1
|
Singal AK, Wong RJ, Dasarathy S, Abdelmalek MF, Neuschwander-Tetri BA, Limketkai BN, Petrey J, McClain CJ. ACG Clinical Guideline: Malnutrition and Nutritional Recommendations in Liver Disease. Am J Gastroenterol 2025; 120:950-972. [PMID: 40314389 DOI: 10.14309/ajg.0000000000003379] [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: 01/31/2024] [Accepted: 01/29/2025] [Indexed: 05/03/2025]
Abstract
Malnutrition, defined as deficiency, excess, or imbalance of nutrients, is a common complication in patients with liver disease, especially those with cirrhosis. Malnutrition may present as an isolated micronutrient deficiency, such as zinc deficiency, and it commonly presents as frailty and/or sarcopenia in patients with advanced liver disease. Patients with cirrhosis and/or alcohol-associated hepatitis should be assessed for malnutrition because it adversely affects patient outcomes including mortality, as well as waitlist and posttransplant outcomes among liver transplant candidates. The prevalence of malnutrition varies based on the method of assessment and disease severity, being higher in those with advanced liver disease. Among stable outpatients with cirrhosis, counseling should be done to eat small frequent meals, a night-time snack between 7 PM and 10 PM, and 2 or more cups of coffee daily. In selected patients with metabolic dysfunction-associated steatohepatitis, vitamin E 800 IU/d should be provided. Among hospitalized patients with cirrhosis, nutritional supplementation preferably by enteral route should be implemented in those with poor oral intake of daily requirements of proteins and/or calories. Protein intake should not be restricted including patients with decompensated cirrhosis and hepatic encephalopathy. A vegetable source of protein seems to be better tolerated than an animal source of protein in patients with hepatic encephalopathy. Branched chain amino acids augment the efficacy of lactulose and rifaximin in the treatment of hepatic encephalopathy. Level of evidence and strength of recommendations were evaluated using the Grading of Recommendations, Assessment, Development, and Evaluations system. This guideline was developed under the auspices of the American College of Gastroenterology Practice Parameters Committee.
Collapse
Affiliation(s)
- Ashwani K Singal
- Department of Medicine, University of Louisville, Louisville, Kentucky, USA
| | - Robert J Wong
- Division of Gastroenterology and Hepatology, Stanford University School of Medicine, Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA
| | - Srinivasan Dasarathy
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine at Case Western Reserve University, Cleveland, Ohio, USA
- Department of Gastroenterology and Hepatology, Cleveland Clinic, Cleveland, Ohio, USA
| | | | - Brent A Neuschwander-Tetri
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Saint Louis University, Saint Louis, Missouri, USA
| | - Berkeley N Limketkai
- Divisions of Digestive Diseases and Clinical Nutrition, UCLA School of Medicine, Los Angeles, California, USA
| | - Jessica Petrey
- Kornhauser Health Sciences Library, University of Louisville, Louisville, Kentucky, USA; and
| | - Craig J McClain
- Departments of Medicine and Pharmacology & Toxicology, Chief of Research Affairs, Division of Gastroenterology, Hepatology and Nutrition, Associate Vice President for Health Affairs/Research, Associate Vice President for Translational Research, Louisville, Kentucky, USA
| |
Collapse
|
2
|
Fu Y, Hou L, Han K, Zhao C, Hu H, Yin S. The physiological role of copper: Dietary sources, metabolic regulation, and safety concerns. Clin Nutr 2025; 48:161-179. [PMID: 40220473 DOI: 10.1016/j.clnu.2025.03.023] [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: 11/10/2024] [Revised: 02/26/2025] [Accepted: 03/30/2025] [Indexed: 04/14/2025]
Abstract
Copper plays an important physiological role in the body, with both deficiency and excess potentially impacting overall health. The body maintains a stringent copper metabolism mechanism to oversee absorption, utilization, storage, and elimination. Dietary consumption serves as the principal source of copper. The dietary factors may interfere with the absorption and metabolism of copper, leading to fluctuation of copper levels in the body. However, these dietary factors can also be strategically employed to facilitate the precise regulation of copper. This paper delved into the advancements in research concerning copper in food processing, including dietary sources of copper, the regulatory processes of copper metabolism and health implications of copper. The safety and its underlying mechanisms of excess copper were also highlighted. In particular, the paper examines the influence of dietary factors on the absorption and metabolism of copper, aiming to provide direction for accurate copper regulation and the creation of functional foods and pharmaceuticals.
Collapse
Affiliation(s)
- Yuhan Fu
- Department of Nutrition and Food Safety, College of Food Science and Nutritional Engineering, China Agricultural University, No.17 Qinghua East Road, Haidian District, Beijing 100083, China
| | - Lirui Hou
- Department of Nutrition and Food Safety, College of Food Science and Nutritional Engineering, China Agricultural University, No.17 Qinghua East Road, Haidian District, Beijing 100083, China
| | - Kai Han
- Department of Nutrition and Food Safety, College of Food Science and Nutritional Engineering, China Agricultural University, No.17 Qinghua East Road, Haidian District, Beijing 100083, China
| | - Chong Zhao
- Department of Nutrition and Food Safety, College of Food Science and Nutritional Engineering, China Agricultural University, No.17 Qinghua East Road, Haidian District, Beijing 100083, China
| | - Hongbo Hu
- Department of Nutrition and Food Safety, College of Food Science and Nutritional Engineering, China Agricultural University, No.17 Qinghua East Road, Haidian District, Beijing 100083, China.
| | - Shutao Yin
- Department of Nutrition and Food Safety, College of Food Science and Nutritional Engineering, China Agricultural University, No.17 Qinghua East Road, Haidian District, Beijing 100083, China.
| |
Collapse
|
3
|
Xu M, Li M, Benz F, Merchant M, McClain CJ, Song M. Ileum Proteomics Identifies Distinct Pathways Associated with Different Dietary Doses of Copper-Fructose Interactions: Implications for the Gut-Liver Axis and MASLD. Nutrients 2024; 16:2083. [PMID: 38999831 PMCID: PMC11242941 DOI: 10.3390/nu16132083] [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: 05/15/2024] [Revised: 06/23/2024] [Accepted: 06/27/2024] [Indexed: 07/14/2024] Open
Abstract
The interactions of different dietary doses of copper with fructose contribute to the development of metabolic dysfunction-associated steatotic liver disease (MASLD) via the gut-liver axis. The underlying mechanisms remain elusive. The aim of this study was to identify the specific pathways leading to gut barrier dysfunction in the ileum using a proteomics approach in a rat model. Male weanling Sprague Dawley rats were fed diets with adequate copper (CuA), marginal copper (CuM), or supplemented copper (CuS) in the absence or presence of fructose supplementation (CuAF, CuMF, and CuSF) for 4 weeks. Ileum protein was extracted and analyzed with an LC-MS. A total of 2847 differentially expressed proteins (DEPs) were identified and submitted to functional enrichment analysis. As a result, the ileum proteome and signaling pathways that were differentially altered were revealed. Of note, the CuAF is characterized by the enrichment of oxidative phosphorylation and ribosome as analyzed with the KEGG; the CuMF is characterized by an enriched arachidonic acid metabolism pathway; and focal adhesion, the regulation of the actin cytoskeleton, and tight junction were significantly enriched by the CuSF. In conclusion, our proteomics analysis identified the specific pathways in the ileum related to the different dietary doses of copper-fructose interactions, suggesting that distinct mechanisms in the gut are involved in the development of MASLD.
Collapse
Affiliation(s)
- Manman Xu
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, University of Louisville School of Medicine, Louisville, KY 40202, USA; (M.X.); (C.J.M.)
| | - Ming Li
- Department of Medicine, Division of Nephrology and Hypertension, University of Louisville School of Medicine, Louisville, KY 40202, USA; (M.L.); (M.M.)
- Hepatobiology & Toxicology Center, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Frederick Benz
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY 40202, USA;
| | - Michael Merchant
- Department of Medicine, Division of Nephrology and Hypertension, University of Louisville School of Medicine, Louisville, KY 40202, USA; (M.L.); (M.M.)
- Hepatobiology & Toxicology Center, University of Louisville School of Medicine, Louisville, KY 40202, USA
- University of Louisville Alcohol Research Center, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Craig J. McClain
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, University of Louisville School of Medicine, Louisville, KY 40202, USA; (M.X.); (C.J.M.)
