1
|
Gancheva S, Roden M, Castera L. Diabetes as a risk factor for MASH progression. Diabetes Res Clin Pract 2024; 217:111846. [PMID: 39245423 DOI: 10.1016/j.diabres.2024.111846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 08/28/2024] [Accepted: 09/03/2024] [Indexed: 09/10/2024]
Abstract
Non-alcoholic (now: metabolic) steatohepatitis (MASH) is the progressive inflammatory form of metabolic dysfunction-associated steatotic liver disease (MASLD), which often coexists and mutually interacts with type 2 diabetes (T2D), resulting in worse hepatic and cardiovascular outcomes. Understanding the intricate mechanisms of diabetes-related MASH progression is crucial for effective therapeutic strategies. This review delineates the multifaceted pathways involved in this interplay and explores potential therapeutic implications. The synergy between adipose tissue, gut microbiota, and hepatic alterations plays a pivotal role in disease progression. Adipose tissue dysfunction, particularly in the visceral depot, coupled with dysbiosis in the gut microbiota, exacerbates hepatic injury and insulin resistance. Hepatic lipid accumulation, oxidative stress, and endoplasmic reticulum stress further potentiate inflammation and fibrosis, contributing to disease severity. Dietary modification with weight reduction and exercise prove crucial in managing T2D-related MASH. Additionally, various well-known but also novel anti-hyperglycemic medications exhibit potential in reducing liver lipid content and, in some cases, improving MASH histology. Therapies targeting incretin receptors show promise in managing T2D-related MASH, while thyroid hormone receptor-β agonism has proven effective as a treatment of MASH and fibrosis.
Collapse
Affiliation(s)
- Sofiya Gancheva
- Department of Endocrinology and Diabetology, Medical Faculty and University Hospital, Heinrich-Heine University, Düsseldorf, Germany; Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany; German Center for Diabetes Research (DZD e.V.), Partner Düsseldorf, München-Neuherberg, Germany
| | - Michael Roden
- Department of Endocrinology and Diabetology, Medical Faculty and University Hospital, Heinrich-Heine University, Düsseldorf, Germany; Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany; German Center for Diabetes Research (DZD e.V.), Partner Düsseldorf, München-Neuherberg, Germany.
| | - Laurent Castera
- Department of Hepatology, Hôpital Beaujon, Assistance Publique-Hôpitaux de Paris, Clichy, France; Université Paris-Cité, INSERM UMR 1149, Centre de Recherche sur l'Inflammation Paris, Montmartre, Paris, France.
| |
Collapse
|
2
|
Wan L, Li T, Yao M, Zhang B, Zhang W, Zhang J. Linoelaidic acid gavage has more severe consequences on triglycerides accumulation, inflammation and intestinal microbiota in mice than elaidic acid. Food Chem X 2024; 22:101328. [PMID: 38576778 PMCID: PMC10992693 DOI: 10.1016/j.fochx.2024.101328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Revised: 03/19/2024] [Accepted: 03/21/2024] [Indexed: 04/06/2024] Open
Abstract
This work aims to study the effects of oral gavage (0.2 mg/g body weight) of elaidic acid (C18:1-9 t, EA) and linoelaidic acid (C18:2-9 t,12 t, LEA) on lipid metabolism, inflammation and gut homeostasis of mice. Results showed that both EA and LEA gavage significantly increased LDL-c, TC and oxidative stress levels in the liver and serum and may stimulate liver inflammation via NF-κB and MAPK signaling pathway. Compared with EA, LEA gavage significantly promoted TAG accumulation and inflammatory signaling. Serum lipidomics revealed that LEA intake significantly increased the concentration of ∼50 TAGs, while EA gavage primarily caused significant decreases in several SMs. 16S rRNA demonstrated that LEA ingestion markedly changed fecal microbiota by enriching Lactobacillus (phylum Firmicutes), however, EA treatment did not affect it. Overall, LEA gavage has more severe consequences on TAG accumulation, inflammation and microbial structure than EA, highlighting that the number of trans double bonds affects these processes.
Collapse
Affiliation(s)
- Liting Wan
- Key Laboratory of Food Nutrition and Functional Food of Hainan Province, School of Food Science and Engineering, Hainan University, Haikou, 570228, China
| | - Tian Li
- Key Laboratory of Food Nutrition and Functional Food of Hainan Province, School of Food Science and Engineering, Hainan University, Haikou, 570228, China
| | - Mengying Yao
- Key Laboratory of Food Nutrition and Functional Food of Hainan Province, School of Food Science and Engineering, Hainan University, Haikou, 570228, China
| | - Baoshun Zhang
- College of Pharmaceutical Sciences, Southwest University, Chongqing, 400716, China
| | - Weimin Zhang
- Key Laboratory of Food Nutrition and Functional Food of Hainan Province, School of Food Science and Engineering, Hainan University, Haikou, 570228, China
- Key Laboratory of Tropical Fruits and Vegetables Quality and Safety for State Market Regulation, Hainan Institute for Food Control, Haikou, 570228, China
| | - Jiachao Zhang
- Key Laboratory of Food Nutrition and Functional Food of Hainan Province, School of Food Science and Engineering, Hainan University, Haikou, 570228, China
| |
Collapse
|
3
|
Tibori K, Zámbó V, Orosz G, Szelényi P, Sarnyai F, Tamási V, Rónai Z, Csala M, Kereszturi É. Allele-specific effect of various dietary fatty acids and ETS1 transcription factor on SCD1 expression. Sci Rep 2024; 14:177. [PMID: 38167845 PMCID: PMC10761808 DOI: 10.1038/s41598-023-50700-5] [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: 08/28/2023] [Accepted: 12/23/2023] [Indexed: 01/05/2024] Open
Abstract
Overnutrition and genetic predisposition are major risk factors for various metabolic disorders. Stearoyl-CoA desaturase-1 (SCD1) plays a key role in these conditions by synthesizing unsaturated fatty acids (FAs), thereby promoting fat storage and alleviating lipotoxicity. Expression of SCD1 is influenced by various saturated and cis-unsaturated FAs, but the possible role of dietary trans FAs (TFAs) and SCD1 promoter polymorphisms in its regulations has not been addressed. Therefore, we aimed to investigate the impact of the two main TFAs, vaccenate and elaidate, and four common promoter polymorphisms (rs1054411, rs670213, rs2275657, rs2275656) on SCD1 expression in HEK293T and HepG2 cell cultures using luciferase reporter assay, qPCR and immunoblotting. We found that SCD1 protein and mRNA levels as well as SCD1 promoter activity are markedly elevated by elaidate, but not altered by vaccenate. The promoter polymorphisms did not affect the basal transcriptional activity of SCD1. However, the minor allele of rs1054411 increased SCD1 expression in the presence of various FAs. Moreover, this variant was predicted in silico and verified in vitro to reduce the binding of ETS1 transcription factor to SCD1 promoter. Although we could not confirm an association with type 2 diabetes mellitus, the FA-dependent and ETS1-mediated effect of rs1054411 polymorphism deserves further investigation as it may modulate the development of lipid metabolism-related conditions.
