1
|
Sakr HF, Sirasanagandla SR, Das S, Bima AI, Elsamanoudy AZ. Insulin Resistance and Hypertension: Mechanisms Involved and Modifying Factors for Effective Glucose Control. Biomedicines 2023; 11:2271. [PMID: 37626767 PMCID: PMC10452601 DOI: 10.3390/biomedicines11082271] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Revised: 07/18/2023] [Accepted: 08/01/2023] [Indexed: 08/27/2023] Open
Abstract
Factors such as aging, an unhealthy lifestyle with decreased physical activity, snacking, a standard Western diet, and smoking contribute to raising blood pressure to a dangerous level, increasing the risk of coronary artery disease and heart failure. Atherosclerosis, or aging of the blood vessels, is a physiological process that has accelerated in the last decades by the overconsumption of carbohydrates as the primary sources of caloric intake, resulting in increased triglycerides and VLDL-cholesterol and insulin spikes. Classically, medications ranging from beta blockers to angiotensin II blockers and even calcium channel blockers were used alone or in combination with lifestyle modifications as management tools in modern medicine to control arterial blood pressure. However, it is not easy to control blood pressure or the associated complications. A low-carbohydrate, high-fat (LCHF) diet can reduce glucose and insulin spikes, improve insulin sensitivity, and lessen atherosclerosis risk factors. We reviewed articles describing the etiology of insulin resistance (IR) and its impact on arterial blood pressure from databases including PubMed, PubMed Central, and Google Scholar. We discuss how the LCHF diet is beneficial to maintaining arterial blood pressure at normal levels, slowing down the progression of atherosclerosis, and reducing the use of antihypertensive medications. The mechanisms involved in IR associated with hypertension are also highlighted.
Collapse
Affiliation(s)
- Hussein F. Sakr
- Department of Physiology, College of Medicine and Health Sciences, Sultan Qaboos University, Muscat 123, Oman
| | - Srinivasa Rao Sirasanagandla
- Department of Human and Clinical Anatomy, College of Medicine and Health Sciences, Sultan Qaboos University, Muscat 123, Oman; (S.R.S.); (S.D.)
| | - Srijit Das
- Department of Human and Clinical Anatomy, College of Medicine and Health Sciences, Sultan Qaboos University, Muscat 123, Oman; (S.R.S.); (S.D.)
| | - Abdulhadi I. Bima
- Department of Clinical Biochemistry, Faculty of Medicine, King Abdulaziz University, Jeddah 21465, Saudi Arabia; (A.I.B.); (A.Z.E.)
| | - Ayman Z. Elsamanoudy
- Department of Clinical Biochemistry, Faculty of Medicine, King Abdulaziz University, Jeddah 21465, Saudi Arabia; (A.I.B.); (A.Z.E.)
| |
Collapse
|
2
|
Chałaśkiewicz K, Karaś K, Zakłos-Szyda M, Karwaciak I, Pastwińska J, Koziołkiewicz M, Ratajewski M. Trichostatin a inhibits expression of the human SLC2A5 gene via SNAI1/SNAI2 transcription factors and sensitizes colon cancer cells to platinum compounds. Eur J Pharmacol 2023; 949:175728. [PMID: 37062501 DOI: 10.1016/j.ejphar.2023.175728] [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: 12/24/2022] [Revised: 03/29/2023] [Accepted: 04/14/2023] [Indexed: 04/18/2023]
Abstract
GLUT5, a key protein encoded by the SLC2A5 gene, is involved in the uptake of fructose from the intestine. Currently, with the increased consumption of this sugar and the associated increased incidence of obesity, diabetes and cancer, GLUT5 may represent an important molecular target in the prevention and treatment of these diseases. Here, we demonstrate that overexpression of the SNAI1 and SNAI2 transcription factors in cells expressing high levels of SLC2A5 mRNA reduced SLC2A5 gene expression. Furthermore, a histone deacetylase inhibitor, trichostatin A, which induces SNAI1 and SNAI2 expression, inhibits SLC2A5/GLUT5 expression and sensitizes colon cancer cells to cisplatin and oxaliplatin. This finding might have potential relevance for the development of therapeutic treatments aimed at modulating fructose transport or genes involved in this process for use with certain cancers.
Collapse
Affiliation(s)
- Katarzyna Chałaśkiewicz
- Laboratory of Epigenetics, Institute of Medical Biology, Polish Academy of Sciences, Lodowa 106, 93-232, Lodz, Poland; Faculty of Biotechnology and Food Sciences, Institute of Molecular and Industrial Biotechnology, Lodz University of Technology, Stefanowskiego 2/22, 90-537, Lodz, Poland
| | - Kaja Karaś
- Laboratory of Epigenetics, Institute of Medical Biology, Polish Academy of Sciences, Lodowa 106, 93-232, Lodz, Poland
| | - Małgorzata Zakłos-Szyda
- Faculty of Biotechnology and Food Sciences, Institute of Molecular and Industrial Biotechnology, Lodz University of Technology, Stefanowskiego 2/22, 90-537, Lodz, Poland
| | - Iwona Karwaciak
- Laboratory of Epigenetics, Institute of Medical Biology, Polish Academy of Sciences, Lodowa 106, 93-232, Lodz, Poland
| | - Joanna Pastwińska
- Laboratory of Epigenetics, Institute of Medical Biology, Polish Academy of Sciences, Lodowa 106, 93-232, Lodz, Poland
| | - Maria Koziołkiewicz
- Faculty of Biotechnology and Food Sciences, Institute of Molecular and Industrial Biotechnology, Lodz University of Technology, Stefanowskiego 2/22, 90-537, Lodz, Poland
| | - Marcin Ratajewski
- Laboratory of Epigenetics, Institute of Medical Biology, Polish Academy of Sciences, Lodowa 106, 93-232, Lodz, Poland.
| |
Collapse
|
3
|
Wei R, Deng D, Teng Y, Lu C, Luo Z, Abdulai M, Liu H, Xu H, Li L, Hu S, Hu J, Wei S, Zeng X, Han C. Study on the effect of different types of sugar on lipid deposition in goose fatty liver. Poult Sci 2022; 101:101729. [PMID: 35172237 PMCID: PMC8850742 DOI: 10.1016/j.psj.2022.101729] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Revised: 09/15/2021] [Accepted: 11/04/2021] [Indexed: 01/02/2023] Open
|
4
|
Jiang H, Lin Q, Ma L, Luo S, Jiang X, Fang J, Lu Z. Fructose and fructose kinase in cancer and other pathologies. J Genet Genomics 2021; 48:531-539. [PMID: 34326012 DOI: 10.1016/j.jgg.2021.06.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 06/22/2021] [Accepted: 06/22/2021] [Indexed: 12/14/2022]
Abstract
Fructose metabolism and fructose kinase KHK-C/A are key factors in the development of lipid oversynthesis-promoted metabolic disorders and cancer. Here, we summarize and discuss the current knowledge about the specific features of fructose metabolism and the distinct roles of KHK-C and KHK-A in metabolic liver diseases and their relevant metabolic disorders and cancer, and we highlight the specific protein kinase activity of KHK-A in tumor development. In addition, different approaches that have been used to inhibit KHK and the exploration of KHK inhibitors in clinical treatment are introduced.
Collapse
Affiliation(s)
- Hongfei Jiang
- Cancer Institute of The Affiliated Hospital of Qingdao University and Qingdao Cancer Institute, Qingdao 266061, China
| | - Qian Lin
- Cancer Institute of The Affiliated Hospital of Qingdao University and Qingdao Cancer Institute, Qingdao 266061, China
| | - Leina Ma
- Cancer Institute of The Affiliated Hospital of Qingdao University and Qingdao Cancer Institute, Qingdao 266061, China
| | - Shudi Luo
- Zhejiang University Cancer Center, Hangzhou 310029, China
| | - Xiaoming Jiang
- Zhejiang University Cancer Center, Hangzhou 310029, China
| | - Jing Fang
- Cancer Institute of The Affiliated Hospital of Qingdao University and Qingdao Cancer Institute, Qingdao 266061, China.
| | - Zhimin Lu
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou 310029, China; Zhejiang University Cancer Center, Hangzhou 310029, China.
| |
Collapse
|
5
|
Abstract
Mammals undergo regular cycles of fasting and feeding that engage dynamic transcriptional responses in metabolic tissues. Here we review advances in our understanding of the gene regulatory networks that contribute to hepatic responses to fasting and feeding. The advent of sequencing and -omics techniques have begun to facilitate a holistic understanding of the transcriptional landscape and its plasticity. We highlight transcription factors, their cofactors, and the pathways that they impact. We also discuss physiological factors that impinge on these responses, including circadian rhythms and sex differences. Finally, we review how dietary modifications modulate hepatic gene expression programs.
Collapse
Affiliation(s)
- Lara Bideyan
- Department of Pathology and Laboratory Medicine, and Molecular Biology Institute, University of California at Los Angeles, Los Angeles, California 90095, USA.,Department of Biological Chemistry, and Molecular Biology Institute, University of California at Los Angeles, Los Angeles, California 90095, USA
| | - Rohith Nagari
- Department of Pathology and Laboratory Medicine, and Molecular Biology Institute, University of California at Los Angeles, Los Angeles, California 90095, USA.,Department of Biological Chemistry, and Molecular Biology Institute, University of California at Los Angeles, Los Angeles, California 90095, USA
| | - Peter Tontonoz
- Department of Pathology and Laboratory Medicine, and Molecular Biology Institute, University of California at Los Angeles, Los Angeles, California 90095, USA.,Department of Biological Chemistry, and Molecular Biology Institute, University of California at Los Angeles, Los Angeles, California 90095, USA
| |
Collapse
|
6
|
Creus A, Chicco A, Álvarez SM, Giménez MS, de Lombardo YB. Dietary Salvia hispanica L. reduces cardiac oxidative stress of dyslipidemic insulin-resistant rats. Appl Physiol Nutr Metab 2020; 45:761-768. [PMID: 31935117 DOI: 10.1139/apnm-2019-0769] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Salvia hispanica L., commonly known as chia seed, has beneficial effects upon some signs of metabolic syndrome (MS), such as dyslipidemia and insulin resistance. However, its action on cardiac oxidative stress associated with MS remains unknown. The goal of this study was to analyze the possible beneficial effects of chia seed (variety Salba) upon the oxidative stress of left ventricle heart muscle (LV) of a well-established dyslipidemic insulin-resistant rat model induced by feeding them a sucrose-rich diet (SRD). Male Wistar rats received an SRD for 3 months. After that, for 3 additional months, half of the animals continued with the SRD, while the other half received the SRD containing chia as the source of dietary fat instead corn oil (SRD+chia). In the LV of SRD-fed rats, chia seed improved/reverted the depleted activity of antioxidant enzymes glutathione peroxidase, superoxide dismutase (SOD), and catalase, and ameliorated manganese superoxide dismutase messenger RNA (mRNA) levels increasing the expression of the nuclear factor erythroid 2-related factor 2 (Nrf2). Improved the glutathione redox estate, reactive oxygen species, and thiobarbituric acid reactive substances contents normalizing the p47NOX subunit mRNA level. Furthermore, chia normalized hypertension and plasma levels of pro-inflammatory cytokines and oxidative stress biomarkers. The findings show that chia seed intake impacts positively upon oxidative imbalance of LV of dyslipidemic insulin-resistant rats. Novelty Healthy effects of chia seed involve an improvement of cardiac antioxidant defenses through Nrf2 induction. Chia seed intake reduces cardiac oxidative stress markers of dyslipidemic insulin-resistant rats. Dietary chia seed restores cardiac unbalanced redox state of dyslipidemic insulin-resistant rats.
