1
|
Carvalho LCF, Ferreira FM, Dias BV, Azevedo DCD, de Souza GHB, Milagre MM, de Lana M, Vieira PMDA, Carneiro CM, Paula-Gomes SD, Cangussu SD, Costa DC. Silymarin inhibits the lipogenic pathway and reduces worsening of non-alcoholic fatty liver disease (NAFLD) in mice. Arch Physiol Biochem 2024; 130:460-474. [PMID: 36328030 DOI: 10.1080/13813455.2022.2138445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 08/17/2022] [Accepted: 09/08/2022] [Indexed: 11/06/2022]
Abstract
CONTEXT The role of silymarin in hepatic lipid dysfunction and its possible mechanisms of action were investigated. OBJECTIVE To evaluate the effects of silymarin on hepatic and metabolic profiles in mice fed with 30% fructose for 8 weeks. METHODS We evaluated the antioxidant profile of silymarin; mice consumed 30% fructose and were treated with silymarin (120 mg/kg/day or 240 mg/kg/day). We performed biochemical, redox status, and histopathological assays. RT-qPCR was performed to detect ACC-1, ACC-2, FAS, and CS expression, and western blotting to detect PGC-1α levels. RESULTS Silymarin contains high levels of phenolic compounds and flavonoids and exhibited significant antioxidant capacity in vitro. In vivo, the fructose-fed groups showed increased levels of AST, ALT, SOD/CAT, TBARS, hepatic TG, and cholesterol, as well as hypertriglyceridaemia, hypercholesterolaemia, and increased ACC-1 and FAS. Silymarin treatment reduced these parameters and increased mRNA levels and activity of hepatic citrate synthase. CONCLUSIONS These results suggest that silymarin reduces worsening of NAFLD.
Collapse
Affiliation(s)
| | | | - Bruna Vidal Dias
- Laboratório de Bioquímica Metabólica, Universidade Federal de Ouro Preto, Ouro Preto, Brazil
| | | | | | - Matheus Marque Milagre
- Laboratório Doença de Chagas, Universidade Federal de Ouro Preto, UFOP, Ouro Preto, Brazil
| | - Marta de Lana
- Laboratório Doença de Chagas, Universidade Federal de Ouro Preto, UFOP, Ouro Preto, Brazil
| | | | | | - Sílvia de Paula-Gomes
- Laboratório de Bioquímica e Biologia Molecular, Universidade Federal de Ouro Preto, UFOP, Ouro Preto, Brazil
| | - Silvia Dantas Cangussu
- Laboratório de Fisiopatologia Experimental, Universidade Federal de Ouro Preto, UFOP, Ouro Preto, Brazil
| | - Daniela Caldeira Costa
- Laboratório de Bioquímica Metabólica, Universidade Federal de Ouro Preto, Ouro Preto, Brazil
| |
Collapse
|
2
|
Lodge M, Dykes R, Kennedy A. Regulation of Fructose Metabolism in Nonalcoholic Fatty Liver Disease. Biomolecules 2024; 14:845. [PMID: 39062559 PMCID: PMC11274671 DOI: 10.3390/biom14070845] [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: 06/17/2024] [Revised: 07/02/2024] [Accepted: 07/08/2024] [Indexed: 07/28/2024] Open
Abstract
Elevations in fructose consumption have been reported to contribute significantly to an increased incidence of obesity and metabolic diseases in industrial countries. Mechanistically, a high fructose intake leads to the dysregulation of glucose, triglyceride, and cholesterol metabolism in the liver, and causes elevations in inflammation and drives the progression of nonalcoholic fatty liver disease (NAFLD). A high fructose consumption is considered to be toxic to the body, and there are ongoing measures to develop pharmaceutical therapies targeting fructose metabolism. Although a large amount of work has summarized the effects fructose exposure within the intestine, liver, and kidney, there remains a gap in our knowledge regarding how fructose both indirectly and directly influences immune cell recruitment, activation, and function in metabolic tissues, which are essential to tissue and systemic inflammation. The most recent literature demonstrates that direct fructose exposure regulates oxidative metabolism in macrophages, leading to inflammation. The present review highlights (1) the mechanisms by which fructose metabolism impacts crosstalk between tissues, nonparenchymal cells, microbes, and immune cells; (2) the direct impact of fructose on immune cell metabolism and function; and (3) therapeutic targets of fructose metabolism to treat NAFLD. In addition, the review highlights how fructose disrupts liver tissue homeostasis and identifies new therapeutic targets for treating NAFLD and obesity.
Collapse
Affiliation(s)
| | | | - Arion Kennedy
- Department of Molecular and Structural Biochemistry, North Carolina State University, 128 Polk Hall Campus, Box 7622, Raleigh, NC 27695, USA
| |
Collapse
|
3
|
Wu QL, Zeng SX, Peng JY, Yuan Y, Zhu Z, Xie ZC, Huang ZH, Huang JS, Lai JM, Chen JA, Lin MH. Advances in metformin for the treatment of non-alcoholic fatty liver disease in children. Expert Rev Gastroenterol Hepatol 2022; 16:863-877. [PMID: 36039840 DOI: 10.1080/17474124.2022.2118112] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
INTRODUCTION The increased economic and social burdens for NAFLD worldwide make treating such a disease a significant public health issue. Metformin, a kind of insulin sensitizer generally used to treat type 2 diabetes, has been recently found to have efficacy on children's NAFLD in various areas such as glucolipid metabolism, intestinal bacterial metabolism, oxidative stress, and anti-inflammatory response. This article aims to provide an overview of the possible mechanisms of NAFLD in children and the potential therapeutic application of metformin. AREAS COVERED The Cochrane Library, PubMed, Scopus, and EMBASE database was systematically searched on 12 April 2022, using the keywords metformin; non-alcoholic fatty liver disease; and children to identify similar studies. An additional search for recently published research was performed in June 2020. EXPERT OPINION Although metformin has been proved to have an excellent therapeutic effect on children's NAFLD; we can still explore its potential impacts and mechanisms from different angles, such as combined medication. At the same time, we should also pay attention to its side effects.
Collapse
Affiliation(s)
- Qian-Long Wu
- Guangzhou Medical University, Guangzhou, Guangdong province, China
| | - Shu-Xin Zeng
- Guangzhou Medical University, Guangzhou, Guangdong province, China
| | | | | | | | - Zi-Chun Xie
- Guangzhou Medical University, Guangzhou, Guangdong province, China
| | - Ze-Hong Huang
- Guangzhou Medical University, Guangzhou, Guangdong province, China
| | - Jia-Shuan Huang
- Guangzhou Medical University, Guangzhou, Guangdong province, China
| | - Jian-Mei Lai
- Guangzhou Medical University, Guangzhou, Guangdong province, China
| | - Jin-An Chen
- Guangzhou Medical University, Guangzhou, Guangdong province, China
| | - Min-Hua Lin
- Guangzhou Medical University, Guangzhou, Guangdong province, China
| |
Collapse
|
4
|
Buczyńska A, Sidorkiewicz I, Krętowski AJ, Zbucka-Krętowska M, Adamska A. Metformin Intervention—A Panacea for Cancer Treatment? Cancers (Basel) 2022; 14:cancers14051336. [PMID: 35267644 PMCID: PMC8909770 DOI: 10.3390/cancers14051336] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 02/25/2022] [Accepted: 03/04/2022] [Indexed: 02/07/2023] Open
Abstract
The molecular mechanism of action and the individual influence of various metabolic pathways related to metformin intervention are under current investigation. The available data suggest that metformin provides many advantages, exhibiting anti-inflammatory, anti-cancer, hepatoprotective, cardioprotective, otoprotective, radioprotective, and radio-sensitizing properties depending on cellular context. This literature review was undertaken to provide novel evidence concerning metformin intervention, with a particular emphasis on cancer treatment and prevention. Undoubtedly, the pleiotropic actions associated with metformin include inhibiting inflammatory processes, increasing antioxidant capacity, and improving glycemic and lipid metabolism. Consequently, these characteristics make metformin an attractive medicament to translate to human trials, the promising results of which were also summarized in this review.
Collapse
Affiliation(s)
- Angelika Buczyńska
- Clinical Research Centre, Medical University of Bialystok, 15-276 Bialystok, Poland; (I.S.); (A.J.K.)