Collapse
Affiliation(s)
- Kinga Tibori
- Department of Molecular Biology, Semmelweis University, 1085, Budapest, Hungary
| | - Veronika Zámbó
- Department of Molecular Biology, Semmelweis University, 1085, Budapest, Hungary.
| | - Gabriella Orosz
- Department of Molecular Biology, Semmelweis University, 1085, Budapest, Hungary
| | - Péter Szelényi
- Department of Molecular Biology, Semmelweis University, 1085, Budapest, Hungary
| | - Farkas Sarnyai
- Department of Molecular Biology, Semmelweis University, 1085, Budapest, Hungary
| | - Viola Tamási
- Department of Molecular Biology, Semmelweis University, 1085, Budapest, Hungary
| | - Zsolt Rónai
- Department of Molecular Biology, Semmelweis University, 1085, Budapest, Hungary
| | - Miklós Csala
- Department of Molecular Biology, Semmelweis University, 1085, Budapest, Hungary
| | - Éva Kereszturi
- Department of Molecular Biology, Semmelweis University, 1085, Budapest, Hungary.
| |
Collapse
|
4
|
Li N, Yin L, Shang J, Liang M, Liu Z, Yang H, Qiang G, Du G, Yang X. Kaempferol attenuates nonalcoholic fatty liver disease in type 2 diabetic mice via the Sirt1/AMPK signaling pathway. Biomed Pharmacother 2023; 165:115113. [PMID: 37418974 DOI: 10.1016/j.biopha.2023.115113] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 06/28/2023] [Accepted: 06/29/2023] [Indexed: 07/09/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is one of the most common liver diseases with limited treatment options. Moreover, its prevalence is doubled in type 2 diabetes mellitus (T2DM). Kaempferol (KAP) is a flavonoid compound that has been suggested to have beneficial effects on NAFLD, but studies on the mechanism are lacking, especially in the diabetic state. Herein, we investigated the effect of KAP on NAFLD associated with T2DM and its underlying mechanism in vitro and in vivo. The results of in vitro studies indicated that KAP treatment (10-8-10-6 M) significantly reduced lipid accumulation in oleic acid-induced HepG2 cells. Moreover, in the T2DM animal model of db/db mice, we confirmed that KAP (50 mg/kg) significantly reduced lipid accumulation and improved liver injury. Mechanistic studies in vitro and in vivo showed that Sirtuin 1 (Sirt1)/AMP-activated protein kinase (AMPK) signal was involved in KAP regulation of hepatic lipid accumulation. KAP treatment activated Sirt1 and AMPK, upregulated the levels of fatty acid oxidation-related protein proliferator activated receptor gamma coactivator 1α (PGC1α); and downregulated lipid synthesis-related proteins, including acetyl-coA carboxylase (ACC), fatty acid synthase (FASN), and sterol regulatory element-binding protein 1 (SREBP1). Furthermore, the curative effect of KAP on lipid accumulation was abolished by siRNA-mediated knockdown of either Sirt1 or AMPK. Collectively, these findings suggest that KAP may be a potential therapeutic agent for NAFLD associated with T2DM by regulating hepatic lipid accumulation through activation of Sirt1/AMPK signaling.
Collapse
Affiliation(s)
- Na Li
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines and Beijing Key Laboratory of Drug Target and Screening Research, Institute of Materia Medica of Peking Union Medical College, Beijing, China
| | - Lin Yin
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines and Beijing Key Laboratory of Drug Target and Screening Research, Institute of Materia Medica of Peking Union Medical College, Beijing, China; Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan 030032, China
| | - Jiamin Shang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines and Beijing Key Laboratory of Drug Target and Screening Research, Institute of Materia Medica of Peking Union Medical College, Beijing, China
| | - Meidai Liang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines and Beijing Key Laboratory of Drug Target and Screening Research, Institute of Materia Medica of Peking Union Medical College, Beijing, China
| | - Zhaoyu Liu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines and Beijing Key Laboratory of Drug Target and Screening Research, Institute of Materia Medica of Peking Union Medical College, Beijing, China
| | - Haiguang Yang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines and Beijing Key Laboratory of Drug Target and Screening Research, Institute of Materia Medica of Peking Union Medical College, Beijing, China
| | - Guifen Qiang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines and Beijing Key Laboratory of Drug Target and Screening Research, Institute of Materia Medica of Peking Union Medical College, Beijing, China.