- Hepatobiology & Toxicology Center, University of Louisville School of Medicine, Louisville, KY 40202, USA
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY 40202, USA;
- University of Louisville Alcohol Research Center, University of Louisville School of Medicine, Louisville, KY 40202, USA
- Robley Rex Louisville VAMC, Louisville, KY 40206, USA
| | - Ming Song
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, University of Louisville School of Medicine, Louisville, KY 40202, USA; (M.X.); (C.J.M.)
- Hepatobiology & Toxicology Center, University of Louisville School of Medicine, Louisville, KY 40202, USA
| |
Collapse
|
4
|
Li C, Liu Z, Wei W, Chen C, Zhang L, Wang Y, Zhou B, Liu L, Li X, Zhao C. Exploring the Regulatory Effect of LPJZ-658 on Copper Deficiency Combined with Sugar-Induced MASLD in Middle-Aged Mice Based on Multi-Omics Analysis. Nutrients 2024; 16:2010. [PMID: 38999758 PMCID: PMC11243161 DOI: 10.3390/nu16132010] [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: 05/05/2024] [Revised: 06/14/2024] [Accepted: 06/15/2024] [Indexed: 07/14/2024] Open
Abstract
Globally, metabolic dysfunction-associated steatotic liver disease (MASLD), previously termed nonalcoholic fatty liver disease (NAFLD), is one of the most common liver disorders and is strongly associated with copper deficiency. To explore the potential effects and mechanisms of Lactiplantibacillus plantarum LPJZ-658, copper deficiency combined with a high-sugar diet-induced MASLD mouse model was utilized in this study. We fed 40-week-old (middle-aged) male C57BL/6 mice a copper-deficient and high-sugar diet for 16 weeks (CuDS), with supplementary LPJZ-658 for the last 6 weeks (CuDS + LPJZ-658). In this study, we measured body weight, liver weight, and serum biochemical markers. Lipid accumulation, histology, lipidomics, and sphingolipid metabolism-related enzyme expression were investigated to analyze liver function. Untargeted metabolomics was used to analyze the serum and the composition and abundance of intestinal flora. In addition, the correlation between differential liver lipid profiles, serum metabolites, and gut flora at the genus level was measured. The results show that LPJZ-658 significantly improves abnormal liver function and hepatic steatosis. The lipidomics analyses and metabolic pathway analysis identified sphingolipid, retinol, and glycerophospholipid metabolism as the most relevant metabolic pathways that characterized liver lipid dysregulation in the CuDS group. Consistently, RT-qPCR analyses revealed that the enzymes catalyzing sphingolipid metabolism that were significantly upregulated in the CuDS group were downregulated by the LPJZ-658 treatment. In addition, the serum metabolomics results indicated that the linoleic acid, taurine and hypotaurine, and ascorbate and aldarate metabolism pathways were associated with CuDS-induced MASLD. Notably, we found that treatment with LPJZ-658 partially reversed the changes in the differential serum metabolites. Finally, LPJZ-658 effectively regulated intestinal flora abnormalities and was significantly correlated with differential hepatic lipid species and serum metabolites. In conclusion, we elucidated the function and potential mechanisms of LPJZ-658 in alleviating copper deficiency combined with sugar-induced middle-aged MASLD and hope this will provide possible treatment strategies for improving MASLD.
Collapse
Affiliation(s)
- Chunhua Li
- College of Animal Science and Technology, Jilin Agricultural Science and Technology University, Jilin City 132101, China; (C.L.); (Z.L.); (C.C.); (L.Z.); (L.L.)
| | - Ziqi Liu
- College of Animal Science and Technology, Jilin Agricultural Science and Technology University, Jilin City 132101, China; (C.L.); (Z.L.); (C.C.); (L.Z.); (L.L.)
| | - Wei Wei
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China;
| | - Chen Chen
- College of Animal Science and Technology, Jilin Agricultural Science and Technology University, Jilin City 132101, China; (C.L.); (Z.L.); (C.C.); (L.Z.); (L.L.)
| | - Lichun Zhang
- College of Animal Science and Technology, Jilin Agricultural Science and Technology University, Jilin City 132101, China; (C.L.); (Z.L.); (C.C.); (L.Z.); (L.L.)
| | - Yang Wang
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun 130117, China;
| | - Bo Zhou
- Research Unit of Key Technologies for Prevention and Control of Virus Zoonoses, Changchun Veterinary Research Institute, Chinese Academy of Medical Sciences, Changchun 130122, China;
| | - Liming Liu
- College of Animal Science and Technology, Jilin Agricultural Science and Technology University, Jilin City 132101, China; (C.L.); (Z.L.); (C.C.); (L.Z.); (L.L.)
| | - Xiao Li
- Research Unit of Key Technologies for Prevention and Control of Virus Zoonoses, Changchun Veterinary Research Institute, Chinese Academy of Medical Sciences, Changchun 130122, China;
- Academician Workstation of Jilin Province, Changchun University of Chinese Medicine, Changchun 130117, China
| | - Cuiqing Zhao
- College of Animal Science and Technology, Jilin Agricultural Science and Technology University, Jilin City 132101, China; (C.L.); (Z.L.); (C.C.); (L.Z.); (L.L.)
| |
Collapse
|
5
|
Jiang X, Hu R, Huang Y, Xu Y, Zheng Z, Shi Y, Miao J, Liu Y. Fructose aggravates copper-deficiency-induced non-alcoholic fatty liver disease. J Nutr Biochem 2023; 119:109402. [PMID: 37311490 PMCID: PMC11186518 DOI: 10.1016/j.jnutbio.2023.109402] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 05/28/2023] [Accepted: 06/07/2023] [Indexed: 06/15/2023]
Abstract
Non-alcoholic fatty liver disease (NAFLD), is the most common cause of chronic liver disease, affecting 24% of the global population. Accumulating evidence demonstrates that copper deficiency (CuD) is implicated in the development of NAFLD, besides, high fructose consumption by promoting inflammation contributes to NAFLD. However, how CuD and/or fructose (Fru) causes NAFLD is not clearly delineated. The present study aims to investigate the role of CuD and/or fructose supplement on hepatic steatosis and hepatic injury. We established a CuD rat model by feeding weaning male Sprague-Dawley rats for 4 weeks with CuD diet. Fructose was supplemented in drinking water. We found the promoting role of CuD or Fructose (Fru) in the progress of NAFLD, which was aggravated by combination of the two. Furthermore, we presented the alteration of hepatic lipid profiles (including content, composition, and saturation), especially ceramide (Cer), cardiolipin (CL), phosphatidylcholine (PC) and phosphatidylethanolamine (PE) was closely associated with CuD and/or Fru fed induced-NAFLD in rat models. In conclusion, insufficient copper intake or excessive fructose supplement resulted in adverse effects on the hepatic lipid profile, and fructose supplement causes a further hepatic injury in CuD-induced NAFLD, which illuminated a better understanding of NAFLD.
Collapse
Affiliation(s)
- Xin Jiang
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, P.R. China
| | - Ruixiang Hu
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Jinan University, Guangzhou, P.R. China
| | - Yipu Huang
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, P.R. China
| | - Yi Xu
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, P.R. China
| | - Zhirui Zheng
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, P.R. China
| | - Yuansen Shi
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, P.R. China
| | - Ji Miao
- Division of Endocrinology, Boston Children's Hospital, Boston, Massachusetts, USA; Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA.
| | - Yun Liu
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, P.R. China.
