department of Bioinformatics and Biostatistics, Duke University, Durham, NC
| | | | | | | | - Ayako Suzuki
- Division of Gastroenterology and Hepatology, Duke University, Durham, NC
| | - Hans L. Tillmann
- Section of Medical Genetics, Department of Medicine, Duke University, Durham, NC
| | | | - Anna Mae Diehl
- Division of Gastroenterology and Hepatology, Duke University, Durham, NC
| |
Collapse
|
63
|
Abstract
NAFLD affects a large proportion of the US population and its incidence and prevalence are increasing to epidemic proportions around the world. As with other liver diseases that cause cirrhosis, NAFLD increases the risk of liver cancer, a disease with poor outcomes and limited therapeutic options. The incidences of hepatocellular carcinoma (HCC) and intrahepatic cholangiocarcinoma are also rising, and HCC is now the leading cause of obesity-related cancer deaths in middle-aged men in the USA. In this Review, we summarize the correlations between liver cancer and NAFLD-related cirrhosis, and the role of the metabolic syndrome in the development of liver cancer from diverse aetiologies, including HCV-mediated cirrhosis. Recent advances in understanding the progression of NAFLD to HCC from preclinical models will also be discussed. Targeted genetic manipulation of certain metabolic or stress-response pathways, including one-carbon metabolism, NF-κB, PTEN and microRNAs, has been valuable in elucidating the pathways that regulate carcinogenesis in NAFLD. Although tremendous advances have occurred in the identification of diagnostic and therapeutic opportunities to reduce the progression of NAFLD, considerable gaps in our knowledge remain with regard to the mechanisms by which NAFLD and its risk factors promote liver cancer.
Collapse
Affiliation(s)
- Gregory A Michelotti
- Division of Gastroenterology, Department of Medicine, Duke University Medical Center, 595 LaSalle Street, Snyderman Building, Suite 1073, Durham, NC 27710, USA
| | | | | |
Collapse
|
64
|
Martínez-Uña M, Varela-Rey M, Cano A, Fernández-Ares L, Beraza N, Aurrekoetxea I, Martínez-Arranz I, García-Rodríguez JL, Buqué X, Mestre D, Luka Z, Wagner C, Alonso C, Finnell RH, Lu SC, Martínez-Chantar ML, Aspichueta P, Mato JM. Excess S-adenosylmethionine reroutes phosphatidylethanolamine towards phosphatidylcholine and triglyceride synthesis. Hepatology 2013; 58:1296-305. [PMID: 23505042 PMCID: PMC3720726 DOI: 10.1002/hep.26399] [Citation(s) in RCA: 99] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2012] [Accepted: 03/10/2013] [Indexed: 01/10/2023]
Abstract
UNLABELLED Methionine adenosyltransferase 1A (MAT1A) and glycine N-methyltransferase (GNMT) are the primary genes involved in hepatic S-adenosylmethionine (SAMe) synthesis and degradation, respectively. Mat1a ablation in mice induces a decrease in hepatic SAMe, activation of lipogenesis, inhibition of triglyceride (TG) release, and steatosis. Gnmt-deficient mice, despite showing a large increase in hepatic SAMe, also develop steatosis. We hypothesized that as an adaptive response to hepatic SAMe accumulation, phosphatidylcholine (PC) synthesis by way of the phosphatidylethanolamine (PE) N-methyltransferase (PEMT) pathway is stimulated in Gnmt(-/-) mice. We also propose that the excess PC thus generated is catabolized, leading to TG synthesis and steatosis by way of diglyceride (DG) generation. We observed that Gnmt(-/-) mice present with normal hepatic lipogenesis and increased TG release. We also observed that the flux from PE to PC is stimulated in the liver of Gnmt(-/-) mice and that this results in a reduction in PE content and a marked increase in DG and TG. Conversely, reduction of hepatic SAMe following the administration of a methionine-deficient diet reverted the flux from PE to PC of Gnmt(-/-) mice to that of wildtype animals and normalized DG and TG content preventing the development of steatosis. Gnmt(-/-) mice with an additional deletion of perilipin2, the predominant lipid droplet protein, maintain high SAMe levels, with a concurrent increased flux from PE to PC, but do not develop liver steatosis. CONCLUSION These findings indicate that excess SAMe reroutes PE towards PC and TG synthesis and lipid sequestration.