| | - Guanhua Du
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines and Beijing Key Laboratory of Drug Target and Screening Research, Institute of Materia Medica of Peking Union Medical College, Beijing, China
| | - Xiuying Yang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines and Beijing Key Laboratory of Drug Target and Screening Research, Institute of Materia Medica of Peking Union Medical College, Beijing, China.
| |
Collapse
|
5
|
Jia F, Hu X, Kimura T, Tanaka N. Impact of Dietary Fat on the Progression of Liver Fibrosis: Lessons from Animal and Cell Studies. Int J Mol Sci 2021; 22:10303. [PMID: 34638640 PMCID: PMC8508674 DOI: 10.3390/ijms221910303] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 09/16/2021] [Indexed: 02/07/2023] Open
Abstract
Previous studies have revealed that a high-fat diet is one of the key contributors to the progression of liver fibrosis, and increasing studies are devoted to analyzing the different influences of diverse fat sources on the progression of non-alcoholic steatohepatitis. When we treated three types of isocaloric diets that are rich in cholesterol, saturated fatty acid (SFA) and trans fatty acid (TFA) with hepatitis C virus core gene transgenic mice that spontaneously developed hepatic steatosis without apparent fibrosis, TFA and cholesterol-rich diet, but not SFA-rich diet, displayed distinct hepatic fibrosis. This review summarizes the recent advances in animal and cell studies regarding the effects of these three types of fat on liver fibrogenesis.
Collapse
Affiliation(s)
- Fangping Jia
- Department of Metabolic Regulation, Shinshu University School of Medicine, Matsumoto 390-8621, Japan;
| | - Xiao Hu
- Department of Pathophysiology, Hebei Medical University, Shijiazhuang 050017, China;
| | - Takefumi Kimura
- Department of Gastroenterology, Shinshu University School of Medicine, Matsumoto 390-8621, Japan;
| | - Naoki Tanaka
- Department of Metabolic Regulation, Shinshu University School of Medicine, Matsumoto 390-8621, Japan;
- International Relations Office, Shinshu University School of Medicine, Matsumoto 390-8621, Japan
- Research Center for Social Systems, Shinshu University, Matsumoto 390-8621, Japan
| |
Collapse
|
6
|
Lavandera JV, Reus V, Saín J, Bernal CA, González MA. Dietary n-9, n-6 and n-3 fatty acids modulate the oxidative stress in brain and liver of mice. Effect of trans fatty acids supplementation. MEDITERRANEAN JOURNAL OF NUTRITION AND METABOLISM 2021. [DOI: 10.3233/mnm-200508] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
BACKGROUND: Arachidonic (20:4n-6) and docosahexaenoic (22:6n-3) acids interaction affects brain structure and function. Unsaturated fatty acids (UFAs) generate oxygenated lipid-derived eicosanoids which modulate the inflammatory response. The presence of trans fatty acids (TFA) in neuronal membranes can favor to generation of pro-oxidant metabolites. OBJECTIVE: This study evaluated the effect of supplementation with TFA to diets containing different proportions of FA, on the oxidative stress (OS) generation and the inflammatory response in mice brain and liver. METHODS: CF1 mice were fed diets (16 weeks) with olive (O), corn (C) or rapeseed (R) oils. OS parameters and gene expression of some key liver and brain enzymes involved in OS production were evaluated. RESULTS: In brain and liver, lipoperoxidation was increased and catalase activity was decreased in C. In brain, glutathione was diminished by supplementation with TFA in all diets and histological sections showed lymphocytes in O and C. In liver, decreased amount of lipid vacuoles and increased of cyclooxygenase-1 (COX-1) and PPARγ mRNA levels were observed in R and Rt. IL-1b and IL-6 in serum were augmented in O and Ot. CONCLUSIONS: Rapeseed oil could have protective effects on the development of OS and inflammation, while TFA supplementation did not showed marked effects on these parameters.
Collapse
Affiliation(s)
- Jimena Verónica Lavandera
- Cátedra de Bromatología y Nutrición, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Santa Fe, Argentina
| | - Verónica Reus
- Facultad de Ciencias Médicas, Universidad Nacional del Litoral, Santa Fe, Argentina
| | - Juliana Saín
- Cátedra de Bromatología y Nutrición, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Santa Fe, Argentina
| | - Claudio Adrian Bernal
- Cátedra de Bromatología y Nutrición, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Santa Fe, Argentina
| | - Marcela Aida González
- Cátedra de Bromatología y Nutrición, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina
| |
Collapse
|
7
|
Suzuki-Kemuriyama N, Abe A, Uno K, Ogawa S, Watanabe A, Sano R, Yuki M, Miyajima K, Nakae D. A trans fatty acid substitute enhanced development of liver proliferative lesions induced in mice by feeding a choline-deficient, methionine-lowered, L-amino acid-defined, high-fat diet. Lipids Health Dis 2020; 19:251. [PMID: 33317575 PMCID: PMC7737357 DOI: 10.1186/s12944-020-01423-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Accepted: 11/22/2020] [Indexed: 02/06/2023] Open
Abstract
Background Nonalcoholic steatohepatitis (NASH) is a form of liver disease characterized by steatosis, necroinflammation, and fibrosis, resulting in cirrhosis and cancer. Efforts have focused on reducing the intake of trans fatty acids (TFAs) because of potential hazards to human health and the increased risk for NASH. However, the health benefits of reducing dietary TFAs have not been fully elucidated. Here, the effects of TFAs vs. a substitute on NASH induced in mice by feeding a choline-deficient, methionine-lowered, L-amino acid-defined, high-fat diet (CDAA-HF) were investigated. Methods Mice were fed CDAA-HF containing shortening with TFAs (CDAA-HF-T(+)), CDAA-HF containing shortening without TFAs (CDAA-HF-T(−)), or a control chow for 13 or 26 weeks. Results At week 13, NASH was induced in mice by feeding CDAA-HF-T(+) containing TFAs or CDAA-HF-T(−) containing no TFAs, but rather mostly saturated fatty acids (FAs), as evidenced by elevated serum transaminase activity and liver changes, including steatosis, inflammation, and fibrosis. CDAA-HF-T(−) induced a greater extent of hepatocellular apoptosis at week 13. At week 26, proliferative (preneoplastic and non-neoplastic) nodular lesions were more pronounced in mice fed CDAA-HF-T(−) than CDAA-HF-T(+). Conclusions Replacement of dietary TFAs with a substitute promoted the development of proliferation lesions in the liver of a mouse NASH model, at least under the present conditions. Attention should be paid regarding use of TFA substitutes in foods for human consumption, and a balance of FAs is likely more important than the particular types of FAs. Supplementary Information The online version contains supplementary material available at 10.1186/s12944-020-01423-3.