| |
Collapse
|
6
|
Kerkadi A, Alkudsi DS, Hamad S, Alkeldi HM, Salih R, Agouni A. The Association between Zinc and Copper Circulating Levels and Cardiometabolic Risk Factors in Adults: A Study of Qatar Biobank Data. Nutrients 2021; 13:nu13082729. [PMID: 34444889 PMCID: PMC8398315 DOI: 10.3390/nu13082729] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 07/26/2021] [Accepted: 07/28/2021] [Indexed: 11/16/2022] Open
Abstract
Cardiometabolic risk (CMR) factors increase the likelihood of developing cardiovascular diseases (CVD). In Qatar, 24% of the total deaths are attributed to CVDs. Several nutritional disturbances have been linked to high risk of CVD. Many studies have discussed the effects of zinc (Zn) and copper (Cu) on CMR factors; however, evidence has been controversial. This investigated the association between CMR factors and the status of Zn and Cu, in addition to Zn/Cu ratio. A total of 575 Qatari men and women aged 18 years and older were obtained from Qatar Biobank. Plasma levels of Zn and Cu were determined using inductively coupled plasma mass spectrometry (ICP-MS). Anthropometric data and CMR factors were determined using standard methods. Adjusted associations between trace minerals and CMR were estimated by logistic regression. Partial correlation was performed to test the strength of the associations. Zn was not strongly correlated (p-value ˃ 0.01) or significantly associated with CMR factors and metabolic syndrome (MetS). Cu levels correlated positively with body mass index (BMI) (0.23; p ˂ 0.001), pulse rate (PR) (0.18; p ˂ 0.001), total cholesterol (0.13; p = 0.01), and high-density lipoproteins (HDL) (0.27; p ˂ 0.001); and negatively with diastolic blood pressure (DBP) (−0.13; p = 0.01). High plasma Cu significantly decreased the risk of metabolic syndrome (MetS) (0.121; p ˂ 0.001). Furthermore, Zn/Cu ratio positively correlated with waist circumference (0.13; p = 0.01), systolic blood pressure (0.13; p ˂ 0.01), and DBP (0.14; p ˂ 0.01); and negatively with BMI (−0.19; p ˂ 0.001), PR (−0.17; p ˂ 0.001), and HDL (−0.27; p ˂ 0.001). High Zn/Cu ratio increased the prevalence of low HDL (4.508; p ˂ 0.001) and MetS (5.570; p ˂ 0.01). These findings suggest that high plasma Cu levels are associated with a protective effect on DBP, HDL and MetS and that high plasma Zn/Cu ratio is associated with the risk of having low HDL and MetS.
Collapse
Affiliation(s)
- Abdelhamid Kerkadi
- Human Nutrition Department, College of Health Sciences, QU Health, Qatar University, Doha P.O. Box 2713, Qatar; (D.S.A.); (S.H.); (H.M.A.); (R.S.)
- Correspondence: ; Tel.: +974-4403-4806; Fax: +974-4403-4801
| | - Dana Samir Alkudsi
- Human Nutrition Department, College of Health Sciences, QU Health, Qatar University, Doha P.O. Box 2713, Qatar; (D.S.A.); (S.H.); (H.M.A.); (R.S.)
| | - Sara Hamad
- Human Nutrition Department, College of Health Sciences, QU Health, Qatar University, Doha P.O. Box 2713, Qatar; (D.S.A.); (S.H.); (H.M.A.); (R.S.)
| | - Hanan Mohamed Alkeldi
- Human Nutrition Department, College of Health Sciences, QU Health, Qatar University, Doha P.O. Box 2713, Qatar; (D.S.A.); (S.H.); (H.M.A.); (R.S.)
| | - Reem Salih
- Human Nutrition Department, College of Health Sciences, QU Health, Qatar University, Doha P.O. Box 2713, Qatar; (D.S.A.); (S.H.); (H.M.A.); (R.S.)
| | - Abdelali Agouni
- Pharmaceutical Sciences Department, College of Pharmacy, QU Health, Qatar University, Doha P.O. Box 2713, Qatar;
- Biomedical and Pharmaceutical Research Unit (BPRU), QU Health, Qatar University, Doha P.O. Box 2713, Qatar
| |
Collapse
|
7
|
Song M, Yuan F, Li X, Ma X, Yin X, Rouchka EC, Zhang X, Deng Z, Prough RA, McClain CJ. Analysis of sex differences in dietary copper-fructose interaction-induced alterations of gut microbial activity in relation to hepatic steatosis. Biol Sex Differ 2021; 12:3. [PMID: 33407877 PMCID: PMC7789350 DOI: 10.1186/s13293-020-00346-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 12/07/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Inadequate copper intake and increased fructose consumption represent two important nutritional problems in the USA. Dietary copper-fructose interactions alter gut microbial activity and contribute to the development of nonalcoholic fatty liver disease (NAFLD). The aim of this study is to determine whether dietary copper-fructose interactions alter gut microbial activity in a sex-differential manner and whether sex differences in gut microbial activity are associated with sex differences in hepatic steatosis. METHODS Male and female weanling Sprague-Dawley (SD) rats were fed ad libitum with an AIN-93G purified rodent diet with defined copper content for 8 weeks. The copper content is 6 mg/kg and 1.5 mg/kg in adequate copper diet (CuA) and marginal copper diet (CuM), respectively. Animals had free access to either deionized water or deionized water containing 10% fructose (F) (w/v) as the only drink during the experiment. Body weight, calorie intake, plasma alanine aminotransferase, aspartate aminotransferase, and liver histology as well as liver triglyceride were evaluated. Fecal microbial contents were analyzed by 16S ribosomal RNA (16S rRNA) sequencing. Fecal and cecal short-chain fatty acids (SCFAs) were determined by gas chromatography-mass spectrometry (GC-MS). RESULTS Male and female rats exhibit similar trends of changes in the body weight gain and calorie intake in response to dietary copper and fructose, with a generally higher level in male rats. Several female rats in the CuAF group developed mild steatosis, while no obvious steatosis was observed in male rats fed with CuAF or CuMF diets. Fecal 16S rRNA sequencing analysis revealed distinct alterations of the gut microbiome in male and female rats. Linear discriminant analysis (LDA) effect size (LEfSe) identified sex-specific abundant taxa in different groups. Further, total SCFAs, as well as, butyrate were decreased in a more pronounced manner in female CuMF rats than in male rats. Of note, the decreased SCFAs are concomitant with the reduced SCFA producers, but not correlated to hepatic steatosis. CONCLUSIONS Our data demonstrated sex differences in the alterations of gut microbial abundance, activities, and hepatic steatosis in response to dietary copper-fructose interaction in rats. The correlation between sex differences in metabolic phenotypes and alterations of gut microbial activities remains elusive.
Collapse
Affiliation(s)
- Ming Song
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, University of Louisville School of Medicine, Louisville, KY 40202 USA
- Hepatobiology&Toxicology Program, University of Louisville, Louisville, KY 40202 USA
| | - Fang Yuan
- Hepatobiology&Toxicology Program, University of Louisville, Louisville, KY 40202 USA
- University of Louisville Alcohol Research Center, University of Louisville, Louisville, KY 40202 USA
- Department of Chemistry, University of Louisville, Louisville, KY 40208 USA
- Center for Regulatory and Environmental Analytical Metabolomics, University of Louisville, Louisville, KY 40208 USA
| | - Xiaohong Li
- KBRIN Bioinformatics Core, Louisville, KY 40292 USA
| | - Xipeng Ma
- Hepatobiology&Toxicology Program, University of Louisville, Louisville, KY 40202 USA
- University of Louisville Alcohol Research Center, University of Louisville, Louisville, KY 40202 USA
- Department of Chemistry, University of Louisville, Louisville, KY 40208 USA
- Center for Regulatory and Environmental Analytical Metabolomics, University of Louisville, Louisville, KY 40208 USA
| | - Xinmin Yin
- Hepatobiology&Toxicology Program, University of Louisville, Louisville, KY 40202 USA
- University of Louisville Alcohol Research Center, University of Louisville, Louisville, KY 40202 USA
- Department of Chemistry, University of Louisville, Louisville, KY 40208 USA
- Center for Regulatory and Environmental Analytical Metabolomics, University of Louisville, Louisville, KY 40208 USA
| | | | - Xiang Zhang
- Hepatobiology&Toxicology Program, University of Louisville, Louisville, KY 40202 USA
- University of Louisville Alcohol Research Center, University of Louisville, Louisville, KY 40202 USA
- Department of Chemistry, University of Louisville, Louisville, KY 40208 USA
- Center for Regulatory and Environmental Analytical Metabolomics, University of Louisville, Louisville, KY 40208 USA
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY 40202 USA
| | - Zhongbin Deng
- Hepatobiology&Toxicology Program, University of Louisville, Louisville, KY 40202 USA
- Department of Microbiology & Immunology, Brown Cancer Center, University of Louisville, Louisville, KY 40202 USA
- James Graham Brown Cancer Center, University of Louisville, Louisville, KY 40202 USA
| | - Russell A. Prough
- Hepatobiology&Toxicology Program, University of Louisville, Louisville, KY 40202 USA
- Department of Biochemistry and Molecular Genetics, University of Louisville School of Medicine, Louisville, KY 40202 USA
| | - Craig J. McClain
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, University of Louisville School of Medicine, Louisville, KY 40202 USA
- Hepatobiology&Toxicology Program, University of Louisville, Louisville, KY 40202 USA
- University of Louisville Alcohol Research Center, University of Louisville, Louisville, KY 40202 USA
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY 40202 USA
- Robley Rex Veterans Affairs Medical Center, Louisville, KY 40206 USA
| |
Collapse
|
8
|
Klevay LM. Cholesterotropic and cuprotropic chemicals. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2020; 100:4057. [PMID: 32323867 DOI: 10.1002/jsfa.10426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
A dozen or so chemicals modify both cholesterol and copper metabolism. Ascorbic acid and cadmium, etc., inhibit copper metabolism and raise cholesterol. Calcium and clofibrate, etc., enhance copper and lower cholesterol. Perhaps the doses of dietary cholesterol and fructose in this experiment were too severe to permit fenofibrate to lower cholesterol in a manner similar to clofibrate. © 2020 Society of Chemical Industry.
