1
|
Berdowska I, Matusiewicz M, Fecka I. A Comprehensive Review of Metabolic Dysfunction-Associated Steatotic Liver Disease: Its Mechanistic Development Focusing on Methylglyoxal and Counterbalancing Treatment Strategies. Int J Mol Sci 2025; 26:2394. [PMID: 40141037 PMCID: PMC11942149 DOI: 10.3390/ijms26062394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 02/21/2025] [Accepted: 03/04/2025] [Indexed: 03/28/2025] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is a multifactorial disorder characterized by excessive lipid accumulation in the liver which dysregulates the organ's function. The key contributor to MASLD development is insulin resistance (IR) which affects many organs (including adipose tissue, skeletal muscles, and the liver), whereas the molecular background is associated with oxidative, nitrosative, and carbonyl stress. Among molecules responsible for carbonyl stress effects, methylglyoxal (MGO) seems to play a major pathological function. MGO-a by-product of glycolysis, fructolysis, and lipolysis (from glycerol and fatty acids-derived ketone bodies)-is implicated in hyperglycemia, hyperlipidemia, obesity, type 2 diabetes, hypertension, and cardiovascular diseases. Its causative effect in the stimulation of prooxidative and proinflammatory pathways has been well documented. Since metabolic dysregulation leading to these pathologies promotes MASLD, the role of MGO in MASLD is addressed in this review. Potential MGO participation in the mechanism of MASLD development is discussed in regard to its role in different signaling routes leading to pathological events accelerating the disorder. Moreover, treatment strategies including approved and potential therapies in MASLD are overviewed and discussed in this review. Among them, medications aimed at attenuating MGO-induced pathological processes are addressed.
Collapse
Affiliation(s)
- Izabela Berdowska
- Department of Medical Biochemistry, Faculty of Medicine, Wroclaw Medical University, Chałubińskiego 10, 50-368 Wrocław, Poland;
| | - Małgorzata Matusiewicz
- Department of Medical Biochemistry, Faculty of Medicine, Wroclaw Medical University, Chałubińskiego 10, 50-368 Wrocław, Poland;
| | - Izabela Fecka
- Department of Pharmacognosy and Herbal Medicines, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211A, 50-556 Wrocław, Poland
| |
Collapse
|
2
|
Cely I, Blencowe M, Shu L, Diamante G, Ahn IS, Zhang G, LaGuardia J, Liu R, Saleem Z, Wang S, Davis R, Lusis AJ, Yang X. Glo1 reduction in mice results in age- and sex-dependent metabolic dysfunction. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.24.634754. [PMID: 39896461 PMCID: PMC11785252 DOI: 10.1101/2025.01.24.634754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
Objectives Advanced glycation end products (AGEs) have been implicated as an important mediator of metabolic disorders including obesity, insulin resistance, and coronary artery disease. Glyoxalase 1 (Glo1) is a critical enzyme in the clearance of toxic dicarbonyl such as methylglyoxal, precursors of AGEs. The role of AGE-independent mechanisms that underly Glo1-induced metabolic disorders have yet to be elucidated. Methods We performed a longitudinal study of female and male Glo1 heterozygous knockdown (Glo1 +/- ) mice with ~50% gene expression and screened metabolic phenotypes such as body weight, adiposity, glycemic control and plasma lipids. We also evaluated atherosclerotic burden, AGE levels, and gene expression profiles across cardiometabolic tissues (liver, adipose, muscle, kidney and aorta) to identify pathway perturbations and potential regulatory genes of Glo1 actions. Results Partial loss of Glo1 resulted in obesity, hyperglycemia, dyslipidemia, and alterations in lipid metabolism in metabolic tissues in an age- and sex-dependent manner. Glo1 +/- females displayed altered glycemic control and increased plasma triglycerides, which aligned with significant perturbations in genes involved in adipogenesis, PPARg, insulin signaling, and fatty acid metabolism pathways in liver and adipose tissues. Conversely, Glo1 +/- males developed increased skeletal muscle mass and visceral adipose depots along with changes in lipid metabolism pathways. For both cohorts, most phenotypes manifested after 14 weeks of age. Evaluation of methylglyoxal-derived AGEs demonstrated changes in only male skeletal muscle but not in female tissues, which cannot explain the broad metabolic changes observed in Glo1 +/- mice. Transcriptional profiles suggest that altered glucose and lipid metabolism may be partially explained by alternative detoxification of methylglyoxal to metabolites such as pyruvate. Moreover, transcription factor (TF) analysis of the tissue-specific gene expression data identified TFs involved in cardiometabolic diseases such as Hnf4a (all tissues) and Arntl (aorta, liver, and kidney) which are female-biased regulators and whose targets are altered in response to Glo1 +/- . Conclusions Our results indicate that Glo1 reduction perturbs metabolic health and metabolic pathways in a sex- and age-dependent manner without significant changes in AGEs across metabolic tissues. Rather, tissue-specific gene expression analysis suggests that key transcription factors such as Hfn4a and Arntl as well as metabolite changes from alternative methylglyoxal detoxification such as pyruvate, likely contribute to metabolic dysregulation in Glo1 +/- mice.
Collapse
Affiliation(s)
- Ingrid Cely
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, USA
- Molecular Toxicology Interdepartmental Program, University of California Los Angeles, Los Angeles, CA, USA
| | - Montgomery Blencowe
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, USA
- Molecular, Cellular, and Integrative Physiology Interdepartmental Ph.D. Program, University of California, Los Angeles, Los Angeles, CA, United States of America
| | - Le Shu
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, USA
- Molecular, Cellular, and Integrative Physiology Interdepartmental Ph.D. Program, University of California, Los Angeles, Los Angeles, CA, United States of America
| | - Graciel Diamante
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, USA
| | - In Sook Ahn
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Guanglin Zhang
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Jonnby LaGuardia
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Ruoshui Liu
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Zara Saleem
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Susanna Wang
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Richard Davis
- Departments of Medicine, Human Genetics, and Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Aldons J. Lusis
- Departments of Medicine, Human Genetics, and Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Medicine, Division of Cardiology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
- Department of Medicine, Human Genetics, and Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Xia Yang
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, USA
- Molecular, Cellular, and Integrative Physiology Interdepartmental Ph.D. Program, University of California, Los Angeles, Los Angeles, CA, United States of America
- Bioinformatics Interdepartmental Program, University of California, Los Angeles, Los Angeles, CA, USA
- Institute for Quantitative and Computational Biosciences, University of California, Los Angeles, Los Angeles, CA, USA
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
3
|
Dobariya P, Xie W, Rao SP, Xie J, Seelig DM, Vince R, Lee MK, More SS. Deletion of Glyoxalase 1 Exacerbates Acetaminophen-Induced Hepatotoxicity in Mice. Antioxidants (Basel) 2024; 13:648. [PMID: 38929087 PMCID: PMC11200933 DOI: 10.3390/antiox13060648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 05/19/2024] [Accepted: 05/23/2024] [Indexed: 06/28/2024] Open
Abstract
Acetaminophen (APAP) overdose triggers a cascade of intracellular oxidative stress events, culminating in acute liver injury. The clinically used antidote, N-acetylcysteine (NAC), has a narrow therapeutic window, and early treatment is essential for a satisfactory therapeutic outcome. For more versatile therapies that can be effective even at late presentation, the intricacies of APAP-induced hepatotoxicity must be better understood. Accumulation of advanced glycation end products (AGEs) and the consequent activation of the receptor for AGEs (RAGE) are considered one of the key mechanistic features of APAP toxicity. Glyoxalase 1 (Glo-1) regulates AGE formation by limiting the levels of methylglyoxal (MEG). In this study, we studied the relevance of Glo-1 in the APAP-mediated activation of RAGE and downstream cell death cascades. Constitutive Glo-1-knockout mice (GKO) and a cofactor of Glo-1, ψ-GSH, were used as tools. Our findings showed elevated oxidative stress resulting from the activation of RAGE and hepatocyte necrosis through steatosis in GKO mice treated with high-dose APAP compared to wild-type controls. A unique feature of the hepatic necrosis in GKO mice was the appearance of microvesicular steatosis as a result of centrilobular necrosis, rather than the inflammation seen in the wild type. The GSH surrogate and general antioxidant ψ-GSH alleviated APAP toxicity irrespective of the Glo-1 status, suggesting that oxidative stress is the primary driver of APAP toxicity. Overall, the exacerbation of APAP hepatotoxicity in GKO mice suggests the importance of this enzyme system in antioxidant defense against the initial stages of APAP overdose.