Collapse
Affiliation(s)
- Noriko Suzuki-Kemuriyama
- Department of Nutritional Science and Food Safety, Faculty of Applied Bioscience, Tokyo University of Agriculture , 1-1-1, Sakuragaoka, Setagaya, Tokyo, 156-8502, Japan
| | - Akari Abe
- Department of Nutritional Science and Food Safety, Graduate School of Applied Bioscience, Tokyo University of Agriculture, 1-1-1 Sakuragaoka, Setagaya, Tokyo, 156-8502, Japan
| | - Kiniko Uno
- Department of Food and Nutritional Science, Graduate School of Applied Bioscience, Tokyo University of Agriculture, 1-1-1, Sakuragaoka, Setagaya, Tokyo, 156-8502, Japan
| | - Shuji Ogawa
- Department of Food and Nutritional Science, Graduate School of Applied Bioscience, Tokyo University of Agriculture, 1-1-1, Sakuragaoka, Setagaya, Tokyo, 156-8502, Japan
| | - Atsushi Watanabe
- Department of Nutritional Science and Food Safety, Graduate School of Applied Bioscience, Tokyo University of Agriculture, 1-1-1 Sakuragaoka, Setagaya, Tokyo, 156-8502, Japan
| | - Ryuhei Sano
- Department of Nutritional Science and Food Safety, Graduate School of Applied Bioscience, Tokyo University of Agriculture, 1-1-1 Sakuragaoka, Setagaya, Tokyo, 156-8502, Japan
| | - Megumi Yuki
- Department of Nutritional Science and Food Safety, Graduate School of Applied Bioscience, Tokyo University of Agriculture, 1-1-1 Sakuragaoka, Setagaya, Tokyo, 156-8502, Japan
| | - Katsuhiro Miyajima
- Department of Nutritional Science and Food Safety, Faculty of Applied Bioscience, Tokyo University of Agriculture , 1-1-1, Sakuragaoka, Setagaya, Tokyo, 156-8502, Japan.,Department of Nutritional Science and Food Safety, Graduate School of Applied Bioscience, Tokyo University of Agriculture, 1-1-1 Sakuragaoka, Setagaya, Tokyo, 156-8502, Japan
| | - Dai Nakae
- Department of Nutritional Science and Food Safety, Faculty of Applied Bioscience, Tokyo University of Agriculture , 1-1-1, Sakuragaoka, Setagaya, Tokyo, 156-8502, Japan. .,Department of Nutritional Science and Food Safety, Graduate School of Applied Bioscience, Tokyo University of Agriculture, 1-1-1 Sakuragaoka, Setagaya, Tokyo, 156-8502, Japan.
| |
Collapse
|
8
|
Xin X, Cai BY, Chen C, Tian HJ, Wang X, Hu YY, Feng Q. High-trans fatty acid and high-sugar diets can cause mice with non-alcoholic steatohepatitis with liver fibrosis and potential pathogenesis. Nutr Metab (Lond) 2020; 17:40. [PMID: 32508961 PMCID: PMC7249374 DOI: 10.1186/s12986-020-00462-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 05/20/2020] [Indexed: 12/14/2022] Open
Abstract
Background and aims Even Non-alcoholic steatohepatitis (NASH) has been becoming the key role in process of liver fibrosis or cirrhosis, no any NASH involving liver fibrosis mice model which consistent with the mechanisms of fatty acid and glucose metabolism disorder was widely accepted. Here, we established a mouse model of nonalcoholic steatohepatitis (NASH) with liver fibrosis using a high-fat, high-carbohydrate diet (HFHC) and analyzed the potential pathogenesis using a transcriptome microarray. Methods Fifty mice were stratified by weight and randomly divided into the HFHC model and control (Con) groups. Ten mice were sacrificed at the beginning of the experiments, 10 mice of HFHC and Con group were euthanized at the end of 20 and 30 weeks. The following analyses were performed: biochemical analysis; histological assessment; evaluation of hepatic type I collagen (Col-I), α-smooth muscle actin (α-SMA) and transforming growth factor-β1 (TGF-β1) protein and mRNA expression levels; and transcriptomic gene chip analysis. Results Compared with the Con group at each time point, the body weight and liver wet weight of the HFHC model group of mice were significantly higher. At 30th weeks, alanine aminotransferase (ALT), aspartate aminotransferase (AST), fasting blood glucose (FBG) and fasting insulin (FINS) levels or activities and the triglyceride (TG) and hydroxyproline (HYP) content in the HFHC model group were significantly elevated. Severe steatosis was present in the liver tissues contributed from the HFHC group of mice. Typically, substantial perisinusoidal fibrosis with a cage-like structure and bridging formations were observed in the mice liver in HFHC group. Col-I, α-SMA and TGF-β1 protein and mRNA expression levels in liver tissues of HFHC mice dramatically increased over time. Compared with the Con group, the HFHC group had 151 differentially expressed genes that were involved in 41 signaling pathways. Conclusions After keeping 30 weeks HFHC diet treatment, the mice exhibited substantial liver fibrosis, hepatic steatosis, ballooning degeneration and inflammation. Basing on the transcriptome microarray assays, the experimental NASH involving liver fibrosis potentially related to dramatically changed ECM-receptor interaction, Toll-like receptor signaling and other signaling pathways.