Collapse
Affiliation(s)
- Agustina Creus
- Department of Biochemistry, School of Biochemistry, University of Litoral, Ciudad Universitaria, Paraje El Pozo, CC 242, (3000) Santa Fe, Argentina
| | - Adriana Chicco
- Department of Biochemistry, School of Biochemistry, University of Litoral, Ciudad Universitaria, Paraje El Pozo, CC 242, (3000) Santa Fe, Argentina
| | - Silvina M Álvarez
- Laboratory of Molecular Biochemistry, Faculty of Chemistry, Biochemistry and Pharmacy, National University of San Luis, Avenida Ejercito de Los Andes 950, and IMIBIO-SL CONICET, San Luis, Argentina
| | - María S Giménez
- Laboratory of Molecular Biochemistry, Faculty of Chemistry, Biochemistry and Pharmacy, National University of San Luis, Avenida Ejercito de Los Andes 950, and IMIBIO-SL CONICET, San Luis, Argentina
| | - Yolanda Bolzón de Lombardo
- Department of Biochemistry, School of Biochemistry, University of Litoral, Ciudad Universitaria, Paraje El Pozo, CC 242, (3000) Santa Fe, Argentina
| |
Collapse
|
7
|
Rodrigo S, Panadero MI, Fauste E, Rodríguez L, Roglans N, Álvarez-Millán JJ, Otero P, Laguna JC, Bocos C. Effects of Maternal Fructose Intake on Perinatal ER-Stress: A Defective XBP1s Nuclear Translocation Affects the ER-stress Resolution. Nutrients 2019; 11:nu11081935. [PMID: 31426466 PMCID: PMC6723662 DOI: 10.3390/nu11081935] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 08/13/2019] [Accepted: 08/15/2019] [Indexed: 12/31/2022] Open
Abstract
Endoplasmic reticulum (ER) homeostasis is crucial to appropriate cell functioning, and when disturbed, a safeguard system called unfolded protein response (UPR) is activated. Fructose consumption modifies ER homeostasis and has been related to metabolic syndrome. However, fructose sweetened beverages intake is allowed during gestation. Therefore, we investigate whether maternal fructose intake affects the ER status and induces UPR. Thus, administrating liquid fructose (10% w/v) to pregnant rats partially activated the ER-stress in maternal and fetal liver and placenta. In fact, a fructose-induced increase in the levels of pIRE1 (phosphorylated inositol requiring enzyme-1) and its downstream effector, X-box binding protein-1 spliced form (XBP1s), was observed. XBP1s is a key transcription factor, however, XBP1s nuclear translocation and the expression of its target genes were reduced in the liver of the carbohydrate-fed mothers, and specifically diminished in the fetal liver and placenta in the fructose-fed mothers. These XBP1s target genes belong to the ER-associated protein degradation (ERAD) system, used to buffer ER-stress and to restore ER-homeostasis. It is known that XBP1s needs to form a complex with diverse proteins to migrate into the nucleus. Since methylglyoxal (MGO) content, a precursor of advanced glycation endproducts (AGE), was augmented in the three tissues in the fructose-fed mothers and has been related to interfere with the functioning of many proteins, the role of MGO in XBP1s migration should not be discarded. In conclusion, maternal fructose intake produces ER-stress, but without XBP1s nuclear migration. Therefore, a complete activation of UPR that would resolve ER-stress is lacking. A state of fructose-induced oxidative stress is probably involved.
Collapse
Affiliation(s)
- Silvia Rodrigo
- Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Montepríncipe, Boadilla del Monte, 28668 Madrid, Spain
| | - María I Panadero
- Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Montepríncipe, Boadilla del Monte, 28668 Madrid, Spain
| | - Elena Fauste
- Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Montepríncipe, Boadilla del Monte, 28668 Madrid, Spain
| | - Lourdes Rodríguez
- Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Montepríncipe, Boadilla del Monte, 28668 Madrid, Spain
| | - Núria Roglans
- Facultad de Farmacia, Universidad de Barcelona, CIBERobn, IBUB, Avda. Joan XXIII 27-31, 08028 Barcelona, Spain
| | | | - Paola Otero
- Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Montepríncipe, Boadilla del Monte, 28668 Madrid, Spain
| | - Juan C Laguna
- Facultad de Farmacia, Universidad de Barcelona, CIBERobn, IBUB, Avda. Joan XXIII 27-31, 08028 Barcelona, Spain
| | - Carlos Bocos
- Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Montepríncipe, Boadilla del Monte, 28668 Madrid, Spain.
| |
Collapse
|
8
|
Zhu H, Wang Z, Wu Y, Jiang H, Zhou F, Xie X, Wang R, Hua C. Untargeted metabonomics reveals intervention effects of chicory polysaccharide in a rat model of non-alcoholic fatty liver disease. Int J Biol Macromol 2019; 128:363-375. [PMID: 30690116 DOI: 10.1016/j.ijbiomac.2019.01.141] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Revised: 01/18/2019] [Accepted: 01/20/2019] [Indexed: 01/01/2023]
Abstract
In the current study, serum metabolomics techniques were used to evaluate the potential mechanism of the effect of chicory polysaccharides in non-alcoholic fatty liver disease (NAFLD) rats. A rat model of NAFLD was constructed according to the histopathological data and biochemical parameters, while the underlying mechanisms of high-fat diet (HFD) induced NAFLD and the therapeutic effects of chicory polysaccharides (CP) were studied by the adoption of serum metabolomics. The serum metabolites were analyzed by GC/MS. Multivariate statistical approaches such as principal component analysis, revealed significant differences with HFD model and CP groups against the control. Results indicated that CP plays a regulatory role in the occurrence of NAFLD. Meantime, a total of 65 candidate biomarkers were screened and identified. Cluster analysis, enrichment analysis and metabolic pathway analysis of differential metabolites also indicated that amino acid metabolism and fatty acid biosynthesis in NAFLD rats, the β-oxidation and urea cycle of very long-chain fatty acids were mainly disturbed when compared against the control group. The corresponding metabolic pathways in the CP group were relieved compared against the NAFLD rats. These results showed that untargeted metabonomics helps to explain intervention effects of chicory polysaccharide with the rat model of NAFLD.
Collapse
Affiliation(s)
- Hua Zhu
- School of Life Sciences, Nanjing Normal University, Nanjing 210023, People's Republic of China; School of Food Science, Nanjing Xiaozhuang University, Nanjing 211171, People's Republic of China; Jiangsu Provincial Key Construction Laboratory of Special Biomass Byproduct Resource Utilization, Nanjing 211171, People's Republic of China
| | - Zhenjiong Wang
- School of Food Science, Nanjing Xiaozhuang University, Nanjing 211171, People's Republic of China; Jiangsu Provincial Key Construction Laboratory of Special Biomass Byproduct Resource Utilization, Nanjing 211171, People's Republic of China.
| | - Yulong Wu
- School of Food Science, Nanjing Xiaozhuang University, Nanjing 211171, People's Republic of China; Jiangsu Provincial Key Construction Laboratory of Special Biomass Byproduct Resource Utilization, Nanjing 211171, People's Republic of China
| | - Haitao Jiang
- School of Food Science, Nanjing Xiaozhuang University, Nanjing 211171, People's Republic of China; Jiangsu Provincial Key Construction Laboratory of Special Biomass Byproduct Resource Utilization, Nanjing 211171, People's Republic of China
| | - Feng Zhou
- School of Food Science, Nanjing Xiaozhuang University, Nanjing 211171, People's Republic of China; Jiangsu Provincial Key Construction Laboratory of Special Biomass Byproduct Resource Utilization, Nanjing 211171, People's Republic of China
| | - Xiaohong Xie
- School of Life Sciences, Nanjing Normal University, Nanjing 210023, People's Republic of China; School of Food Science, Nanjing Xiaozhuang University, Nanjing 211171, People's Republic of China; Jiangsu Provincial Key Construction Laboratory of Special Biomass Byproduct Resource Utilization, Nanjing 211171, People's Republic of China
| | - Renlei Wang
- Biology Department, Jiangsu Second Normal University, Nanjing 210013, People's Republic of China
| | - Chun Hua
- School of Food Science, Nanjing Xiaozhuang University, Nanjing 211171, People's Republic of China; Jiangsu Provincial Key Construction Laboratory of Special Biomass Byproduct Resource Utilization, Nanjing 211171, People's Republic of China.
| |
Collapse
|
9
|
Zecchinati F, Barranco MM, Arana MR, Tocchetti GN, Domínguez CJ, Perdomo VG, Ruiz ML, Mottino AD, García F, Villanueva SSM. Reversion of down-regulation of intestinal multidrug resistance-associated protein 2 in fructose-fed rats by geraniol and vitamin C: Potential role of inflammatory response and oxidative stress. J Nutr Biochem 2019; 68:7-15. [PMID: 31005848 DOI: 10.1016/j.jnutbio.2019.03.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 12/28/2018] [Accepted: 03/12/2019] [Indexed: 12/13/2022]
Abstract
Intestinal multidrug resistance-associated protein 2 is an ABC transporter that limits the absorption of xenobiotics ingested orally, thus acting as essential component of the intestinal biochemical barrier. Metabolic Syndrome (MetS) is a pathological condition characterized by dyslipidemia, hyperinsulinemia, insulin resistance, chronic inflammation, and oxidative stress (OS). In a previous study we demonstrated that MetS-like conditions induced by fructose in drinking water (10% v/v, during 21 days), significantly reduced the expression and activity of intestinal Mrp2 in rats. We here evaluated the potential beneficial effect of geraniol or vitamin C supplementation, natural compounds with anti-inflammatory and anti-oxidant properties, in reverse fructose-induced Mrp2 alterations. After MetS-like conditions were induced (21 days), animals were cotreated with geraniol or vitamin C or vehicle for another 14 days. Decreased expression of Mrp2 protein and mRNA due to fructose administration was reversed by geraniol and by vitamin C, consistent with restoration of Mrp2 activity evaluated in everted intestinal sacs. Concomitantly, increased intestinal IL-1β and IL-6 levels induced by fructose were totally and partially counterbalanced, respectively, by geraniol administration. The intestinal redox unbalance generated by fructose was improved by geraniol and vitamin C, as evidenced by decreasing lipid peroxidation products and activity of Superoxide Dismutase and by normalizing glutathione reduced/oxidized glutathione ratio. The restoration effects exhibited by geraniol and vitamin C suggest that local inflammatory response and OS generated under MetS-like conditions represent important mediators of the intestinal Mrp2 down-regulation. Additionally, both agents could be considered of potential therapeutic value to preserve Mrp2 function under MetS conditions.
Collapse
Affiliation(s)
- Felipe Zecchinati
- Instituto de Fisiología Experimental (IFISE-CONICET)-Facultad de Ciencias Bioquímicas y Farmacéuticas-Universidad Nacional de Rosario, Rosario, Santa Fe, Argentina
| | - Maria Manuela Barranco
- Laboratorio de Fisiología Metabólica-Facultad de Ciencias Médicas-Universidad Nacional de Rosario-CONICET, Rosario, Santa Fe, Argentina
| | - Maite Rocío Arana
- Instituto de Fisiología Experimental (IFISE-CONICET)-Facultad de Ciencias Bioquímicas y Farmacéuticas-Universidad Nacional de Rosario, Rosario, Santa Fe, Argentina
| | - Guillermo Nicolás Tocchetti
- Instituto de Fisiología Experimental (IFISE-CONICET)-Facultad de Ciencias Bioquímicas y Farmacéuticas-Universidad Nacional de Rosario, Rosario, Santa Fe, Argentina
| | - Camila Juliana Domínguez
- Instituto de Fisiología Experimental (IFISE-CONICET)-Facultad de Ciencias Bioquímicas y Farmacéuticas-Universidad Nacional de Rosario, Rosario, Santa Fe, Argentina
| | - Virginia Gabriela Perdomo
- Instituto de Biología Molecular y Celular de Rosario (IBR-CONICET)-Facultad de Ciencias Bioquímicas y Farmacéuticas-Universidad Nacional de Rosario, Rosario, Santa Fe, Argentina
| | - María Laura Ruiz
- Instituto de Fisiología Experimental (IFISE-CONICET)-Facultad de Ciencias Bioquímicas y Farmacéuticas-Universidad Nacional de Rosario, Rosario, Santa Fe, Argentina
| | - Aldo Domingo Mottino
- Instituto de Fisiología Experimental (IFISE-CONICET)-Facultad de Ciencias Bioquímicas y Farmacéuticas-Universidad Nacional de Rosario, Rosario, Santa Fe, Argentina
| | - Fabiana García
- Laboratorio de Fisiología Metabólica-Facultad de Ciencias Médicas-Universidad Nacional de Rosario-CONICET, Rosario, Santa Fe, Argentina
| | - Silvina Stella Maris Villanueva
- Instituto de Fisiología Experimental (IFISE-CONICET)-Facultad de Ciencias Bioquímicas y Farmacéuticas-Universidad Nacional de Rosario, Rosario, Santa Fe, Argentina.
| |
Collapse
|
10
|
Abd Eldaim MA, Ibrahim FM, Orabi SH, Hassan A, El Sabagh HS. l-Carnitine-induced amelioration of HFD-induced hepatic dysfunction is accompanied by a reduction in hepatic TNF-α and TGF-β1. Biochem Cell Biol 2018; 96:713-725. [PMID: 29677453 DOI: 10.1139/bcb-2018-0074] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/02/2023] Open
Abstract
In this study, we evaluated the possible mechanisms through which l-carnitine ameliorates the adverse effects from obesity in rats, induced with a high-fat diet (HFD). For this, 56 albino Wister rats were randomly assigned to 7 groups. The control group was fed a basal diet and injected with saline. The second group was fed the basal diet and injected with l-carnitine (200 mg/kg body mass, by intraperitoneal injection; i.p.). The third group were fed the HFD. The fourth group was fed the HFD and injected with l-carnitine (200 mg/kg body mass, i.p.) for 8 weeks. The fifth group was fed the HFD for 10 weeks. The sixth group were fed the HFD for 10 weeks and were also injected with l-carnitine (200 mg/kg body mass, i.p.) during the final 2 weeks. The seventh group was fed the HFD diet for 8 weeks then the basal diet for 2 weeks. The HFD induced significantly increased levels of hyperglycemia, lipid peroxidation, pathological changes, TNF-α and TGF-β1 protein expression in hepatic tissue, food intake, body weight gain, serum levels of total and non-high-density lipoprotein cholesterol, ketone bodies, triacylglycerol, urea, creatinine, AST, and ALT. However, the HFD diet significantly decreased serum levels of high-density lipoprotein (HDL) and hepatic levels of reduced glutathione. l-Carnitine ameliorated the effects of the HFD on the above-mentioned parameters. This study indicated that l-carnitine had protective and curative effects against HFD-induced hepatosteatosis by reducing hepatic oxidative stress and protein expression of TNF-α and TGF-β1.