Collapse
Affiliation(s)
- Prakashkumar Dobariya
- Center for Drug Design, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, USA; (P.D.); (W.X.); (S.P.R.); (J.X.); (R.V.)
| | - Wei Xie
- Center for Drug Design, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, USA; (P.D.); (W.X.); (S.P.R.); (J.X.); (R.V.)
| | - Swetha Pavani Rao
- Center for Drug Design, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, USA; (P.D.); (W.X.); (S.P.R.); (J.X.); (R.V.)
| | - Jiashu Xie
- Center for Drug Design, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, USA; (P.D.); (W.X.); (S.P.R.); (J.X.); (R.V.)
| | - Davis M. Seelig
- Comparative Pathology Shared Resource, Masonic Cancer Center, University of Minnesota, St. Paul, MN 55108, USA;
- College of Veterinary Medicine, University of Minnesota, St. Paul, MN 55108, USA
| | - Robert Vince
- Center for Drug Design, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, USA; (P.D.); (W.X.); (S.P.R.); (J.X.); (R.V.)
| | - Michael K. Lee
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA;
- Institute for Translational Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA
| | - Swati S. More
- Center for Drug Design, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, USA; (P.D.); (W.X.); (S.P.R.); (J.X.); (R.V.)
| |
Collapse
|
4
|
Peter A, Schleicher E, Kliemank E, Szendroedi J, Königsrainer A, Häring HU, Nawroth PP, Fleming T. Accumulation of Non-Pathological Liver Fat Is Associated with the Loss of Glyoxalase I Activity in Humans. Metabolites 2024; 14:209. [PMID: 38668337 PMCID: PMC11051733 DOI: 10.3390/metabo14040209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 04/03/2024] [Accepted: 04/05/2024] [Indexed: 04/28/2024] Open
Abstract
The underlying molecular mechanisms for the development of non-alcoholic fatty liver (NAFL) and its progression to advanced liver diseases remain elusive. Glyoxalase 1 (Glo1) loss, leading to elevated methylglyoxal (MG) and dicarbonyl stress, has been implicated in various diseases, including obesity-related conditions. This study aimed to investigate changes in the glyoxalase system in individuals with non-pathological liver fat. Liver biopsies were obtained from 30 individuals with a narrow range of BMI (24.6-29.8 kg/m2). Whole-body insulin sensitivity was assessed using HOMA-IR. Liver biopsies were analyzed for total triglyceride content, Glo1 and Glo2 mRNA, protein expression, and activity. Liquid chromatography-tandem mass spectrometry determined liver dicarbonyl content and oxidation and glycation biomarkers. Liver Glo1 activity showed an inverse correlation with HOMA-IR and liver triglyceride content, but not BMI. Despite reduced Glo1 activity, no associations were found with elevated liver dicarbonyls or glycation markers. A sex dimorphism was observed in Glo1, with females exhibiting significantly lower liver Glo1 protein expression and activity, and higher liver MG-H1 content compared to males. This study demonstrates that increasing liver fat, even within a non-pathological range, is associated with reduced Glo1 activity.
Collapse
Affiliation(s)
- Andreas Peter
- German Centre for Diabetes Research (DZD), Helmholtz Centre Munich, 85764 Munich, Germany
- Institute for Clinical Chemistry and Pathobiochemistry, Department for Diagnostic Laboratory Medicine, University Hospital Tübingen, 72016 Tübingen, Germany
- Institute for Diabetes Research and Metabolic Diseases, Helmholtz Centre Munich, University of Tübingen, 72016 Tübingen, Germany
| | - Erwin Schleicher
- German Centre for Diabetes Research (DZD), Helmholtz Centre Munich, 85764 Munich, Germany
- Institute for Clinical Chemistry and Pathobiochemistry, Department for Diagnostic Laboratory Medicine, University Hospital Tübingen, 72016 Tübingen, Germany
| | - Elisabeth Kliemank
- German Centre for Diabetes Research (DZD), Helmholtz Centre Munich, 85764 Munich, Germany
- Department of Medicine I and Clinical Chemistry, Heidelberg University Hospital, INF 410, 69120 Heidelberg, Germany
| | - Julia Szendroedi
- German Centre for Diabetes Research (DZD), Helmholtz Centre Munich, 85764 Munich, Germany
- Department of Medicine I and Clinical Chemistry, Heidelberg University Hospital, INF 410, 69120 Heidelberg, Germany
- Joint Heidelberg-IDC Translational Diabetes Program, Internal Medicine I, Heidelberg University Hospital, 69120 Heidelberg, Germany
| | - Alfred Königsrainer
- Department of General, Visceral and Transplant Surgery, Eberhard-Karls-University Tübingen, 72016 Tübingen, Germany
| | - Hans-Ulrich Häring
- German Centre for Diabetes Research (DZD), Helmholtz Centre Munich, 85764 Munich, Germany
- Institute for Diabetes Research and Metabolic Diseases, Helmholtz Centre Munich, University of Tübingen, 72016 Tübingen, Germany
- Division of Diabetology, Endocrinology and Nephrology, Department of Internal Medicine IV, Eberhard-Karls-University Tübingen, 72016 Tübingen, Germany
| | - Peter P. Nawroth
- German Centre for Diabetes Research (DZD), Helmholtz Centre Munich, 85764 Munich, Germany
- Department of Medicine I and Clinical Chemistry, Heidelberg University Hospital, INF 410, 69120 Heidelberg, Germany
- Institute for Immunology, University Hospital of Heidelberg, INF 305, 69120 Heidelberg, Germany
| | - Thomas Fleming
- German Centre for Diabetes Research (DZD), Helmholtz Centre Munich, 85764 Munich, Germany
- Department of Medicine I and Clinical Chemistry, Heidelberg University Hospital, INF 410, 69120 Heidelberg, Germany
- Joint Heidelberg-IDC Translational Diabetes Program, Internal Medicine I, Heidelberg University Hospital, 69120 Heidelberg, Germany
| |
Collapse
|
5
|
Lai SWT, Bhattacharya S, Lopez Gonzalez EDJ, Shuck SC. Methylglyoxal-Derived Nucleoside Adducts Drive Vascular Dysfunction in a RAGE-Dependent Manner. Antioxidants (Basel) 2024; 13:85. [PMID: 38247509 PMCID: PMC10812505 DOI: 10.3390/antiox13010085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 12/29/2023] [Accepted: 01/04/2024] [Indexed: 01/23/2024] Open
Abstract
Diabetic kidney disease (DKD) is a leading cause of death in patients with diabetes. An early precursor to DKD is endothelial cell dysfunction (ECD), which often precedes and exacerbates vascular disease progression. We previously discovered that covalent adducts formed on DNA, RNA, and proteins by the reactive metabolic by-product methylglyoxal (MG) predict DKD risk in patients with type 1 diabetes up to 16 years pre-diagnosis. However, the mechanisms by which MG adducts contribute to vascular disease onset and progression remain unclear. Here, we report that the most predominant MG-induced nucleoside adducts, N2-(1-carboxyethyl)-deoxyguanosine (CEdG) and N2-(1-carboxyethyl)-guanosine (CEG), drive endothelial dysfunction. Following CEdG or CEG exposure, primary human umbilical vein endothelial cells (HUVECs) undergo endothelial dysfunction, resulting in enhanced monocyte adhesion, increased reactive oxygen species production, endothelial permeability, impaired endothelial homeostasis, and exhibit a dysfunctional transcriptomic signature. These effects were discovered to be mediated through the receptor for advanced glycation end products (RAGE), as an inhibitor for intracellular RAGE signaling diminished these dysfunctional phenotypes. Therefore, we found that not only are MG adducts biomarkers for DKD, but that they may also have a role as potential drivers of vascular disease onset and progression and a new therapeutic modality.
Collapse
Affiliation(s)
- Seigmund Wai Tsuen Lai
- Department of Diabetes and Cancer Metabolism, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA; (S.W.T.L.); (E.D.J.L.G.)
| | - Supriyo Bhattacharya
- Department of Computational and Quantitative Medicine, City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA;
| | - Edwin De Jesus Lopez Gonzalez
- Department of Diabetes and Cancer Metabolism, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA; (S.W.T.L.); (E.D.J.L.G.)
| | - Sarah C. Shuck
- Department of Diabetes and Cancer Metabolism, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA; (S.W.T.L.); (E.D.J.L.G.)
| |
Collapse
|
6
|
Dobariya P, Xie W, Rao SP, Xie J, Seelig DM, Vince R, Lee MK, More SS. Deletion of Glyoxalase 1 exacerbates acetaminophen-induced hepatotoxicity in mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.21.572856. [PMID: 38187538 PMCID: PMC10769331 DOI: 10.1101/2023.12.21.572856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Acetaminophen (APAP) overdose triggers a cascade of intracellular oxidative stress events culminating in acute liver injury. The clinically used antidote, N-acetylcysteine (NAC) has a narrow therapeutic window and early treatment is essential for satisfactory therapeutic outcome. For more versatile therapies that can be effective even at late-presentation, the intricacies of APAP-induced hepatotoxicity must be better understood. Accumulation of advanced glycation end-products (AGEs) and consequent activation of the receptor for AGEs (RAGE) are considered one of the key mechanistic features of APAP toxicity. Glyoxalase-1 (Glo-1) regulates AGE formation by limiting the levels of methylglyoxal (MEG). In this study, we studied the relevance of Glo-1 in APAP mediated activation of RAGE and downstream cell-death cascades. Constitutive Glo-1 knockout mice (GKO) and a cofactor of Glo-1, ψ-GSH, were employed as tools. Our findings show elevated oxidative stress, activation of RAGE and hepatocyte necrosis through steatosis in GKO mice treated with high-dose APAP compared to wild type controls. A unique feature of the hepatic necrosis in GKO mice is the appearance of microvesicular steatosis as a result of centrilobular necrosis, rather than inflammation seen in wild type. The GSH surrogate and general antioxidant, ψ-GSH alleviated APAP toxicity irrespective of Glo-1 status, suggesting that oxidative stress being the primary driver of APAP toxicity. Overall, exacerbation of APAP hepatotoxicity in GKO mice suggests the importance of this enzyme system in antioxidant defense against initial stages of APAP overdose.