Collapse
Affiliation(s)
- Xin Xin
- Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine, 528 Zhangheng Road, Pudong New Area, Shanghai, 201203 China
| | - Bei-Yu Cai
- Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine, 528 Zhangheng Road, Pudong New Area, Shanghai, 201203 China
| | - Cheng Chen
- Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine, 528 Zhangheng Road, Pudong New Area, Shanghai, 201203 China
| | - Hua-Jie Tian
- Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine, 528 Zhangheng Road, Pudong New Area, Shanghai, 201203 China
| | - Xin Wang
- Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine, 528 Zhangheng Road, Pudong New Area, Shanghai, 201203 China
| | - Yi-Yang Hu
- Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine, 528 Zhangheng Road, Pudong New Area, Shanghai, 201203 China.,Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shanghai, 201203 China.,Key Laboratory of Liver and Kidney Diseases, Shanghai University of Traditional Chinese Medicine, Ministry of Education, Shanghai, 201203 China
| | - Qin Feng
- Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine, 528 Zhangheng Road, Pudong New Area, Shanghai, 201203 China.,Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shanghai, 201203 China.,Key Laboratory of Liver and Kidney Diseases, Shanghai University of Traditional Chinese Medicine, Ministry of Education, Shanghai, 201203 China
| |
Collapse
|
9
|
Oteng AB, Kersten S. Mechanisms of Action of trans Fatty Acids. Adv Nutr 2020; 11:697-708. [PMID: 31782488 PMCID: PMC7231579 DOI: 10.1093/advances/nmz125] [Citation(s) in RCA: 136] [Impact Index Per Article: 27.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 09/03/2019] [Accepted: 10/31/2019] [Indexed: 12/11/2022] Open
Abstract
Human studies have established a positive association between the intake of industrial trans fatty acids and the development of cardiovascular diseases, leading several countries to enact laws that restrict the presence of industrial trans fatty acids in food products. However, trans fatty acids cannot be completely eliminated from the human diet since they are also naturally present in meat and dairy products of ruminant animals. Moreover, bans on industrial trans fatty acids have not yet been instituted in all countries. The epidemiological evidence against trans fatty acids by far overshadows mechanistic insights that may explain how trans fatty acids achieve their damaging effects. This review focuses on the mechanisms that underlie the deleterious effects of trans fatty acids by juxtaposing effects of trans fatty acids against those of cis-unsaturated fatty acids and saturated fatty acids (SFAs). This review also carefully explores the argument that ruminant trans fatty acids have differential effects from industrial trans fatty acids. Overall, in vivo and in vitro studies demonstrate that industrial trans fatty acids promote inflammation and endoplasmic reticulum (ER) stress, although to a lesser degree than SFAs, whereas cis-unsaturated fatty acids are protective against ER stress and inflammation. Additionally, industrial trans fatty acids promote fat storage in the liver at the expense of adipose tissue compared with cis-unsaturated fatty acids and SFAs. In cultured hepatocytes and adipocytes, industrial trans fatty acids, but not cis-unsaturated fatty acids or SFAs, stimulate the cholesterol synthesis pathway by activating sterol regulatory element binding protein (SREBP) 2-mediated gene regulation. Interestingly, although industrial and ruminant trans fatty acids show similar effects on human plasma lipoproteins, in preclinical models, only industrial trans fatty acids promote inflammation, ER stress, and cholesterol synthesis. Overall, clearer insight into the molecular mechanisms of action of trans fatty acids may create new therapeutic windows for the treatment of diseases characterized by disrupted lipid metabolism.
Collapse
Affiliation(s)
- Antwi-Boasiako Oteng
- Nutrition, Metabolism and Genomics Group, Division of Human Nutrition and Health, Wageningen University, Wageningen, The Netherlands
| | - Sander Kersten
- Nutrition, Metabolism and Genomics Group, Division of Human Nutrition and Health, Wageningen University, Wageningen, The Netherlands
| |
Collapse
|
10
|
Ocker M. Fibroblast growth factor signaling in non-alcoholic fatty liver disease and non-alcoholic steatohepatitis: Paving the way to hepatocellular carcinoma. World J Gastroenterol 2020; 26:279-290. [PMID: 31988589 PMCID: PMC6969880 DOI: 10.3748/wjg.v26.i3.279] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 12/17/2019] [Accepted: 01/01/2020] [Indexed: 02/06/2023] Open
Abstract
Metabolic disorders are increasingly leading to non-alcoholic fatty liver disease, subsequent steatohepatitis, cirrhosis and hepatocellular carcinoma. Fibroblast growth factors and their receptors play an important role in maintaining metabolic homeostasis also in the liver and disorders in signaling have been identified to contribute to those pathophysiologic conditions leading to hepatic lipid accumulation and chronic inflammation. While specific and well tolerated inhibitors of fibroblast growth factor receptor activity are currently developed for (non-liver) cancer therapy, treatment of non-alcoholic fatty liver disease and non-alcoholic steatohepatitis is still limited. Fibroblast growth factor-mimicking or restoring approaches have recently evolved as a novel therapeutic option and the impact of such interactions with the fibroblast growth factor receptor signaling network during non-alcoholic fatty liver disease/non-alcoholic steatohepatitis development is reviewed here.