- Correspondence: (A.B.); (A.A.); Tel.: +48-85-746-8513 (A.B.); +48-85-746-8660 (A.A.)
| | - Iwona Sidorkiewicz
- Clinical Research Centre, Medical University of Bialystok, 15-276 Bialystok, Poland; (I.S.); (A.J.K.)
| | - Adam Jacek Krętowski
- Clinical Research Centre, Medical University of Bialystok, 15-276 Bialystok, Poland; (I.S.); (A.J.K.)
- Department of Endocrinology, Diabetology and Internal Medicine, Medical University of Bialystok, 15-276 Bialystok, Poland
| | - Monika Zbucka-Krętowska
- Department of Gynecological Endocrinology and Adolescent Gynecology, Medical University of Bialystok, 15-276 Bialystok, Poland;
| | - Agnieszka Adamska
- Department of Endocrinology, Diabetology and Internal Medicine, Medical University of Bialystok, 15-276 Bialystok, Poland
- Correspondence: (A.B.); (A.A.); Tel.: +48-85-746-8513 (A.B.); +48-85-746-8660 (A.A.)
| |
Collapse
|
5
|
Omar ZMM, Ahmed AAN, El-Bakry MH, Ahmed MA, Hasan A. Metformin versus Silymarin as Hepatoprotective Agents in Mice Fibrotic Model Caused by Carbon Tetrachloride. ANNALES PHARMACEUTIQUES FRANÇAISES 2022; 80:659-668. [PMID: 35093389 DOI: 10.1016/j.pharma.2022.01.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 12/11/2021] [Accepted: 01/12/2022] [Indexed: 02/07/2023]
Abstract
OBJECTIVES To study metformin hepatoprotective effects compared to silymarin on hepatic fibrosis caused by carbon tetrachloride (CCl4) in mice. MATERIAL AND METHODS liver fibrosis in mice was achieved by intraperitoneal injection of 2 ml/kg of CCl4 dilution in olive oil [1:9 (v/v)] twice a week for 7 weeks followed by oral treatment with metformin (250 mg/kg/day) or silymarin (100 mg/kg/day) (a standard hepatoprotective drug). The changes that follow liver fibrosis were assessed by measurement of hepatic enzymes (ALT, AST and ALP), histopathological examination using hematoxylin and eosin stain, special stains, and α-smooth muscle actin (α-SMA) immunostaining, measuring oxidative stress markers (MDA, GSH, NOx and MnSOD) and transforming growth factor-beta 1 (TGF-β1) in liver. RESULTS liver fibrosis was obviously developed in mice after intraperitoneal injection with CCl4 for 7 weeks. Both silymarin and metformin treatment exhibited a significant decrease in the fibrotic changes and similarly an increase in endogenous antioxidants. Interestingly there is a significant difference between silymarin and metformin regarding both efficacy and potency. CONCLUSION These findings highlight the anti-inflammatory, antioxidant and antifibrotic effects of metformin in CCl4-induced hepatic fibrosis in mice, but silymarin is more beneficial.
Collapse
Affiliation(s)
| | | | | | - Mohammed Ahmed Ahmed
- Department of Pathology, Faculty of Medicine, Al- Azhar University, Assiut, Egypt
| | - Abdulkarim Hasan
- Department of Pathology, Faculty of Medicine, Al- Azhar University, Cairo, Egypt.
| |
Collapse
|
6
|
Ma Y, Lee G, Heo SY, Roh YS. Oxidative Stress Is a Key Modulator in the Development of Nonalcoholic Fatty Liver Disease. Antioxidants (Basel) 2021; 11:antiox11010091. [PMID: 35052595 PMCID: PMC8772974 DOI: 10.3390/antiox11010091] [Citation(s) in RCA: 90] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 12/27/2021] [Accepted: 12/28/2021] [Indexed: 12/14/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is the most common chronic liver disease worldwide, and scientific studies consistently report that NAFLD development can be accelerated by oxidative stress. Oxidative stress can induce the progression of NAFLD to NASH by stimulating Kupffer cells, hepatic stellate cells, and hepatocytes. Therefore, studies are underway to identify the role of antioxidants in the treatment of NAFLD. In this review, we have summarized the origins of reactive oxygen species (ROS) in cells, the relationship between ROS and NAFLD, and have discussed the use of antioxidants as therapeutic agents for NAFLD.
Collapse
Affiliation(s)
- Yuanqiang Ma
- College of Pharmacy and Medical Research Center, Chungbuk National University, Cheongju 28160, Korea; (Y.M.); (G.L.)
| | - Gyurim Lee
- College of Pharmacy and Medical Research Center, Chungbuk National University, Cheongju 28160, Korea; (Y.M.); (G.L.)
| | - Su-Young Heo
- College of Veterinary Medicine, Jeonbuk National University, Jeonju 54896, Korea
- Correspondence: (S.-Y.H.); (Y.-S.R.)
| | - Yoon-Seok Roh
- College of Pharmacy and Medical Research Center, Chungbuk National University, Cheongju 28160, Korea; (Y.M.); (G.L.)
- Correspondence: (S.-Y.H.); (Y.-S.R.)
| |
Collapse
|
7
|
Mandwie M, Karunia J, Niaz A, Keay KA, Musumeci G, Rennie C, McGrath K, Al-Badri G, Castorina A. Metformin Treatment Attenuates Brain Inflammation and Rescues PACAP/VIP Neuropeptide Alterations in Mice Fed a High-Fat Diet. Int J Mol Sci 2021; 22:ijms222413660. [PMID: 34948457 PMCID: PMC8706124 DOI: 10.3390/ijms222413660] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 12/16/2021] [Accepted: 12/17/2021] [Indexed: 12/28/2022] Open
Abstract
High-fat diet (HFD)-induced comorbid cognitive and behavioural impairments are thought to be the result of persistent low-grade neuroinflammation. Metformin, a first-line medication for the treatment of type-2 diabetes, seems to ameliorate these comorbidities, but the underlying mechanism(s) are not clear. Pituitary adenylate cyclase-activating peptide (PACAP) and vasoactive intestinal peptide (VIP) are neuroprotective peptides endowed with anti-inflammatory properties. Alterations to the PACAP/VIP system could be pivotal during the development of HFD-induced neuroinflammation. To unveil the pathogenic mechanisms underlying HFD-induced neuroinflammation and assess metformin’s therapeutic activities, (1) we determined if HFD-induced proinflammatory activity was present in vulnerable brain regions associated with the development of comorbid behaviors, (2) investigated if the PACAP/VIP system is altered by HFD, and (3) assessed if metformin rescues such diet-induced neurochemical alterations. C57BL/6J male mice were divided into two groups to receive either standard chow (SC) or HFD for 16 weeks. A further HFD group received metformin (HFD + M) (300 mg/kg BW daily for 5 weeks) via oral gavage. Body weight, fasting glucose, and insulin levels were measured. After 16 weeks, the proinflammatory profile, glial activation markers, and changes within the PI3K/AKT intracellular pathway and the PACAP/VIP system were evaluated by real-time qPCR and/or Western blot in the hypothalamus, hippocampus, prefrontal cortex, and amygdala. Our data showed that HFD causes widespread low-grade neuroinflammation and gliosis, with regional-specific differences across brain regions. HFD also diminished phospho-AKT(Ser473) expression and caused significant disruptions to the PACAP/VIP system. Treatment with metformin attenuated these neuroinflammatory signatures and reversed PI3K/AKT and PACAP/VIP alterations caused by HFD. Altogether, our findings demonstrate that metformin treatment rescues HFD-induced neuroinflammation in vulnerable brain regions, most likely by a mechanism involving the reinstatement of PACAP/VIP system homeostasis. Data also suggests that the PI3K/AKT pathway, at least in part, mediates some of metformin’s beneficial effects.