Collapse
Affiliation(s)
- Mabrouk Attia Abd Eldaim
- Department of Biochemistry and Chemistry of Nutrition, Faculty of Veterinary Medicine, Menoufia University, Sheben Elkom, Egypt
- Division of Applied Biosciences, Graduate School of Agriculture, Kyoto University, 606-8502, Japan
| | - Fatma Mohamed Ibrahim
- Department of Biochemistry and Chemistry of Nutrition, Faculty of Veterinary Medicine, University of Sadat City, Sadat City, Egypt
| | - Saher Hassan Orabi
- Department of Biochemistry and Chemistry of Nutrition, Faculty of Veterinary Medicine, University of Sadat City, Sadat City, Egypt
| | - Azza Hassan
- Department of Pathology, Faculty of Veterinary Medicine, Cairo University, Cairo, Egypt
| | - Hesham Saad El Sabagh
- Department of Toxicology and Fronsic Medicine, Faculty of Veterinary Medicine, University of Sadat City, Sadat City, Egypt
| |
Collapse
|
11
|
Selenscig D, Ferreira MDR, Chicco A, Lombardo YB. Dietary fish oil ameliorates adipose tissue dysfunction in insulin-resistant rats fed a sucrose-rich diet improving oxidative stress, peroxisome proliferator-activated receptor γ and uncoupling protein 2. Food Funct 2018; 9:2496-2507. [PMID: 29645025 DOI: 10.1039/c7fo01993a] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
This work aims to assess the possible beneficial effects of dietary fish oil (FO) on the pre-existing adipose tissue dysfunction through the improvement or reversion of the mechanisms underlying oxidative stress and pro-inflammatory cytokines in dyslipemic insulin-resistant rats. Wistar rats were fed a sucrose rich diet (SRD) for 6 months. After that half of the animals continued with the SRD until month 8 while in the other half corn oil was replaced by FO for 2 months (SRD + FO). A reference group consumed a control diet all the time. In an epididymal fat pad, we analyzed antioxidant and oxidant enzyme activities, ROS content, glutathione redox state, the protein level of peroxisome proliferator-activated receptor gamma (PPARγ) and the expression and protein levels of uncoupling protein 2 (UCP2) as well as oxidative stress biomarkers and TNF-α and IL-6 plasma levels. Besides these, insulin sensitivity and the composition of fatty acid phospholipids of adipose tissue were measured. Compared with the SRD the SRD + FO fed group showed a decrease of fat pad weight and the antioxidant and oxidant enzyme activities and ROS content returned to control values along with normal plasma TNF-α and IL-6 levels. FO normalized both the decrease of PPARγ protein and the increase of protein and expression of UCP2. Furthermore, FO increased the n-3/n-6 fatty acid ratio in the adipose tissue phospholipids and normalized dyslipidemia and insulin resistance. Finally, these findings reinforce the view that dietary FO may exert a beneficial effect in ameliorating the dyslipidemia and insulin resistance in this animal model.
Collapse
Affiliation(s)
- Dante Selenscig
- Department of Biochemistry, School of Biochemistry, University of Litoral, Ciudad Universitaria, Paraje El Pozo, CC 242 (3000) Santa Fe and (CONICET) Consejo Nacional de Investigaciones Científicas y Técnicas, Santa Fe, Argentina.
| | | | | | | |
Collapse
|
12
|
Trouwborst I, Bowser SM, Goossens GH, Blaak EE. Ectopic Fat Accumulation in Distinct Insulin Resistant Phenotypes; Targets for Personalized Nutritional Interventions. Front Nutr 2018; 5:77. [PMID: 30234122 PMCID: PMC6131567 DOI: 10.3389/fnut.2018.00077] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Accepted: 08/15/2018] [Indexed: 12/13/2022] Open
Abstract
Cardiometabolic diseases are one of the leading causes for disability and mortality in the Western world. The prevalence of these chronic diseases is expected to rise even further in the next decades. Insulin resistance (IR) and related metabolic disturbances are linked to ectopic fat deposition, which is the storage of excess lipids in metabolic organs such as liver and muscle. Notably, a vicious circle exists between IR and ectopic fat, together increasing the risk for the development of cardiometabolic diseases. Nutrition is a key-determining factor for both IR and ectopic fat deposition. The macronutrient composition of the diet may impact metabolic processes related to ectopic fat accumulation and IR. Interestingly, however, the metabolic phenotype of an individual may determine the response to a certain diet. Therefore, population-based nutritional interventions may not always lead to the most optimal (cardiometabolic) outcomes at the individual level, and differences in the metabolic phenotype may underlie conflicting findings related to IR and ectopic fat in dietary intervention studies. Detailed metabolic phenotyping will help to better understand the complex relationship between diet and metabolic regulation, and to optimize intervention outcomes. A subgroup-based approach that integrates, among others, tissue-specific IR, cardiometabolic parameters, anthropometrics, gut microbiota, age, sex, ethnicity, and psychological factors may thereby increase the efficacy of dietary interventions. Nevertheless, the implementation of more personalized nutrition may be complex, costly, and time consuming. Future studies are urgently warranted to obtain insight into a more personalized approach to nutritional interventions, taking into account the metabolic phenotype to ultimately improve insulin sensitivity and reduce the risk for cardiometabolic diseases.
Collapse
Affiliation(s)
- Inez Trouwborst
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, Netherlands
| | - Suzanne M Bowser
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, Netherlands
| | - Gijs H Goossens
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, Netherlands
| | - Ellen E Blaak
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, Netherlands
| |
Collapse
|
13
|
Stanhope KL, Goran MI, Bosy-Westphal A, King JC, Schmidt LA, Schwarz JM, Stice E, Sylvetsky AC, Turnbaugh PJ, Bray GA, Gardner CD, Havel PJ, Malik V, Mason AE, Ravussin E, Rosenbaum M, Welsh JA, Allister-Price C, Sigala DM, Greenwood MRC, Astrup A, Krauss RM. Pathways and mechanisms linking dietary components to cardiometabolic disease: thinking beyond calories. Obes Rev 2018; 19:1205-1235. [PMID: 29761610 PMCID: PMC6530989 DOI: 10.1111/obr.12699] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2017] [Revised: 03/09/2018] [Accepted: 03/31/2018] [Indexed: 12/11/2022]
Abstract
Calories from any food have the potential to increase risk for obesity and cardiometabolic disease because all calories can directly contribute to positive energy balance and fat gain. However, various dietary components or patterns may promote obesity and cardiometabolic disease by additional mechanisms that are not mediated solely by caloric content. Researchers explored this topic at the 2017 CrossFit Foundation Academic Conference 'Diet and Cardiometabolic Health - Beyond Calories', and this paper summarizes the presentations and follow-up discussions. Regarding the health effects of dietary fat, sugar and non-nutritive sweeteners, it is concluded that food-specific saturated fatty acids and sugar-sweetened beverages promote cardiometabolic diseases by mechanisms that are additional to their contribution of calories to positive energy balance and that aspartame does not promote weight gain. The challenges involved in conducting and interpreting clinical nutritional research, which preclude more extensive conclusions, are detailed. Emerging research is presented exploring the possibility that responses to certain dietary components/patterns are influenced by the metabolic status, developmental period or genotype of the individual; by the responsiveness of brain regions associated with reward to food cues; or by the microbiome. More research regarding these potential 'beyond calories' mechanisms may lead to new strategies for attenuating the obesity crisis.
Collapse
Affiliation(s)
- K L Stanhope
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, CA, USA
| | - M I Goran
- Department of Preventive Medicine, Diabetes and Obesity Research Institute, University of Southern California, Los Angeles, CA, USA
| | - A Bosy-Westphal
- Institute of Human Nutrition and Food Science, Christian-Albrechts-Universität zu Kiel, Kiel, Germany
| | - J C King
- Children's Hospital Oakland Research Institute, Oakland, CA, USA
| | - L A Schmidt
- Philip R. Lee Institute for Health Policy Studies, University of California, San Francisco, San Francisco, CA, USA
- California Clinical and Translational Science Institute, University of California, San Francisco, San Francisco, CA, USA
- Department of Anthropology, History, and Social Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - J-M Schwarz
- Touro University, Vallejo, CA, USA
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - E Stice
- Oregon Research Institute, Eugene, OR, USA
| | - A C Sylvetsky
- Department of Exercise and Nutrition Sciences, Milken Institute School of Public Health, The George Washington University, Washington, DC, USA
| | - P J Turnbaugh
- Department of Microbiology and Immunology, G.W. Hooper Research Foundation, University of California, San Francisco, San Francisco, CA, USA
| | - G A Bray
- Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, LA, USA
| | - C D Gardner
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - P J Havel
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, CA, USA
- Department of Nutrition, University of California, Davis, Davis, CA, USA
| | - V Malik
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - A E Mason
- Department of Psychiatry, Osher Center for Integrative Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - E Ravussin
- Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, LA, USA
| | - M Rosenbaum
- Division of Molecular Genetics, Department of Pediatrics, Columbia University, New York, NY, USA
| | - J A Welsh
- Department of Pediatrics, Emory University School of Medicine, Wellness Department, Children's Healthcare of Atlanta, Nutrition and Health Sciences Doctoral Program, Laney Graduate School, Emory University, Atlanta, GA, USA
| | - C Allister-Price
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, CA, USA
| | - D M Sigala
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, CA, USA
| | - M R C Greenwood
- Department of Nutrition, University of California, Davis, Davis, CA, USA
| | - A Astrup
- Department of Nutrition, Exercise, and Sports, Faculty of Sciences, University of Copenhagen, Copenhagen, Denmark
| | - R M Krauss
- Children's Hospital Oakland Research Institute, Oakland, CA, USA
| |
Collapse
|
14
|
Carvalho DV, Santos FA, de Lima RP, Viana AFSC, Fonseca SGC, Nunes PIG, de Melo TS, Gallão MI, de Brito ES. Influence of low molecular weight compounds associated to cashew ( Anacardium occidentale L.) fiber on lipid metabolism, glycemia and insulinemia of normal mice. ACTA ACUST UNITED AC 2018. [DOI: 10.1016/j.bcdf.2017.12.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
|
15
|
Seraphim DCC, Punaro GR, Fernandes TDO, Ginoza M, Lopes GS, Higa EMS. Assessment of fructose overload in the metabolic profile and oxidative/nitrosative stress in the kidney of senescent female rats. Exp Gerontol 2017; 99:53-60. [PMID: 28939527 DOI: 10.1016/j.exger.2017.09.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Revised: 09/16/2017] [Accepted: 09/18/2017] [Indexed: 11/27/2022]
Abstract
The aging process is a complex phenomenon that leads the body to several changes, affecting its integrity and resulting in chronic pathologies, which compromises health and quality of life of elderly people. Animals supplemented with fructose have been used as an experimental model for induction of insulin resistance. The objective of this study was to evaluate the metabolic effects and the levels of oxidative/nitrosative stress in the kidney of senescent rats with a high fructose intake. The animals were allocated into 4 groups: young control (Y), aged control (A), young fructose (YF) and aged fructose (AF). Groups Y and A received water and groups YF and AF received fructose (100g/L) in the water, both ad libitum. After 12weeks of high fructose intake, the animals were sacrificed to collect their kidneys, blood and the thoracic aorta. The results are presented as mean±SE, analyzed by the One-Way ANOVA test with Newman-Keuls post-test; significant at p<0.05. The fructose overload caused metabolic dysfunctions and insulin resistance, confirming the efficacy of the chosen model. In this study, we observed a body weight gain in the studied groups (except in the elderly fructose group), and an increase in general caloric intake, diuresis and adipose tissue; insulin resistance, increased fasting glucose, triglycerides and cholesterol in the fructose groups. We also found a loss of renal function, increased oxidative/nitrosative stress and inflammation, and a reduction of antioxidants and a lower vasodepressor response in the studied groups, especially those who consumed fructose. In summary, our data showed that aging or high fructose intake contributed to the increase of oxidative/nitrosative stress in animals, demonstrating that at the dose and the period of fructose treatment utilized in this study, fructose was not able to aggravate several aspects which were already altered by aging. We believe that the high fructose intake simulates most of the effects of aging, and this understanding would be useful to prevent or minimize many of the alterations caused by this condition.