Collapse
Affiliation(s)
- Prakashkumar Dobariya
- Center for Drug Design, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota 55455, USA
| | - Wei Xie
- Center for Drug Design, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota 55455, USA
| | - Swetha Pavani Rao
- Center for Drug Design, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota 55455, USA
| | - Jiashu Xie
- Center for Drug Design, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota 55455, USA
| | - Davis M. Seelig
- Comparative Pathology Shared Resource, Masonic Cancer Center, University of Minnesota, St. Paul, Minnesota 55108, USA
- College of Veterinary Medicine, University of Minnesota, St. Paul, Minnesota 55108, USA
| | - Robert Vince
- Center for Drug Design, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota 55455, USA
| | - Michael K. Lee
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota 55455, USA
- Institute for Translational Neuroscience, University of Minnesota, Minneapolis, Minnesota 55455, USA
| | - Swati S. More
- Center for Drug Design, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota 55455, USA
| |
Collapse
|
7
|
Santacroce G, Gentile A, Soriano S, Novelli A, Lenti MV, Di Sabatino A. Glutathione: Pharmacological aspects and implications for clinical use in non-alcoholic fatty liver disease. Front Med (Lausanne) 2023; 10:1124275. [PMID: 37035339 PMCID: PMC10075255 DOI: 10.3389/fmed.2023.1124275] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 02/27/2023] [Indexed: 04/11/2023] Open
Abstract
Glutathione is a tripeptide synthesized at cytosolic level, that exists in cells in a reduced form (thiol-reduced-GSH-) and in an oxidized form (disulfide-oxidized). The antioxidant function of GSH has led to speculation about its therapeutic role in numerous chronic diseases characterized by altered redox balance and reduced GSH levels, including, for instance, neurodegenerative disorders, cancer, and chronic liver diseases. Among these latter, non-alcoholic fatty liver disease (NAFLD), characterized by lipid accumulation in hepatocytes, in the absence of alcohol abuse or other steatogenic factors, is one of the most prevalent. The umbrella term NAFLD includes the pure liver fat accumulation, the so-called hepatic steatosis or non-alcoholic fatty liver, and the progressive form with inflammation, also known as non-alcoholic steatohepatitis, which is related to the increase in oxidative stress and reactive oxygen species, eventually leading to liver fibrosis. Although the pathogenetic role of oxidative stress in these diseases is well established, there is still limited evidence on the therapeutic role of GSH in such conditions. Hence, the aim of this review is to depict the current molecular and pharmacological knowledge on glutathione, focusing on the available studies related to its therapeutic activity in NAFLD.
Collapse
Affiliation(s)
- Giovanni Santacroce
- Department of Internal Medicine and Medical Therapeutics, University of Pavia, Pavia, Italy
- First Department of Internal Medicine, San Matteo Hospital Foundation, Pavia, Italy
| | - Antonella Gentile
- Department of Internal Medicine and Medical Therapeutics, University of Pavia, Pavia, Italy
- First Department of Internal Medicine, San Matteo Hospital Foundation, Pavia, Italy
| | - Simone Soriano
- Department of Internal Medicine and Medical Therapeutics, University of Pavia, Pavia, Italy
- First Department of Internal Medicine, San Matteo Hospital Foundation, Pavia, Italy
| | - Andrea Novelli
- Department of Health Sciences, Clinical Pharmacology and Oncology Section, Università Degli Studi di Firenze, Firenze, Italy
| | - Marco Vincenzo Lenti
- Department of Internal Medicine and Medical Therapeutics, University of Pavia, Pavia, Italy
- First Department of Internal Medicine, San Matteo Hospital Foundation, Pavia, Italy
| | - Antonio Di Sabatino
- Department of Internal Medicine and Medical Therapeutics, University of Pavia, Pavia, Italy
- First Department of Internal Medicine, San Matteo Hospital Foundation, Pavia, Italy
- *Correspondence: Antonio Di Sabatino,
| |
Collapse
|
8
|
Chua D, Low ZS, Cheam GX, Ng AS, Tan NS. Utility of Human Relevant Preclinical Animal Models in Navigating NAFLD to MAFLD Paradigm. Int J Mol Sci 2022; 23:14762. [PMID: 36499091 PMCID: PMC9737809 DOI: 10.3390/ijms232314762] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 11/15/2022] [Accepted: 11/23/2022] [Indexed: 11/29/2022] Open
Abstract
Fatty liver disease is an emerging contributor to disease burden worldwide. The past decades of work established the heterogeneous nature of non-alcoholic fatty liver disease (NAFLD) etiology and systemic contributions to the pathogenesis of the disease. This called for the proposal of a redefinition in 2020 to that of metabolic dysfunction-associated fatty liver disease (MAFLD) to better reflect the current understanding of the disease. To date, several clinical cohort studies comparing NAFLD and MAFLD hint at the relevancy of the new nomenclature in enriching for patients with more severe hepatic injury and extrahepatic comorbidities. However, the underlying systemic pathogenesis is still not fully understood. Preclinical animal models have been imperative in elucidating key biological mechanisms in various contexts, including intrahepatic disease progression, interorgan crosstalk and systemic dysregulation. Furthermore, they are integral in developing novel therapeutics against MAFLD. However, substantial contextual variabilities exist across different models due to the lack of standardization in several aspects. As such, it is crucial to understand the strengths and weaknesses of existing models to better align them to the human condition. In this review, we consolidate the implications arising from the change in nomenclature and summarize MAFLD pathogenesis. Subsequently, we provide an updated evaluation of existing MAFLD preclinical models in alignment with the new definitions and perspectives to improve their translational relevance.
Collapse
Affiliation(s)
- Damien Chua
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, 11 Mandalay Road, Singapore 308232, Singapore
| | - Zun Siong Low
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, 11 Mandalay Road, Singapore 308232, Singapore
| | - Guo Xiang Cheam
- School of Biological Sciences, Nanyang Technological University Singapore, 60 Nanyang Drive, Singapore 637551, Singapore
| | - Aik Seng Ng
- Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DU, UK
| | - Nguan Soon Tan
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, 11 Mandalay Road, Singapore 308232, Singapore
- School of Biological Sciences, Nanyang Technological University Singapore, 60 Nanyang Drive, Singapore 637551, Singapore
| |
Collapse
|
9
|
Diamanti K, Cavalli M, Pereira MJ, Pan G, Castillejo-López C, Kumar C, Mundt F, Komorowski J, Deshmukh AS, Mann M, Korsgren O, Eriksson JW, Wadelius C. Organ-specific metabolic pathways distinguish prediabetes, type 2 diabetes, and normal tissues. Cell Rep Med 2022; 3:100763. [PMID: 36198307 PMCID: PMC9589007 DOI: 10.1016/j.xcrm.2022.100763] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Revised: 07/02/2022] [Accepted: 09/13/2022] [Indexed: 11/28/2022]
Abstract
Environmental and genetic factors cause defects in pancreatic islets driving type 2 diabetes (T2D) together with the progression of multi-tissue insulin resistance. Mass spectrometry proteomics on samples from five key metabolic tissues of a cross-sectional cohort of 43 multi-organ donors provides deep coverage of their proteomes. Enrichment analysis of Gene Ontology terms provides a tissue-specific map of altered biological processes across healthy, prediabetes (PD), and T2D subjects. We find widespread alterations in several relevant biological pathways, including increase in hemostasis in pancreatic islets of PD, increase in the complement cascade in liver and pancreatic islets of PD, and elevation in cholesterol biosynthesis in liver of T2D. Our findings point to inflammatory, immune, and vascular alterations in pancreatic islets in PD that are hypotheses to be tested for potential contributions to hormonal perturbations such as impaired insulin and increased glucagon production. This multi-tissue proteomic map suggests tissue-specific metabolic dysregulations in T2D.