Collapse
Affiliation(s)
- Mawj Mandwie
- Laboratory of Cellular and Molecular Neuroscience (LCMN), School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW 2007, Australia; (M.M.); (J.K.); (A.N.); (G.A.-B.)
| | - Jocelyn Karunia
- Laboratory of Cellular and Molecular Neuroscience (LCMN), School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW 2007, Australia; (M.M.); (J.K.); (A.N.); (G.A.-B.)
| | - Aram Niaz
- Laboratory of Cellular and Molecular Neuroscience (LCMN), School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW 2007, Australia; (M.M.); (J.K.); (A.N.); (G.A.-B.)
| | - Kevin A. Keay
- Laboratory of Neural Structure and Function, School of Medical Science (Neuroscience), University of Sydney, Sydney, NSW 2006, Australia;
| | - Giuseppe Musumeci
- Department of Biomedical and Biotechnological Sciences, Anatomy, Histology and Movement Sciences Section, School of Medicine, University of Catania, 95125 Catania, Italy;
| | - Claire Rennie
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW 2007, Australia; (C.R.); (K.M.)
| | - Kristine McGrath
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW 2007, Australia; (C.R.); (K.M.)
| | - Ghaith Al-Badri
- Laboratory of Cellular and Molecular Neuroscience (LCMN), School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW 2007, Australia; (M.M.); (J.K.); (A.N.); (G.A.-B.)
| | - Alessandro Castorina
- Laboratory of Cellular and Molecular Neuroscience (LCMN), School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW 2007, Australia; (M.M.); (J.K.); (A.N.); (G.A.-B.)
- Laboratory of Neural Structure and Function, School of Medical Science (Neuroscience), University of Sydney, Sydney, NSW 2006, Australia;
- Correspondence:
| |
Collapse
|
8
|
Heida A, Gruben N, Catrysse L, Koehorst M, Koster M, Kloosterhuis NJ, Gerding A, Havinga R, Bloks VW, Bongiovanni L, Wolters JC, van Dijk T, van Loo G, de Bruin A, Kuipers F, Koonen DPY, van de Sluis B. The hepatocyte IKK:NF-κB axis promotes liver steatosis by stimulating de novo lipogenesis and cholesterol synthesis. Mol Metab 2021; 54:101349. [PMID: 34626855 PMCID: PMC8581577 DOI: 10.1016/j.molmet.2021.101349] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 09/20/2021] [Accepted: 09/30/2021] [Indexed: 02/06/2023] Open
Abstract
OBJECTIVE Obesity-related chronic inflammation plays an important role in the development of Metabolic Associated Fatty Liver Disease (MAFLD). Although the contribution of the pro-inflammatory NF-κB signaling pathway to the progression from simple steatosis to non-alcoholic steatohepatitis (NASH) is well-established, its role as an initiator of hepatic steatosis and the underlying mechanism remains unclear. Here, we investigated the hypothesis that the hepatocytic NF-κB signaling pathway acts as a metabolic regulator, thereby promoting hepatic steatosis development. METHODS A murine model expressing a constitutively active form of IKKβ in hepatocytes (Hep-IKKβca) was used to activate hepatocyte NF-κB. In addition, IKKβca was also expressed in hepatocyte A20-deficient mice (IKKβca;A20LKO). A20 is an NF-κB-target gene that inhibits the activation of the NF-κB signaling pathway upstream of IKKβ. These mouse models were fed a sucrose-rich diet for 8 weeks. Hepatic lipid levels were measured and using [1-13C]-acetate de novo lipogenesis and cholesterol synthesis rate were determined. Gene expression analyses and immunoblotting were used to study the lipogenesis and cholesterol synthesis pathways. RESULTS Hepatocytic NF-κB activation by expressing IKKβca in hepatocytes resulted in hepatic steatosis without inflammation. Ablation of hepatocyte A20 in Hep-IKKβca mice (IKKβca;A20LKO mice) exacerbated hepatic steatosis, characterized by macrovesicular accumulation of triglycerides and cholesterol, and increased plasma cholesterol levels. Both De novo lipogenesis (DNL) and cholesterol synthesis were found elevated in IKKβca;A20LKO mice. Phosphorylation of AMP-activated kinase (AMPK) - a suppressor in lipogenesis and cholesterol synthesis - was decreased in IKKβca;A20LKO mice. This was paralleled by elevated protein levels of hydroxymethylglutaryl-CoA synthase 1 (HMGCS1) and reduced phosphorylation of HMG-CoA reductase (HMGCR) both key enzymes in the cholesterol synthesis pathway. Whereas inflammation was not observed in young IKKβca;A20LKO mice sustained hepatic NF-κB activation resulted in liver inflammation, together with elevated hepatic and plasma cholesterol levels in middle-aged mice. CONCLUSIONS The hepatocytic IKK:NF-κB axis is a metabolic regulator by controlling DNL and cholesterol synthesis, independent of its central role in inflammation. The IKK:NF-κB axis controls the phosphorylation levels of AMPK and HMGCR and the protein levels of HMGCS1. Chronic IKK-mediated NF-κB activation may contribute to the initiation of hepatic steatosis and cardiovascular disease risk in MAFLD patients.
Collapse
Affiliation(s)
- Andries Heida
- Departments of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Nanda Gruben
- Departments of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Leen Catrysse
- VIB Inflammation Research Center, Ghent University, Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Martijn Koehorst
- Departments of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Mirjam Koster
- Departments of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Niels J Kloosterhuis
- Departments of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Albert Gerding
- Departments of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Rick Havinga
- Departments of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Vincent W Bloks
- Departments of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Laura Bongiovanni
- Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, the Netherlands; Faculty of Veterinary Medicine, University of Teramo, Teramo, Italy
| | - Justina C Wolters
- Departments of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Theo van Dijk
- Departments of Laboratory Medicine, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Geert van Loo
- VIB Inflammation Research Center, Ghent University, Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Alain de Bruin
- Departments of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands; Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, the Netherlands
| | - Folkert Kuipers
- Departments of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands; Departments of Laboratory Medicine, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Debby P Y Koonen
- Departments of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands.
| | - Bart van de Sluis
- Departments of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands.
| |
Collapse
|
9
|
Chen H, Zhou S, Zhu M, Wang B, Chen W, Zheng L, Wang M, Feng W. Gold Nanoparticles Modified With Polyethyleneimine Disturbed the Activity of Drug-Metabolic Enzymes and Induced Inflammation-Mediated Liver Injury in Mice. Front Pharmacol 2021; 12:706791. [PMID: 34335268 PMCID: PMC8321413 DOI: 10.3389/fphar.2021.706791] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Accepted: 06/21/2021] [Indexed: 12/17/2022] Open
Abstract
Gold nanoparticles (GNPs) have been used as a potential bioactive platform for drug delivery due to their unique optical and thermal characteristics. Liver is the main organ in orchestrating physiological homeostasis through metabolization of drugs and detoxification of exogenous substances. Therefore, it is crucial to deeply understand the mechanism of nanoparticle–liver interaction and the potential hepatic effects of GNPs in vivo. In this study, we studied the hepatic impacts of the intravenously injected polyethyleneimine (PEI)-modified GNPs (PEI-GNPs) on the expression of hepatic drug-metabolic enzymes and sterol responsive element binding protein 1c (SREBP-1c)-mediated de novo lipogenesis in mice for 24 h and 1 week. PEI-GNP accumulation in the liver is associated with increased liver inflammation, as evidenced by the gene expression of pro-inflammatory cytokines. Moreover, the GNP-induced hepatotoxicity in mice is partly due to liver inflammation–triggered disruption in the function of drug-metabolic enzymes, including hepatic uptake and efflux transporters, cytochrome P450 (CYP450), and UDP-glucuronosyltransferases (UGTs). The study provides evidence that it is necessary to consider the nanomaterial–liver interaction and manipulate the surface chemistry of GNPs prior to biomedical application of nanoparticles.