Collapse
Affiliation(s)
- Deborah Chianelli Costalonga Seraphim
- Translational Medicine, Department of Medicine, Universidade Federal de Sao Paulo, Sao Paulo, Brazil; Laboratory of Nitric Oxide and Oxidative Stress, Nephrology Division, Universidade Federal de Sao Paulo, Sao Paulo, Brazil
| | - Giovana Rita Punaro
- Nephrology Division, Universidade Federal de Sao Paulo, Sao Paulo, Brazil; Laboratory of Nitric Oxide and Oxidative Stress, Nephrology Division, Universidade Federal de Sao Paulo, Sao Paulo, Brazil
| | | | - Milton Ginoza
- Nephrology Division, Universidade Federal de Sao Paulo, Sao Paulo, Brazil
| | | | - Elisa Mieko Suemitsu Higa
- Translational Medicine, Department of Medicine, Universidade Federal de Sao Paulo, Sao Paulo, Brazil; Nephrology Division, Universidade Federal de Sao Paulo, Sao Paulo, Brazil; Emergency Division, Universidade Federal de Sao Paulo, Sao Paulo, Brazil; Laboratory of Nitric Oxide and Oxidative Stress, Nephrology Division, Universidade Federal de Sao Paulo, Sao Paulo, Brazil.
| |
Collapse
|
16
|
Abbasi Oshaghi E, Goodarzi MT, Higgins V, Adeli K. Role of resveratrol in the management of insulin resistance and related conditions: Mechanism of action. Crit Rev Clin Lab Sci 2017; 54:267-293. [DOI: 10.1080/10408363.2017.1343274] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Ebrahim Abbasi Oshaghi
- Department of Clinical Biochemistry, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Mohammad Taghi Goodarzi
- Department of Clinical Biochemistry, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Victoria Higgins
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Khosrow Adeli
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
- Department of Biochemistry, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
17
|
Green CL, Mitchell SE, Derous D, Wang Y, Chen L, Han JDJ, Promislow DEL, Lusseau D, Douglas A, Speakman JR. The effects of graded levels of calorie restriction: IX. Global metabolomic screen reveals modulation of carnitines, sphingolipids and bile acids in the liver of C57BL/6 mice. Aging Cell 2017; 16:529-540. [PMID: 28139067 PMCID: PMC5418186 DOI: 10.1111/acel.12570] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/23/2016] [Indexed: 12/12/2022] Open
Abstract
Calorie restriction (CR) remains the most robust intervention to extend lifespan and improve health span. Using a global mass spectrometry-based metabolomic approach, we identified 193 metabolites that were significantly differentially expressed (SDE) in the livers of C57BL/6 mice, fed graded levels of CR (10, 20, 30 and 40% CR) compared to mice fed ad libitum for 12 h a day. The differential expression of metabolites also varied with the different feeding groups. Pathway analysis revealed that graded CR had an impact on carnitine synthesis and the carnitine shuttle pathway, sphingosine-1-phosphate (S1P) signalling and methionine metabolism. S1P, sphingomyelin and L-carnitine were negatively correlated with body mass, leptin, insulin-like growth factor- 1 (IGF-1) and major urinary proteins (MUPs). In addition, metabolites which showed a graded effect, such as ceramide, S1P, taurocholic acid and L-carnitine, responded in the opposite direction to previously observed age-related changes. We suggest that the modulation of this set of metabolites may improve liver processes involved in energy release from fatty acids. S1P also negatively correlated with catalase activity and body temperature, and positively correlated with food anticipatory activity. Injecting mice with S1P or an S1P receptor 1 agonist did not precipitate changes in body temperature, physical activity or food intake suggesting that these correlations were not causal relationships.
Collapse
Affiliation(s)
- Cara L. Green
- Institute of Biological and Environmental Sciences; University of Aberdeen; Aberdeen UK
| | - Sharon E. Mitchell
- Institute of Biological and Environmental Sciences; University of Aberdeen; Aberdeen UK
| | - Davina Derous
- Institute of Biological and Environmental Sciences; University of Aberdeen; Aberdeen UK
| | - Yingchun Wang
- State Key Laboratory of Molecular Developmental Biology; Institute of Genetics and Developmental Biology; Chinese Academy of Sciences; Chaoyang Beijing China
| | - Luonan Chen
- Key Laboratory of Systems Biology, Innovation Center for Cell Signaling Network; Institute of Biochemistry and Cell Biology; Shanghai Institute of Biological Sciences; Chinese Academy of Sciences; Shanghai China
| | - Jing-Dong J. Han
- Key Laboratory of Computational Biology; Chinese Academy of Sciences-Max Planck Partner Institute for Computational Biology; Shanghai Institutes for Biological Sciences; Chinese Academy of Sciences; Shanghai China
| | - Daniel E. L. Promislow
- Department of Pathology and Department of Biology; University of Washington; Seattle WA USA
| | - David Lusseau
- Institute of Biological and Environmental Sciences; University of Aberdeen; Aberdeen UK
| | - Alex Douglas
- Institute of Biological and Environmental Sciences; University of Aberdeen; Aberdeen UK
| | - John R. Speakman
- Institute of Biological and Environmental Sciences; University of Aberdeen; Aberdeen UK
- State Key Laboratory of Molecular Developmental Biology; Institute of Genetics and Developmental Biology; Chinese Academy of Sciences; Chaoyang Beijing China
| |
Collapse
|
18
|
Pereira CD, Passos E, Severo M, Vitó I, Wen X, Carneiro F, Gomes P, Monteiro R, Martins MJ. Ingestion of a natural mineral-rich water in an animal model of metabolic syndrome: effects in insulin signalling and endoplasmic reticulum stress. Horm Mol Biol Clin Investig 2017; 26:135-50. [PMID: 26741353 DOI: 10.1515/hmbci-2015-0033] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2015] [Accepted: 11/18/2015] [Indexed: 12/15/2022]
Abstract
BACKGROUND High-fructose and/or low-mineral diets are relevant in metabolic syndrome (MS) development. Insulin resistance (IR) represents a central mechanism in MS development. Glucocorticoid signalling dysfunction and endoplasmic reticulum (ER) and oxidative stresses strongly contribute to IR and associate with MS. We have described that natural mineral-rich water ingestion delays fructose-induced MS development, modulates fructose effects on the redox state and glucocorticoid signalling and increases sirtuin 1 expression. Here, we investigated mineral-rich water ingestion effects on insulin signalling and ER homeostasis of fructose-fed rats. MATERIALS AND METHODS Adult male Sprague-Dawley rats had free access to standard-chow diet and different drinking solutions (8 weeks): tap water (CONT), 10%-fructose/tap water (FRUCT) or 10%-fructose/mineral-rich water (FRUCTMIN). Hepatic and adipose (visceral, VAT) insulin signalling and hepatic ER homeostasis (Western blot or PCR) as well as hepatic lipid accumulation were evaluated. RESULTS Hepatic p-IRS1Ser307/IRS1 (tendency), p-IRS1Ser307, total JNK and (activated IRE1α)/(activated JNK) decreased with fructose ingestion, while p-JNK tended to increase; mineral-rich water ingestion, totally or partially, reverted all these effects. Total PERK, p-eIF2α (tendency) and total IRS1 (tendency) decreased in both fructose-fed groups. p-ERK/ERK and total IRE1α increasing tendencies in FRUCT became significant in FRUCTMIN (similar pattern for lipid area). Additionally, unspliced-XBP1 increased with mineral-rich water. In VAT, total ERK fructose-induced increase was partially prevented in FRUCTMIN. CONCLUSIONS Mineral-rich water modulation of fructose-induced effects on insulin signalling and ER homeostasis matches the better metabolic profile previously reported. Increased p-ERK/ERK, adding to decreased IRE1α activation, and increased unspliced-XBP1 and lipid area may protect against oxidative stress and IR development in FRUCTMIN.
Collapse
|
19
|
Gopal K, Ussher JR. Sugar-sweetened beverages and vascular function: food for thought. Am J Physiol Heart Circ Physiol 2017; 312:H285-H288. [DOI: 10.1152/ajpheart.00783.2016] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Keshav Gopal
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada
- Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, Canada; and
- Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada
| | - John R. Ussher
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada
- Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, Canada; and
- Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
20
|
Pimentel CFMG, Lai M. Nutrition Interventions for Chronic Liver Diseases and Nonalcoholic Fatty Liver Disease. Med Clin North Am 2016; 100:1303-1327. [PMID: 27745596 DOI: 10.1016/j.mcna.2016.06.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The progressively increasing rates of obesity have led to a worldwide epidemic of nonalcoholic fatty liver disease (NAFLD), the hepatic manifestation of the metabolic syndrome. It is currently the most common cause of liver disease worldwide and projected to be the leading indication for liver transplantation in the United States by 2020. NAFLD is associated with both liver-related and overall mortality. Undoubtedly, nutrition interventions are key in the treatment of NAFLD, to reverse the disease, and prevent disease progression, complications, and associated comorbidities, including cardiovascular disease and diabetes.
Collapse
Affiliation(s)
| | - Michelle Lai
- Gastroenterology, Liver Center, Beth Israel Deaconess Medical Center, Harvard University, 110 Francis St, Boston, MA 02115, USA
| |
Collapse
|
21
|
Saad AF, Dickerson J, Kechichian TB, Yin H, Gamble P, Salazar A, Patrikeev I, Motamedi M, Saade GR, Costantine MM. High-fructose diet in pregnancy leads to fetal programming of hypertension, insulin resistance, and obesity in adult offspring. Am J Obstet Gynecol 2016; 215:378.e1-6. [PMID: 27060421 DOI: 10.1016/j.ajog.2016.03.038] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Accepted: 03/21/2016] [Indexed: 12/18/2022]
Abstract
BACKGROUND Consumption of fructose-rich diets in the United States is on the rise and thought to be associated with obesity and cardiometabolic diseases. OBJECTIVE We sought to determine the effects of antenatal exposure to high-fructose diet on offspring's development of metabolic syndrome-like phenotype and other cardiovascular disease risk factors later in life. STUDY DESIGN Pregnant C57BL/6J dams were randomly allocated to fructose solution (10% wt/vol, n = 10) or water (n = 10) as the only drinking fluid from day 1 of pregnancy until delivery. After weaning, pups were started on regular chow, and evaluated at 1 year of life. We measured percent visceral adipose tissue and liver fat infiltrates using computed tomography, and blood pressure using CODA nonivasive monitor. Intraperitoneal glucose tolerance testing with corresponding insulin concentrations were obtained. Serum concentrations of glucose, insulin, triglycerides, total cholesterol, leptin, and adiponectin were measured in duplicate using standardized assays. Fasting homeostatic model assessment was also calculated to assess insulin resistance. P values <.05 were considered statistically significant. RESULTS Maternal weight, pup number, and average weight at birth were similar between the 2 groups. Male and female fructose group offspring had higher peak glucose and area under the intraperitoneal glucose tolerance testing curve compared with control, and higher mean arterial pressure compared to control. Female fructose group offspring were heavier and had higher percent visceral adipose tissue, liver fat infiltrates, homeostatic model assessment of insulin resistance scores, insulin area under the intraperitoneal glucose tolerance testing curve, and serum concentrations of leptin, and lower concentrations of adiponectin compared to female control offspring. No significant differences in these parameters were noted in male offspring. Serum concentrations of triglycerides or total cholesterol were not different between the 2 groups for either gender. CONCLUSION Maternal intake of high fructose leads to fetal programming of adult obesity, hypertension, and metabolic dysfunction, all risk factors for cardiovascular disease. This fetal programming is more pronounced in female offspring. Limiting intake of high fructose-enriched diets in pregnancy may have significant impact on long-term health.
Collapse
|
22
|
Dietary Salba (Salvia hispanica L.) ameliorates the adipose tissue dysfunction of dyslipemic insulin-resistant rats through mechanisms involving oxidative stress, inflammatory cytokines and peroxisome proliferator-activated receptor γ. Eur J Nutr 2016; 57:83-94. [DOI: 10.1007/s00394-016-1299-5] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Accepted: 08/13/2016] [Indexed: 10/21/2022]
|
23
|
Montgomery MK, Fiveash CE, Braude JP, Osborne B, Brown SHJ, Mitchell TW, Turner N. Disparate metabolic response to fructose feeding between different mouse strains. Sci Rep 2015; 5:18474. [PMID: 26690387 PMCID: PMC4686880 DOI: 10.1038/srep18474] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Accepted: 11/19/2015] [Indexed: 02/06/2023] Open
Abstract
Diets enriched in fructose (FR) increase lipogenesis in the liver, leading to hepatic lipid accumulation and the development of insulin resistance. Previously, we have shown that in contrast to other mouse strains, BALB/c mice are resistant to high fat diet-induced metabolic deterioration, potentially due to a lack of ectopic lipid accumulation in the liver. In this study we have compared the metabolic response of BALB/c and C57BL/6 (BL6) mice to a fructose-enriched diet. Both strains of mice increased adiposity in response to FR-feeding, while only BL6 mice displayed elevated hepatic triglyceride (TAG) accumulation and glucose intolerance. The lack of hepatic TAG accumulation in BALB/c mice appeared to be linked to an altered balance between lipogenic and lipolytic pathways, while the protection from fructose-induced glucose intolerance in this strain was likely related to low levels of ER stress, a slight elevation in insulin levels and an altered profile of diacylglycerol species in the liver. Collectively these findings highlight the multifactorial nature of metabolic defects that develop in response to changes in the intake of specific nutrients and the divergent response of different mouse strains to dietary challenges.