Collapse
Affiliation(s)
- Matthias Ocker
- Department of Gastroenterology (CBF), Charité University Medicine Berlin, Berlin 10117, Germany
| |
Collapse
|
11
|
Tamer F, Ulug E, Akyol A, Nergiz-Unal R. The potential efficacy of dietary fatty acids and fructose induced inflammation and oxidative stress on the insulin signaling and fat accumulation in mice. Food Chem Toxicol 2020; 135:110914. [DOI: 10.1016/j.fct.2019.110914] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 10/17/2019] [Accepted: 10/23/2019] [Indexed: 02/09/2023]
|
12
|
Boland ML, Oró D, Tølbøl KS, Thrane ST, Nielsen JC, Cohen TS, Tabor DE, Fernandes F, Tovchigrechko A, Veidal SS, Warrener P, Sellman BR, Jelsing J, Feigh M, Vrang N, Trevaskis JL, Hansen HH. Towards a standard diet-induced and biopsy-confirmed mouse model of non-alcoholic steatohepatitis: Impact of dietary fat source. World J Gastroenterol 2019; 25:4904-4920. [PMID: 31543682 PMCID: PMC6737317 DOI: 10.3748/wjg.v25.i33.4904] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 06/28/2019] [Accepted: 07/19/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND The trans-fat containing AMLN (amylin liver non-alcoholic steatohepatitis, NASH) diet has been extensively validated in C57BL/6J mice with or without the Lepob/Lepob (ob/ob) mutation in the leptin gene for reliably inducing metabolic and liver histopathological changes recapitulating hallmarks of NASH. Due to a recent ban on trans-fats as food additive, there is a marked need for developing a new diet capable of promoting a compatible level of disease in ob/ob and C57BL/6J mice.
AIM To develop a biopsy-confirmed mouse model of NASH based on an obesogenic diet with trans-fat substituted by saturated fat.
METHODS Male ob/ob mice were fed AMLN diet or a modified AMLN diet with trans-fat (Primex shortening) substituted by equivalent amounts of palm oil [Gubra amylin NASH, (GAN) diet] for 8, 12 and 16 wk. C57BL/6J mice were fed the same diets for 28 wk. AMLN and GAN diets had similar caloric content (40% fat kcal), fructose (22%) and cholesterol (2%) level.
RESULTS The GAN diet was more obesogenic compared to the AMLN diet and impaired glucose tolerance. Biopsy-confirmed steatosis, lobular inflammation, hepatocyte ballooning, fibrotic liver lesions and hepatic transcriptome changes were similar in ob/ob mice fed the GAN or AMLN diet. C57BL/6J mice developed a mild to moderate fibrotic NASH phenotype when fed the same diets.
CONCLUSION Substitution of Primex with palm oil promotes a similar phenotype of biopsy-confirmed NASH in ob/ob and C57BL/6J mice, making GAN diet-induced obese mouse models suitable for characterizing novel NASH treatments.
Collapse
Affiliation(s)
- Michelle L Boland
- Cardiovascular, Renal and Metabolic Diseases, MedImmune, Gaithersburg, MD 20878, United States
- Pharmacology, Gubra, Hørsholm DK-2970, Denmark
| | - Denise Oró
- Pharmacology, Gubra, Hørsholm DK-2970, Denmark
| | | | | | | | - Taylor S Cohen
- Cardiovascular, Renal and Metabolic Diseases, MedImmune, Gaithersburg, MD 20878, United States
| | - David E Tabor
- Cardiovascular, Renal and Metabolic Diseases, MedImmune, Gaithersburg, MD 20878, United States
| | - Fiona Fernandes
- Cardiovascular, Renal and Metabolic Diseases, MedImmune, Gaithersburg, MD 20878, United States
| | - Andrey Tovchigrechko
- Cardiovascular, Renal and Metabolic Diseases, MedImmune, Gaithersburg, MD 20878, United States
| | | | - Paul Warrener
- Cardiovascular, Renal and Metabolic Diseases, MedImmune, Gaithersburg, MD 20878, United States
| | - Bret R Sellman
- Cardiovascular, Renal and Metabolic Diseases, MedImmune, Gaithersburg, MD 20878, United States
| | | | | | - Niels Vrang
- Pharmacology, Gubra, Hørsholm DK-2970, Denmark
| | - James L Trevaskis
- Cardiovascular, Renal and Metabolic Diseases, MedImmune, Gaithersburg, MD 20878, United States
| | | |
Collapse
|
13
|
Andrade N, Andrade S, Silva C, Rodrigues I, Guardão L, Guimarães JT, Keating E, Martel F. Chronic consumption of the dietary polyphenol chrysin attenuates metabolic disease in fructose-fed rats. Eur J Nutr 2019; 59:151-165. [PMID: 30631887 DOI: 10.1007/s00394-019-01895-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Accepted: 01/05/2019] [Indexed: 02/07/2023]
Abstract
PURPOSE Metabolic syndrome (MS) is a major public health issue worldwide and fructose consumption has been associated with MS development. Recently, we showed that the dietary polyphenol chrysin is an effective inhibitor of fructose uptake by human intestinal epithelial cells. Therefore, our aim was to investigate if chrysin interferes with the development of MS induced by fructose in an animal model. METHODS Adult male Sprague-Dawley rats (220-310 g) were randomly divided into four groups: (A) tap water (control), (B) tap water and a daily dose of chrysin (100 mg/kg) by oral administration (chrysin) (C) 10% fructose in tap water (fructose), and (D) 10% fructose in tap water and a daily dose of chrysin (100 mg/kg) by oral administration (fructose + chrysin). All groups were fed ad libitum with standard laboratory chow diet and dietary manipulation lasted 18 weeks. RESULTS Fructose-feeding for 18 weeks induced an increase in serum triacylglycerols, insulin and angiotensin II levels and in hepatic fibrosis and these changes did not occur in fructose + chrysin rats. Moreover, the increase in both systolic and diastolic blood pressure which was found in fructose-fed animals from week 14th onwards was not observed in fructose + chrysin animals. In contrast, the increase in energy consumption, liver/body, heart/body and right kidney/body weight ratios, serum proteins, serum leptin and liver triacylglycerols observed in fructose-fed rats was not affected by chrysin. CONCLUSIONS Chrysin was able to protect against some of the MS features induced by fructose-feeding.