Collapse
Affiliation(s)
- Maite Martínez-Uña
- Department of Physiology, University of the Basque Country UPV/EHU, Medical School, Bilbao, Spain
| | - Marta Varela-Rey
- CIC bioGUNE, Ciberehd, Parque Tecnológico de Bizkaia, Bizkaia, Spain
| | | | - Larraitz Fernández-Ares
- Department of Physiology, University of the Basque Country UPV/EHU, Medical School, Bilbao, Spain
| | - Naiara Beraza
- CIC bioGUNE, Ciberehd, Parque Tecnológico de Bizkaia, Bizkaia, Spain
| | - Igor Aurrekoetxea
- Department of Physiology, University of the Basque Country UPV/EHU, Medical School, Bilbao, Spain
| | | | | | - Xabier Buqué
- Department of Physiology, University of the Basque Country UPV/EHU, Medical School, Bilbao, Spain
| | - Daniela Mestre
- Department of Physiology, University of the Basque Country UPV/EHU, Medical School, Bilbao, Spain
| | - Zigmund Luka
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, Tennesse
| | - Conrad Wagner
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, Tennesse
| | | | - Richard H Finnell
- Department of Nutritional Sciences, Dell Pediatric Institute, The University of Texas at Austin, Austin, Texas
| | - Shelly C Lu
- Division of Gastroenterology and Liver Diseases, USC Research Center for Liver Diseases, The Southern California Research Center for Alcoholic and Pancreatic Diseases & Cirrhosis, Keck School of Medicine USC, Los Angeles, California
| | | | - Patricia Aspichueta
- Department of Physiology, University of the Basque Country UPV/EHU, Medical School, Bilbao, Spain
| | - José M Mato
- CIC bioGUNE, Ciberehd, Parque Tecnológico de Bizkaia, Bizkaia, Spain
| |
Collapse
|
65
|
Involvement of lipid droplets in hepatic responses to lipopolysaccharide treatment in mice. Biochim Biophys Acta Mol Cell Biol Lipids 2013; 1831:1357-67. [DOI: 10.1016/j.bbalip.2013.04.015] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2012] [Revised: 04/05/2013] [Accepted: 04/30/2013] [Indexed: 01/07/2023]
|
66
|
Huidobro C, Fernandez AF, Fraga MF. The role of genetics in the establishment and maintenance of the epigenome. Cell Mol Life Sci 2013; 70:1543-73. [PMID: 23474979 PMCID: PMC11113764 DOI: 10.1007/s00018-013-1296-2] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2013] [Revised: 02/05/2013] [Accepted: 02/05/2013] [Indexed: 12/19/2022]
Abstract
Epigenetic mechanisms play an important role in gene regulation during development. DNA methylation, which is probably the most important and best-studied epigenetic mechanism, can be abnormally regulated in common pathologies, but the origin of altered DNA methylation remains unknown. Recent research suggests that these epigenetic alterations could depend, at least in part, on genetic mutations or polymorphisms in DNA methyltransferases and certain genes encoding enzymes of the one-carbon metabolism pathway. Indeed, the de novo methyltransferase 3B (DNMT3B) has been recently found to be mutated in several types of cancer and in the immunodeficiency, centromeric region instability and facial anomalies syndrome (ICF), in which these mutations could be related to the loss of global DNA methylation. In addition, mutations in glycine-N-methyltransferase (GNMT) could be associated with a higher risk of hepatocellular carcinoma and liver disease due to an unbalanced S-adenosylmethionine (SAM)/S-adenosylhomocysteine (SAH) ratio, which leads to aberrant methylation reactions. Also, genetic variants of chromatin remodeling proteins and histone tail modifiers are involved in genetic disorders like α thalassemia X-linked mental retardation syndrome, CHARGE syndrome, Cockayne syndrome, Rett syndrome, systemic lupus erythematous, Rubinstein-Taybi syndrome, Coffin-Lowry syndrome, Sotos syndrome, and facioescapulohumeral syndrome, among others. Here, we review the potential genetic alterations with a possible role on epigenetic factors and discuss their contribution to human disease.