Collapse
Affiliation(s)
- Hanqing Chen
- Department of Gastroenterology, Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Shuang Zhou
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Meilin Zhu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing, China.,Institute of Physical Science and Information Technology, Anhui University, Hefei, China
| | - Bing Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing, China
| | - Wei Chen
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Lingna Zheng
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing, China
| | - Meng Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing, China
| | - Weiyue Feng
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing, China
| |
Collapse
|
10
|
Vasques-Monteiro IML, Silva-Veiga FM, Miranda CS, de Andrade Gonçalves ÉCB, Daleprane JB, Souza-Mello V. A rise in Proteobacteria is an indicator of gut-liver axis-mediated nonalcoholic fatty liver disease in high-fructose-fed adult mice. Nutr Res 2021; 91:26-35. [PMID: 34130208 DOI: 10.1016/j.nutres.2021.04.008] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Revised: 04/17/2021] [Accepted: 04/30/2021] [Indexed: 01/12/2023]
Abstract
Current evidence suggests that high fructose intake results in gut dysbiosis, leading to endotoxemia and NAFLD onset. Thus, the hypothesis of the study was that an enhanced Proteobacteria proportion in the cecal microbiota could be the most prominent trigger of NAFLD through enhanced endotoxin (LPS) in adult high-fructose-fed C57BL/6 mice. Male C57BL/6 mice received a control diet (n = 10, C: 76% of energy as carbohydrates, 0% as fructose) or high-fructose diet (n = 10, HFRU: 76% of energy as carbohydrate, 50% as fructose) for 12 weeks. Outcomes included biochemical analyses, 16S rDNA PCR amplification, hepatic stereology, and RT-qPCR. The groups showed similar body masses during the whole experiment. However, the HFRU group showed greater water intake and blood pressure than the C group. The HFRU group showed a significantly lower amount of Bacteroidetes and a predominant rise in Proteobacteria, implying increased LPS. The HFRU group also showed enhanced de novo lipogenesis (Chrebp expression), while beta-oxidation was decreased (Ppar-alpha expression). These results agree with the deposition of fat droplets within hepatocytes and the enhanced hepatic triacylglycerol concentrations, as observed in the photomicrographs, where the HFRU group had a higher volume density of steatosis than the C group. Thus, we confirmed that a rise in the Proteobacteria phylum proportion was the most prominent alteration in gut-liver axis-induced hepatic steatosis in HFRU-fed C57BL/6 mice. Gut dysbiosis and fatty liver were observed even in the absence of overweight in this dietary adult mouse model.
Collapse
Affiliation(s)
- Isabela Macedo Lopes Vasques-Monteiro
- Laboratory of Morphometry, Metabolism, and Cardiovascular Diseases, Biomedical Center, Institute of Biology, Rio de Janeiro State University, Rio de Janeiro, Brazil; Laboratory of bioactive compounds, LABBIO, School of Nutrition, Federal University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Flávia Maria Silva-Veiga
- Laboratory of Morphometry, Metabolism, and Cardiovascular Diseases, Biomedical Center, Institute of Biology, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Carolline Santos Miranda
- Laboratory of Morphometry, Metabolism, and Cardiovascular Diseases, Biomedical Center, Institute of Biology, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | | | - Julio Beltrame Daleprane
- Laboratory for Studies of Interactions Between Nutrition and Genetics, LEING, Institute of Nutrition, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Vanessa Souza-Mello
- Laboratory of Morphometry, Metabolism, and Cardiovascular Diseases, Biomedical Center, Institute of Biology, Rio de Janeiro State University, Rio de Janeiro, Brazil.
| |
Collapse
|
11
|
Duarte-Alonso A, Cu-Cañetas TE, Avila-Nava A, Sansores-España D, Acevedo-Fernández JJ, Sandoval-Peraza M, Chel-Guerrero L, Torre-Villalvazo I. A Cecropia peltata ethanolic extract reduces insulin resistance and hepatic steatosis in rats fed a high-fat diet. JOURNAL OF ETHNOPHARMACOLOGY 2020; 261:113087. [PMID: 32534116 DOI: 10.1016/j.jep.2020.113087] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 05/23/2020] [Accepted: 06/04/2020] [Indexed: 06/11/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Cecropia peltata L. (CP) leaves have been used in Latin American traditional medicine by its purported hypoglycemic, anti-inflammatory and antioxidant properties. PURPOSE The aim of this study was to evaluate the metabolic effects of an ethanolic extract of CP leaves in rats fed a high-fat diet and 10% of sugar in water (HFD). METHODS Male Wistar rats were randomly divided into four groups: group 1 was fed a control diet; groups 2, 3 and 4 were fed a HFD. In addition, group 3 was co-administered with 10 mg/kg/day of CP extract (HFD + CP) and group 4 with a solution of 5 mg/kg/day metformin (HFD + M) for 90 days. RESULTS Body weight gain and serum triglycerides were significantly decreased in the HFD + CP group compared with the HFD and HFD + M groups. Glucose tolerance increased in the HFD + CP compared with the HFD group. Administration with CP extract reduced adipose tissue lipolysis and lipid accumulation in liver of HFD + CP rats with respect to HFD and HFD + M groups. Histological examinations showed that the area of the adipocytes in WAT and the area of lipid vesicles in BAT were significantly smaller in the HFD + CP group than in the HFD and HFD + M groups. CONCLUSION Administration of a CP extract prevented glucose intolerance and hepatic lipid accumulation in rats fed a HFD in association with reduced adipocyte hypertrophy, demonstrating potential antidiabetic properties.
Collapse
Affiliation(s)
- Andrea Duarte-Alonso
- Escuela de Salud, Universidad Modelo, Carretera a Cholul 176, C.P. 97305, Mérida, Yucatán, Mexico.
| | | | - Azalia Avila-Nava
- Hospital Regional de Alta Especialidad de La Península de Yucatán, Calle 7, No. 433, Fracc. Altabrisa, Mérida, C.P. 97130, Yucatán, Mexico.
| | - Delia Sansores-España
- Escuela de Salud, Universidad Modelo, Carretera a Cholul 176, C.P. 97305, Mérida, Yucatán, Mexico.
| | - Juan José Acevedo-Fernández
- Departamento de Fisiología y Fisiopatología, Facultad de Medicina, Universidad Autónoma Del Estado de Morelos, Calle Leñeros S/n, Col. Los Volcanes, Cuernavaca Mor, C.P. 62350, Mexico.
| | - Mukthar Sandoval-Peraza
- Escuela de Ciencias de La Salud. Universidad Del Valle de México, Calle 79 No 500 Col. Dzityá. Altura Km 9.5 de La Carretera a Progreso, C.P. 97302, Mérida, Yucatán, Mexico.
| | - Luis Chel-Guerrero
- Facultad de Ingeniería Química, Universidad Autónoma de Yucatán, Periférico Norte Km. 33.5, Tablaje Catastral 13615, Colonia Chuburná de Hidalgo Inn, 97203, Mérida, Yucatán, Mexico.
| | - Ivan Torre-Villalvazo
- Departamento de Fisiología de La Nutrición, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Av. Vasco de Quiroga No. 15, Belisario Dominguez Sección XVI, 14080, Ciudad de México, Mexico.
| |
Collapse
|
12
|
Spezani R, da Silva RR, Martins FF, de Souza Marinho T, Aguila MB, Mandarim-de-Lacerda CA. Intermittent fasting, adipokines, insulin sensitivity, and hypothalamic neuropeptides in a dietary overload with high-fat or high-fructose diet in mice. J Nutr Biochem 2020; 83:108419. [PMID: 32580132 DOI: 10.1016/j.jnutbio.2020.108419] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 03/14/2020] [Accepted: 05/14/2020] [Indexed: 02/06/2023]
Abstract
The intermittent fasting (IF) might have benefits on metabolism and food intake. Twelve-week old C57BL/6 J mice were fed a control diet (C, 10% kcal fat), a high-fat diet (HF, 50% kcal fat) or a high-fructose diet (HFru, 50% kcal fructose) for 8 weeks, then half of the animals in each group underwent IF (24 h fed, 24 h fasting) for an additional 4 weeks. Although food intake on the fed day remained the same for all groups, all fasting groups showed a reduction in body mass compared to their counterparts. IF reduced total cholesterol, triacylglycerol, fasting glucose, fasting insulin resistance index, and plasma leptin, but increased plasma adiponectin. IF reduced Leptin gene expression in the HF-IF group, but increased proinflammatory markers in the hypothalamus, also in the C-IF group. Both groups HFru-IF and C-IF, showed alterations in the leptin signaling pathway (Leptin, OBRb, and SOCS3), mainly in the HFru-IF group, suggesting leptin resistance. NPY and POMC neuropeptides labeled the neurons of the hypothalamus by immunofluorescence, corroborating qualitatively other quantitative findings of the study. In conclusion, current results are convincing in demonstrating the IF effect on central regulation of food intake control, as shown by NPY and POMC neuropeptide expressions, resulting in a lower weight gain. Besides, IF improves glycemia, lipid metabolism, and consequently insulin and leptin resistance. However, there is increased expression of inflammatory markers in mouse hypothalamus challenged by the HF and HFru diets, which in the long term may induce adverse effects.