Collapse
Affiliation(s)
- Nelson Andrade
- Unit of Biochemistry, Department of Biomedicine, Faculty of Medicine of Porto, University of Porto, Porto, Portugal
- Instituto de Investigação e Inovação em Saúde (I3S), University of Porto, Porto, Portugal
| | - Sara Andrade
- Unit of Biochemistry, Department of Biomedicine, Faculty of Medicine of Porto, University of Porto, Porto, Portugal
- Instituto de Investigação e Inovação em Saúde (I3S), University of Porto, Porto, Portugal
| | - Claúdia Silva
- Unit of Biochemistry, Department of Biomedicine, Faculty of Medicine of Porto, University of Porto, Porto, Portugal
- Instituto de Investigação e Inovação em Saúde (I3S), University of Porto, Porto, Portugal
| | - Ilda Rodrigues
- Unit of Biochemistry, Department of Biomedicine, Faculty of Medicine of Porto, University of Porto, Porto, Portugal
| | - Luísa Guardão
- Unit of Biochemistry, Department of Biomedicine, Faculty of Medicine of Porto, University of Porto, Porto, Portugal
| | - João T Guimarães
- Unit of Biochemistry, Department of Biomedicine, Faculty of Medicine of Porto, University of Porto, Porto, Portugal
- Department of Clinical Pathology, São João Hospital Centre, Porto, Portugal
- Institute of Public Health, University of Porto, Porto, Portugal
| | - Elisa Keating
- Unit of Biochemistry, Department of Biomedicine, Faculty of Medicine of Porto, University of Porto, Porto, Portugal
- CINTESIS, Center for Research in Health Technologies and Information Systems, University of Porto, Porto, Portugal
| | - Fátima Martel
- Unit of Biochemistry, Department of Biomedicine, Faculty of Medicine of Porto, University of Porto, Porto, Portugal.
- Instituto de Investigação e Inovação em Saúde (I3S), University of Porto, Porto, Portugal.
| |
Collapse
|
14
|
Ge Y, Liu W, Tao H, Zhang Y, Liu L, Liu Z, Qiu B, Xu T. Effect of industrial trans-fatty acids-enriched diet on gut microbiota of C57BL/6 mice. Eur J Nutr 2018; 58:2625-2638. [PMID: 30120538 DOI: 10.1007/s00394-018-1810-2] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2018] [Accepted: 08/13/2018] [Indexed: 02/08/2023]
Abstract
PURPOSE Previous studies have shown that industrially originated trans-fatty acids (iTFAs) are associated with several chronic diseases, but the underlying mechanisms remain unknown. Because gut microbiota play a critical role in human health, diet competent induced gut microbiota dysbiosis may contributing to disease pathogenesis. Therefore, the present study examined the impact of iTFA on gut microbiota, help understanding the underling mechanism of iTFA-associated chronic diseases. METHODS Forty male 8-week-old mice were divided into 4 groups and randomly assigned to diets containing soybean oil (non-iTFA) or partially hydrogenated soybean oil (iTFA). The intervention groups were: (1) low soybean oil (LS); (2) high soybean oil (HS); (3) low partially hydrogenated oil (LH) and (4) high partially hydrogenated oil (HH). The gut microbiota profiles were determined by 16S rRNA gene sequencing. Physiological parameters and the inflammatory status of the small intestine and other tissues were analyzed. Short-chain fatty acid levels in feces were measured using gas chromatography. RESULTS The intake of iTFA increased the abundance of well-documented 'harmful' bacteria, such as Proteobacteria and Desulfovibrionaceae (P < 0.05), whereas it decreased relative abundance of 'beneficial' bacteria, such as Bacteroidetes, Lachnospiraceae, Bacteroidales S24-7 (P < 0.05). Surprisingly, the intake of iTFA increased the abundance of the probiotic Lactobacillaceae (P < 0.05). Additionally, the intake of iTFA induced increase of inflammatory parameters, as well as a numerical decrease of fecal butyric acid and valeric acid. CONCLUSIONS This study, to our knowledge, is the first to demonstrate that the consumption of iTFA resulted in a significant dysbiosis of gut microbiota, which may contribute to the development of chronic diseases associated with iTFA.
Collapse
Affiliation(s)
- Yueting Ge
- Institute of Agro-Food Science and Technology, Shandong Academy of Agricultural Sciences/Key Laboratory of Agro-Products Processing Technology of Shandong Province/Key Laboratory of Novel Food Resources Processing, Ministry of Agriculture, Jinan, China.,The Laboratory of Food Nutrition and Functional Factors, School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Wei Liu
- Institute of Agro-Food Science and Technology, Shandong Academy of Agricultural Sciences/Key Laboratory of Agro-Products Processing Technology of Shandong Province/Key Laboratory of Novel Food Resources Processing, Ministry of Agriculture, Jinan, China.
| | - Haiteng Tao
- State Key Laboratory of Biobased Material and Green Papermaking, College of Food Science and Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan, China
| | - Yu Zhang
- Institute of Agro-Food Science and Technology, Shandong Academy of Agricultural Sciences/Key Laboratory of Agro-Products Processing Technology of Shandong Province/Key Laboratory of Novel Food Resources Processing, Ministry of Agriculture, Jinan, China
| | - Lina Liu
- Institute of Agro-Food Science and Technology, Shandong Academy of Agricultural Sciences/Key Laboratory of Agro-Products Processing Technology of Shandong Province/Key Laboratory of Novel Food Resources Processing, Ministry of Agriculture, Jinan, China
| | - Zhenhua Liu
- School of Public Health and Health Sciences, University of Massachusetts, Amherst, USA
| | - Bin Qiu
- Institute of Agro-Food Science and Technology, Shandong Academy of Agricultural Sciences/Key Laboratory of Agro-Products Processing Technology of Shandong Province/Key Laboratory of Novel Food Resources Processing, Ministry of Agriculture, Jinan, China.