Collapse
Affiliation(s)
- Leslie M Klevay
- School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND, USA
| |
Collapse
|
9
|
Abstract
Copper accumulation and deficiency are reciprocally connected to lipid metabolism. In Wilson disease (WD), which is caused by a genetic loss of function of the copper-transporting P-type ATPase beta, copper accumulates mainly in the liver and lipid metabolism is dysregulated. The underlying mechanisms linking copper and lipid metabolism in WD are not clear. Copper may impair metabolic machinery by direct binding to protein and lipid structures or by generating reactive oxygen species with consequent damage to cellular organelles vital to energy metabolism. In the liver, copper overload results in mitochondrial impairment, down-regulation of lipid metabolism, and the development of steatosis with an etiology not fully elucidated. Little is known regarding the effect of copper overload on extrahepatic energy homeostasis. This review aims to discuss alterations in hepatic energy metabolism associated with WD, highlights potential mechanisms involved in the development of hepatic and systemic dysregulation of lipid metabolism, and reviews current knowledge on the effects of copper overload on extrahepatic energy metabolism.
Collapse
Affiliation(s)
- Tagreed A. Mazi
- Department of Nutrition, University of California Davis, Davis, CA, USA,Department of Community Health Sciences, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Noreene M. Shibata
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, University of California Davis, Sacramento, CA, USA
| | - Valentina Medici
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, University of California Davis, Sacramento, CA, USA,Corresponding author. (V. Medici)
| |
Collapse
|
10
|
Wooton-Kee CR, Robertson M, Zhou Y, Dong B, Sun Z, Kim KH, Liu H, Xu Y, Putluri N, Saha P, Coarfa C, Moore DD, Nuotio-Antar AM. Metabolic dysregulation in the Atp7b-/- Wilson's disease mouse model. Proc Natl Acad Sci U S A 2020; 117:2076-2083. [PMID: 31924743 PMCID: PMC6994990 DOI: 10.1073/pnas.1914267117] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Inactivating mutations in the copper transporter Atp7b result in Wilson's disease. The Atp7b-/- mouse develops hallmarks of Wilson's disease. The activity of several nuclear receptors decreased in Atp7b-/- mice, and nuclear receptors are critical for maintaining metabolic homeostasis. Therefore, we anticipated that Atp7b-/- mice would exhibit altered progression of diet-induced obesity, fatty liver, and insulin resistance. Following 10 wk on a chow or Western-type diet (40% kcal fat), parameters of glucose and lipid homeostasis were measured. Hepatic metabolites were measured by liquid chromatography-mass spectrometry and correlated with transcriptomic data. Atp7b-/- mice fed a chow diet presented with blunted body-weight gain over time, had lower fat mass, and were more glucose tolerant than wild type (WT) littermate controls. On the Western diet, Atp7b-/- mice exhibited reduced body weight, adiposity, and hepatic steatosis compared with WT controls. Atp7b-/- mice fed either diet were more insulin sensitive than WT controls; however, fasted Atp7b-/- mice exhibited hypoglycemia after administration of insulin due to an impaired glucose counterregulatory response, as evidenced by reduced hepatic glucose production. Coupling gene expression with metabolomic analyses, we observed striking changes in hepatic metabolic profiles in Atp7b-/- mice, including increases in glycolytic intermediates and components of the tricarboxylic acid cycle. In addition, the active phosphorylated form of AMP kinase was significantly increased in Atp7b-/- mice relative to WT controls. Alterations in hepatic metabolic profiles and nuclear receptor signaling were associated with improved glucose tolerance and insulin sensitivity as well as with impaired fasting glucose production in Atp7b-/- mice.
Collapse
Affiliation(s)
- Clavia Ruth Wooton-Kee
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030;
| | - Matthew Robertson
- Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030
| | - Ying Zhou
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030
- Integrative Molecular and Biomedical Sciences Graduate Program, Baylor College of Medicine, Houston, TX 77030
| | - Bingning Dong
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030
| | - Zhen Sun
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030
| | - Kang Ho Kim
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030
| | - Hailan Liu
- Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030
| | - Yong Xu
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030
- Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030
| | - Nagireddy Putluri
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030
| | - Pradip Saha
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030
| | - Cristian Coarfa
- Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030
| | - David D Moore
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030;
- Integrative Molecular and Biomedical Sciences Graduate Program, Baylor College of Medicine, Houston, TX 77030
| | | |
Collapse
|
11
|
Morrell A, Tripet BP, Eilers BJ, Tegman M, Thompson D, Copié V, Burkhead JL. Copper modulates sex-specific fructose hepatoxicity in nonalcoholic fatty liver disease (NALFD) Wistar rat models. J Nutr Biochem 2019; 78:108316. [PMID: 31986483 DOI: 10.1016/j.jnutbio.2019.108316] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 11/07/2019] [Accepted: 12/06/2019] [Indexed: 02/08/2023]
Abstract
This study aimed to characterize the impact of dietary copper on the biochemical and hepatic metabolite changes associated with fructose toxicity in a Wistar rat model of fructose-induced liver disease. Twenty-four male and 24 female, 6-week-old, Wister rats were separated into four experimental dietary treatment groups (6 males and 6 females per group), as follows: (1) a control diet: containing no fructose with adequate copper (i.e., CuA/0% Fruct); (2) a diet regimen identical to the control and supplemented with 30% w/v fructose in the animals' drinking water (CuA/30% Fruct); (3) a diet identical to the control diet but deficient in copper content (CuD/0% Fruct) and (4) a diet identical to the control diet but deficient in copper content and supplemented with 30% w/v fructose in the drinking water (CuD/30% Fruct). The animals were fed the four diet regimens for 5 weeks, followed by euthanization and assessment of histology, elemental profiles and identification and quantitation of liver metabolites. Results from 1H nuclear magnetic resonance metabolomics revealed mechanistic insights into copper modulation of fructose hepatotoxicity through identification of distinct metabolic phenotypes that were highly correlated with diet and sex. This study also identified previously unknown sex-specific responses to both fructose supplementation and restricted copper intake, while the presence of adequate dietary copper promoted most pronounced fructose-induced metabolite changes.
Collapse
Affiliation(s)
- Austin Morrell
- Department of Biological Sciences, University of Alaska Anchorage, Anchorage, AK
| | - Brian P Tripet
- Department of Chemistry and Biochemistry, Montana State University, Bozeman, MT
| | - Brian J Eilers
- Department of Chemistry and Biochemistry, Montana State University, Bozeman, MT
| | - Megan Tegman
- Department of Chemistry and Biochemistry, Montana State University, Bozeman, MT
| | - Damon Thompson
- Department of Biological Sciences, University of Alaska Anchorage, Anchorage, AK
| | - Valérie Copié
- Department of Chemistry and Biochemistry, Montana State University, Bozeman, MT.
| | - Jason L Burkhead
- Department of Biological Sciences, University of Alaska Anchorage, Anchorage, AK.
| |
Collapse
|
12
|
Rodriguez-Ramiro I, Dell'Aquila C, Ward J, Neal A, Bruggraber S, Shewry P, Fairweather-Tait S. Estimation of the iron bioavailability in green vegetables using an in vitro digestion/Caco-2 cell model. Food Chem 2019; 301:125292. [DOI: 10.1016/j.foodchem.2019.125292] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 07/27/2019] [Accepted: 07/28/2019] [Indexed: 02/02/2023]
|
13
|
Léveillé M, Estall JL. Mitochondrial Dysfunction in the Transition from NASH to HCC. Metabolites 2019; 9:E233. [PMID: 31623280 PMCID: PMC6836234 DOI: 10.3390/metabo9100233] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 09/26/2019] [Accepted: 10/11/2019] [Indexed: 02/06/2023] Open
Abstract
The liver constantly adapts to meet energy requirements of the whole body. Despite its remarkable adaptative capacity, prolonged exposure of liver cells to harmful environmental cues (such as diets rich in fat, sugar, and cholesterol) results in the development of chronic liver diseases (including non-alcoholic fatty liver disease (NAFLD) and non-alcoholic steatohepatitis (NASH)) that can progress to hepatocellular carcinoma (HCC). The pathogenesis of these diseases is extremely complex, multifactorial, and poorly understood. Emerging evidence suggests that mitochondrial dysfunction or maladaptation contributes to detrimental effects on hepatocyte bioenergetics, reactive oxygen species (ROS) homeostasis, endoplasmic reticulum (ER) stress, inflammation, and cell death leading to NASH and HCC. The present review highlights the potential contribution of altered mitochondria function to NASH-related HCC and discusses how agents targeting this organelle could provide interesting treatment strategies for these diseases.