Collapse
Affiliation(s)
- Renata Spezani
- Laboratory of Morphometry, Metabolism and Cardiovascular Disease, Biomedical Centre, Institute of Biology, The University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Renata Ribeiro da Silva
- Laboratory of Morphometry, Metabolism and Cardiovascular Disease, Biomedical Centre, Institute of Biology, The University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Fabiane Ferreira Martins
- Laboratory of Morphometry, Metabolism and Cardiovascular Disease, Biomedical Centre, Institute of Biology, The University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Thatiany de Souza Marinho
- Laboratory of Morphometry, Metabolism and Cardiovascular Disease, Biomedical Centre, Institute of Biology, The University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Marcia Barbosa Aguila
- Laboratory of Morphometry, Metabolism and Cardiovascular Disease, Biomedical Centre, Institute of Biology, The University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Carlos A Mandarim-de-Lacerda
- Laboratory of Morphometry, Metabolism and Cardiovascular Disease, Biomedical Centre, Institute of Biology, The University of the State of Rio de Janeiro, Rio de Janeiro, Brazil.
| |
Collapse
|
13
|
Sakamuri A, Sakamuri SSVP, Kona SR, Jeyapal S, Ibrahim A. Diets with low n-6:n-3 PUFA ratio protects rats from fructose-induced dyslipidemia and associated hepatic changes: Comparison between 18:3 n-3 and long-chain n-3 PUFA. Prostaglandins Leukot Essent Fatty Acids 2020; 155:102082. [PMID: 32169807 DOI: 10.1016/j.plefa.2020.102082] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 02/07/2020] [Accepted: 02/18/2020] [Indexed: 12/17/2022]
Abstract
In the present study, we investigated the impact of substituting alpha-linolenic acid (ALA) or long-chain n-3 PUFA (eicosapentaenoic acid and docosahexaenoic acid) for linoleic acid and hence decreasing n-6:n-3 PUFA ratio on high-fructose diet-induced hypertriglyceridemia and associated hepatic changes. Weanling male Wistar rats were divided into four groups and fed with starch-diet (n-6:n-3 PUFA ratio 215:1) and high-fructose diets with different n-6:n-3 PUFA ratio (215:1, 2:1 with ALA and 5:1 with long-chain n-3 PUFA) for twenty-four weeks. Substitution of linoleic acid with ALA (n-6:n-3 PUFA ratio of 2) or long-chain n-3 PUFA (n-6:n-3 PUFA ratio of 5) protected the rats from fructose-induced dyslipidemia, hepatic oxidative stress and corrected lipogenic and proinflammatory gene expression. Both ALA and long-chain n-3 PUFA supplementation also reversed the fructose-induced upregulation of 11β-hydroxysteroid dehydrogenase type 1 (11β-HSD1) gene, which is involved in the generation of active glucocorticoids in tissues. Although both ALA and LC n-3 PUFA prevented fructose-induced dyslipidemia to a similar extent, compared to ALA, LC n-3 PUFA is more effective in preventing hepatic oxidative stress and inflammation.
Collapse
Affiliation(s)
- Anil Sakamuri
- Department of lipid chemistry, National Institute of Nutrition, Hyderabad, India
| | - Siva S V P Sakamuri
- Department of lipid chemistry, National Institute of Nutrition, Hyderabad, India
| | - Suryam Reddy Kona
- Department of lipid chemistry, National Institute of Nutrition, Hyderabad, India
| | - Sugeedha Jeyapal
- Department of lipid chemistry, National Institute of Nutrition, Hyderabad, India
| | - Ahamed Ibrahim
- Department of lipid chemistry, National Institute of Nutrition, Hyderabad, India.
| |
Collapse
|
14
|
Figueiredo ID, Lima TFO, Inácio MD, Costa MC, Assis RP, Brunetti IL, Baviera AM. Lycopene Improves the Metformin Effects on Glycemic Control and Decreases Biomarkers of Glycoxidative Stress in Diabetic Rats. Diabetes Metab Syndr Obes 2020; 13:3117-3135. [PMID: 32982345 PMCID: PMC7495351 DOI: 10.2147/dmso.s265944] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 08/07/2020] [Indexed: 12/16/2022] Open
Abstract
INTRODUCTION Oxidative stress and exacerbated generation of advanced glycation end products (AGEs) participate in the onset of diabetic complications. Lycopene is a potent antioxidant; evidence accounts for its ability to mitigate diabetic disturbances, including the deleterious events of advanced glycation. Therefore, this carotenoid has emerged as a candidate to be used in combination with antidiabetic drugs, such as metformin, attempting to counteract the glycoxidative stress. This study investigated the effects of the treatments with lycopene or metformin, alone or in combination, on glycoxidative stress biomarkers and antioxidant defenses in diabetic rats. METHODS Streptozotocin-induced diabetic rats were treated for 35 days with lycopene (45 mg/kg) or metformin (250 mg/kg), alone or as mixtures in yoghurt. Plasma levels of glucose, triglycerides, cholesterol, thiobarbituric acid reactive substances and protein carbonyl groups (biomarkers of oxidative damage), fluorescent AGEs (biomarkers of advanced glycation), and paraoxonase 1 activity (antioxidant enzyme) were assessed. Changes in the hepatic and renal levels of glycoxidative damage biomarkers and the activities of antioxidant enzymes were investigated. RESULTS The combination of lycopene with metformin maintained the beneficial effects of the isolated treatments, improving the glucose tolerance and lipid profile, lessening biomarkers of oxidative damage, and increasing the paraoxonase 1 activity. Besides, the combined therapy caused further decreases in postprandial glycemia, plasma levels of cholesterol and AGEs, avoided lipid peroxidation (plasma, kidney), and increased antioxidant defenses, mainly the activity of superoxide dismutase (liver, kidney), indicating the maintenance of the lycopene effects. CONCLUSION Lycopene combined with metformin may act synergistically in the control of postprandial glycemia, dyslipidemia and glycoxidative stress, as well as increased antioxidant defenses, arising as a promising therapeutic strategy to mitigate diabetic complications.
Collapse
Affiliation(s)
- Ingrid Delbone Figueiredo
- Department of Clinical Analysis, School of Pharmaceutical Sciences, São Paulo State University (Unesp), Araraquara, São Paulo, Brazil
| | - Tayra Ferreira Oliveira Lima
- Department of Clinical Analysis, School of Pharmaceutical Sciences, São Paulo State University (Unesp), Araraquara, São Paulo, Brazil
| | - Maiara Destro Inácio
- Department of Clinical Analysis, School of Pharmaceutical Sciences, São Paulo State University (Unesp), Araraquara, São Paulo, Brazil
| | - Mariana Campos Costa
- Department of Clinical Analysis, School of Pharmaceutical Sciences, São Paulo State University (Unesp), Araraquara, São Paulo, Brazil
| | - Renata Pires Assis
- Department of Clinical Analysis, School of Pharmaceutical Sciences, São Paulo State University (Unesp), Araraquara, São Paulo, Brazil
| | - Iguatemy Lourenço Brunetti
- Department of Clinical Analysis, School of Pharmaceutical Sciences, São Paulo State University (Unesp), Araraquara, São Paulo, Brazil
| | - Amanda Martins Baviera
- Department of Clinical Analysis, School of Pharmaceutical Sciences, São Paulo State University (Unesp), Araraquara, São Paulo, Brazil
- Correspondence: Amanda Martins Baviera Department of Clinical Analysis, School of Pharmaceutical Sciences, São Paulo State University Rodovia Araraquara Jaú, Km 01 – s/n, Campos Ville, Araraquara, São PauloCEP 14800-903, BrazilTel +55 16 3301 5717Fax +55 16 3322 0073 Email
| |
Collapse
|
15
|
Maia-Ceciliano TC, Dutra RR, Aguila MB, Mandarim-De-Lacerda CA. The deficiency and the supplementation of vitamin D and liver: Lessons of chronic fructose-rich diet in mice. J Steroid Biochem Mol Biol 2019; 192:105399. [PMID: 31175967 DOI: 10.1016/j.jsbmb.2019.105399] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2018] [Revised: 03/25/2019] [Accepted: 06/05/2019] [Indexed: 01/01/2023]
Abstract
The fructose added to soft drinks and processed food, as well as frequent detection of vitamin D deficiency in the body, are two insults increasingly considered to cause lesions in target organs. We studied the liver after a chronic high-fructose diet deficient and supplemented with vitamin D. Sixty C57BL/6 mature male mice were allocated into six groups (n = 10) for ten weeks: control (C), control deficient in vitamin D (CDD), control supplemented with vitamin D (CDS), fructose (F), fructose deficient in vitamin D (FDD), and fructose supplemented with vitamin D (FDS). The gene expressions of vitamin D receptor and CYP27B1 and 25 hydroxyvitamin D plasma level ensured that the diets caused vitamin D deficiency or supplementation. Body mass did not change, but blood pressure (BP) increased in CDD, F, and FDD, whereas BP was controlled in FDS. Insulinemia, insulin tolerance and resistance were seen in both vitamin D deficiency and fructose groups but improved with vitamin D supplementation. The steatosis and fibrosis were observed in the CDD, F and FDD groups. Also, F and FDD showed activation of stellate cells (HSC). Lipogenesis and inflammation gene expressions were enhanced in the CDD, F and FDD groups, but diminished with vitamin D supplementation. In conclusion, we demonstrated the adverse effects of vitamin D deficiency on metabolism, liver steatosis and, combined with fructose intake, liver interstitial fibrosis with hepatic stellate cell activation, and alteration of the lipogenesis, beta-oxidation, and liver inflammation. All these data improved when vitamin D was supplemented in the animals.