Collapse
Affiliation(s)
- Klev Diamanti
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Marco Cavalli
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Maria J Pereira
- Department of Medical Sciences, Clinical Diabetes and Metabolism, Uppsala University, Uppsala, Sweden
| | - Gang Pan
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Casimiro Castillejo-López
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Chanchal Kumar
- Translational Science & Experimental Medicine, Early Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden; Karolinska Institutet/AstraZeneca Integrated CardioMetabolic Center (KI/AZ ICMC), Department of Medicine, Novum, Huddinge, Sweden
| | - Filip Mundt
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark; Department of Oncology-Pathology, Karolinska Institute, Stockholm, Sweden
| | - Jan Komorowski
- Science for Life Laboratory, Department of Cell and Molecular Biology, Uppsala University, Uppsala, Sweden; Institute of Computer Science, Polish Academy of Sciences, Warsaw, Poland; Washington National Primate Research Center, Seattle, WA, USA; Swedish Collegium for Advanced Study, Uppsala, Sweden
| | - Atul S Deshmukh
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark; Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Science, University of Copenhagen, Copenhagen, Denmark
| | - Matthias Mann
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark; Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Olle Korsgren
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden; Department of Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Jan W Eriksson
- Department of Medical Sciences, Clinical Diabetes and Metabolism, Uppsala University, Uppsala, Sweden
| | - Claes Wadelius
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
10
|
Lai SWT, Lopez Gonzalez EDJ, Zoukari T, Ki P, Shuck SC. Methylglyoxal and Its Adducts: Induction, Repair, and Association with Disease. Chem Res Toxicol 2022; 35:1720-1746. [PMID: 36197742 PMCID: PMC9580021 DOI: 10.1021/acs.chemrestox.2c00160] [Citation(s) in RCA: 68] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Metabolism is an essential part of life that provides energy for cell growth. During metabolic flux, reactive electrophiles are produced that covalently modify macromolecules, leading to detrimental cellular effects. Methylglyoxal (MG) is an abundant electrophile formed from lipid, protein, and glucose metabolism at intracellular levels of 1-4 μM. MG covalently modifies DNA, RNA, and protein, forming advanced glycation end products (MG-AGEs). MG and MG-AGEs are associated with the onset and progression of many pathologies including diabetes, cancer, and liver and kidney disease. Regulating MG and MG-AGEs is a potential strategy to prevent disease, and they may also have utility as biomarkers to predict disease risk, onset, and progression. Here, we review recent advances and knowledge surrounding MG, including its production and elimination, mechanisms of MG-AGEs formation, the physiological impact of MG and MG-AGEs in disease onset and progression, and the latter in the context of its receptor RAGE. We also discuss methods for measuring MG and MG-AGEs and their clinical application as prognostic biomarkers to allow for early detection and intervention prior to disease onset. Finally, we consider relevant clinical applications and current therapeutic strategies aimed at targeting MG, MG-AGEs, and RAGE to ultimately improve patient outcomes.
Collapse
Affiliation(s)
- Seigmund Wai Tsuen Lai
- Department of Diabetes and Cancer Metabolism, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope Comprehensive Cancer Center, Duarte, California 91010, United States
| | - Edwin De Jesus Lopez Gonzalez
- Department of Diabetes and Cancer Metabolism, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope Comprehensive Cancer Center, Duarte, California 91010, United States
| | - Tala Zoukari
- Department of Diabetes and Cancer Metabolism, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope Comprehensive Cancer Center, Duarte, California 91010, United States
| | - Priscilla Ki
- Department of Diabetes and Cancer Metabolism, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope Comprehensive Cancer Center, Duarte, California 91010, United States
| | - Sarah C Shuck
- Department of Diabetes and Cancer Metabolism, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope Comprehensive Cancer Center, Duarte, California 91010, United States
| |
Collapse
|
11
|
Prisco SZ, Hartweck L, Keen JL, Vogel N, Kazmirczak F, Eklund M, Hemnes AR, Brittain EL, Prins KW. Glyoxylase-1 combats dicarbonyl stress and right ventricular dysfunction in rodent pulmonary arterial hypertension. Front Cardiovasc Med 2022; 9:940932. [PMID: 36093169 PMCID: PMC9452736 DOI: 10.3389/fcvm.2022.940932] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 08/01/2022] [Indexed: 01/06/2023] Open
Abstract
Background Heightened glycolytic flux is associated with right ventricular (RV) dysfunction in pulmonary arterial hypertension (PAH). Methylglyoxal, a glycolysis byproduct, is a highly reactive dicarbonyl that has toxic effects via non-enzymatic post-translational modifications (protein glycation). Methylglyoxal is degraded by the glyoxylase system, which includes the rate-limiting enzyme glyoxylase-1 (GLO1), to combat dicarbonyl stress. However, the potential consequences of excess protein glycation on RV function are unknown. Methods Bioinformatics analysis of previously identified glycated proteins predicted how protein glycation regulated cardiac biology. Methylglyoxal treatment of H9c2 cardiomyocytes evaluated the consequences of excess protein glycation on mitochondrial respiration. The effects of adeno-associated virus serotype 9-mediated (AAV9) GLO1 expression on RV function in monocrotaline rats were quantified with echocardiography and hemodynamic studies. Immunoblots and immunofluorescence were implemented to probe the effects of AAV-Glo1 on total protein glycation and fatty acid oxidation (FAO) and fatty acid binding protein levels. Results In silico analyses highlighted multiple mitochondrial metabolic pathways may be affected by protein glycation. Exogenous methylglyoxal minimally altered mitochondrial respiration when cells metabolized glucose, however methylglyoxal depressed FAO. AAV9-Glo1 increased RV cardiomyocyte GLO1 expression, reduced total protein glycation, partially restored mitochondrial density, and decreased lipid accumulation. In addition, AAV9-Glo1 increased RV levels of FABP4, a fatty acid binding protein, and hydroxyacyl-CoA dehydrogenase trifunctional multienzyme complex subunits alpha and beta (HADHA and HADHB), the two subunits of the mitochondrial trifunctional protein for FAO. Finally, AAV9-Glo1 blunted RV fibrosis and improved RV systolic and diastolic function. Conclusion Excess protein glycation promotes RV dysfunction in preclinical PAH, potentially through suppression of FAO.
Collapse
Affiliation(s)
- Sasha Z. Prisco
- Cardiovascular Division, Department of Medicine, Lillehei Heart Institute, University of Minnesota, Minneapolis, MN, United States
| | - Lynn Hartweck
- Cardiovascular Division, Department of Medicine, Lillehei Heart Institute, University of Minnesota, Minneapolis, MN, United States
| | - Jennifer L. Keen
- Pulmonary and Critical Care, Department of Medicine, University of Minnesota, Minneapolis, MN, United States
| | - Neal Vogel
- Cardiovascular Division, Department of Medicine, Lillehei Heart Institute, University of Minnesota, Minneapolis, MN, United States
| | - Felipe Kazmirczak
- Cardiovascular Division, Department of Medicine, Lillehei Heart Institute, University of Minnesota, Minneapolis, MN, United States
| | - Megan Eklund
- Cardiovascular Division, Department of Medicine, Lillehei Heart Institute, University of Minnesota, Minneapolis, MN, United States
| | - Anna R. Hemnes
- Division of Allergy Pulmonary and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Evan L. Brittain
- Division of Cardiovascular Medicine and Vanderbilt Translational and Clinical Cardiovascular Research Center, Nashville, TN, United States
| | - Kurt W. Prins
- Cardiovascular Division, Department of Medicine, Lillehei Heart Institute, University of Minnesota, Minneapolis, MN, United States
| |
Collapse
|
12
|
Michel M, Hess C, Kaps L, Kremer WM, Hilscher M, Galle PR, Moehler M, Schattenberg JM, Wörns MA, Labenz C, Nagel M. Elevated serum levels of methylglyoxal are associated with impaired liver function in patients with liver cirrhosis. Sci Rep 2021; 11:20506. [PMID: 34654829 PMCID: PMC8519993 DOI: 10.1038/s41598-021-00119-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 10/06/2021] [Indexed: 12/15/2022] Open
Abstract
Methylglyoxal (MGO) is a highly reactive dicarbonyl species that forms advanced glycation end products (AGEs). The binding of these AGEs to their receptor (RAGE) causes and sustains severe inflammation. Systemic inflammation is postulated to be a major driver in the progression of liver cirrhosis. However, the role of circulating MGO levels in liver cirrhosis remains unknown. In this study, we investigated the serum levels of two dicarbonyl species, MGO and glyoxal (GO) using tandem mass spectrometry (HPLC-MS/MS) and evaluated their association with disease severity. A total of 51 inpatients and outpatients with liver cirrhosis of mixed etiology and different disease stages were included. Elevated MGO levels were seen in an advanced stage of liver cirrhosis (p < 0.001). High MGO levels remained independently associated with impaired liver function, as assessed by the model for end-stage liver disease (MELD) (β = 0.448, p = 0.002) and acute decompensation (AD) (β = 0.345, p = 0.005) scores. Furthermore, MGO was positively correlated with markers of systemic inflammation (IL-6, p = 0.004) and the development of ascites (p = 0.013). In contrast, no changes were seen in GO serum levels. Circulating levels of MGO are elevated in advanced stages of liver cirrhosis and are associated with impaired liver function and liver-related parameters.