Collapse
Affiliation(s)
- M K Montgomery
- Department of Pharmacology, School of Medical Sciences, UNSW Australia, Sydney, NSW, Australia
| | - C E Fiveash
- Department of Pharmacology, School of Medical Sciences, UNSW Australia, Sydney, NSW, Australia
| | - J P Braude
- Department of Pharmacology, School of Medical Sciences, UNSW Australia, Sydney, NSW, Australia
| | - B Osborne
- Department of Pharmacology, School of Medical Sciences, UNSW Australia, Sydney, NSW, Australia
| | - S H J Brown
- School of Health Sciences, University of Wollongong, Wollongong, NSW, Australia.,Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, NSW, Australia
| | - T W Mitchell
- School of Health Sciences, University of Wollongong, Wollongong, NSW, Australia.,Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, NSW, Australia
| | - N Turner
- Department of Pharmacology, School of Medical Sciences, UNSW Australia, Sydney, NSW, Australia
| |
Collapse
|
24
|
Stanhope KL. Sugar consumption, metabolic disease and obesity: The state of the controversy. Crit Rev Clin Lab Sci 2015; 53:52-67. [PMID: 26376619 DOI: 10.3109/10408363.2015.1084990] [Citation(s) in RCA: 411] [Impact Index Per Article: 45.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
The impact of sugar consumption on health continues to be a controversial topic. The objective of this review is to discuss the evidence and lack of evidence that allows the controversy to continue, and why resolution of the controversy is important. There are plausible mechanisms and research evidence that supports the suggestion that consumption of excess sugar promotes the development of cardiovascular disease (CVD) and type 2 diabetes (T2DM) both directly and indirectly. The direct pathway involves the unregulated hepatic uptake and metabolism of fructose, leading to liver lipid accumulation, dyslipidemia, decreased insulin sensitivity and increased uric acid levels. The epidemiological data suggest that these direct effects of fructose are pertinent to the consumption of the fructose-containing sugars, sucrose and high fructose corn syrup (HFCS), which are the predominant added sugars. Consumption of added sugar is associated with development and/or prevalence of fatty liver, dyslipidemia, insulin resistance, hyperuricemia, CVD and T2DM, often independent of body weight gain or total energy intake. There are diet intervention studies in which human subjects exhibited increased circulating lipids and decreased insulin sensitivity when consuming high sugar compared with control diets. Most recently, our group has reported that supplementing the ad libitum diets of young adults with beverages containing 0%, 10%, 17.5% or 25% of daily energy requirement (Ereq) as HFCS increased lipid/lipoprotein risk factors for CVD and uric acid in a dose-response manner. However, un-confounded studies conducted in healthy humans under a controlled, energy-balanced diet protocol that enables determination of the effects of sugar with diets that do not allow for body weight gain are lacking. Furthermore, recent reports conclude that there are no adverse effects of consuming beverages containing up to 30% Ereq sucrose or HFCS, and the conclusions from several meta-analyses suggest that fructose has no specific adverse effects relative to any other carbohydrate. Consumption of excess sugar may also promote the development of CVD and T2DM indirectly by causing increased body weight and fat gain, but this is also a topic of controversy. Mechanistically, it is plausible that fructose consumption causes increased energy intake and reduced energy expenditure due to its failure to stimulate leptin production. Functional magnetic resonance imaging (fMRI) of the brain demonstrates that the brain responds differently to fructose or fructose-containing sugars compared with glucose or aspartame. Some epidemiological studies show that sugar consumption is associated with body weight gain, and there are intervention studies in which consumption of ad libitum high-sugar diets promoted increased body weight gain compared with consumption of ad libitum low- sugar diets. However, there are no studies in which energy intake and weight gain were compared in subjects consuming high or low sugar, blinded, ad libitum diets formulated to ensure both groups consumed a comparable macronutrient distribution and the same amounts of fiber. There is also little data to determine whether the form in which added sugar is consumed, as beverage or as solid food, affects its potential to promote weight gain. It will be very challenging to obtain the funding to conduct the clinical diet studies needed to address these evidence gaps, especially at the levels of added sugar that are commonly consumed. Yet, filling these evidence gaps may be necessary for supporting the policy changes that will help to turn the food environment into one that does not promote the development of obesity and metabolic disease.
Collapse
Affiliation(s)
- Kimber L Stanhope
- a Department of Molecular Biosciences , School of Veterinary Medicine and.,b Department of Nutrition , University of California , Davis , CA , USA
| |
Collapse
|
25
|
Di Bartolomeo F, Van den Ende W. Fructose and Fructans: Opposite Effects on Health? PLANT FOODS FOR HUMAN NUTRITION (DORDRECHT, NETHERLANDS) 2015; 70:227-37. [PMID: 25904233 DOI: 10.1007/s11130-015-0485-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
Fructans are fructose-based oligo-and polysaccharides of natural origin. Fructan and fructose species are sometimes confused by the great public, although they clearly have different biochemical and physiological properties. This review discusses aspects of the use of fructose and fructans in foods in the context of human health, with possible differential effects on cellular autophagy in cells of the human body. Although there are uncertainties on the daily levels of ingested fructose to be considered harmful to human health, there is an emerging consensus on the benefits of the use of fructans in functional foods, sustaining health via direct immunomodulatory and antioxidant effects or through indirect, prebiotic mechanisms.
Collapse
Affiliation(s)
- Francesca Di Bartolomeo
- Institute of Biochemistry, Graz University of Technology, Petersgasse 12/2, 8010, Graz, Austria
| | | |
Collapse
|
26
|
Yan Z, Fan R, Yin S, Zhao X, Liu J, Li L, Zhang W, Ge L. Protective effects of Ginkgo biloba leaf polysaccharide on nonalcoholic fatty liver disease and its mechanisms. Int J Biol Macromol 2015; 80:573-80. [PMID: 26047899 DOI: 10.1016/j.ijbiomac.2015.05.054] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2014] [Revised: 05/27/2015] [Accepted: 05/30/2015] [Indexed: 12/11/2022]
Abstract
A water-soluble polysaccharide fraction extracted from the leaf of Ginkgo biloba was named GBLP. The protective effect of GBLP on nonalcoholic fatty liver disease (NAFLD) was observed and underlying mechanism was explored. Wistar male rats were randomly divided into five groups, namely, normal control group, model control group and GBLP groups (100, 200 and 400 mg/kg/d). A rat model of NAFLD was established in male Wistar rats by feeding with high-fat diet (HFD) for 8 weeks. On day 57, the intragastric administration of GBLP started once daily for 4 weeks. The results showed that GBLP supplementation significantly and dose-dependently lowered the weight gain of body, liver index and serum lipid parameters in HFD-fed rat. Meanwhile, GBLP attenuated HFD-induced liver injury through reducing hepatic steatosis, TG accumulation, serum ALT, AST and ALP levels. GBLP had a positive effect on obesity-associated insulin resistance (IR) via reducing serum glucose and insulin levels. Furthermore, GBLP enhanced the activities of antioxidant enzymes and reduced MDA levels in serum and liver. These results indicate that GBLP can play a certain protective role against HFD-induced NAFLD, and the protective effects may be associated with attenuating IR, preserving liver function, enhancing antioxidant defense system, and reducing lipid peroxidation.
Collapse
Affiliation(s)
- Zhengui Yan
- College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai'an 271018, PR China.
| | - Ruifeng Fan
- College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai'an 271018, PR China
| | - Shaojie Yin
- College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai'an 271018, PR China
| | - Xiaona Zhao
- College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai'an 271018, PR China
| | - Jianzhu Liu
- College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai'an 271018, PR China
| | - Liuhui Li
- College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai'an 271018, PR China
| | - Wenqi Zhang
- College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai'an 271018, PR China
| | - Lijiang Ge
- College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai'an 271018, PR China
| |
Collapse
|
27
|
D'Alessandro ME, Selenscig D, Illesca P, Chicco A, Lombardo YB. Time course of adipose tissue dysfunction associated with antioxidant defense, inflammatory cytokines and oxidative stress in dyslipemic insulin resistant rats. Food Funct 2015; 6:1299-309. [DOI: 10.1039/c4fo00903g] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Time course of adipose tissue dysfunction in dyslipemic insulin resistant rats.
Collapse
Affiliation(s)
| | - Dante Selenscig
- Department of Biochemistry
- School of Biochemistry
- University of Litoral
- Santa Fe
- Argentina
| | - Paola Illesca
- Department of Biochemistry
- School of Biochemistry
- University of Litoral
- Santa Fe
- Argentina
| | - Adriana Chicco
- Department of Biochemistry
- School of Biochemistry
- University of Litoral
- Santa Fe
- Argentina
| | - Yolanda B. Lombardo
- Department of Biochemistry
- School of Biochemistry
- University of Litoral
- Santa Fe
- Argentina
| |
Collapse
|
28
|
Consumption of sucrose, but not high fructose corn syrup, leads to increased adiposity and dyslipidaemia in the pregnant and lactating rat. J Dev Orig Health Dis 2014; 6:38-46. [PMID: 25523154 DOI: 10.1017/s2040174414000610] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Excess consumption of added sugars, including sucrose and high fructose corn syrup (HFCS-55), have been implicated in the global epidemics of obesity and type 2 diabetes. This study aimed to investigate and compare the impact of maternal consumption of sucrose or HFCS-55 during pregnancy and lactation on the metabolic health of the dam and her offspring at birth. Female Albino Wistar rats were given access to chow and water, in addition to a sucrose or HFCS-55 beverage (10% w/v) before, and during pregnancy and lactation. Maternal glucose tolerance was determined throughout the study, and a postmortem was conducted on dams following lactation, and on offspring within 24 h of birth. Sucrose and HFCS-55 consumption resulted in increased total energy intake compared with controls, however the increase from sucrose consumption was accompanied by a compensatory decrease in chow consumption. There was no effect of sucrose or HFCS-55 consumption on body weight, however sucrose consumption resulted in increased adiposity and elevated total plasma cholesterol in the dam, while HFCS-55 consumption resulted in increased plasma insulin and decreased plasma non-esterified fatty acids (NEFA). Maternal HFCS-55 consumption was associated with decreased relative liver weight and plasma NEFA in the offspring at birth. There was no effect of either treatment on pup weight at birth. These findings suggest that both sucrose and HFCS-55 consumption during pregnancy and lactation have the potential to impact negatively on maternal metabolic health, which may have adverse consequences for the long-term health of the offspring.
Collapse
|
29
|
Beneficial effects of soluble dietary Jerusalem artichoke (Helianthus tuberosus) in the prevention of the onset of type 2 diabetes and non-alcoholic fatty liver disease in high-fructose diet-fed rats. Br J Nutr 2014; 112:709-17. [PMID: 24968200 DOI: 10.1017/s0007114514001421] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Jerusalem artichoke (JA) has the potential to attenuate lipid disturbances and insulin resistance (IR), but the underlying mechanisms are not well understood. In the present study, we elucidated the physiological responses and mechanisms of JA intervention with a comprehensive transcriptome analysis. Wistar rats were fed a control diet, a 60 % fructose-enriched diet (FRU), or a FRU with 10 % JA (n 6-7) for 4 weeks. An oral glucose tolerance test was carried out on day 21. Liver samples were collected for biochemical and global gene expression analyses (GeneChip® Rat Genome 230 2.0 Array, Affymetrix). Fructose feeding resulted in IR and hepatic TAG accumulation; dietary JA supplementation significantly improved these changes. Transcriptomic profiling revealed that the expression of malic enzyme 1 (Me1), associated with fatty acid synthesis; decorin (Dcn), related to fibrosis; and cytochrome P450, family 1, subfamily a, polypeptide 2 (Cyp1a2) and nicotinamide phosphoribosyltransferase (Nampt), associated with inflammation, was differentially altered by the FRU, whereas dietary JA supplementation significantly improved the expression of these genes. We established for the first time the molecular mechanisms driving the beneficial effects of JA in the prevention of type 2 diabetes and non-alcoholic fatty liver disease. We propose that 10 % JA supplementation may be beneficial for the prevention of the onset of these diseases.
Collapse
|
30
|
Regnault TRH, Gentili S, Sarr O, Toop CR, Sloboda DM. Fructose, pregnancy and later life impacts. Clin Exp Pharmacol Physiol 2013; 40:824-37. [DOI: 10.1111/1440-1681.12162] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2013] [Revised: 08/08/2013] [Accepted: 08/14/2013] [Indexed: 02/06/2023]
Affiliation(s)
- Timothy RH Regnault
- Department of Obstetrics and Gynaecology; Children's Health Research Institute; Western University; London ON Canada
| | - Sheridan Gentili
- School of Pharmacy and Medical Sciences; Sansom Institute for Health Research; University of South Australia; Adelaide SA Australia
| | - Ousseynou Sarr
- Department of Obstetrics and Gynaecology; Children's Health Research Institute; Western University; London ON Canada
| | - Carla R Toop
- School of Pharmacy and Medical Sciences; Sansom Institute for Health Research; University of South Australia; Adelaide SA Australia
| | - Deborah M Sloboda
- Department of Biochemistry and Biomedical Sciences; Faculty of Health Sciences; McMaster University; Hamilton ON Canada
| |
Collapse
|
31
|
Wei Y, Wang D, Moran G, Estrada A, Pagliassotti MJ. Fructose-induced stress signaling in the liver involves methylglyoxal. Nutr Metab (Lond) 2013; 10:32. [PMID: 23566306 PMCID: PMC3635994 DOI: 10.1186/1743-7075-10-32] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2012] [Accepted: 04/01/2013] [Indexed: 11/10/2022] Open
Abstract
Background Fructose produces hepatic insulin resistance in humans and animals. We have proposed that the selective metabolism of fructose by the liver can, under conditions of elevated fructose delivery, inflict a metabolic insult that is localized to the hepatocyte. The present study was designed to identify potential cellular effectors of this insult. Methods Primary hepatocytes were incubated with 8 mM glucose and 0.12% inulin (G, n = 6) or 8 mM glucose, 0.12% inulin and 28 mU of inulinase (GF, n = 6) in the presence or absence of insulin for 0, 2, or 4 h. Results GF produced fructose concentrations of ~0.7 mM over the 4 h experiment. GF induced phosphorylation of MKK7 and JNK, phosphorylation of serine307 on IRS-1, and reduced tyrosine phosphorylation of IRS-1 and -2. GF increased ceramide levels and reactive oxygen species (ROS); however inhibitors of ceramide synthesis or ROS accumulation did not prevent GF-mediated changes in MKK7, JNK or IRS proteins. GF increased cellular methylglyoxal concentrations and a selective increase in methylglyoxal recapitulated the GF-induced changes in MKK7, JNK and IRS proteins. Conclusions We hypothesize that GF-mediated changes in stress signaling involve methylglyoxal in primary hepatocytes.