Collapse
Affiliation(s)
- Covadonga Huidobro
- Cancer Epigenetics Laboratory, Institute of Oncology of Asturias (IUOPA-HUCA), University of Oviedo, Oviedo, Spain
| | - Agustin F. Fernandez
- Cancer Epigenetics Laboratory, Institute of Oncology of Asturias (IUOPA-HUCA), University of Oviedo, Oviedo, Spain
| | - Mario F. Fraga
- Cancer Epigenetics Laboratory, Institute of Oncology of Asturias (IUOPA-HUCA), University of Oviedo, Oviedo, Spain
- Department of Immunology and Oncology, Centro Nacional de Biotecnología (CNB-CSIC), Madrid, Spain
| |
Collapse
|
67
|
|
68
|
Guzmán C, Benet M, Pisonero-Vaquero S, Moya M, García-Mediavilla MV, Martínez-Chantar ML, González-Gallego J, Castell JV, Sánchez-Campos S, Jover R. The human liver fatty acid binding protein (FABP1) gene is activated by FOXA1 and PPARα; and repressed by C/EBPα: Implications in FABP1 down-regulation in nonalcoholic fatty liver disease. Biochim Biophys Acta Mol Cell Biol Lipids 2013; 1831:803-18. [PMID: 23318274 DOI: 10.1016/j.bbalip.2012.12.014] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2012] [Revised: 11/22/2012] [Accepted: 12/27/2012] [Indexed: 01/24/2023]
Abstract
Liver fatty acid binding protein (FABP1) prevents lipotoxicity of free fatty acids and regulates fatty acid trafficking and partition. Our objective is to investigate the transcription factors controlling the human FABP1 gene and their regulation in nonalcoholic fatty liver disease (NAFLD). Adenovirus-mediated expression of multiple transcription factors in HepG2 cells and cultured human hepatocytes demonstrated that FOXA1 and PPARα are among the most effective activators of human FABP1, whereas C/EBPα is a major dominant repressor. Moreover, FOXA1 and PPARα induced re-distribution of FABP1 protein and increased cytoplasmic expression. Reporter assays demonstrated that the major basal activity of the human FABP1 promoter locates between -96 and -229bp, where C/EBPα binds to a composite DR1-C/EBP element. Mutation of this element at -123bp diminished basal reporter activity, abolished repression by C/EBPα and reduced transactivation by HNF4α. Moreover, HNF4α gene silencing by shRNA in HepG2 cells caused a significant down-regulation of FABP1 mRNA expression. FOXA1 activated the FABP1 promoter through binding to a cluster of elements between -229 and -592bp, whereas PPARα operated through a conserved proximal element at -59bp. Finally, FABP1, FOXA1 and PPARα were concomitantly repressed in animal models of NAFLD and in human nonalcoholic fatty livers, whereas C/EBPα was induced or did not change. We conclude that human FABP1 has a complex mechanism of regulation where C/EBPα displaces HNF4α and hampers activation by FOXA1 and PPARα. Alteration of expression of these transcription factors in NAFLD leads to FABP1 gen repression and could exacerbate lipotoxicity and disease progression.
Collapse
Affiliation(s)
- Carla Guzmán
- Experimental Hepatology Unit, IIS Hospital La Fe, Valencia, Spain
| | | | | | | | | | | | | | | | | | | |
Collapse
|
69
|
Anstee QM, Day CP. S-adenosylmethionine (SAMe) therapy in liver disease: a review of current evidence and clinical utility. J Hepatol 2012; 57:1097-109. [PMID: 22659519 DOI: 10.1016/j.jhep.2012.04.041] [Citation(s) in RCA: 165] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2012] [Revised: 04/12/2012] [Accepted: 04/15/2012] [Indexed: 12/13/2022]
Abstract
S-adenosyl-L-methionine (SAMe; AdoMet) is an important, metabolically pleiotropic molecule that participates in multiple cellular reactions as the precursor for the synthesis of glutathione and principle methyl donor required for methylation of nucleic acids, phospholipids, histones, biogenic amines, and proteins. SAMe synthesis is depressed in chronic liver disease and so there has been considerable interest in the utility of SAMe to ameliorate disease severity. Despite encouraging pre-clinical data confirming that SAMe depletion can exacerbate liver injury and supporting a hepatoprotective role for SAMe therapy, to date no large, high-quality randomised clinical trials have been performed that establish clinical utility in specific disease states. Here, we offer an in-depth review of the published scientific literature relating to the physiological and pathophysiological roles of SAMe and its therapeutic use in liver disease, critically assessing implications for clinical practice and offering recommendations for further research.