Collapse
Affiliation(s)
- Maurice Michel
- I. Department of Medicine, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, 55131, Mainz, Germany.
| | - Cornelius Hess
- Institute of Forensic Medicine, Forensic Toxicology, University Medical Center of the Johannes Gutenberg University Mainz, 55131, Mainz, Germany
| | - Leonard Kaps
- I. Department of Medicine, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, 55131, Mainz, Germany
| | - Wolfgang M Kremer
- I. Department of Medicine, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, 55131, Mainz, Germany
| | - Max Hilscher
- I. Department of Medicine, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, 55131, Mainz, Germany
| | - Peter R Galle
- I. Department of Medicine, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, 55131, Mainz, Germany
| | - Markus Moehler
- I. Department of Medicine, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, 55131, Mainz, Germany
| | - Jörn M Schattenberg
- I. Department of Medicine, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, 55131, Mainz, Germany
| | - Marcus-Alexander Wörns
- I. Department of Medicine, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, 55131, Mainz, Germany
| | - Christian Labenz
- I. Department of Medicine, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, 55131, Mainz, Germany
| | - Michael Nagel
- I. Department of Medicine, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, 55131, Mainz, Germany.
| |
Collapse
|
13
|
Petriv N, Neubert L, Vatashchuk M, Timrott K, Suo H, Hochnadel I, Huber R, Petzold C, Hrushchenko A, Yatsenko AS, Shcherbata HR, Wedemeyer H, Lichtinghagen R, Falfushynska H, Lushchak V, Manns MP, Bantel H, Semchyshyn H, Yevsa T. Increase of α-dicarbonyls in liver and receptor for advanced glycation end products on immune cells are linked to nonalcoholic fatty liver disease and liver cancer. Oncoimmunology 2021; 10:1874159. [PMID: 33628620 PMCID: PMC7889131 DOI: 10.1080/2162402x.2021.1874159] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is the most common primary malignancy of the liver with a very poor prognosis and constantly growing incidence. Among other primary risks of HCC, metabolic disorders and obesity have been extensively investigated over recent decades. The latter can promote nonalcoholic fatty liver disease (NAFLD) leading to the inflammatory form of nonalcoholic steatohepatitis (NASH), that, in turn, promotes HCC. Molecular determinants of this pathogenic progression, however, remain largely undefined. In this study, we have focussed on the investigation of α-dicarbonyl compounds (α-dC), highly reactive and tightly associated with overweight-induced metabolic disorders, and studied their potential role in NAFLD and progression toward HCC using murine models. NAFLD was induced using high-fat diet (HFD). Autochthonous HCC was induced using transposon-based stable intrahepatic overexpression of oncogenic NRASG12V in mice lacking p19Arf tumor suppressor. Our study demonstrates that the HFD regimen and HCC resulted in strong upregulation of α-dC in the liver, heart, and muscles. In addition, an increase in α-dC was confirmed in sera of NAFLD and NASH patients. Furthermore, higher expression of the receptor for advanced glycation products (RAGE) was detected exclusively on immune cells and not on stroma cells in livers of mice with liver cancer progression. Our work confirms astable interplay of liver inflammation, carbonyl stress mediated by α-dC, and upregulated RAGE expression on CD8+ Tand natural killer (NK) cells in situ in NAFLD and HCC, as key factors/determinants in liver disease progression. The obtained findings underline the role of α-dC and RAGE+CD8+ Tand RAGE+ NK cells as biomarkers and candidates for a local therapeutic intervention in NAFLD and malignant liver disease.
Collapse
Affiliation(s)
- Nataliia Petriv
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Lavinia Neubert
- Institute of Pathology, Hannover Medical School, Hannover, Germany
| | - Myroslava Vatashchuk
- Department of Biochemistry and Biotechnology, Vasyl Stefanyk Precarpathian National University, Ivano-Frankivsk, Ukraine
| | - Kai Timrott
- Department of General-, Visceral and Transplantation Surgery, Hannover Medical School, Hannover, Germany
| | - Huizhen Suo
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Inga Hochnadel
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - René Huber
- Department of Clinical Chemistry, Hannover Medical School, Hannover, Germany
| | | | - Anastasiia Hrushchenko
- Department of Biochemistry and Biotechnology, Vasyl Stefanyk Precarpathian National University, Ivano-Frankivsk, Ukraine
| | - Andriy S Yatsenko
- Gene Expression and Signaling Group, Institute of Cell Biochemistry, Hannover Medical School, Germany
| | - Halyna R Shcherbata
- Gene Expression and Signaling Group, Institute of Cell Biochemistry, Hannover Medical School, Germany
| | - Heiner Wedemeyer
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Ralf Lichtinghagen
- Department of Clinical Chemistry, Hannover Medical School, Hannover, Germany
| | - Halina Falfushynska
- Department of Biochemistry, Ternopil Volodymyr Hnatiuk National Pedagogical University, Ternopil, Ukraine
| | - Volodymyr Lushchak
- Department of Biochemistry and Biotechnology, Vasyl Stefanyk Precarpathian National University, Ivano-Frankivsk, Ukraine
| | - Michael P Manns
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Heike Bantel
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Halyna Semchyshyn
- Department of Biochemistry and Biotechnology, Vasyl Stefanyk Precarpathian National University, Ivano-Frankivsk, Ukraine
| | - Tetyana Yevsa
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| |
Collapse
|
14
|
Morgenstern J, Campos Campos M, Nawroth P, Fleming T. The Glyoxalase System-New Insights into an Ancient Metabolism. Antioxidants (Basel) 2020; 9:antiox9100939. [PMID: 33019494 PMCID: PMC7600140 DOI: 10.3390/antiox9100939] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 09/24/2020] [Accepted: 09/28/2020] [Indexed: 02/07/2023] Open
Abstract
The glyoxalase system was discovered over a hundred years ago and since then it has been claimed to provide the role of an indispensable enzyme system in order to protect cells from a toxic byproduct of glycolysis. This review gives a broad overview of what has been postulated in the last 30 years of glyoxalase research, but within this context it also challenges the concept that the glyoxalase system is an exclusive tool of detoxification and that its substrate, methylglyoxal, is solely a detrimental burden for every living cell due to its toxicity. An overview of consequences of a complete loss of the glyoxalase system in various model organisms is presented with an emphasis on the role of alternative detoxification pathways of methylglyoxal. Furthermore, this review focuses on the overlooked posttranslational modification of Glyoxalase 1 and its possible implications for cellular maintenance under various (patho-)physiological conditions. As a final note, an intriguing point of view for the substrate methylglyoxal is offered, the concept of methylglyoxal (MG)-mediated hormesis.
Collapse
Affiliation(s)
- Jakob Morgenstern
- Department of Internal Medicine I and Clinical Chemistry, University Hospital Heidelberg, 69120 Heidelberg, Germany; (M.C.C.); (P.N.); (T.F.)
- Correspondence:
| | - Marta Campos Campos
- Department of Internal Medicine I and Clinical Chemistry, University Hospital Heidelberg, 69120 Heidelberg, Germany; (M.C.C.); (P.N.); (T.F.)
| | - Peter Nawroth
- Department of Internal Medicine I and Clinical Chemistry, University Hospital Heidelberg, 69120 Heidelberg, Germany; (M.C.C.); (P.N.); (T.F.)
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany
- Institute for Diabetes and Cancer at Helmholtz Zentrum Munich, 85764 Neuherberg, Germany
| | - Thomas Fleming
- Department of Internal Medicine I and Clinical Chemistry, University Hospital Heidelberg, 69120 Heidelberg, Germany; (M.C.C.); (P.N.); (T.F.)
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany
| |
Collapse
|
15
|
Han B, Jian Y, Xia X, Hu W, Zhang L, Zhou P. Studying the effects of sea cucumber ovum powder on nonalcoholic fatty liver disease by proteomics techniques in a rat model. Food Funct 2020; 11:6139-6147. [PMID: 32573635 DOI: 10.1039/d0fo00741b] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Sea cucumber is a valuable marine food that has antioxidant, anti-diabetic, and anti-obesity functionalities. Sea cucumber ovum (SCO) may contain functional components, however, it is considered to be a waste product during industrial processing. In order to make good use of SCO, this work investigated the effects of freeze-dried SCO powder on NAFLD, using a rat model, through iBT labeling proteomics techniques, tracking changes in the hepatic protein profiles of rats whose diets were supplemented with SCO powder. Male rats were fed with standard food, a high fat diet (HFD), or a HFD supplemented with 150 mg per kg BW or 450 mg per kg BW SCO powder for 6 weeks. Compared with the HFD, low-dose SCO supplementation in the diet could significantly reduce body weight gain and liver weight. Furthermore, in total, 5922 proteins were identified, and 767 proteins were found to be significantly different proteins (p < 0.05) among all four groups. Most of the significantly different proteins were related to apoptosis and lipid metabolism. Fadd, Dci, and Aif1 have been identified as key proteins in the pathways related to apoptosis, lipid metabolism, and inflammation. The results in this study provide an overview of the SCO-induced changes in the liver proteome of NAFLD, which may help us to understand the molecular mechanism behind the effects of SCO on the alleviation of NAFLD.