Collapse
Affiliation(s)
- Yuren Wei
- Department of Food Science and Human Nutrition, Colorado State University, 234 Gifford, Fort Collins, CO 80523-1571, USA
| | - Dong Wang
- Department of Food Science and Human Nutrition, Colorado State University, 234 Gifford, Fort Collins, CO 80523-1571, USA
| | - Gretchen Moran
- Department of Food Science and Human Nutrition, Colorado State University, 234 Gifford, Fort Collins, CO 80523-1571, USA
| | - Andrea Estrada
- Department of Food Science and Human Nutrition, Colorado State University, 234 Gifford, Fort Collins, CO 80523-1571, USA
| | - Michael J Pagliassotti
- Department of Food Science and Human Nutrition, Colorado State University, 234 Gifford, Fort Collins, CO 80523-1571, USA
| |
Collapse
|
32
|
Antioxidant treatment prevents the development of fructose-induced abdominal adipose tissue dysfunction. Clin Sci (Lond) 2013; 125:87-97. [DOI: 10.1042/cs20120470] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
In the present study, we tested the effect of OS (oxidative stress) inhibition in rats fed on an FRD [fructose-rich diet; 10% (w/v) in drinking water] for 3 weeks. Normal adult male rats received a standard CD (commercial diet) or an FRD without or with an inhibitor of NADPH oxidase, APO (apocynin; 5 mM in drinking water; CD-APO and FRD-APO). We thereafter measured plasma OS and metabolic-endocrine markers, AAT (abdominal adipose tissue) mass and cell size, FA (fatty acid) composition (content and release), OS status, LEP (leptin) and IRS (insulin receptor substrate)-1/IRS-2 mRNAs, ROS (reactive oxygen species) production, NADPH oxidase activity and LEP release by isolated AAT adipocytes. FRD-fed rats had larger AAT mass without changes in body weight, and higher plasma levels of TAG (triacylglycerol), FAs, TBARS (thiobarbituric acid-reactive substance) and LEP. Although no significant changes in glucose and insulin plasma levels were observed in these animals, their HOMA-IR (homoeostasis model assessment of insulin resistance) values were significantly higher than those of CD. The AAT from FRD-fed rats had larger adipocytes, higher saturated FA content, higher NADPH oxidase activity, greater ROS production, a distorted FA content/release pattern, lower insulin sensitivity together with higher and lower mRNA content of LEP and IRS-1-/2 respectively, and released a larger amount of LEP. The development of all the clinical, OS, metabolic, endocrine and molecular changes induced by the FRD were significantly prevented by APO co-administration. The fact that APO treatment prevented both changes in NADPH oxidase activity and the development of all the FRD-induced AAT dysfunctions in normal rats strongly suggests that OS plays an important role in the FRD-induced MS (metabolic syndrome) phenotype.
Collapse
|
33
|
Inhibitors of Fatty Acid Synthesis Induce PPAR α -Regulated Fatty Acid β -Oxidative Genes: Synergistic Roles of L-FABP and Glucose. PPAR Res 2013; 2013:865604. [PMID: 23533380 PMCID: PMC3600304 DOI: 10.1155/2013/865604] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2012] [Accepted: 12/21/2012] [Indexed: 12/21/2022] Open
Abstract
While TOFA (acetyl CoA carboxylase inhibitor) and C75 (fatty acid synthase inhibitor) prevent lipid accumulation by inhibiting fatty acid synthesis, the mechanism of action is not simply accounted for by inhibition of the enzymes alone.
Liver fatty acid binding protein (L-FABP), a mediator of long chain fatty acid signaling to peroxisome
proliferator-activated receptor-α (PPARα) in the nucleus, was found to bind
TOFA and its activated CoA thioester, TOFyl-CoA, with high affinity while binding C75 and C75-CoA
with lower affinity. Binding of TOFA and C75-CoA significantly altered L-FABP secondary structure. High (20 mM) but not physiological
(6 mM) glucose conferred on both TOFA and C75 the ability to induce PPARα transcription of the fatty
acid β-oxidative enzymes CPT1A, CPT2, and ACOX1 in cultured primary hepatocytes from wild-type (WT) mice.
However, L-FABP gene ablation abolished the effects of TOFA and C75 in the context of high glucose. These effects were not associated
with an increased cellular level of unesterified fatty acids but rather by increased intracellular glucose. These findings suggested that L-FABP may function as an intracellular fatty acid synthesis inhibitor binding protein
facilitating TOFA and C75-mediated induction of PPARα in the context of high glucose at levels similar to those in uncontrolled diabetes.
Collapse
|
34
|
Petrescu AD, Huang H, Martin GG, McIntosh AL, Storey SM, Landrock D, Kier AB, Schroeder F. Impact of L-FABP and glucose on polyunsaturated fatty acid induction of PPARα-regulated β-oxidative enzymes. Am J Physiol Gastrointest Liver Physiol 2013; 304:G241-56. [PMID: 23238934 PMCID: PMC3566512 DOI: 10.1152/ajpgi.00334.2012] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Liver fatty acid binding protein (L-FABP) is the major soluble protein that binds very-long-chain n-3 polyunsaturated fatty acids (n-3 PUFAs) in hepatocytes. However, nothing is known about L-FABP's role in n-3 PUFA-mediated peroxisome proliferator activated receptor-α (PPARα) transcription of proteins involved in long-chain fatty acid (LCFA) β-oxidation. This issue was addressed in cultured primary hepatocytes from wild-type, L-FABP-null, and PPARα-null mice with these major findings: 1) PUFA-mediated increase in the expression of PPARα-regulated LCFA β-oxidative enzymes, LCFA/LCFA-CoA binding proteins (L-FABP, ACBP), and PPARα itself was L-FABP dependent; 2) PPARα transcription, robustly potentiated by high glucose but not maltose, a sugar not taken up, correlated with higher protein levels of these LCFA β-oxidative enzymes and with increased LCFA β-oxidation; and 3) high glucose altered the potency of n-3 relative to n-6 PUFA. This was not due to a direct effect of glucose on PPARα transcriptional activity nor indirectly through de novo fatty acid synthesis from glucose. Synergism was also not due to glucose impacting other signaling pathways, since it was observed only in hepatocytes expressing both L-FABP and PPARα. Ablation of L-FABP or PPARα as well as treatment with MK886 (PPARα inhibitor) abolished/reduced PUFA-mediated PPARα transcription of these genes, especially at high glucose. Finally, the PUFA-enhanced L-FABP distribution into nuclei with high glucose augmentation of the L-FABP/PPARα interaction reveals not only the importance of L-FABP for PUFA induction of PPARα target genes in fatty acid β-oxidation but also the significance of a high glucose enhancement effect in diabetes.
Collapse
Affiliation(s)
- Anca D. Petrescu
- 1Department of Physiology and Pharmacology, Texas A&M University, TVMC, College Station, Texas; and
| | - Huan Huang
- 1Department of Physiology and Pharmacology, Texas A&M University, TVMC, College Station, Texas; and
| | - Gregory G. Martin
- 1Department of Physiology and Pharmacology, Texas A&M University, TVMC, College Station, Texas; and
| | - Avery L. McIntosh
- 1Department of Physiology and Pharmacology, Texas A&M University, TVMC, College Station, Texas; and
| | - Stephen M. Storey
- 1Department of Physiology and Pharmacology, Texas A&M University, TVMC, College Station, Texas; and
| | - Danilo Landrock
- 1Department of Physiology and Pharmacology, Texas A&M University, TVMC, College Station, Texas; and
| | - Ann B. Kier
- 2Department of Pathobiology, Texas A&M University, TVMC, College Station, Texas
| | - Friedhelm Schroeder
- 1Department of Physiology and Pharmacology, Texas A&M University, TVMC, College Station, Texas; and
| |
Collapse
|
35
|
Tapia E, Cristóbal M, García-Arroyo FE, Soto V, Monroy-Sánchez F, Pacheco U, Lanaspa MA, Roncal-Jiménez CA, Cruz-Robles D, Ishimoto T, Madero M, Johnson RJ, Sánchez-Lozada LG. Synergistic effect of uricase blockade plus physiological amounts of fructose-glucose on glomerular hypertension and oxidative stress in rats. Am J Physiol Renal Physiol 2013; 304:F727-36. [PMID: 23303409 DOI: 10.1152/ajprenal.00485.2012] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Fructose in sweetened beverages (SB) increases the risk for metabolic and cardiorenal disorders, and these effects are in part mediated by a secondary increment in uric acid (UA). Rodents have an active uricase, thus requiring large doses of fructose to increase plasma UA and to induce metabolic syndrome and renal hemodynamic changes. We therefore hypothesized that the effects of fructose in rats might be enhanced in the setting of uricase inhibition. Four groups of male Sprague-Dawley rats (n = 7/group) were studied during 8 wk: water + vehicle (V), water + oxonic acid (OA; 750 mg/k BW), sweetened beverage (SB; 11% fructose-glucose combination) + V, and SB + OA. Systemic blood pressure, plasma UA, triglycerides (TG), glucose and insulin, glomerular hemodynamics, renal structural damage, renal cortex and liver UA, TG, markers of oxidative stress, mitDNA, fructokinase, and fatty liver synthase protein expressions were evaluated at the end of the experiment. Chronic hyperuricemia and SB induced features of the metabolic syndrome, including hypertension, hyperuricemia, hyperglycemia, and systemic and hepatic TG accumulation. OA alone also induced glomerular hypertension, and SB alone induced insulin resistance. SB + OA induced a combined phenotype including metabolic and renal alterations induced by SB or OA alone and in addition also acted synergistically on systemic and glomerular pressure, plasma glucose, hepatic TG, and oxidative stress. These findings explain why high concentrations of fructose are required to induce greater metabolic changes and renal disease in rats whereas humans, who lack uricase, appear to be much more sensitive to the effects of fructose.
Collapse
Affiliation(s)
- Edilia Tapia
- Laboratory of Renal Physiopathology, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Nojima K, Sugimoto K, Ueda H, Babaya N, Ikegami H, Rakugi H. Analysis of hepatic gene expression profile in a spontaneous mouse model of type 2 diabetes under a high sucrose diet. Endocr J 2013; 60:261-74. [PMID: 23131898 DOI: 10.1507/endocrj.ej12-0258] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Both genetic factors and diabetogenic environmental factors, such as a high-sucrose diet (HSD), are involved in the development of type 2 diabetes. In this study, the Nagoya-Shibata-Yasuda (NSY) mouse, an animal model of type 2 diabetes and C3H mice used as controls, were fed a HSD, a high-fat diet (HFD) or a regular diet (RD) from weaning. In C3H mice, HFD significantly increased body weight gain, but maintained glucose tolerance. In contrast, in NSY mice, HSD resulted in increased body weight gain and liver steatosis and increased glucose intolerance to a greater extent than HFD. Furthermore, we performed DNA microarray analysis to detect differences in hepatic gene expression levels in both strains under HSD. We then performed RT-PCR analysis on selected genes to evaluate basal expression level under RD and changes under HSD conditions. HSD-fed NSY, but not C3H mice, exhibited increased hepatic expression levels of Pparg2, an isoform of Pparg as well as G0s2, a target of Pparg, which are known to be adipocyte-specific genes. Compared to RD-fed C3H mice, hepatic expression levels of Kat2b (transcriptional regulation), Hsd3b5 (steroid hormone metabolism) and Cyp7b1 (bile acid metabolism) were initially lower in RD-fed NSY mice, and were further decreased in HSD-fed NSY mice. Expression of Metallothionein (Mt1) and Metallothionein 2 (Mt2) was significantly lower in NSY mice compared to C3H mice, irrespective of dietary condition. These data suggest that elucidation of this heterogeneity in response to HSD might contribute to further understanding of the gene-environment interactions leading to diabetes in humans.