Collapse
Affiliation(s)
- Mélissa Léveillé
- Institut de Recherches Cliniques de Montréal (IRCM), Montreal, Quebec, QC H2W 1R7, Canada.
- Faculty of Medicine, University of Montreal, Montreal, Quebec, QC H3G 2M1, Canada.
| | - Jennifer L Estall
- Institut de Recherches Cliniques de Montréal (IRCM), Montreal, Quebec, QC H2W 1R7, Canada.
- Faculty of Medicine, University of Montreal, Montreal, Quebec, QC H3G 2M1, Canada.
- Division of Experimental Medicine, McGill University, Montreal, Quebec, QC H4A 3J1, Canada.
| |
Collapse
|
14
|
Yang H, Liu CN, Wolf RM, Ralle M, Dev S, Pierson H, Askin F, Steele KE, Magnuson TH, Schweitzer MA, Wong GW, Lutsenko S. Obesity is associated with copper elevation in serum and tissues. Metallomics 2019; 11:1363-1371. [PMID: 31249997 DOI: 10.1039/c9mt00148d] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Copper misbalance has been linked to fat accumulation in animals and experimental systems; however, information about copper homeostasis in human obesity is limited. In this study, the copper status of obese individuals was evaluated by measuring their levels of copper and cuproproteins in serum, adipose and hepatic tissues. The analysis of serum trace elements showed significant positive and element-specific correlation between copper and BMI after controlling for gender, age, and ethnicity. Serum copper also positively correlated with leptin, insulin, and the leptin/BMI ratio. When compared to lean controls, obese patients had elevated circulating cuproproteins, such as semucarbazide-sensitive amine oxidase (SSAO) and ceruloplasmin, and higher SSAO activity and copper levels in visceral fat. Although hepatic steatosis reduces copper levels in the liver, obese patients with no or mild steatosis have higher copper content in the liver compared to lean controls. In conclusion, obese patients evaluated in this study had altered copper status. Strong positive correlations of copper levels with BMI and leptin suggest that copper and/or cuproproteins may be functionally linked to fat accumulation.
Collapse
Affiliation(s)
- Haojun Yang
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Chin-Nung Liu
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Risa M Wolf
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA and Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Martina Ralle
- Department of Genetics, Oregon Health & Science University, Portland, Oregon, USA
| | - Som Dev
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Hannah Pierson
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Frederic Askin
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Kimberley E Steele
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Thomas H Magnuson
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Michael A Schweitzer
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - G William Wong
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA. and Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Svetlana Lutsenko
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
15
|
Abstract
Many metals have biological functions and play important roles in human health. Copper (Cu) is an essential metal that supports normal cellular physiology. Significant research efforts have focused on identifying the molecules and pathways involved in dietary Cu uptake in the digestive tract. The lack of an adequate in vitro model for assessing Cu transport processes in the gut has led to contradictory data and gaps in our understanding of the mechanisms involved in dietary Cu acquisition. The recent development of organoid technology has provided a tractable model system for assessing the detailed mechanistic processes involved in Cu utilization and transport in the context of nutrition. Enteroid (intestinal epithelial organoid)-based studies have identified new links between intestinal Cu metabolism and dietary fat processing. Evidence for a metabolic coupling between the dietary uptake of Cu and uptake of fat (which were previously thought to be independent) is a new and exciting finding that highlights the utility of these three-dimensional primary culture systems. This review has three goals: (a) to critically discuss the roles of key Cu transport enzymes in dietary Cu uptake; (b) to assess the use, utility, and limitations of organoid technology in research into nutritional Cu transport and Cu-based diseases; and (c) to highlight emerging connections between nutritional Cu homeostasis and fat metabolism.
Collapse
Affiliation(s)
- Hannah Pierson
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA; ,
| | - Haojun Yang
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA; ,
| | - Svetlana Lutsenko
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA; ,
| |
Collapse
|
16
|
Song M, Vos MB, McClain CJ. Copper-Fructose Interactions: A Novel Mechanism in the Pathogenesis of NAFLD. Nutrients 2018; 10:E1815. [PMID: 30469339 PMCID: PMC6266129 DOI: 10.3390/nu10111815] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Revised: 11/08/2018] [Accepted: 11/16/2018] [Indexed: 12/13/2022] Open
Abstract
Compelling epidemiologic data support the critical role of dietary fructose in the epidemic of obesity, metabolic syndrome and nonalcoholic fatty liver disease (NAFLD). The metabolic effects of fructose on the development of metabolic syndrome and NAFLD are not completely understood. High fructose intake impairs copper status, and copper-fructose interactions have been well documented in rats. Altered copper-fructose metabolism leads to exacerbated experimental metabolic syndrome and NAFLD. A growing body of evidence has demonstrated that copper levels are low in NAFLD patients. Moreover, hepatic and serum copper levels are inversely correlated with the severity of NAFLD. Thus, high fructose consumption and low copper availability are considered two important risk factors in NAFLD. However, the causal effect of copper-fructose interactions as well as the effects of fructose intake on copper status remain to be evaluated in humans. The aim of this review is to summarize the role of copper-fructose interactions in the pathogenesis of the metabolic syndrome and discuss the potential underlying mechanisms. This review will shed light on the role of copper homeostasis and high fructose intake and point to copper-fructose interactions as novel mechanisms in the fructose induced NAFLD.
Collapse
Affiliation(s)
- Ming Song
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, University of Louisville School of Medicine, Louisville, KY 40202, USA.
- Hepatobiology&Toxicology Center, University of Louisville School of Medicine, Louisville, KY 40202, USA.
| | - Miriam B Vos
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30307, USA.
- Children's Healthcare of Atlanta, Atlanta, GA 30322, USA.
| | - Craig J McClain
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, University of Louisville School of Medicine, Louisville, KY 40202, USA.
- Hepatobiology&Toxicology Center, University of Louisville School of Medicine, Louisville, KY 40202, USA.
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY 40202, USA.
- University of Louisville Alcohol Research Center, University of Louisville School of Medicine, Louisville, KY 40202, USA.
- Robley Rex Veterans Affairs Medical Center, Louisville, KY 40206, USA.
| |
Collapse
|
17
|
DiNicolantonio JJ, Mangan D, O’Keefe JH. The fructose–copper connection: Added sugars induce fatty liver and insulin resistance via copper deficiency. JOURNAL OF INSULIN RESISTANCE 2018. [DOI: 10.4102/jir.v3i1.43] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
|
18
|
Lembede BW, Erlwanger KH, Nkomozepi P, Chivandi E. Terminalia Sericea aqueous leaf extract protects growing wistar rats against fructose-induced fatty liver disease. JOURNAL OF COMPLEMENTARY & INTEGRATIVE MEDICINE 2018; 16:jcim-2018-0035. [PMID: 29927747 DOI: 10.1515/jcim-2018-0035] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 05/23/2018] [Indexed: 12/16/2022]
Abstract
Background Terminalia sericea (T. sericea) is traditionally used to treat stomach ailments, infections, hypertension and diabetes mellitus. Previous in vitro studies have reported that T. sericea has lipolytic properties. This study interrogated the effects of T. sericea on linear growth, development of fatty liver disease, viscera morphometry and health of growing rats fed a 12% fructose solution (FS). Methods Thirty 21-day old male Wistar rat pups were randomly allocated to five treatments: group I - plain gelatine cubes (PGC) + plain tap water (PW), group II - 12% FS + PGC, group III - gelatine cubes containing fenofibrate (Feno) at a dose of 100 mg/kg body + FS, group IV - gelatine cubes containing the low dose (100 mg/kg body mass per day) of the T. sericea extract (TsL) + FS, group V - gelatine cubes containing the high dose (400 mg/kg body mass per day) of the T. sericea extract (TsH) + FS. Following 12 weeks of feeding, the rats were fasted overnight, euthanized and plasma and viscera harvested for analysis. Results Consumption of fructose resulted in significantly increased (p<0.05) liver lipid content and caused macrovesicular steatosis. The T. sericea extracts at 400 mg/kg per day suppressed the fructose-induced liver lipid accumulation and macrovesicular steatosis similarly to 100 mg/kg per day of Feno. Conclusions These findings suggest that the aqueous T. sericea leaf extract at 400 mg/kg per day could potentially protect against fructose-induced lipid accumulation as well as macrovesicular steatosis.