Collapse
Affiliation(s)
- Binsong Han
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China.
| | | | | | | | | | | |
Collapse
|
16
|
From personalised nutrition to precision medicine: the rise of consumer genomics and digital health. Proc Nutr Soc 2020; 79:300-310. [PMID: 32468984 DOI: 10.1017/s0029665120006977] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Advances in genomics generated the concept that a better understanding of individual characteristics, e.g. genotype, will lead to improved tailoring of pharmaceutical and nutritional therapies. Subsequent developments in proteomics and metabolomics, in addition to wearable technologies for tracking parameters, such as dietary intakes, physical activity, heart rate and blood glucose, have further driven this idea. Alongside these innovations, there has been a rapid rise in companies offering direct-to-consumer genetic and/or microbiome testing, in combination with the marketing of personalised nutrition services. Key scientific questions include how disparate datasets are integrated, how accurate are current predictions and how these may be developed in the future. In this regard, lessons can be learned from systems biology, which aims both to integrate data from different levels of organisation (e.g. genomic, proteomic and metabolomic) and predict the emergent behaviours of biological systems or organisms as a whole. The present paper reviews the origins and recent advancement of 'big data' and systems approaches in medicine and nutrition. Conclusions are that systems integration of multiple technologies has generated mechanistic insights and informed the evolution of precision medicine and personalised nutrition. Pertinent ethical issues include who is entitled to access new technologies and how commercial companies are storing, using and/or re-mining consumer data. Questions about efficacy (both long-term behavioural change and health outcomes), cost-benefit and impacts on health inequalities remain to be fully addressed.
Collapse
|
17
|
Huang B, Yao Y, Li Y, Yang H, Liu H, Liu H, Li D, Shu W, Chen M. Proteomics approach to investigate dynamic protein profile involved in high fat diet-induced fatty liver disease in rats. J Toxicol Pathol 2019; 32:223-232. [PMID: 31719749 PMCID: PMC6831498 DOI: 10.1293/tox.2018-0045] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2018] [Accepted: 03/19/2019] [Indexed: 12/15/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a disorder of the liver found worldwide. The
molecular mechanisms underlying NAFLD initiation and progression, however, remain poorly
understood. In this study, fluorescence difference gel electrophoresis (DIGE) combined
with mass spectrometry was performed to profile the intracellular processes in the rat
liver at the proteome level when rats were fed a high-fat diet for 8 weeks. Dynamic
changes of 27 protein spots were observed. Among them, upregulation of 14 spots and
downregulation of 13 spots were observed during the eight weeks of the high fat
diet-induction period. These spots were analyzed by matrix-assisted laser
desorption/ionization tandem time-of-flight mass spectrometry (MALDI-TOF/TOF), and
ultimately 24 proteins were identified with more than 95% confidence. Gene ontology (GO)
annotation indicated that these proteins were implicated in the metabolism of
carbohydrates, lipids, and amino acids. Four proteins were validated by western blot.
Further functional studies on these dynamically changing proteins may lead to a better
understanding of the mechanisms of high fat diet-induced fatty liver disease.
Collapse
Affiliation(s)
- Baohua Huang
- State Key Laboratory for Chemistry and Molecular Engineering of Medical Resources, Guangxi Normal University, 15 Yucai Road, Guilin city, Guangxi Province, 541004, China
| | - Yanling Yao
- State Key Laboratory for Chemistry and Molecular Engineering of Medical Resources, Guangxi Normal University, 15 Yucai Road, Guilin city, Guangxi Province, 541004, China
| | - Yaping Li
- State Key Laboratory for Chemistry and Molecular Engineering of Medical Resources, Guangxi Normal University, 15 Yucai Road, Guilin city, Guangxi Province, 541004, China
| | - Hua Yang
- State Key Laboratory for Chemistry and Molecular Engineering of Medical Resources, Guangxi Normal University, 15 Yucai Road, Guilin city, Guangxi Province, 541004, China
| | - Huchen Liu
- State Key Laboratory for Chemistry and Molecular Engineering of Medical Resources, Guangxi Normal University, 15 Yucai Road, Guilin city, Guangxi Province, 541004, China
| | - Heng Liu
- Department of Cell Biology and Genetics, Guangxi Medical University, 22 Shuangyong Road, Nanning city, Guangxi Province, 530021, China
| | - Dongming Li
- Department of Cell Biology and Genetics, Guangxi Medical University, 22 Shuangyong Road, Nanning city, Guangxi Province, 530021, China
| | - Wei Shu
- Department of Cell Biology and Genetics, Guangxi Medical University, 22 Shuangyong Road, Nanning city, Guangxi Province, 530021, China
| | - Ming Chen
- State Key Laboratory for Chemistry and Molecular Engineering of Medical Resources, Guangxi Normal University, 15 Yucai Road, Guilin city, Guangxi Province, 541004, China
| |
Collapse
|
18
|
Moore JB, Thorne JL. Predicting and reducing hepatic lipotoxicity in non-alcoholic fatty liver disease. Lab Anim (NY) 2019; 48:143-144. [PMID: 31000821 DOI: 10.1038/s41684-019-0291-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
| | - James L Thorne
- School of Food Science & Nutrition, University of Leeds, Leeds, UK
| |
Collapse
|
19
|
Cha SH, Hwang Y, Heo SJ, Jun HS. Indole-4-carboxaldehyde Isolated from Seaweed, Sargassum thunbergii, Attenuates Methylglyoxal-Induced Hepatic Inflammation. Mar Drugs 2019; 17:E486. [PMID: 31438528 PMCID: PMC6780312 DOI: 10.3390/md17090486] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 07/19/2019] [Accepted: 08/16/2019] [Indexed: 12/16/2022] Open
Abstract
Glucose degradation is aberrantly increased in hyperglycemia, which causes various harmful effects on the liver. Glyoxalase-1 (Glo-1) is a ubiquitous cellular enzyme that participates in the detoxification of methylglyoxal (MGO), a cytotoxic byproduct of glycolysis that induces protein modification (advanced glycation end-products, AGEs) and inflammation. Here, we investigated the anti-inflammatory effect of indole-4-carboxaldehyde (ST-I4C), which was isolated from the edible seaweed Sargassum thunbergii, on MGO-induced inflammation in HepG2 cells, a human hepatocyte cell line. ST-I4C attenuated the MGO-induced expression of inflammatory-related genes, such as tumor necrosis factor (TNF)-α and IFN-γ by activating nuclear factor-kappa B (NF-κB) without toxicity in HepG2 cells. In addition, ST-I4C reduced the MGO-induced AGE formation and the expression of the receptor for AGE (RAGE). Interestingly, both the mRNA and protein expression levels of Glo-1 increased following ST-I4C treatment, and the decrease in Glo-1 mRNA expression caused by MGO exposure was rescued by ST-I4C pretreatment. These results suggest that ST-I4C shows anti-inflammatory activity against MGO-induced inflammation in human hepatocytes by preventing an increase in the pro-inflammatory gene expression and AGE formation. Therefore, it represents a potential therapeutic agent for the prevention of hepatic steatosis.
Collapse
Affiliation(s)
- Seon-Heui Cha
- Department of Marine Biomedical Sciences, Hanseo University, Chungcheongnam-do 31962, Korea
| | - Yongha Hwang
- Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon 21999, Korea
- College of Pharmacy, Gachon University, Incheon 21999, Korea
| | - Soo-Jin Heo
- Jeju International Marine Science Center for Research & Education, Korea Institute of Ocean Science & Technology (KIOST), Jeju 63349, Korea
| | - Hee-Sook Jun
- Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon 21999, Korea.
- College of Pharmacy, Gachon University, Incheon 21999, Korea.
- Gachon Medical and Convergence Institute, Gachon Gil Medical Center, Incheon 21999, Korea.
| |
Collapse
|
20
|
Parker BL, Calkin AC, Seldin MM, Keating MF, Tarling EJ, Yang P, Moody SC, Liu Y, Zerenturk EJ, Needham EJ, Miller ML, Clifford BL, Morand P, Watt MJ, Meex RCR, Peng KY, Lee R, Jayawardana K, Pan C, Mellett NA, Weir JM, Lazarus R, Lusis AJ, Meikle PJ, James DE, de Aguiar Vallim TQ, Drew BG. An integrative systems genetic analysis of mammalian lipid metabolism. Nature 2019; 567:187-193. [PMID: 30814737 PMCID: PMC6656374 DOI: 10.1038/s41586-019-0984-y] [Citation(s) in RCA: 91] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Accepted: 01/23/2019] [Indexed: 12/16/2022]
Abstract
Dysregulation of lipid homeostasis is a precipitating event in the pathogenesis and progression of hepatosteatosis and metabolic syndrome. These conditions are highly prevalent in developed societies and currently have limited options for diagnostic and therapeutic intervention. Here, using a proteomic and lipidomic-wide systems genetic approach, we interrogated lipid regulatory networks in 107 genetically distinct mouse strains to reveal key insights into the control and network structure of mammalian lipid metabolism. These include the identification of plasma lipid signatures that predict pathological lipid abundance in the liver of mice and humans, defining subcellular localization and functionality of lipid-related proteins, and revealing functional protein and genetic variants that are predicted to modulate lipid abundance. Trans-omic analyses using these datasets facilitated the identification and validation of PSMD9 as a previously unknown lipid regulatory protein. Collectively, our study serves as a rich resource for probing mammalian lipid metabolism and provides opportunities for the discovery of therapeutic agents and biomarkers in the setting of hepatic lipotoxicity.