Collapse
Affiliation(s)
- Thais C Maia-Ceciliano
- Laboratory of Morphometry, Metabolism and Cardiovascular Disease, Institute of Biology, The University of the State of Rio de Janeiro, Rio de Janeiro, Brazil.
| | - Rafaela R Dutra
- Laboratory of Morphometry, Metabolism and Cardiovascular Disease, Institute of Biology, The University of the State of Rio de Janeiro, Rio de Janeiro, Brazil.
| | - Marcia B Aguila
- Laboratory of Morphometry, Metabolism and Cardiovascular Disease, Institute of Biology, The University of the State of Rio de Janeiro, Rio de Janeiro, Brazil.
| | - Carlos A Mandarim-De-Lacerda
- Laboratory of Morphometry, Metabolism and Cardiovascular Disease, Institute of Biology, The University of the State of Rio de Janeiro, Rio de Janeiro, Brazil.
| |
Collapse
|
16
|
Metformin attenuates the onset of non-alcoholic fatty liver disease and affects intestinal microbiota and barrier in small intestine. Sci Rep 2019; 9:6668. [PMID: 31040374 PMCID: PMC6491483 DOI: 10.1038/s41598-019-43228-0] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Accepted: 04/18/2019] [Indexed: 02/07/2023] Open
Abstract
The antidiabetic drug metformin has been proposed to affect non-alcoholic fatty liver disease (NAFLD) through its effects on intestinal microbiota and barrier function. However, so far most studies focused on long-term effects and more progressed disease stages. The aim of this study was to assess in two experimental settings, if the onset of NAFLD is associated with changes of intestinal microbiota and barrier function and to determine effects of metformin herein. C57Bl/6J mice were fed a liquid control diet (C) or fat-, fructose- and cholesterol-rich diet (FFC) for four days or six weeks ±300 mg/kg BW/day metformin (Met). Markers of liver health, intestinal barrier function and microbiota composition were assessed. Metformin treatment markedly attenuated FFC-induced NAFLD in both experiments with markers of inflammation and lipidperoxidation in livers of FFC + Met-fed mice being almost at the level of controls. Metformin treatment attenuated the loss of tight junction proteins in small intestine and the increase of bacterial endotoxin levels in portal plasma. Changes of intestinal microbiota found in FFC-fed mice were also significantly blunted in FFC + Met-fed mice. Taken together, protective effects of metformin on the onset of NAFLD are associated with changes of intestinal microbiota composition and lower translocation of bacterial endotoxins.
Collapse
|
17
|
Green CJ, Marjot T, Tomlinson JW, Hodson L. Of mice and men: Is there a future for metformin in the treatment of hepatic steatosis? Diabetes Obes Metab 2019; 21:749-760. [PMID: 30456918 DOI: 10.1111/dom.13592] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 11/06/2018] [Accepted: 11/15/2018] [Indexed: 02/06/2023]
Abstract
Non-alcoholic fatty liver disease (NAFLD) encompasses a spectrum of liver diseases, of which the first stage is steatosis. It is one of the most common liver diseases in developed countries and there is a clear association between type 2 diabetes (T2DM) and NAFLD. It is estimated that 70% of people with T2DM have NAFLD and yet there is currently no licensed pharmacological agent to treat it. Whilst lifestyle modification may ameliorate liver fat, it is often difficult to achieve or sustain; thus, there is great interest in pharmacological treatments for NAFLD. Metformin is the first-line medication in the management of T2DM and evidence from animal and human studies has suggested that it may be useful in reducing liver fat via inhibition of lipogenesis and increased fatty acid oxidation. Findings from the majority of studies undertaken in rodent models clearly suggest that metformin may be a powerful therapeutic agent specifically to reduce liver fat accumulation; data from human studies are less convincing. In the present review we discuss the evidence for the specific effects of metformin treatment on liver fat accumulation in animal and human studies, as well as the underlying proposed mechanisms, to try and understand and reconcile the difference in findings between rodent and human work in this area.
Collapse
Affiliation(s)
- Charlotte J Green
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Churchill Hospital, Oxford, UK
| | - Thomas Marjot
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Churchill Hospital, Oxford, UK
| | - Jeremy W Tomlinson
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Churchill Hospital, Oxford, UK
- Oxford NIHR Biomedical Research Centre, Churchill Hospital, Oxford, UK
| | - Leanne Hodson
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Churchill Hospital, Oxford, UK
- Oxford NIHR Biomedical Research Centre, Churchill Hospital, Oxford, UK
| |
Collapse
|
18
|
Bargut TCL, Martins FF, Santos LP, Aguila MB, Mandarim-de-Lacerda CA. Administration of eicosapentaenoic and docosahexaenoic acids may improve the remodeling and browning in subcutaneous white adipose tissue and thermogenic markers in brown adipose tissue in mice. Mol Cell Endocrinol 2019; 482:18-27. [PMID: 30552919 DOI: 10.1016/j.mce.2018.12.003] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 11/14/2018] [Accepted: 12/07/2018] [Indexed: 12/16/2022]
Abstract
The role of eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) in browning and thermogenesis has not been fully elucidated. Thus, we meant to evaluate the effect of EPA and DHA, administered alone or combined, with the activation of browning markers in subcutaneous white adipose tissue (sWAT), and thermogenic markers in brown adipose tissue (BAT). C57BL/6 adult male mice received a control diet or a high-fructose diet (HFru) for eight weeks, but after the first three weeks, HFru was divided into new groups: HFru, HFru + EPA, HFru + DHA, and HFru-EPA + DHA. EPA and DHA diminished adipocyte hypertrophy, recovered markers of browning in sWAT and thermogenic factors in the BAT, and improved gene expressions linked with mitochondrial biogenesis and lipid metabolism. Importantly, EPA and DHA administrated alone showed stronger results than the combination of EPA + DHA. The results suggest that EPA and DHA might be useful as adjuvant strategies to treat metabolic-associated disorders.
Collapse
Affiliation(s)
| | - Fabiane Ferreira Martins
- Laboratory of Morphometry, Metabolism and Cardiovascular Diseases, Biomedical Center, Institute of Biology, State University of Rio de Janeiro, Rio de Janeiro, Brazil.
| | - Larissa Pereira Santos
- Laboratory of Morphometry, Metabolism and Cardiovascular Diseases, Biomedical Center, Institute of Biology, State University of Rio de Janeiro, Rio de Janeiro, Brazil.
| | - Marcia Barbosa Aguila
- Laboratory of Morphometry, Metabolism and Cardiovascular Diseases, Biomedical Center, Institute of Biology, State University of Rio de Janeiro, Rio de Janeiro, Brazil.
| | - Carlos A Mandarim-de-Lacerda
- Laboratory of Morphometry, Metabolism and Cardiovascular Diseases, Biomedical Center, Institute of Biology, State University of Rio de Janeiro, Rio de Janeiro, Brazil.