| | - Tongcheng Xu
- Institute of Agro-Food Science and Technology, Shandong Academy of Agricultural Sciences/Key Laboratory of Agro-Products Processing Technology of Shandong Province/Key Laboratory of Novel Food Resources Processing, Ministry of Agriculture, Jinan, China
| |
Collapse
|
15
|
Jeyapal S, Kona SR, Mullapudi SV, Putcha UK, Gurumurthy P, Ibrahim A. Substitution of linoleic acid with α-linolenic acid or long chain n-3 polyunsaturated fatty acid prevents Western diet induced nonalcoholic steatohepatitis. Sci Rep 2018; 8:10953. [PMID: 30026586 PMCID: PMC6053361 DOI: 10.1038/s41598-018-29222-y] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Accepted: 06/21/2018] [Indexed: 02/06/2023] Open
Abstract
Imbalance in the n-6 polyunsaturated fatty acids (PUFA) and n-3 PUFA in the Western diet may increase the risk of nonalcoholic fatty liver disease (NAFLD). This study investigates the impact of substitution of linoleic acid with α-linolenic acid (ALA) or long chain (LC) n-3 PUFA and hence decreasing n-6:n-3 fatty acid ratio on high fat, high fructose (HFHF) diet induced nonalcoholic steatohepatitis (NASH). Male Sprague-Dawley rats were divided into four groups and fed control diet, HFHF diet (n-6:n-3 ratio of 200), HFHF diet with ALA (n-6:n-3 ratio of 2) or HFHF diet with LC n-3 PUFA (n-6:n-3 ratio of 5) for 24 weeks. Rats fed HFHF diet with n-6:n-3 ratio of 200 resulted in hepatic steatosis, induced glucose intolerance, insulin resistance and oxidative stress accompanied by increase in markers of inflammation, plasma lipids and aminotransferase levels. Histopathological examination of liver further confirmed the establishment of NASH. ALA and LC n-3 PUFA supplementation prevented hepatic steatosis and dyslipidemia by inhibiting lipogenesis and increasing insulin sensitivity. Furthermore, n-3 PUFA supplementation attenuated hepatic oxidative stress by restoring antioxidant status, decreased inflammation and preserved hepatic architecture. These finding suggest that decreasing n-6:n-3 ratio prevented HFHF induced NASH by attenuating oxidative stress and inflammation.
Collapse
Affiliation(s)
- Sugeedha Jeyapal
- Department of Lipid Chemistry, National Institute of Nutrition, Hyderabad, India
| | - Suryam Reddy Kona
- Department of Lipid Chemistry, National Institute of Nutrition, Hyderabad, India
| | | | - Uday Kumar Putcha
- Department of Pathology, National Institute of Nutrition, Hyderabad, India
| | | | - Ahamed Ibrahim
- Department of Lipid Chemistry, National Institute of Nutrition, Hyderabad, India.
| |
Collapse
|
16
|
Lambertz J, Berger T, Mak TW, van Helden J, Weiskirchen R. Lipocalin-2 in Fructose-Induced Fatty Liver Disease. Front Physiol 2017; 8:964. [PMID: 29234288 PMCID: PMC5712346 DOI: 10.3389/fphys.2017.00964] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Accepted: 11/13/2017] [Indexed: 12/18/2022] Open
Abstract
The intake of excess dietary fructose most often leads to non-alcoholic fatty liver disease (NAFLD). Fructose is metabolized mainly in the liver and its chronic consumption results in lipogenic gene expression in this organ. However, precisely how fructose is involved in NAFLD progression is still not fully understood, limiting therapy. Lipocalin-2 (LCN2) is a small secreted transport protein that binds to fatty acids, phospholipids, steroids, retinol, and pheromones. LCN2 regulates lipid and energy metabolism in obesity and is upregulated in response to insulin. We previously discovered that LCN2 has a hepatoprotective effect during hepatic insult, and that its upregulation is a marker of liver damage and inflammation. To investigate if LCN2 has impact on the metabolism of fructose and thereby arising liver damage, we fed wild type and Lcn2−/− mice for 4 or 8 weeks on diets that were enriched in fructose either by adding this sugar to the drinking water (30% w/v), or by feeding a chow containing 60% (w/w) fructose. Body weight and daily intake of food and water of these mice was then measured. Fat content in liver sections was visualized using Oil Red O stain, and expression levels of genes involved in fat and sugar metabolism were measured by qRT-PCR and Western blot analysis. We found that fructose-induced steatosis and liver damage was more prominent in female than in male mice, but that the most severe hepatic damage occurred in female mice lacking LCN2. Unexpectedly, consumption of elevated fructose did not induce de novo lipogenesis or fat accumulation. We conclude that LCN2 acts in a lipid-independent manner to protect the liver against fructose-induced damage.
Collapse
Affiliation(s)
- Jessica Lambertz
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry, RWTH University Hospital Aachen, Aachen, Germany
| | - Thorsten Berger
- The Campbell Family Institute for Breast Cancer Research, University Health Network, Toronto, ON, Canada
| | - Tak W Mak
- The Campbell Family Institute for Breast Cancer Research, University Health Network, Toronto, ON, Canada.,Ontario Cancer Institute, University Health Network, Toronto, ON, Canada
| | | | - Ralf Weiskirchen
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry, RWTH University Hospital Aachen, Aachen, Germany
| |
Collapse
|