Collapse
Affiliation(s)
- Benjamin L Parker
- Metabolic Systems Biology Laboratory, Charles Perkins Centre, School of Life and Environmental Sciences, University of Sydney, Sydney, New South Wales, Australia
| | - Anna C Calkin
- Lipid Metabolism & Cardiometabolic Disease Laboratory, Baker Heart & Diabetes Institute, Melbourne, Victoria, Australia.
- Central Clinical School, Department of Medicine, Monash University, Melbourne, Victoria, Australia.
| | - Marcus M Seldin
- Department of Human Genetics/Medicine, David Geffen School of Medicine, University of California Los Angeles (UCLA), Los Angeles, CA, USA
| | - Michael F Keating
- Lipid Metabolism & Cardiometabolic Disease Laboratory, Baker Heart & Diabetes Institute, Melbourne, Victoria, Australia
- Central Clinical School, Department of Medicine, Monash University, Melbourne, Victoria, Australia
- Molecular Metabolism & Ageing Laboratory, Baker Heart & Diabetes Institute, Melbourne, Victoria, Australia
| | - Elizabeth J Tarling
- Department of Medicine, Division of Cardiology, University of California Los Angeles (UCLA), Los Angeles, CA, USA
- Molecular Biology Institute, University of California Los Angeles (UCLA), Los Angeles, CA, USA
| | - Pengyi Yang
- Charles Perkins Centre, School of Mathematics and Statistics, University of Sydney, Sydney, New South Wales, Australia
| | - Sarah C Moody
- Lipid Metabolism & Cardiometabolic Disease Laboratory, Baker Heart & Diabetes Institute, Melbourne, Victoria, Australia
- Molecular Metabolism & Ageing Laboratory, Baker Heart & Diabetes Institute, Melbourne, Victoria, Australia
| | - Yingying Liu
- Lipid Metabolism & Cardiometabolic Disease Laboratory, Baker Heart & Diabetes Institute, Melbourne, Victoria, Australia
- Molecular Metabolism & Ageing Laboratory, Baker Heart & Diabetes Institute, Melbourne, Victoria, Australia
| | - Eser J Zerenturk
- Lipid Metabolism & Cardiometabolic Disease Laboratory, Baker Heart & Diabetes Institute, Melbourne, Victoria, Australia
- Molecular Metabolism & Ageing Laboratory, Baker Heart & Diabetes Institute, Melbourne, Victoria, Australia
| | - Elise J Needham
- Metabolic Systems Biology Laboratory, Charles Perkins Centre, School of Life and Environmental Sciences, University of Sydney, Sydney, New South Wales, Australia
| | - Matthew L Miller
- Molecular Biology Institute, University of California Los Angeles (UCLA), Los Angeles, CA, USA
| | - Bethan L Clifford
- Department of Medicine, Division of Cardiology, University of California Los Angeles (UCLA), Los Angeles, CA, USA
| | - Pauline Morand
- Department of Biological Chemistry, David Geffen School of Medicine, University of California Los Angeles (UCLA), Los Angeles, CA, USA
| | - Matthew J Watt
- Department of Physiology, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne, Victoria, Australia
| | - Ruth C R Meex
- Department of Physiology, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne, Victoria, Australia
| | - Kang-Yu Peng
- Metabolomics Laboratory, Baker Heart & Diabetes Institute, Melbourne, Victoria, Australia
| | | | - Kaushala Jayawardana
- Metabolomics Laboratory, Baker Heart & Diabetes Institute, Melbourne, Victoria, Australia
| | - Calvin Pan
- Department of Human Genetics/Medicine, David Geffen School of Medicine, University of California Los Angeles (UCLA), Los Angeles, CA, USA
| | - Natalie A Mellett
- Metabolomics Laboratory, Baker Heart & Diabetes Institute, Melbourne, Victoria, Australia
| | - Jacquelyn M Weir
- Metabolomics Laboratory, Baker Heart & Diabetes Institute, Melbourne, Victoria, Australia
| | - Ross Lazarus
- Metabolomics Laboratory, Baker Heart & Diabetes Institute, Melbourne, Victoria, Australia
| | - Aldons J Lusis
- Department of Human Genetics/Medicine, David Geffen School of Medicine, University of California Los Angeles (UCLA), Los Angeles, CA, USA
| | - Peter J Meikle
- Metabolomics Laboratory, Baker Heart & Diabetes Institute, Melbourne, Victoria, Australia
| | - David E James
- Metabolic Systems Biology Laboratory, Charles Perkins Centre, School of Life and Environmental Sciences, University of Sydney, Sydney, New South Wales, Australia
| | - Thomas Q de Aguiar Vallim
- Department of Medicine, Division of Cardiology, University of California Los Angeles (UCLA), Los Angeles, CA, USA.
- Molecular Biology Institute, University of California Los Angeles (UCLA), Los Angeles, CA, USA.
- Department of Biological Chemistry, David Geffen School of Medicine, University of California Los Angeles (UCLA), Los Angeles, CA, USA.
| | - Brian G Drew
- Central Clinical School, Department of Medicine, Monash University, Melbourne, Victoria, Australia.
- Molecular Metabolism & Ageing Laboratory, Baker Heart & Diabetes Institute, Melbourne, Victoria, Australia.
| |
Collapse
|
21
|
Liu L, Hu Q, Wu H, Wang X, Gao C, Chen G, Yao P, Gong Z. Dietary DHA/EPA Ratio Changes Fatty Acid Composition and Attenuates Diet-Induced Accumulation of Lipid in the Liver of ApoE -/- Mice. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:6256802. [PMID: 30538803 PMCID: PMC6261399 DOI: 10.1155/2018/6256802] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Accepted: 09/25/2018] [Indexed: 02/07/2023]
Abstract
Diets containing various docosahexaenoic acid (DHA)/eicosapentaenoic acid (EPA) ratios protect against liver damage in mice fed with a high-fat diet (HFD). However, it is unclear whether these beneficial roles of DHA and EPA are associated with alterations of fatty acid (FA) composition in the liver. This study evaluated the positive impacts of n-6/n-3 polyunsaturated fatty acids (PUFAs) containing different DHA/EPA ratios on HFD-induced liver disease and alterations of the hepatic FA composition. ApoE-/- mice were fed with HFDs with various ratios of DHA/EPA (2 : 1, 1 : 1, and 1 : 2) and an n-6/n-3 ratio of 4 : 1 for 12 weeks. After treatment, the serum and hepatic FA compositions, serum biochemical parameters, liver injury, and hepatic lipid metabolism-related gene expression were determined. Our results demonstrated that dietary DHA/EPA changed serum and hepatic FA composition by increasing contents of n-6 and n-3 PUFAs and decreasing amounts of monounsaturated fatty acids (MUFAs) and the n-6/n-3 ratio. Among the three DHA/EPA groups, the DHA/EPA 2 : 1 group tended to raise n-3 PUFAs concentration and lower the n-6/n-3 ratio in the liver, whereas DHA/EPA 1 : 2 tended to raise n-6 PUFAs concentration and improve the n-6/n-3 ratio. DHA/EPA supplementation reduced the hepatic impairment of lipid homeostasis, oxidative stress, and the inflammatory responses in HFD-fed mice. The DHA/EPA 2 : 1 group had lower serum levels of total cholesterol, triglycerides, and low-density lipoprotein cholesterol and higher levels of adiponectin than HFD group. The DHA/EPA 1 : 2 group had elevated serum levels of aspartate aminotransferase, alanine aminotransferase, and alkaline phosphatase, without significant change the expression of genes for inflammation or hepatic lipid metabolism among the three DHA/EPA groups. The results suggest that DHA/EPA-enriched diet with an n-6/n-3 ratio of 4 : 1 may reverse HFD-induced nonalcoholic fatty liver disease to some extent by increasing n-6 and n-3 PUFAs and decreasing the amount of MUFAs and the n-6/n-3 ratio.