Collapse
Affiliation(s)
- Quentin M Anstee
- Liver Research Group, Institute of Cellular Medicine, The Medical School, Newcastle University, Framlington Place, Newcastle-Upon-Tyne NE2 4HH, UK.
| | | |
Collapse
|
70
|
Abstract
S-adenosylmethionine (AdoMet, also known as SAM and SAMe) is the principal biological methyl donor synthesized in all mammalian cells but most abundantly in the liver. Biosynthesis of AdoMet requires the enzyme methionine adenosyltransferase (MAT). In mammals, two genes, MAT1A that is largely expressed by normal liver and MAT2A that is expressed by all extrahepatic tissues, encode MAT. Patients with chronic liver disease have reduced MAT activity and AdoMet levels. Mice lacking Mat1a have reduced hepatic AdoMet levels and develop oxidative stress, steatohepatitis, and hepatocellular carcinoma (HCC). In these mice, several signaling pathways are abnormal that can contribute to HCC formation. However, injury and HCC also occur if hepatic AdoMet level is excessive chronically. This can result from inactive mutation of the enzyme glycine N-methyltransferase (GNMT). Children with GNMT mutation have elevated liver transaminases, and Gnmt knockout mice develop liver injury, fibrosis, and HCC. Thus a normal hepatic AdoMet level is necessary to maintain liver health and prevent injury and HCC. AdoMet is effective in cholestasis of pregnancy, and its role in other human liver diseases remains to be better defined. In experimental models, it is effective as a chemopreventive agent in HCC and perhaps other forms of cancer as well.
Collapse
Affiliation(s)
- Shelly C Lu
- Division of Gastroenterology and Liver Diseases, USC Research Center for Liver Diseases, Southern California Research Center for ALPD and Cirrhosis, Keck School of Medicine, Los Angeles, California 90033, USA.
| | | |
Collapse
|
71
|
Tehlivets O, Malanovic N, Visram M, Pavkov-Keller T, Keller W. S-adenosyl-L-homocysteine hydrolase and methylation disorders: yeast as a model system. Biochim Biophys Acta Mol Basis Dis 2012; 1832:204-15. [PMID: 23017368 PMCID: PMC3787734 DOI: 10.1016/j.bbadis.2012.09.007] [Citation(s) in RCA: 106] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2012] [Revised: 09/14/2012] [Accepted: 09/18/2012] [Indexed: 12/14/2022]
Abstract
S-adenosyl-L-methionine (AdoMet)-dependent methylation is central to the regulation of many biological processes: more than 50 AdoMet-dependent methyltransferases methylate a broad spectrum of cellular compounds including nucleic acids, proteins and lipids. Common to all AdoMet-dependent methyltransferase reactions is the release of the strong product inhibitor S-adenosyl-L-homocysteine (AdoHcy), as a by-product of the reaction. S-adenosyl-L-homocysteine hydrolase is the only eukaryotic enzyme capable of reversible AdoHcy hydrolysis to adenosine and homocysteine and, thus, relief from AdoHcy inhibition. Impaired S-adenosyl-L-homocysteine hydrolase activity in humans results in AdoHcy accumulation and severe pathological consequences. Hyperhomocysteinemia, which is characterized by elevated levels of homocysteine in blood, also exhibits a similar phenotype of AdoHcy accumulation due to the reversal of the direction of the S-adenosyl-L-homocysteine hydrolase reaction. Inhibition of S-adenosyl-L-homocysteine hydrolase is also linked to antiviral effects. In this review the advantages of yeast as an experimental system to understand pathologies associated with AdoHcy accumulation will be discussed.