Collapse
Affiliation(s)
- Busisani W Lembede
- School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown, Johannesburg2193, Republic of South Africa
| | - Kennedy H Erlwanger
- School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown, Johannesburg2193, Republic of South Africa
| | - Pilani Nkomozepi
- Department of Human Anatomy and Physiology, Faculty of Health Sciences, University of Johannesburg, Doornfontein, Johannesburg, Republic of South Africa
| | - Eliton Chivandi
- School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown, Johannesburg2193, Republic of South Africa
| |
Collapse
|
19
|
Song M, Li X, Zhang X, Shi H, Vos MB, Wei X, Wang Y, Gao H, Rouchka EC, Yin X, Zhou Z, Prough RA, Cave MC, McClain CJ. Dietary copper-fructose interactions alter gut microbial activity in male rats. Am J Physiol Gastrointest Liver Physiol 2018; 314:G119-G130. [PMID: 29025734 PMCID: PMC5866377 DOI: 10.1152/ajpgi.00378.2016] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Revised: 09/18/2017] [Accepted: 09/20/2017] [Indexed: 02/08/2023]
Abstract
Dietary copper-fructose interactions contribute to the development of nonalcoholic fatty liver disease (NAFLD). Gut microbiota play critical roles in the pathogenesis of NAFLD. The aim of this study was to determine the effect of different dietary doses of copper and their interactions with high fructose on gut microbiome. Male weanling Sprague-Dawley rats were fed diets with adequate copper (6 ppm CuA), marginal copper (1.5 ppm CuM) (low copper), or supplemented copper (20 ppm CuS) (high copper) for 4 wk. Deionized water or deionized water containing 30% fructose (wt/vol) was given ad libitum. Copper status, liver enzymes, gut barrier function, and gut microbiome were evaluated. Both low- and high-copper diets led to liver injury in high-fructose-fed rats, and this was associated with gut barrier dysfunction, as shown by the markedly decreased tight junction proteins and increased gut permeability. 16S rDNA sequencing analysis revealed distinct alterations of the gut microbiome associated with dietary low- and high-copper/high-fructose feeding. The common features of the alterations of the gut microbiome were the increased abundance of Firmicutes and the depletion of Akkermansia. However, they differed mainly within the phylum Firmicutes. Our data demonstrated that a complex interplay among host, microbes, and dietary copper-fructose interaction regulates gut microbial metabolic activity, which may contribute to the development of liver injury and hepatic steatosis. The distinct alterations of gut microbial activity, which were associated with the different dietary doses of copper and fructose, imply that separate mechanism(s) may be involved. NEW & NOTEWORTHY First, dietary low- and high-copper/high-fructose-induced liver injury are associated with distinct alterations of gut microbiome. Second, dietary copper level plays a critical role in maintaining the gut barrier integrity, likely by acting on the intestinal tight junction proteins and the protective commensal bacteria Akkermansia. Third, the alterations of gut microbiome induced by dietary low and high copper with or without fructose differ mainly within the phylum Firmicutes.
Collapse
Affiliation(s)
- Ming Song
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, University of Louisville School of Medicine , Louisville, Kentucky
- Hepatobiology and Toxicology Center, University of Louisville School of Medicine , Louisville, Kentucky
| | - Xiaohong Li
- Bioinformatics Core. University of Louisville School of Medicine , Louisville, Kentucky
| | - Xiang Zhang
- Department of Chemistry, University of Louisville School of Medicine , Louisville, Kentucky
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine , Louisville, Kentucky
- Hepatobiology and Toxicology Center, University of Louisville School of Medicine , Louisville, Kentucky
- University of Louisville Alcohol Research Center, University of Louisville School of Medicine , Louisville, Kentucky
- Center for Regulatory and Environmental Analytical Metabolomics, University of Louisville School of Medicine , Louisville, Kentucky
| | - Hongxue Shi
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine , Louisville, Kentucky
| | - Miriam B Vos
- Department of Pediatrics, Emory University School of Medicine, and Children's Healthcare of Atlanta, Atlanta, Georgia
| | - Xiaoli Wei
- Department of Chemistry, University of Louisville School of Medicine , Louisville, Kentucky
- Center for Regulatory and Environmental Analytical Metabolomics, University of Louisville School of Medicine , Louisville, Kentucky
| | - Yuhua Wang
- College of Food Science and Engineering, Jilin Agricultural University , Changchun , China
| | - Hong Gao
- Genomics Facility, University of Louisville School of Medicine , Louisville, Kentucky
| | - Eric C Rouchka
- Bioinformatics Core. University of Louisville School of Medicine , Louisville, Kentucky
| | - Xinmin Yin
- Department of Chemistry, University of Louisville School of Medicine , Louisville, Kentucky
- Center for Regulatory and Environmental Analytical Metabolomics, University of Louisville School of Medicine , Louisville, Kentucky
| | - Zhanxiang Zhou
- Center for Translational Biomedical Research, University of North Carolina at Greensboro, Kannapolis, North Carolina
- Department of Nutrition, University of North Carolina at Greensboro, Kannapolis, North Carolina
| | - Russell A Prough
- Department of Biochemistry and Molecular Genetics, University of Louisville School of Medicine , Louisville, Kentucky
| | - Matthew C Cave
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, University of Louisville School of Medicine , Louisville, Kentucky
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine , Louisville, Kentucky
- Hepatobiology and Toxicology Center, University of Louisville School of Medicine , Louisville, Kentucky
- University of Louisville Alcohol Research Center, University of Louisville School of Medicine , Louisville, Kentucky
- Department of Biochemistry and Molecular Genetics, University of Louisville School of Medicine , Louisville, Kentucky
- Robley Rex Louisville Veterans Afairs Medical Center , Louisville, Kentucky
| | - Craig J McClain
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, University of Louisville School of Medicine , Louisville, Kentucky
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine , Louisville, Kentucky
- Hepatobiology and Toxicology Center, University of Louisville School of Medicine , Louisville, Kentucky
- University of Louisville Alcohol Research Center, University of Louisville School of Medicine , Louisville, Kentucky
- Robley Rex Louisville Veterans Afairs Medical Center , Louisville, Kentucky
| |
Collapse
|
20
|
Abstract
OBJECTIVE Animal models and studies in adults have demonstrated that copper restriction increases severity of liver injury in nonalcoholic fatty liver disease (NAFLD). This has not been studied in children. We aimed to determine if lower tissue copper is associated with increased NAFLD severity in children. METHODS This was a retrospective study of pediatric patients who had a liver biopsy including a hepatic copper quantitation. The primary outcome compared hepatic copper concentration in NAFLD versus non-NAFLD. Secondary outcomes compared hepatic copper levels against steatosis, fibrosis, lobular inflammation, balloon degeneration, and NAFLD activity score (NAS). RESULTS The study analysis included 150 pediatric subjects (102 with NAFLD and 48 non-NAFLD). After adjusting for age, body mass index z score, gamma glutamyl transferase, alanine aminotransferase, and total bilirubin, NAFLD subjects had lower levels of hepatic copper than non-NAFLD (P = 0.005). In addition, tissue copper concentration decreased as steatosis severity increased (P < 0.001). Copper levels were not associated with degree of fibrosis, lobular inflammation, portal inflammation, or balloon degeneration. CONCLUSIONS In this cohort of pediatric subjects with NAFLD, we observed decreased tissue copper levels in subjects with NAFLD when compared with non-NAFLD subjects. In addition, tissue copper levels were lower in subjects with nonalcoholic steatohepatitis, a more severe form of the disease, when compared with steatosis alone. Further studies are needed to explore the relationship between copper levels and NAFLD progression.