Collapse
Affiliation(s)
- Liang Liu
- Key Laboratory for Deep Processing of Major Grain and Oil, Ministry of Education, Wuhan 430023, China
- College of Food Science and Engineering, Wuhan Polytechnic University, Wuhan 430023, China
- Hubei Key Laboratory for Processing and Transformation of Agricultural Products (Wuhan Polytechnic University), Wuhan 430023, China
| | - Qinling Hu
- Key Laboratory for Deep Processing of Major Grain and Oil, Ministry of Education, Wuhan 430023, China
- College of Food Science and Engineering, Wuhan Polytechnic University, Wuhan 430023, China
| | - Huihui Wu
- Key Laboratory for Deep Processing of Major Grain and Oil, Ministry of Education, Wuhan 430023, China
- College of Food Science and Engineering, Wuhan Polytechnic University, Wuhan 430023, China
| | - Xiujing Wang
- Key Laboratory for Deep Processing of Major Grain and Oil, Ministry of Education, Wuhan 430023, China
- College of Food Science and Engineering, Wuhan Polytechnic University, Wuhan 430023, China
| | - Chao Gao
- National Institute for Nutrition and Health, Chinese Center for Disease Control and Prevention, Beijing 100050, China
| | - Guoxun Chen
- Department of Nutrition, University of Tennessee at Knoxville, Knoxville 37996, USA
| | - Ping Yao
- Department of Nutrition and Food Hygiene, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Zhiyong Gong
- Key Laboratory for Deep Processing of Major Grain and Oil, Ministry of Education, Wuhan 430023, China
- College of Food Science and Engineering, Wuhan Polytechnic University, Wuhan 430023, China
- Hubei Key Laboratory for Processing and Transformation of Agricultural Products (Wuhan Polytechnic University), Wuhan 430023, China
| |
Collapse
|
22
|
Maldonado EM, Fisher CP, Mazzatti DJ, Barber AL, Tindall MJ, Plant NJ, Kierzek AM, Moore JB. Multi-scale, whole-system models of liver metabolic adaptation to fat and sugar in non-alcoholic fatty liver disease. NPJ Syst Biol Appl 2018; 4:33. [PMID: 30131870 PMCID: PMC6102210 DOI: 10.1038/s41540-018-0070-3] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Revised: 07/30/2018] [Accepted: 07/31/2018] [Indexed: 12/11/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a serious public health issue associated with high fat, high sugar diets. However, the molecular mechanisms mediating NAFLD pathogenesis are only partially understood. Here we adopt an iterative multi-scale, systems biology approach coupled to in vitro experimentation to investigate the roles of sugar and fat metabolism in NAFLD pathogenesis. The use of fructose as a sweetening agent is controversial; to explore this, we developed a predictive model of human monosaccharide transport, signalling and metabolism. The resulting quantitative model comprising a kinetic model describing monosaccharide transport and insulin signalling integrated with a hepatocyte-specific genome-scale metabolic network (GSMN). Differential kinetics for the utilisation of glucose and fructose were predicted, but the resultant triacylglycerol production was predicted to be similar for monosaccharides; these predictions were verified by in vitro data. The role of physiological adaptation to lipid overload was explored through the comprehensive reconstruction of the peroxisome proliferator activated receptor alpha (PPARα) regulome integrated with a hepatocyte-specific GSMN. The resulting qualitative model reproduced metabolic responses to increased fatty acid levels and mimicked lipid loading in vitro. The model predicted that activation of PPARα by lipids produces a biphasic response, which initially exacerbates steatosis. Our data support the evidence that it is the quantity of sugar rather than the type that is critical in driving the steatotic response. Furthermore, we predict PPARα-mediated adaptations to hepatic lipid overload, shedding light on potential challenges for the use of PPARα agonists to treat NAFLD.
Collapse
Affiliation(s)
- Elaina M. Maldonado
- School of Biosciences and Medicine, University of Surrey, Guildford, Surrey, GU2 7XH UK
| | - Ciarán P. Fisher
- Certara UK Limited, Simcyp Division, Level 2-Acero, 1 Concourse Way, Sheffield, S1 2BJ UK
| | | | - Amy L. Barber
- School of Biosciences and Medicine, University of Surrey, Guildford, Surrey, GU2 7XH UK
| | - Marcus J. Tindall
- Department of Mathematics and Statistics, University of Reading, Berkshire, RG6 6AX UK
- Institute of Cardiovascular and Metabolic Research, University of Reading, Berkshire, RG6 6UR UK
| | - Nicholas J. Plant
- School of Biosciences and Medicine, University of Surrey, Guildford, Surrey, GU2 7XH UK
- Faculty of Biological Sciences, University of Leeds, Leeds, West Yorkshire, LS2 9JT UK
| | - Andrzej M. Kierzek
- School of Biosciences and Medicine, University of Surrey, Guildford, Surrey, GU2 7XH UK
- Certara UK Limited, Simcyp Division, Level 2-Acero, 1 Concourse Way, Sheffield, S1 2BJ UK
| | - J. Bernadette Moore
- School of Biosciences and Medicine, University of Surrey, Guildford, Surrey, GU2 7XH UK
- School of Food Science & Nutrition, University of Leeds, Leeds, West Yorkshire, LS2 9JT UK
| |
Collapse
|
23
|
Spanos C, Maldonado EM, Fisher CP, Leenutaphong P, Oviedo-Orta E, Windridge D, Salguero FJ, Bermúdez-Fajardo A, Weeks ME, Evans C, Corfe BM, Rabbani N, Thornalley PJ, Miller MH, Wang H, Dillon JF, Quaglia A, Dhawan A, Fitzpatrick E, Moore JB. Correction to: Proteomic identification and characterization of hepatic glyoxalase 1 dysregulation in non-alcoholic fatty liver disease. Proteome Sci 2018; 16:13. [PMID: 29983642 PMCID: PMC6019799 DOI: 10.1186/s12953-018-0142-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Accepted: 06/04/2018] [Indexed: 11/29/2022] Open
Affiliation(s)
- Christos Spanos
- 1Department of Nutritional Sciences, Faculty of Health and Medical Sciences, University of Surrey, Guildford, Surrey, GU2 7XH UK
| | - Elaina M Maldonado
- 1Department of Nutritional Sciences, Faculty of Health and Medical Sciences, University of Surrey, Guildford, Surrey, GU2 7XH UK
| | - Ciarán P Fisher
- 1Department of Nutritional Sciences, Faculty of Health and Medical Sciences, University of Surrey, Guildford, Surrey, GU2 7XH UK
| | - Petchpailin Leenutaphong
- 1Department of Nutritional Sciences, Faculty of Health and Medical Sciences, University of Surrey, Guildford, Surrey, GU2 7XH UK
| | - Ernesto Oviedo-Orta
- 1Department of Nutritional Sciences, Faculty of Health and Medical Sciences, University of Surrey, Guildford, Surrey, GU2 7XH UK
| | - David Windridge
- 1Department of Nutritional Sciences, Faculty of Health and Medical Sciences, University of Surrey, Guildford, Surrey, GU2 7XH UK
| | - Francisco J Salguero
- 1Department of Nutritional Sciences, Faculty of Health and Medical Sciences, University of Surrey, Guildford, Surrey, GU2 7XH UK
| | - Alexandra Bermúdez-Fajardo
- 1Department of Nutritional Sciences, Faculty of Health and Medical Sciences, University of Surrey, Guildford, Surrey, GU2 7XH UK
| | - Mark E Weeks
- 2Institute of Child Health, University College London, WC1N 1EH, London, UK
| | - Caroline Evans
- 3Biological and Systems Engineering Group, ChELSI Institute, Department of Chemical and Biological Engineering, University of Sheffield, S1 3JD, Sheffield, UK
| | - Bernard M Corfe
- 4Molecular Gastroenterology Research Group, Department of Oncology and Insigneo Institute for in silico Medicine, University of Sheffield, S10 2RX, Sheffield, UK
| | - Naila Rabbani
- Clinical Sciences Research Laboratories, Warwick Medical School, University of Warwick, University Hospital, Coventry, CV2 2DX UK
| | - Paul J Thornalley
- Clinical Sciences Research Laboratories, Warwick Medical School, University of Warwick, University Hospital, Coventry, CV2 2DX UK
| | - Michael H Miller
- Medical Research Institute, University of Dundee, Ninewells Hospital and Medical School, Dundee, DD1 9SY UK
| | - Huan Wang
- Medical Research Institute, University of Dundee, Ninewells Hospital and Medical School, Dundee, DD1 9SY UK
| | - John F Dillon
- Medical Research Institute, University of Dundee, Ninewells Hospital and Medical School, Dundee, DD1 9SY UK
| | - Alberto Quaglia
- 7Paediatric Liver, GI and Nutrition Centre, King's College London School of Medicine, London, SE5 9RS UK
| | - Anil Dhawan
- 7Paediatric Liver, GI and Nutrition Centre, King's College London School of Medicine, London, SE5 9RS UK
| | - Emer Fitzpatrick
- 7Paediatric Liver, GI and Nutrition Centre, King's College London School of Medicine, London, SE5 9RS UK
| | - J Bernadette Moore
- 1Department of Nutritional Sciences, Faculty of Health and Medical Sciences, University of Surrey, Guildford, Surrey, GU2 7XH UK.,8School of Food Science and Nutrition, University of Leeds, Leeds, LS2 9JT UK
| |
Collapse
|