Collapse
Affiliation(s)
- Oksana Tehlivets
- Institute of Molecular Biosciences, University of Graz, Graz, Austria.
| | | | | | | | | |
Collapse
|
72
|
Buqué X, Cano A, Miquilena-Colina ME, García-Monzón C, Ochoa B, Aspichueta P. High insulin levels are required for FAT/CD36 plasma membrane translocation and enhanced fatty acid uptake in obese Zucker rat hepatocytes. Am J Physiol Endocrinol Metab 2012; 303:E504-14. [PMID: 22693206 DOI: 10.1152/ajpendo.00653.2011] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
In myocytes and adipocytes, insulin increases fatty acid translocase (FAT)/CD36 translocation to the plasma membrane (PM), enhancing fatty acid (FA) uptake. Evidence links increased hepatic FAT/CD36 protein amount and gene expression with hyperinsulinemia in animal models and patients with fatty liver, but whether insulin regulates FAT/CD36 expression, amount, distribution, and function in hepatocytes is currently unknown. To investigate this, FAT/CD36 protein content in isolated hepatocytes, subfractions of organelles, and density-gradient isolated membrane subfractions was analyzed in obese and lean Zucker rats by Western blotting in liver sections by immunohistochemistry and in hepatocytes by immunocytochemistry. The uptake of oleate and oleate incorporation into lipids were assessed in hepatocytes at short time points (30-600 s). We found that FAT/CD36 protein amount at the PM was higher in hepatocytes from obese rats than from lean controls. In obese rat hepatocytes, decreased cytoplasmatic content of FAT/CD36 and redistribution from low- to middle- to middle- to high-density subfractions of microsomes were found. Hallmarks of obese Zucker rat hepatocytes were increased amount of FAT/CD36 protein at the PM and enhanced FA uptake and incorporation into triglycerides, which were maintained only when exposed to hyperinsulinemic conditions (80 mU/l). In conclusion, high insulin levels are required for FAT/CD36 translocation to the PM in obese rat hepatocytes to enhance FA uptake and triglyceride synthesis. These results suggest that the hyperinsulinemia found in animal models and patients with insulin resistance and fatty liver might contribute to liver fat accumulation by inducing FAT/CD36 functional presence at the PM of hepatocytes.
Collapse
Affiliation(s)
- Xabier Buqué
- Department of Physiology, Faculty of Medicine and Dentistry, University of the Basque Country UPV/EHU, Leioa, Spain
| | | | | | | | | | | |
Collapse
|
73
|
Ramírez-Torres A, Barceló-Batllori S, Fernández-Vizarra E, Navarro MA, Arnal C, Guillén N, Acín S, Osada J. Proteomics and gene expression analyses of mitochondria from squalene-treated apoE-deficient mice identify short-chain specific acyl-CoA dehydrogenase changes associated with fatty liver amelioration. J Proteomics 2012; 75:2563-75. [DOI: 10.1016/j.jprot.2012.02.025] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2012] [Revised: 02/06/2012] [Accepted: 02/20/2012] [Indexed: 02/07/2023]
|
74
|
Abstract
PURPOSE OF REVIEW Choline is an essential nutrient and the liver is a central organ responsible for choline metabolism. Hepatosteatosis and liver cell death occur when humans are deprived of choline. In the last few years, there have been significant advances in our understanding of the mechanisms that influence choline requirements in humans and in our understanding of choline's effects on liver function. These advances are useful in elucidating why nonalcoholic fatty liver disease (NAFLD) occurs and progresses sometimes to hepatocarcinogenesis. RECENT FINDINGS Humans eating low-choline diets develop fatty liver and liver damage. This dietary requirement for choline is modulated by estrogen and by single-nucleotide polymorphisms in specific genes of choline and folate metabolism. The spectrum of choline's effects on liver range from steatosis to development of hepatocarcinomas, and several mechanisms for these effects have been identified. They include abnormal phospholipid synthesis, defects in lipoprotein secretion, oxidative damage caused by mitochondrial dysfunction, and endoplasmic reticulum stress. Furthermore, the hepatic steatosis phenotype can be characterized more fully via metabolomic signatures and is influenced by the gut microbiome. Importantly, the intricate connection between liver function, one-carbon metabolism, and energy metabolism is just beginning to be elucidated. SUMMARY Choline influences liver function, and the dietary requirement for this nutrient varies depending on an individual's genotype and estrogen status. Understanding these individual differences is important for gastroenterologists seeking to understand why some individuals develop NAFLD and others do not, and why some patients tolerate total parenteral nutrition and others develop liver dysfunction.
Collapse
|