Collapse
|
21
|
Morrell A, Tallino S, Yu L, Burkhead JL. The role of insufficient copper in lipid synthesis and fatty-liver disease. IUBMB Life 2017; 69:263-270. [PMID: 28271632 PMCID: PMC5619695 DOI: 10.1002/iub.1613] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Accepted: 02/08/2017] [Indexed: 12/12/2022]
Abstract
The essential transition metal copper is important in lipid metabolism, redox balance, iron mobilization, and many other critical processes in eukaryotic organisms. Genetic diseases where copper homeostasis is disrupted, including Menkes disease and Wilson disease, indicate the importance of copper balance to human health. The severe consequences of insufficient copper supply are illustrated by Menkes disease, caused by mutation in the X-linked ATP7A gene encoding a protein that transports copper from intestinal epithelia into the bloodstream and across the blood-brain barrier. Inadequate copper supply to the body due to poor diet quality or malabsorption can disrupt several molecular level pathways and processes. Though much of the copper distribution machinery has been described and consequences of disrupted copper handling have been characterized in human disease as well as animal models, physiological consequences of sub-optimal copper due to poor nutrition or malabsorption have not been extensively studied. Recent work indicates that insufficient copper may be important in a number of common diseases including obesity, ischemic heart disease, and metabolic syndrome. Specifically, marginal copper deficiency (CuD) has been reported as a potential etiologic factor in diseases characterized by disrupted lipid metabolism such as non-alcoholic fatty-liver disease (NAFLD). In this review, we discuss the available data suggesting that a significant portion of the North American population may consume insufficient copper, the potential mechanisms by which CuD may promote lipid biosynthesis, and the interaction between CuD and dietary fructose in the etiology of NAFLD. © 2016 IUBMB Life, 69(4):263-270, 2017.
Collapse
Affiliation(s)
- Austin Morrell
- University of Alaska Anchorage, Department of Biological Sciences Anchorage, Alaska
| | - Savannah Tallino
- University of Alaska Anchorage, Department of Biological Sciences Anchorage, Alaska
| | - Lei Yu
- University of Washington School of Medicine, Seattle, Washington
| | - Jason L. Burkhead
- University of Alaska Anchorage, Department of Biological Sciences Anchorage, Alaska
| |
Collapse
|
22
|
Wei X, Song M, Yin X, Schuschke DA, Koo I, McClain CJ, Zhang X. Effects of Dietary Different Doses of Copper and High Fructose Feeding on Rat Fecal Metabolome. J Proteome Res 2015. [PMID: 26216400 DOI: 10.1021/acs.jproteome.5b00596] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The gut microbiota plays a critical role in the pathogenesis of nonalcoholic fatty liver disease (NAFLD). Increased fructose consumption and inadequate copper intake are two critical risk factors in the development of NAFLD. To gain insight into the role of gut microbiota, fecal metabolites, obtained from rats exposed to different dietary levels of copper with and without high fructose intake for 4 weeks, were analyzed by comprehensive two-dimensional gas chromatography time-of-flight mass spectrometry (GC × GC-TOF MS). In parallel, liver tissues were assessed by histology and triglyceride assay. Our data showed that high fructose feeding led to obvious hepatic steatosis in both marginal copper deficient rats and copper supplementation rats. Among the 38 metabolites detected with significant abundance alteration between groups, short chain fatty acids were markedly decreased with excessive fructose intake irrespective of copper levels. C15:0 and C17:0 long chain fatty acids, produced only by bacteria, were increased by either high copper level or high fructose intake. In addition, increased fecal urea and malic acid paralleled the increased hepatic fat accumulation. Collectively, GC × GC-TOF MS analysis of rat fecal samples revealed distinct fecal metabolome profiles associated with the dietary high fructose and copper level, with some metabolites possibly serving as potential noninvasive biomarkers of fructose induced-NAFLD.
Collapse
Affiliation(s)
| | | | | | | | | | - Craig J McClain
- Robley Rex Louisville VAMC, Louisville, Kentucky 40292, United States
| | | |
Collapse
|
23
|
Stättermayer AF, Traussnigg S, Dienes HP, Aigner E, Stauber R, Lackner K, Hofer H, Stift J, Wrba F, Stadlmayr A, Datz C, Strasser M, Maieron A, Trauner M, Ferenci P. Hepatic steatosis in Wilson disease--Role of copper and PNPLA3 mutations. J Hepatol 2015; 63:156-163. [PMID: 25678388 DOI: 10.1016/j.jhep.2015.01.034] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2014] [Revised: 01/18/2015] [Accepted: 01/21/2015] [Indexed: 12/13/2022]
Abstract
BACKGROUND & AIMS The earliest characteristic alterations of the liver pathology in Wilson disease (WD) include steatosis, which is sometimes indistinguishable from non-alcoholic fatty liver disease (NAFLD). Steatosis in WD may reflect copper-induced mitochondrial dysfunction. A genetic polymorphism in rs738409, in the patatin-like phospholipase domain-containing 3 gene (PNPLA3), is strongly associated with appearance of in NAFLD. This study evaluated the role of PNPLA3 and hepatic copper content for development of steatosis in patients with WD. METHODS Liver biopsies obtained at diagnosis and the PNPLA3 genotype were analyzed in 98 Caucasian patients with WD (male: 52 [53.1%]; mean age: 27.6 years [CI 95%: 24.8-30.4, range: 5.8-61.5]). Steatosis was graded as percentage of lipid containing hepatocytes by an expert hepatopathologist unaware of the results of genetic testing. RESULTS Moderate/severe steatosis (>33% of hepatocytes) was observed in 28 patients (pediatric: n=13/26 [50.0%], adult: n=15/72 [20.8%]; p=0.01). Forty-six patients (46.9%; pediatric: n=7, adult: n=39; p=0.022) had cirrhosis. Multivariate logistic regression identified PNPLA3 G allele (OR: 2.469, CI 95%: 1.203-5.068; p=0.014) and pediatric age (OR: 4.348; 1.577-11.905; p=0.004) as independent variables associated with moderate/severe steatosis. In contrast, hepatic copper content did not impact on moderate/severe steatosis (OR: 1.000, CI 95%: 1.000-1.001; p=0.297). CONCLUSIONS Steatosis is common in WD and the PNPLA3 G allele contributes to its pathogenesis. The role of hepatic copper concentration and ATP7B mutations in steatosis development deserve further investigations.
Collapse
Affiliation(s)
- Albert Friedrich Stättermayer
- Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Stefan Traussnigg
- Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Hans-Peter Dienes
- Institute for Clinical Pathology, Medical University of Vienna, Vienna, Austria
| | - Elmar Aigner
- Department of Internal Medicine I, Paracelsus Private Medical University, Salzburg, Austria
| | - Rudolf Stauber
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Karoline Lackner
- Institute of Pathology, Medical University of Graz, Graz, Austria
| | - Harald Hofer
- Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Judith Stift
- Institute for Clinical Pathology, Medical University of Vienna, Vienna, Austria
| | - Friedrich Wrba
- Institute for Clinical Pathology, Medical University of Vienna, Vienna, Austria
| | | | - Christian Datz
- Department of Internal Medicine, KH Oberndorf, Oberndorf, Austria
| | - Michael Strasser
- Department of Internal Medicine I, Paracelsus Private Medical University, Salzburg, Austria
| | - Andreas Maieron
- Department of Internal Medicine IV, KH der Elisabethinen, Linz, Austria
| | - Michael Trauner
- Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Peter Ferenci
- Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
24
|
Song M, Schuschke DA, Zhou Z, Zhong W, Zhang J, Zhang X, Wang Y, McClain CJ. Kupffer cell depletion protects against the steatosis, but not the liver damage, induced by marginal-copper, high-fructose diet in male rats. Am J Physiol Gastrointest Liver Physiol 2015; 308:G934-45. [PMID: 25813056 PMCID: PMC4451322 DOI: 10.1152/ajpgi.00285.2014] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2014] [Accepted: 03/24/2015] [Indexed: 01/31/2023]
Abstract
High-fructose feeding impairs copper status and leads to low copper availability, which is a novel mechanism in obesity-related fatty liver. Copper deficiency-associated hepatic iron overload likely plays an important role in fructose-induced liver injury. Excess iron in the liver is distributed throughout hepatocytes and Kupffer cells (KCs). The aim of this study was to examine the role of KCs in the pathogenesis of nonalcoholic fatty liver disease induced by a marginal-copper high-fructose diet (CuMF). Male weanling Sprague-Dawley rats were fed either a copper-adequate or a marginally copper-deficient diet for 4 wk. Deionized water or deionized water containing 30% fructose (wt/vol) was also given ad libitum. KCs were depleted by intravenous administration of gadolinium chloride (GdCl3) before and/or in the middle of the experimental period. Hepatic triglyceride accumulation was completely eliminated with KC depletion in CuMF consumption rats, which was associated with the normalization of elevated plasma monocyte chemoattractant protein-1 (MCP-1) and increased hepatic sterol regulatory element binding protein-1 expression. However, hepatic copper and iron content were not significantly affected by KC depletion. In addition, KC depletion reduced body weight and epididymal fat weight as well as adipocyte size. Plasma endotoxin and gut permeability were markedly increased in CuMF rats. Moreover, MCP-1 was robustly increased in the culture medium when isolated KCs from CuMF rats were treated with LPS. Our data suggest that KCs play a critical role in the development of hepatic steatosis induced by marginal-copper high-fructose diet.