Collapse
Affiliation(s)
- Koji Nojima
- Department of Geriatric Medicine and Nephrology, Osaka University Graduate School of Medicine, Osaka, Japan.
| | | | | | | | | | | |
Collapse
|
37
|
Grayson BE, Seeley RJ, Sandoval DA. Wired on sugar: the role of the CNS in the regulation of glucose homeostasis. Nat Rev Neurosci 2013; 14:24-37. [PMID: 23232606 PMCID: PMC4231433 DOI: 10.1038/nrn3409] [Citation(s) in RCA: 82] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Obesity and type 2 diabetes mellitus (T2DM)--disorders of energy homeostasis and glucose homeostasis, respectively--are tightly linked and the incidences of both conditions are increasing in parallel. The CNS integrates information regarding peripheral nutrient and hormonal changes and processes this information to regulate energy homeostasis. Recent findings indicate that some of the neural circuits and mechanisms underlying energy balance are also essential for the regulation of glucose homeostasis. We propose that disruption of these overlapping pathways links the metabolic disturbances associated with obesity and T2DM. A better understanding of these converging mechanisms may lead to therapeutic strategies that target both T2DM and obesity.
Collapse
Affiliation(s)
- Bernadette E Grayson
- Metabolic Diseases Institute, University of Cincinnati, Cincinnati, Ohio 45237, USA
| | | | | |
Collapse
|
38
|
Effect of pioglitazone on the fructose-induced abdominal adipose tissue dysfunction. PPAR Res 2012; 2012:259093. [PMID: 23091482 PMCID: PMC3469242 DOI: 10.1155/2012/259093] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2012] [Accepted: 08/23/2012] [Indexed: 12/31/2022] Open
Abstract
Aim. To test the potential role of PPARγ in the endocrine abdominal tissue dysfunction induced by feeding normal rats with a fructose rich diet (FRD) during three weeks. Methodology. Adult normal male rats received a standard commercial diet (CD) or FRD, (10% in drinking water) without or with pioglitazone (PIO) (i.p. 0.25 mg/Kg BW/day; CD-PIO and FRD-PIO). Thereafter, we measured circulating metabolic, endocrine, and oxidative stress (OS) markers, abdominal adipose tissue (AAT) mass, leptin (LEP) and plasminogen activator inhibitor-1 (PAI-1) tissue content/expression, and leptin release by isolated adipocytes incubated with different concentrations of insulin. Results. Plasma glucose, insulin, triglyceride, TBARS, LEP, and PAI-1 levels were higher in FRD rats; PIO coadministration fully prevented all these increments. AAT adipocytes from FRD rats were larger, secreted a higher amount of LEP, and displayed decreased sensitivity to insulin stimulation; these effects were significantly ameliorated by PIO. Whereas AAT LEP and PAI-1 (mRNA) concentrations increased significantly in FRD rats, those of insulin-receptor-substrate- (IRS-) 1 and IRS-2 were reduced. PIO coadministration prevented FRD effects on LEP, PAI-1, and IRS-2 (fully) and IRS-1 (partially) mRNAs in AAT. Conclusion. PPARγ would play a relevant role in the development of the FRD-induced metabolic-endocrine dysfunction.
Collapse
|
39
|
Storey SM, McIntosh AL, Huang H, Martin GG, Landrock KK, Landrock D, Payne HR, Kier AB, Schroeder F. Loss of intracellular lipid binding proteins differentially impacts saturated fatty acid uptake and nuclear targeting in mouse hepatocytes. Am J Physiol Gastrointest Liver Physiol 2012; 303:G837-50. [PMID: 22859366 PMCID: PMC3469595 DOI: 10.1152/ajpgi.00489.2011] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The liver expresses high levels of two proteins with high affinity for long-chain fatty acids (LCFAs): liver fatty acid binding protein (L-FABP) and sterol carrier protein-2 (SCP-2). Real-time confocal microscopy of cultured primary hepatocytes from gene-ablated (L-FABP, SCP-2/SCP-x, and L-FABP/SCP-2/SCP-x null) mice showed that the loss of L-FABP reduced cellular uptake of 12-N-methyl-(7-nitrobenz-2-oxa-1,3-diazo)-aminostearic acid (a fluorescent-saturated LCFA analog) by ∼50%. Importantly, nuclear targeting of the LCFA was enhanced when L-FABP was upregulated (SCP-2/SCP-x null) but was significantly reduced when L-FABP was ablated (L-FABP null), thus impacting LCFA nuclear targeting. These effects were not associated with a net decrease in expression of key membrane proteins involved in LCFA or glucose transport. Since hepatic LCFA uptake and metabolism are closely linked to glucose uptake, the effect of glucose on L-FABP-mediated LCFA uptake and nuclear targeting was examined. Increasing concentrations of glucose decreased cellular LCFA uptake and even more extensively decreased LCFA nuclear targeting. Loss of L-FABP exacerbated the decrease in LCFA nuclear targeting, while loss of SCP-2 reduced the glucose effect, resulting in enhanced LCFA nuclear targeting compared with control. Simply, ablation of L-FABP decreases LCFA uptake and even more extensively decreases its nuclear targeting.
Collapse
Affiliation(s)
- Stephen M. Storey
- 1Department of Physiology and Pharmacology, Texas A & M University, College Station, Texas; and
| | - Avery L. McIntosh
- 1Department of Physiology and Pharmacology, Texas A & M University, College Station, Texas; and
| | - Huan Huang
- 1Department of Physiology and Pharmacology, Texas A & M University, College Station, Texas; and
| | - Gregory G. Martin
- 1Department of Physiology and Pharmacology, Texas A & M University, College Station, Texas; and
| | - Kerstin K. Landrock
- 1Department of Physiology and Pharmacology, Texas A & M University, College Station, Texas; and
| | - Danilo Landrock
- 2Department of Pathobiology, Texas A & M University, College Station, Texas
| | - H. Ross Payne
- 2Department of Pathobiology, Texas A & M University, College Station, Texas
| | - Ann B. Kier
- 2Department of Pathobiology, Texas A & M University, College Station, Texas
| | - Friedhelm Schroeder
- 1Department of Physiology and Pharmacology, Texas A & M University, College Station, Texas; and
| |
Collapse
|
40
|
The role of nutrients in the development, progression, and treatment of nonalcoholic fatty liver disease. J Clin Gastroenterol 2012; 46:457-67. [PMID: 22469640 DOI: 10.1097/mcg.0b013e31824cf51e] [Citation(s) in RCA: 88] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is the most common cause of liver disease in adults and children and is currently the third most common indication for liver transplantation in North America. Its pathogenesis is thought to be secondary to multiple "hits" derived from the dietary components, adipose tissue, immune system, and intestinal microbiota. Lack of physical activity may contribute as well. Nutrients may exert their effect directly or through alteration of the intestinal microbiota. Research focusing on specific dietary components predisposing to NAFLD has shown conflicting results. Total energy intake, and macronutrients, has been linked to the development of NAFLD. Fructose not only contributes to hepatic steatosis but may trigger inflammatory signals as well. Polyunsaturated fatty acids are thought to exert anti-inflammatory effects. The role of vitamins as well as minerals in this field is actively being investigated. In this review, we discuss the evidence-linking macronutrients (such as carbohydrates and fat in general and fructose, fiber, short chain fatty acids, polyunsaturated fatty, and choline specifically) and micronutrients (such as vitamin E and C and minerals) with the development and treatment of NAFLD. We also discuss the literature on physical activity and NAFLD.
Collapse
|
41
|
Abstract
Despite a lack of consistent diagnostic criteria, the metabolic syndrome (MetS) is increasingly evident in children and adolescents, portending a tsunami of chronic disease and mortality as this generation ages. The diagnostic criteria for MetS apply absolute cutoffs to continuous variables and fail to take into account aging, pubertal changes, and race/ethnicity. We attempt to define MetS mechanistically to determine its specific etiologies and to identify targets for therapy. Whereas the majority of studies document a relationship of visceral fat to insulin resistance, ectopic liver fat correlates better with dysfunctional insulin dynamics from which the rest of MetS derives. In contrast to the systemic metabolism of glucose, the liver is the primary metabolic clearinghouse for 4 specific foodstuffs that have been associated with the development of MetS: trans-fats, branched-chain amino acids, ethanol, and fructose. These 4 substrates (1) are not insulin regulated and (2) deliver metabolic intermediates to hepatic mitochondria without an appropriate "pop-off" mechanism for excess substrate, enhancing lipogenesis and ectopic adipose storage. Excessive fatty acid derivatives interfere with hepatic insulin signal transduction. Reactive oxygen species accumulate, which cannot be quenched by adjacent peroxisomes; these reactive oxygen species reach the endoplasmic reticulum, leading to a compensatory process termed the "unfolded protein response," driving further insulin resistance and eventually insulin deficiency. No obvious drug target exists in this pathway; thus, the only rational therapeutic approaches remain (1) altering hepatic substrate availability (dietary modification), (2) reducing hepatic substrate flux (high fiber), or (3) increasing mitochondrial efficiency (exercise).
Collapse
Affiliation(s)
- Andrew A. Bremer
- Department of Pediatrics, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Michele Mietus-Snyder
- Department of Pediatrics and Children’s National Obesity Institute, Children’s National Medical Center, Washington, District of Columbia; and
| | - Robert H. Lustig
- Department of Pediatrics, University of California, San Francisco, San Francisco, California
| |
Collapse
|
42
|
Janevski M, Ratnayake S, Siljanovski S, McGlynn MA, Cameron-Smith D, Lewandowski P. Fructose containing sugars modulate mRNA of lipogenic genes ACC and FAS and protein levels of transcription factors ChREBP and SREBP1c with no effect on body weight or liver fat. Food Funct 2011; 3:141-9. [PMID: 22159273 DOI: 10.1039/c1fo10111k] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
The aim of this study was to determine the effects of high-glucose, high-fructose and high-sucrose diets on weight gain, liver lipid metabolism and gene expression of proteins involved with hepatic fat metabolism. Rats were fed a diet containing either 60% glucose, 60% fructose, 60% sucrose, or a standard chow for 28 days. Results indicated that high-fructose and high-sucrose diets were associated with higher mRNA levels of gene transcripts involved with fat synthesis; ACC, FAS and ChREBP, with no change in SREBP-1C mRNA. The protein level of ChREBP and SREBP1c was similar in liver homogenates from all groups, but were higher in nuclear fractions from the liver of high-fructose and high-sucrose fed rats. The mRNA level of gene transcripts involved with fat oxidation was the same in all three diets, whilst a high-fructose diet was associated with greater amount of mRNA of the fat transporter CD36. Despite the changes in mRNA of lipogenic proteins, the body weight of animals from each group was the same and the livers from rats fed high-fructose and high-sucrose diets did not contain more fat than control diet livers. In conclusion, changing the composition of the principal monosaccharide in the diet to a fructose containing sugar elicits changes in the level of hepatic mRNA of lipogenic and fat transport proteins and protein levels of their transcriptional regulators; however this is not associated with any changes in body weight or liver fat content.
Collapse
Affiliation(s)
- Mile Janevski
- School of Medicine, Deakin University, Pigdons Road, Waurn Ponds, Victoria 3217, Australia.
| | | | | | | | | | | |
Collapse
|
43
|
Nomura K, Yamanouchi T. The role of fructose-enriched diets in mechanisms of nonalcoholic fatty liver disease. J Nutr Biochem 2011; 23:203-8. [PMID: 22129639 DOI: 10.1016/j.jnutbio.2011.09.006] [Citation(s) in RCA: 149] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2011] [Accepted: 09/25/2011] [Indexed: 02/08/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) currently affects 20%-30% of adults and 10% of children in industrialized countries, and its prevalence is increasing worldwide. Although NAFLD is a benign form of liver dysfunction, it can proceed to a more serious condition, nonalcoholic steatohepatitis (NASH), which may lead to liver cirrhosis and hepatocellular carcinoma. NAFLD is accompanied by obesity, metabolic syndrome and diabetes mellitus, and evidence suggests that fructose, a major caloric sweetener in the diet, plays a significant role in its pathogenesis. Inflammatory progression to NASH is proposed to occur by a two-hit process. The first "hit" is hepatic fat accumulation owing to increased hepatic de novo lipogenesis, inhibition of fatty acid beta oxidation, impaired triglyceride clearance and decreased very-low-density lipoprotein export. The mechanisms of the second "hit" are still largely unknown, but recent studies suggest several possibilities, including inflammation caused by oxidative stress associated with lipid peroxidation, cytokine activation, nitric oxide and reactive oxygen species, and endogenous toxins of fructose metabolites.