| |
Collapse
|
19
|
Nguyen TT, Ung TT, Li S, Lian S, Xia Y, Park SY, Do Jung Y. Metformin inhibits lithocholic acid-induced interleukin 8 upregulation in colorectal cancer cells by suppressing ROS production and NF-kB activity. Sci Rep 2019; 9:2003. [PMID: 30765814 PMCID: PMC6376015 DOI: 10.1038/s41598-019-38778-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Accepted: 10/22/2018] [Indexed: 12/29/2022] Open
Abstract
Metformin, an inexpensive, well-tolerated oral agent that is a commonly used first-line treatment for type 2 diabetes, has become the focus of intense research as a potential anticancer agent. In this study, we describe the inhibitory effect of metformin in interleukin 8 (IL-8) upregulation by lithocholic acid (LCA) in HCT116 colorectal cancer (CRC) cells. Pharmacological inhibition studies indicated that reactive oxygen species (ROS) were involved in LCA-induced IL-8 upregulation through activation of the transcription factor NF-κB. Metformin was demonstrated to block LCA-stimulated ROS production, in turn suppressing NF-κB signaling that was critical for IL-8 upregulation. An NADPH oxidase assay proved that the inhibitory effect of metformin on ROS production was derived from its strong suppression of NADPH oxidase, a key producer of ROS in cells. Compared with conditioned media (CM) derived from HCT116 cells treated with LCA, CM derived from HCT116 cells pretreated with metformin and then treated with LCA lost all stimulatory effect on endothelial cell proliferation and tubelike formation. In conclusion, metformin inhibited NADPH oxidase, which in turn suppressed ROS production and NF-κB activation to prevent IL-8 upregulation stimulated by LCA; this prevention thus obstructed endothelial cell proliferation and tubelike formation.
Collapse
Affiliation(s)
- Thi Thinh Nguyen
- Department of Biochemistry, Chonnam National University Medical School, Seoyang Ro 264, Hwasun, Jeonnam, 58138, Korea
| | - Trong Thuan Ung
- Department of Biochemistry, Chonnam National University Medical School, Seoyang Ro 264, Hwasun, Jeonnam, 58138, Korea
| | - Shinan Li
- Department of Biochemistry, Chonnam National University Medical School, Seoyang Ro 264, Hwasun, Jeonnam, 58138, Korea
| | - Sen Lian
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Yong Xia
- Department of Urology, New York University School of Medicine, New York, NY, 10010, USA
| | - Sun Young Park
- Department of Biochemistry, Chonnam National University Medical School, Seoyang Ro 264, Hwasun, Jeonnam, 58138, Korea
| | - Young Do Jung
- Department of Biochemistry, Chonnam National University Medical School, Seoyang Ro 264, Hwasun, Jeonnam, 58138, Korea.
| |
Collapse
|
20
|
Guimarães J, Bargut TCL, Mandarim-de-Lacerda CA, Aguila MB. Medium-chain triglyceride reinforce the hepatic damage caused by fructose intake in mice. Prostaglandins Leukot Essent Fatty Acids 2019; 140:64-71. [PMID: 30553406 DOI: 10.1016/j.plefa.2018.11.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Revised: 11/06/2018] [Accepted: 11/08/2018] [Indexed: 01/16/2023]
Abstract
We aimed to investigate the effects of medium-chain triglyceride oil on the high fructose diet-provoked hepatic abnormalities in mice. We used C57bl/6 mice of 3-months-old divided into four groups for 12 weeks: control (C), control with MCT (C-MCT), fructose (F), and fructose with MCT (F-MCT). We investigated food and water intake, body mass, blood pressure, glucose tolerance, plasma and liver biochemistry, hepatic protein and gene expression. There were no changes in body mass, food intake and glucose tolerance among the groups. The F group presented increased water intake and blood pressure associated with hepatic steatosis and elevated de novo lipogenesis, beta-oxidation, mitochondrial biogenesis and inflammation in the liver. Surprisingly, the C-MCT group also showed hepatic steatosis and inflammation in the liver, and the F-MCT group had no exacerbations of fructose-induced abnormalities, showing marked hepatic steatosis, lipogenesis de novo and hepatic inflammation. The MCT oil groups also presented increased beta-oxidation and mitochondrial biogenesis. In conclusion, MCT oil showed detrimental hepatic effects and should be used with caution, especially in the presence of hepatic alterations.
Collapse
Affiliation(s)
- Janaina Guimarães
- Laboratory of Morphometry, Metabolism and Cardiovascular Diseases, Biomedical Center, Institute of Biology, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Carlos Alberto Mandarim-de-Lacerda
- Laboratory of Morphometry, Metabolism and Cardiovascular Diseases, Biomedical Center, Institute of Biology, State University of Rio de Janeiro, Rio de Janeiro, Brazil.
| | - Marcia Barbosa Aguila
- Laboratory of Morphometry, Metabolism and Cardiovascular Diseases, Biomedical Center, Institute of Biology, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
21
|
Borges CC, Bringhenti I, Mandarim-de-Lacerda CA, Aguila MB. Vitamin D deficiency aggravates the liver metabolism and inflammation in ovariectomized mice. Biomed Pharmacother 2018; 107:878-888. [DOI: 10.1016/j.biopha.2018.08.075] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Revised: 08/04/2018] [Accepted: 08/15/2018] [Indexed: 02/06/2023] Open
|
22
|
BORGES CC, SALLES AF, BRINGHENTI I, MANDARIM-DE-LACERDA CA, AGUILA MB. Vitamin D Deficiency Increases Lipogenesis and Reduces Beta-Oxidation in the Liver of Diet-Induced Obese Mice. J Nutr Sci Vitaminol (Tokyo) 2018; 64:106-115. [DOI: 10.3177/jnsv.64.106] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Affiliation(s)
- Celina Carvalho BORGES
- Laboratory of Morphometry, Metabolism and Cardiovascular Disease, Biomedical Center, Institute of Biology, The University of the State of Rio de Janeiro
| | - Andreza Fernandes SALLES
- Laboratory of Morphometry, Metabolism and Cardiovascular Disease, Biomedical Center, Institute of Biology, The University of the State of Rio de Janeiro
| | - Isabele BRINGHENTI
- Laboratory of Morphometry, Metabolism and Cardiovascular Disease, Biomedical Center, Institute of Biology, The University of the State of Rio de Janeiro
| | - Carlos Alberto MANDARIM-DE-LACERDA
- Laboratory of Morphometry, Metabolism and Cardiovascular Disease, Biomedical Center, Institute of Biology, The University of the State of Rio de Janeiro
| | - Marcia Barbosa AGUILA
- Laboratory of Morphometry, Metabolism and Cardiovascular Disease, Biomedical Center, Institute of Biology, The University of the State of Rio de Janeiro
| |
Collapse
|
23
|
Kim TH, Choi D, Kim JY, Lee JH, Koo SH. Fast food diet-induced non-alcoholic fatty liver disease exerts early protective effect against acetaminophen intoxication in mice. BMC Gastroenterol 2017; 17:124. [PMID: 29179698 PMCID: PMC5704433 DOI: 10.1186/s12876-017-0680-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2017] [Accepted: 11/15/2017] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Acetaminophen (APAP) is a readily available and safe painkiller. However, its overdose is the most common cause of acute liver injury (ALI). Many predisposing factors contribute to susceptibility to APAP-induced ALI. Non-alcoholic fatty liver disease (NAFLD), the major cause of chronic liver disease, is considered an important predictor of APAP-induced ALI, although the exact mechanism controversial. In this study, we aimed to elucidate the effects of NAFLD on APAP-induced ALI. METHODS Two groups of mice, normal chow (NC) diet-fed and fast food (FF) diet-fed mice for 14 weeks, were further divided into two subgroups: intraperitoneally injected with either saline (NC-S and FF-S groups) or APAP (NC-A and FF-A groups). Biochemical tests, histological analysis, quantitative PCR, and western blotting were conducted. RESULTS Alanine aminotransferase (ALT) level (199.0 ± 39.0 vs. 63.8 ± 7.4 IU/L, p < 0.05) and NAFLD activity score (0 vs. 4.5 ± 0.22) were significantly higher in mice in FF-S group than those in NC-S group. ALI features such as ALT level (8447.8 ± 1185.3 vs. 836.6 ± 185.1 IU/L, p < 0.001) and centrizonal necrosis were prominent and mRNA levels of Trib3 (RR, 1.81) was high in mice in the NC-A group. Levels of CYP2E1 and anti-inflammatory molecules such as PPAR-γ, p62, and NRF2 were high in mice in the FF-A group. CONCLUSIONS Our results showed that while the FF diet clearly induced non-alcoholic steatohepatitis and metabolic syndrome, NAFLD also attenuates APAP-induced ALI by inducing anti-inflammatory molecules such as PPAR-γ.