Collapse
Affiliation(s)
- Ming Song
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, University of Louisville School of Medicine, Louisville, Kentucky;
| | - Dale A. Schuschke
- 2Department of Physiology and Biophysics, University of Louisville School of Medicine, Louisville, Kentucky;
| | - Zhanxiang Zhou
- 6Center for Translational Biomedical Research, University of North Carolina at Greensboro, Kannapolis, North Carolina; ,7Department of Nutrition, University of North Carolina at Greensboro, Kannapolis, North Carolina; and
| | - Wei Zhong
- 6Center for Translational Biomedical Research, University of North Carolina at Greensboro, Kannapolis, North Carolina;
| | - Jiayuan Zhang
- 4Department of Chemistry, University of Louisville School of Medicine, Louisville, Kentucky;
| | - Xiang Zhang
- 3Department of Pharmacology and Toxicology, ,4Department of Chemistry, University of Louisville School of Medicine, Louisville, Kentucky;
| | - Yuhua Wang
- 8College of Food Science and Engineering, Jilin Agricultural University, Changchun, China
| | - Craig J. McClain
- 1Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, University of Louisville School of Medicine, Louisville, Kentucky; ,3Department of Pharmacology and Toxicology, ,5Robley Rex Veterans Affairs Medical Center, Louisville, Kentucky;
| |
Collapse
|
25
|
Nutrigenomics analysis reveals that copper deficiency and dietary sucrose up-regulate inflammation, fibrosis and lipogenic pathways in a mature rat model of nonalcoholic fatty liver disease. J Nutr Biochem 2015; 26:996-1006. [PMID: 26033743 DOI: 10.1016/j.jnutbio.2015.04.009] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Revised: 04/08/2015] [Accepted: 04/13/2015] [Indexed: 12/18/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD) prevalence is increasing worldwide, with the affected US population estimated near 30%. Diet is a recognized risk factor in the NAFLD spectrum, which includes nonalcoholic steatohepatitis (NASH) and fibrosis. Low hepatic copper (Cu) was recently linked to clinical NAFLD/NASH severity. Simple sugar consumption including sucrose and fructose is implicated in NAFLD, while consumption of these macronutrients also decreases liver Cu levels. Though dietary sugar and low Cu are implicated in NAFLD, transcript-level responses that connect diet and pathology are not established. We have developed a mature rat model of NAFLD induced by dietary Cu deficiency, human-relevant high sucrose intake (30% w/w) or both factors in combination. Compared to the control diet with adequate Cu and 10% (w/w) sucrose, rats fed either high-sucrose or low-Cu diet had increased hepatic expression of genes involved in inflammation and fibrogenesis, including hepatic stellate cell activation, while the combination of diet factors also increased ATP citrate lyase and fatty acid synthase gene transcription (fold change > 2, P < 0.02). Low dietary Cu decreased hepatic and serum Cu (P ≤ 0.05), promoted lipid peroxidation and induced NAFLD-like histopathology, while the combined factors also induced fasting hepatic insulin resistance and liver damage. Neither low Cu nor 30% sucrose in the diet led to enhanced weight gain. Taken together, transcript profiles, histological and biochemical data indicate that low Cu and high sucrose promote hepatic gene expression and physiological responses associated with NAFLD and NASH, even in the absence of obesity or severe steatosis.
Collapse
|
26
|
Ackerman Z, Skarzinski G, Grozovski M, Oron-Herman M, Sela BA. Effects of Antihypertensive and Triglyceride-lowering Agents on Hepatic Copper Concentrations in Rats with Fatty Liver Disease. Basic Clin Pharmacol Toxicol 2014; 115:545-51. [DOI: 10.1111/bcpt.12283] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Accepted: 06/03/2014] [Indexed: 12/11/2022]
Affiliation(s)
- Zvi Ackerman
- Departments of Medicine; Hadassah-Hebrew University Medical Center; Jerusalem Israel
| | - Galina Skarzinski
- Departments of Medicine; Hadassah-Hebrew University Medical Center; Jerusalem Israel
| | | | - Mor Oron-Herman
- Institute of Chemical Pathology; Sheba Medical Center; Tel Hashomer Israel
| | - Ben-Ami Sela
- Institute of Chemical Pathology; Sheba Medical Center; Tel Hashomer Israel
| |
Collapse
|
27
|
Nestorov J, Glban AM, Mijušković A, Nikolić-Kokić A, Elaković I, Veličković N, Matić G. Long-term fructose-enriched diet introduced immediately after weaning does not induce oxidative stress in the rat liver. Nutr Res 2014; 34:646-52. [PMID: 25150124 DOI: 10.1016/j.nutres.2014.06.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Revised: 05/30/2014] [Accepted: 06/09/2014] [Indexed: 01/19/2023]
Abstract
Increased fructose consumption is correlated with the rising prevalence of obesity, metabolic syndrome, and type 2 diabetes. It is believed that reactive oxygen species contribute to the development and progression of metabolic disturbances, especially those associated with insulin resistance. Dietary fructose produces both pro-oxidative and antioxidative effects, depending upon the experimental conditions, dosage, duration of treatment, and pathophysiological milieu. The effects of fructose overconsumption on young populations, which have an increased risk of developing metabolic disorders in adulthood, have not been fully elucidated. We have previously shown that rats subjected to a long-term fructose-enriched diet immediately after weaning display impaired hepatic insulin sensitivity. In this study, we tested the hypothesis that long-term fructose consumption induces alterations in the redox setting of the liver. Starting from the 21st day after birth, male Wistar rats were maintained for 9 weeks on a standard diet (control) or a fructose-enriched diet that consisted of standard food and 10% fructose solution instead of drinking water. The expression and activity of antioxidant enzymes as well as lipid peroxidation and protein damage markers were measured. The results showed that a fructose-enriched diet led to an increased expression of mitochondrial manganese superoxide dismutase but did not affect antioxidant enzymes activity, lipid peroxidation, thiol content, and the level of protein oxidation. Therefore, our results suggest that the decrease in hepatic insulin sensitivity that was previously observed in rats that were kept on the same diet regime might be attributed to molecular mechanisms other than redox disbalance. A possible fructose-related micronutrient deficiency should be examined.
Collapse
Affiliation(s)
- Jelena Nestorov
- Department of Biochemistry Institute for Biological Research "Siniša Stanković," University of Belgrade, 11060 Belgrade, Serbia.
| | - Alhadi M Glban
- Department of Biochemistry Institute for Biological Research "Siniša Stanković," University of Belgrade, 11060 Belgrade, Serbia
| | - Ana Mijušković
- Department of Physiology, Institute for Biological Research "Siniša Stanković," University of Belgrade, 11060 Belgrade, Serbia
| | - Aleksandra Nikolić-Kokić
- Department of Physiology, Institute for Biological Research "Siniša Stanković," University of Belgrade, 11060 Belgrade, Serbia
| | - Ivana Elaković
- Department of Biochemistry Institute for Biological Research "Siniša Stanković," University of Belgrade, 11060 Belgrade, Serbia
| | - Nataša Veličković
- Department of Biochemistry Institute for Biological Research "Siniša Stanković," University of Belgrade, 11060 Belgrade, Serbia
| | - Gordana Matić
- Department of Biochemistry Institute for Biological Research "Siniša Stanković," University of Belgrade, 11060 Belgrade, Serbia
| |
Collapse
|