Collapse
Affiliation(s)
- Kyoko Nomura
- Department of Hygiene and Public Health, School of Medicine, Teikyo University, Tokyo 1838605, Japan.
| | | |
Collapse
|
44
|
Hepatic oxidative stress in fructose-induced fatty liver is not caused by sulfur amino acid insufficiency. Nutrients 2011; 3:987-1002. [PMID: 22254090 PMCID: PMC3257721 DOI: 10.3390/nu3110987] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2011] [Revised: 10/24/2011] [Accepted: 11/04/2011] [Indexed: 02/08/2023] Open
Abstract
Fructose-sweetened liquid consumption is associated with fatty liver and oxidative stress. In rodent models of fructose-mediated fatty liver, protein consumption is decreased. Additionally, decreased sulfur amino acid intake is known to cause oxidative stress. Studies were designed to test whether oxidative stress in fructose-sweetened liquid-induced fatty liver is caused by decreased ad libitum solid food intake with associated inadequate sulfur amino acid intake. C57BL6 mice were grouped as: control (ad libitum water), fructose (ad libitum 30% fructose-sweetened liquid), glucose (ad libitum 30% glucose-sweetened water) and pair-fed (ad libitum water and sulfur amino acid intake same as the fructose group). Hepatic and plasma thiol-disulfide antioxidant status were analyzed after five weeks. Fructose- and glucose-fed mice developed fatty liver. The mitochondrial antioxidant protein, thioredoxin-2, displayed decreased abundance in the liver of fructose and glucose-fed mice compared to controls. Glutathione/glutathione disulfide redox potential (E(h)GSSG) and abundance of the cytoplasmic antioxidant protein, peroxiredoxin-2, were similar among groups. We conclude that both fructose and glucose-sweetened liquid consumption results in fatty liver and upregulated thioredoxin-2 expression, consistent with mitochondrial oxidative stress; however, inadequate sulfur amino acid intake was not the cause of this oxidative stress.
Collapse
|
45
|
Hein GJ, Chicco A, Lombardo YB. Fish oil normalizes plasma glucose levels and improves liver carbohydrate metabolism in rats fed a sucrose-rich diet. Lipids 2011; 47:141-50. [PMID: 22045300 DOI: 10.1007/s11745-011-3623-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2011] [Accepted: 10/04/2011] [Indexed: 12/23/2022]
Abstract
A sucrose-rich diet (SRD) induces insulin resistance and dyslipidemia with impaired hepatic glucose production and gluconeogenesis, accompanied by altered post-receptor insulin signaling steps. The aim of this study was to examine the effectiveness of fish oil (FO) to reverse or improve the impaired hepatic glucose metabolism once installed in rats fed 8 months a SRD. In the liver of rats fed SRD in which FO replaced corn-oil during the last 2 months, as dietary fat, several key enzyme activities and metabolites involved in glucose metabolisms (phosphorylation, glycolysis, gluconeogenesis and oxidative and non oxidative glucose pathway) were measured. The protein mass levels of IRS-1 and αp85 PI-3K at basal conditions were also analyzed. FO improved the altered activities of some enzymes involved in the glycolytic and oxidative pathways observed in the liver of SRD fed rats but was unable to restore the impaired capacity of glucose phosphorylation. Moreover, FO reversed the increase in PEPCK and G-6-Pase and reduced the G-6-Pase/GK ratio. Glycogen concentration and GSa activity returned to levels similar to those observed in the liver of the control-fed rats. Besides, FO did not modify the altered protein mass levels of IRS-1 and αp85 PI-3K. Finally, dietary FO was effective in reversing or improving the impaired activities of several key enzymes of hepatic carbohydrate metabolism contributing, at least in part, to the normalization of plasma glucose levels in the SRD-fed rats. However, these positive effects of FO were not observed under basal conditions in the early steps of insulin signaling transduction.
Collapse
Affiliation(s)
- Gustavo J Hein
- Department of Biochemistry, School of Biochemistry, University of Litoral, Ciudad Universitaria Paraje El Pozo. CC 242 (3000), Santa Fe, Argentina
| | | | | |
Collapse
|
46
|
Aeberli I, Gerber PA, Hochuli M, Kohler S, Haile SR, Gouni-Berthold I, Berthold HK, Spinas GA, Berneis K. Low to moderate sugar-sweetened beverage consumption impairs glucose and lipid metabolism and promotes inflammation in healthy young men: a randomized controlled trial. Am J Clin Nutr 2011; 94:479-85. [PMID: 21677052 DOI: 10.3945/ajcn.111.013540] [Citation(s) in RCA: 245] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Sugar-sweetened beverages (SSBs) have unfavorable effects on glucose and lipid metabolism if consumed in high quantities by obese subjects, but the effect of lower doses in normal-weight subjects is less clear. OBJECTIVE The aim was to investigate the effects of SSBs consumed in small to moderate quantities for 3 wk on LDL particle distribution and on other parameters of glucose and lipid metabolism as well as on inflammatory markers in healthy young men. DESIGN Twenty-nine subjects were studied in a prospective, randomized, controlled crossover trial. Six 3-wk interventions were assigned in random order as follows: 600 mL SSBs containing 1)40 g fructose/d [medium fructose (MF)], 2) 80 g fructose/d [high fructose (HF)], 3) 40 g glucose/d [medium glucose (MG)], 4) 80 g glucose/d [high glucose (HG)], 5) 80 g sucrose/d [high sucrose (HS)], or 6) dietary advice to consume low amounts of fructose. Outcome parameters were measured at baseline and after each intervention. RESULTS LDL particle size was reduced after HF by -0.51 nm (95% CI: -0.19, -0.82 nm) and after HS by -0.43 nm (95% CI: -0.12, -0.74; P < 0.05 for both). Similarly, a more atherogenic LDL subclass distribution was seen when fructose-containing SSBs were consumed (MF, HF, and HS: P < 0.05). Fasting glucose and high-sensitivity C-reactive protein (hs-CRP) increased significantly after all interventions (by 4-9% and 60-109%, respectively; P < 0.05); leptin increased during interventions with SSBs containing glucose only (MG and HG: P < 0.05). CONCLUSION The present data show potentially harmful effects of low to moderate consumption of SSBs on markers of cardiovascular risk such as LDL particles, fasting glucose, and hs-CRP within just 3 wk in healthy young men, which is of particular significance for young consumers. This trial was registered at clinicaltrials.gov as NCT01021969.
Collapse
Affiliation(s)
- Isabelle Aeberli
- Division of Endocrinology, Diabetes, and Clinical Nutrition, University Hospital Zurich, Zurich, Switzerland
| | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Abdulla MH, Sattar MA, Johns EJ. The Relation between Fructose-Induced Metabolic Syndrome and Altered Renal Haemodynamic and Excretory Function in the Rat. Int J Nephrol 2011; 2011:934659. [PMID: 21785727 PMCID: PMC3139200 DOI: 10.4061/2011/934659] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2011] [Revised: 04/30/2011] [Accepted: 05/15/2011] [Indexed: 11/20/2022] Open
Abstract
This paper explores the possible relationships between dietary fructose and altered neurohumoral regulation of renal haemodynamic and excretory function in this model of metabolic syndrome. Fructose consumption induces hyperinsulinemia, hypertriglyceridaemia, insulin resistance, and hypertension. The pathogenesis of fructose-induced hypertension is dubious and involves numerous pathways acting both singly and together. In addition, hyperinsulinemia and hypertension contribute significantly to progressive renal disease in fructose-fed rats. Moreover, increased activity of the renin-angiotensin and sympathetic nervous systems leading to downregulation of receptors may be responsible for the blunted vascular sensitivity to angiotensin II and catecholamines, respectively. Various approaches have been suggested to prevent the development of fructose-induced hypertension and/or metabolic alteration. In this paper, we address the role played by the renin-angiotensin and sympathetic nervous systems in the haemodynamic alterations that occur due to prolonged consumption of fructose.
Collapse
Affiliation(s)
- Mohammed H Abdulla
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, Minden, 11800 Penang, Malaysia
| | | | | |
Collapse
|
48
|
Johnson PM, Chen SS, Santomango TS, Williams PE, Lacy DB, McGuinness OP. Continuous low-dose fructose infusion does not reverse glucagon-mediated decrease in hepatic glucose utilization. Metabolism 2011; 60:867-73. [PMID: 20940071 PMCID: PMC3736817 DOI: 10.1016/j.metabol.2010.08.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2009] [Revised: 08/18/2010] [Accepted: 08/18/2010] [Indexed: 01/04/2023]
Abstract
An adaptation to continuous total parenteral nutrition (TPN; 75% of nonprotein calories as glucose) is the liver becomes a major consumer of glucose with lactate release as a by-product. The liver is able to further increase liver glucose uptake when a small dose of fructose is acutely infused via the portal system. Glucagon, commonly elevated during inflammatory stress, is a potent inhibitor of glucose uptake by the liver during TPN. The aim was to determine if continuous fructose infusion could overcome the glucagon-mediated decrease in hepatic glucose uptake. Studies were performed in conscious, insulin-treated, chronically catheterized, pancreatectomized dogs that adapted to TPN for 33 hours. They were then assigned to 1 of 4 groups: TPN (C), TPN + fructose (4.4 μmol kg(-1) min(-1); F), TPN + glucagon (0.2 pmol kg(-1) min(-1); GGN), or TPN + fructose and glucagon (F + GGN) for an additional 63 hours (33-96 hours). Insulin, fructose, and glucagon were infused into the portal vein. During that period, all animals received a fixed insulin infusion of 0.4 mU·kg(-1)·min(-1) (33-96 hours); and the glucose infusion rates were adjusted to maintain euglycemia (6.6 mmol/L). Continuous fructose infusion was unable to further enhance net hepatic glucose uptake (in micromoles per kilogram per minute) (31.1 ± 2.8 vs 36.1 ± 5.0; C vs F), nor was it able to overcome glucagon-mediated decrease in net hepatic glucose uptake (10.0 ± 4.4 vs 12.2 ± 3.9; GGN vs F + GGN). In summary, continuous fructose infusion cannot augment liver glucose uptake during TPN; nor can it overcome the inhibitory effects of glucagon.
Collapse
Affiliation(s)
- Paulette M Johnson
- Division of Pediatric Critical Care Medicine, Vanderbilt University School of Medicine, Nashville, TN 37232-0615, USA
| | | | | | | | | | | |
Collapse
|
49
|
Prevention and reversal of diet-induced leptin resistance with a sugar-free diet despite high fat content. Br J Nutr 2011; 106:390-7. [PMID: 21418711 DOI: 10.1017/s000711451100033x] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Chronic consumption of a Western-type diet, containing both elevated sugar and fat, results in leptin resistance. We hypothesised that fructose, as part of the sugar component of Western-type diets, is one causative ingredient in the development of leptin resistance and that removal of this component will prevent leptin resistance despite high fat (HF) content. We fed rats a sugar-free (SF), 30 % HF (SF/HF) diet or a 40 % high-fructose (HFr), 30 % HF (HFr/HF) diet for 134 d. The HFr/HF diet resulted in impaired anorexic and body-weight responses to both peripherally (0·6 mg/kg, assessed on day 65 of the diet) and centrally (1·5 μg/d, assessed on days 129-134) administered leptin, whereas SF/HF-fed rats were fully leptin responsive. At day 70, half the HFr/HF-fed animals were switched to the SF/HF diet, reversing the leptin resistance (assessed 18 d after the diet switch). The HFr/HF diet elevated serum leptin and reduced adiponectin, and levels were restored abruptly at day 3 after switching to the SF/HF diet. These data demonstrate that a diet containing both HFr and fat leads to leptin resistance, while an isoenergetic SF/HF diet does not. Moreover, removal of fructose from this diet reverses the leptin resistance and the elevated leptin, suggesting a cause-and-effect relationship. These data suggest that fructose is the bioactive component of a HF/high-sugar diet that is essential for the induction of leptin resistance.
Collapse
|
50
|
Fructose sensitivity is suppressed in Arabidopsis by the transcription factor ANAC089 lacking the membrane-bound domain. Proc Natl Acad Sci U S A 2011; 108:3436-41. [PMID: 21300879 DOI: 10.1073/pnas.1018665108] [Citation(s) in RCA: 91] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
In living organisms sugars not only provide energy and carbon skeletons but also act as evolutionarily conserved signaling molecules. The three major soluble sugars in plants are sucrose, glucose, and fructose. Information on plant glucose and sucrose signaling is available, but to date no fructose-specific signaling pathway has been reported. In this study, sugar repression of seedling development was used to study fructose sensitivity in the Landsberg erecta (Ler)/Cape Verde Islands (Cvi) recombinant inbred line population, and eight fructose-sensing quantitative trait loci (QTLs) (FSQ1-8) were mapped. Among them, FSQ6 was confirmed to be a fructose-specific QTL by analyzing near-isogenic lines in which Cvi genomic fragments were introgressed in the Ler background. These results indicate the existence of a fructose-specific signaling pathway in Arabidopsis. Further analysis demonstrated that the FSQ6-associated fructose-signaling pathway functions independently of the hexokinase1 (HXK1) glucose sensor. Remarkably, fructose-specific FSQ6 downstream signaling interacts with abscisic acid (ABA)- and ethylene-signaling pathways, similar to HXK1-dependent glucose signaling. The Cvi allele of FSQ6 acts as a suppressor of fructose signaling. The FSQ6 gene was identified using map-based cloning approach, and FSQ6 was shown to encode the transcription factor gene Arabidopsis NAC (petunia No apical meristem and Arabidopsis transcription activation factor 1, 2 and Cup-shaped cotyledon 2) domain containing protein 89 (ANAC089). The Cvi allele of FSQ6/ANAC089 is a gain-of-function allele caused by a premature stop in the third exon of the gene. The truncated Cvi FSQ6/ANAC089 protein lacks a membrane association domain that is present in ANAC089 proteins from other Arabidopsis accessions. As a result, Cvi FSQ6/ANAC089 is constitutively active as a transcription factor in the nucleus.
Collapse
|