Collapse
Affiliation(s)
- Tae Hyung Kim
- Department of Internal Medicine, Korea University College of Medicine, Seoul, South Korea
| | - Dahee Choi
- Division of Life Sciences, Korea University College of Life Sciences & Biotechnology, 145 Anam-Ro Seongbuk-Gu, Seoul, 02841, South Korea
| | - Joo Young Kim
- Department of Pathology, Korea University College of Medicine, Seoul, South Korea
| | - Jeong Hyeon Lee
- Department of Pathology, Korea University College of Medicine, Seoul, South Korea
| | - Seung-Hoi Koo
- Division of Life Sciences, Korea University College of Life Sciences & Biotechnology, 145 Anam-Ro Seongbuk-Gu, Seoul, 02841, South Korea.
| |
Collapse
|
24
|
Riera-Borrull M, García-Heredia A, Fernández-Arroyo S, Hernández-Aguilera A, Cabré N, Cuyàs E, Luciano-Mateo F, Camps J, Menendez JA, Joven J. Metformin Potentiates the Benefits of Dietary Restraint: A Metabolomic Study. Int J Mol Sci 2017; 18:ijms18112263. [PMID: 29143783 PMCID: PMC5713233 DOI: 10.3390/ijms18112263] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Revised: 10/17/2017] [Accepted: 10/25/2017] [Indexed: 12/22/2022] Open
Abstract
Prevention of the metabolic consequences of a chronic energy-dense/high-fat diet (HFD) represents a public health priority. Metformin is a strong candidate to be incorporated in alternative therapeutic approaches. We used a targeted metabolomic approach to assess changes related to the multi-faceted metabolic disturbances provoked by HFD. We evaluated the protective effects of metformin and explored how pro-inflammatory and metabolic changes respond when mice rendered obese, glucose-intolerant and hyperlipidemic were switched to diet reversal with or without metformin. Mice treated with metformin and diet-reversal showed a dramatically improved protection against HFD-induced hepatic steatosis, a beneficial effect that was accompanied by a lowering of liver-infiltrating pro-inflammatory macrophages and lower release of pro-inflammatory cytokines. Metformin combined with diet reversal promoted effective weight loss along with better glucose control, lowered levels of circulating cholesterol and triglycerides, and reduced adipose tissue content. Our findings underscored the ability of metformin to target the contribution of branched chain amino acids to adipose tissue metabolism while suppressing mitochondrial-dependent biosynthesis in hepatic tissue. The relationship between adipose tissue and liver might provide clinical potential for combining metformin and dietary modifications to protect against the metabolic damage occurring upon excessive dietary fat intake.
Collapse
Affiliation(s)
- Marta Riera-Borrull
- Unitat de Recerca Biomèdica, Hospital Universitari Sant Joan, Institut d'Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, 43201 Reus, Spain.
- Centro de Investigaciones Biológicas (CIB-CSIC), 28040 Madrid, Spain.
| | - Anabel García-Heredia
- Unitat de Recerca Biomèdica, Hospital Universitari Sant Joan, Institut d'Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, 43201 Reus, Spain.
| | - Salvador Fernández-Arroyo
- Unitat de Recerca Biomèdica, Hospital Universitari Sant Joan, Institut d'Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, 43201 Reus, Spain.
- Molecular Oncology Group, Girona Biomedical Research Insitiute (IDIBGI), 17190 Girona, Spain.
| | - Anna Hernández-Aguilera
- Unitat de Recerca Biomèdica, Hospital Universitari Sant Joan, Institut d'Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, 43201 Reus, Spain.
| | - Noemí Cabré
- Unitat de Recerca Biomèdica, Hospital Universitari Sant Joan, Institut d'Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, 43201 Reus, Spain.
| | - Elisabet Cuyàs
- Molecular Oncology Group, Girona Biomedical Research Insitiute (IDIBGI), 17190 Girona, Spain.
- ProCURE (Program against Cancer Therapeutic Resistance), Metabolism and Cancer Group, Catalan Institute of Oncology, 17190 Girona, Spain.
| | - Fedra Luciano-Mateo
- Unitat de Recerca Biomèdica, Hospital Universitari Sant Joan, Institut d'Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, 43201 Reus, Spain.
| | - Jordi Camps
- Unitat de Recerca Biomèdica, Hospital Universitari Sant Joan, Institut d'Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, 43201 Reus, Spain.
| | - Javier A Menendez
- Molecular Oncology Group, Girona Biomedical Research Insitiute (IDIBGI), 17190 Girona, Spain.
- ProCURE (Program against Cancer Therapeutic Resistance), Metabolism and Cancer Group, Catalan Institute of Oncology, 17190 Girona, Spain.
| | - Jorge Joven
- Unitat de Recerca Biomèdica, Hospital Universitari Sant Joan, Institut d'Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, 43201 Reus, Spain.
- The Campus of International Excellence Southern Catalonia, 43003 Tarragona, Spain.
| |
Collapse
|
25
|
Motta VF, Bargut TL, Aguila MB, Mandarim-de-Lacerda CA. Treating fructose-induced metabolic changes in mice with high-intensity interval training: insights in the liver, white adipose tissue, and skeletal muscle. J Appl Physiol (1985) 2017; 123:699-709. [PMID: 28495843 DOI: 10.1152/japplphysiol.00154.2017] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Revised: 05/03/2017] [Accepted: 05/04/2017] [Indexed: 01/06/2023] Open
Abstract
Fructose-rich caloric sweeteners induce adverse changes in the metabolism of humans. The study evaluated the effects of high-intensity interval training (HIIT) on a fructose feeding model, focusing on the liver, white adipose tissue (WAT), skeletal muscle, and their interplay. Male C57BL/6 mice were fed for 18 wk one of the following diets: control (C; 5% of total energy from fructose) or fructose (F; 55% of total energy from fructose). In the 10th week, for an additional 8-wk period, the groups were divided into nontrained (NT) or HIIT groups, totaling four groups: C-NT, C-HIIT, F-NT, and F-HIIT. At the end of the experiment, fructose consumption in the F-NT group led to a high systolic blood pressure, high plasma triglycerides, insulin resistance with glucose intolerance, and lower insulin sensitivity. We also observed liver steatosis, adipocyte hypertrophy, and diminished gene expressions of peroxisome proliferator-activated receptor-γ coactivator 1-α and fibronectin type III domain containing 5 (FNDC5; irisin) in this F-NT group. These results were accompanied by decreased gene expressions of nuclear respiratory factor 1 and mitochondrial transcription factor A (markers of mitochondrial biogenesis), and peroxisome proliferator-activated receptor-α and carnitine palmitoyltransferase 1 (markers of β-oxidation). HIIT improved all of these data in the C-HIIT and F-HIIT groups. In conclusion, in mice fed a fructose diet, HIIT improved body mass, blood pressure, glucose metabolism, and plasma triglycerides. Liver, WAT, and skeletal muscle were positively modulated by HIIT, indicating HIIT as a coadjutant treatment for diseases affecting these tissues.NEW & NOTEWORTHY We investigated the effects of high-intensity interval training (HIIT) in mice fed a fructose-rich diet and the resulting severe negative effect on the liver, white adipose tissue (WAT), and skeletal muscle, which reduced the expression of fibronectin type III domain containing 5 (FNDC5, irisin) and PGC1α and, consequently, affected markers of mitochondrial biogenesis and β-oxidation. Because HIIT may block these adverse effects in all of these three tissues, it might be suggested that it functions as a coadjutant treatment in combatting the alterations caused by high-fructose intake.
Collapse
Affiliation(s)
- Victor F Motta
- Laboratory of Morphometry, Metabolism, and Cardiovascular Diseases, Biomedical Center, Institute of Biology, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Thereza L Bargut
- Laboratory of Morphometry, Metabolism, and Cardiovascular Diseases, Biomedical Center, Institute of Biology, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Marcia B Aguila
- Laboratory of Morphometry, Metabolism, and Cardiovascular Diseases, Biomedical Center, Institute of Biology, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Carlos A Mandarim-de-Lacerda
- Laboratory of Morphometry, Metabolism, and Cardiovascular Diseases, Biomedical Center, Institute of Biology, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|