1
|
Müller M, Schubert T, Welke C, Maske T, Patschkowski T, Donhauser E, Heinen-Weiler J, Hormann FL, Heiles S, Schulz TJ, Lengenfelder LA, Landwehrjohann L, Vogt ET, Stratmann B, Hense J, Lüdtke S, Düfer M, Tolstik E, Dierks J, Lorenz K, Huxohl T, Reil JC, Sequeira V, Schopfer FJ, Freeman BA, Rudolph V, Schlomann U, Klinke A. Nitro-oleic acid enhances mitochondrial metabolism and ameliorates heart failure with preserved ejection fraction in mice. Nat Commun 2025; 16:3933. [PMID: 40287424 PMCID: PMC12033319 DOI: 10.1038/s41467-025-59192-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 04/14/2025] [Indexed: 04/29/2025] Open
Abstract
The prevalence of heart failure with preserved ejection fraction (HFpEF) is increasing, while treatment options are inadequate. Hypertension and obesity-related metabolic dysfunction contribute to HFpEF. Nitro-oleic acid (NO2-OA) impacts metabolic syndromes by improving glucose tolerance and adipocyte function. Here we show that treatment with NO2-OA ameliorates diastolic dysfunction and heart failure symptoms in a HFpEF mouse model induced by high-fat diet and inhibition of the endothelial nitric oxide synthase. Proteomic analysis of left ventricular tissue reveals that one-third of identified proteins, predominantly mitochondrial, are upregulated in hearts of NO2-OA-treated HFpEF mice compared to naïve and vehicle-treated HFpEF mice. Increased mitochondrial mass and numbers, and enhanced mitochondrial respiration are linked with this response, as assessed by transmission electron microscopy and high-resolution respirometry. Activation of the 5'-adenosine-monophosphate-activated-protein-kinase (AMPK) signaling pathway mediates the enhancement of mitochondrial dynamics in hearts of NO2-OA-treated HFpEF mice. These findings suggest that targeting mitochondrial function with NO2-OA may represent a promising therapeutic strategy for HFpEF.
Collapse
Affiliation(s)
- Marion Müller
- Clinic for General and Interventional Cardiology/ Angiology, Herz- und Diabeteszentrum NRW, Ruhr-Universität Bochum, Bad Oeynhausen, Germany
- Agnes Wittenborg Institute for Translational Cardiovascular Research (AWIHK), Herz- und Diabeteszentrum NRW, Ruhr-Universität Bochum, Bad Oeynhausen, Germany
| | - Torben Schubert
- Clinic for General and Interventional Cardiology/ Angiology, Herz- und Diabeteszentrum NRW, Ruhr-Universität Bochum, Bad Oeynhausen, Germany
- Agnes Wittenborg Institute for Translational Cardiovascular Research (AWIHK), Herz- und Diabeteszentrum NRW, Ruhr-Universität Bochum, Bad Oeynhausen, Germany
| | - Cornelius Welke
- Clinic for General and Interventional Cardiology/ Angiology, Herz- und Diabeteszentrum NRW, Ruhr-Universität Bochum, Bad Oeynhausen, Germany
- Agnes Wittenborg Institute for Translational Cardiovascular Research (AWIHK), Herz- und Diabeteszentrum NRW, Ruhr-Universität Bochum, Bad Oeynhausen, Germany
| | - Tibor Maske
- Clinic for General and Interventional Cardiology/ Angiology, Herz- und Diabeteszentrum NRW, Ruhr-Universität Bochum, Bad Oeynhausen, Germany
- Agnes Wittenborg Institute for Translational Cardiovascular Research (AWIHK), Herz- und Diabeteszentrum NRW, Ruhr-Universität Bochum, Bad Oeynhausen, Germany
| | - Thomas Patschkowski
- Technology Platform Genomics, Center for Biotechnology (CeBiTec), Bielefeld University, Bielefeld, Germany
| | - Elfi Donhauser
- Clinic for General and Interventional Cardiology/ Angiology, Herz- und Diabeteszentrum NRW, Ruhr-Universität Bochum, Bad Oeynhausen, Germany
- Agnes Wittenborg Institute for Translational Cardiovascular Research (AWIHK), Herz- und Diabeteszentrum NRW, Ruhr-Universität Bochum, Bad Oeynhausen, Germany
| | - Jacqueline Heinen-Weiler
- Medical Imaging Center (MIC), Electron Microscopy Medical Analysis - Core Facility (EMMACF), Med. Fakultät, Ruhr-Universität Bochum, Bochum, Germany
| | - Felix-Levin Hormann
- Leibniz-Institut für Analytische Wissenschaften-ISAS e.V., Dortmund, Germany
| | - Sven Heiles
- Leibniz-Institut für Analytische Wissenschaften-ISAS e.V., Dortmund, Germany
- Faculty of Chemistry, University of Duisburg-Essen, 45141, Essen, Germany
| | - Tina Johanna Schulz
- Clinic for General and Interventional Cardiology/ Angiology, Herz- und Diabeteszentrum NRW, Ruhr-Universität Bochum, Bad Oeynhausen, Germany
- Agnes Wittenborg Institute for Translational Cardiovascular Research (AWIHK), Herz- und Diabeteszentrum NRW, Ruhr-Universität Bochum, Bad Oeynhausen, Germany
| | - Luisa Andrea Lengenfelder
- Clinic for General and Interventional Cardiology/ Angiology, Herz- und Diabeteszentrum NRW, Ruhr-Universität Bochum, Bad Oeynhausen, Germany
- Agnes Wittenborg Institute for Translational Cardiovascular Research (AWIHK), Herz- und Diabeteszentrum NRW, Ruhr-Universität Bochum, Bad Oeynhausen, Germany
| | - Lucia Landwehrjohann
- Clinic for General and Interventional Cardiology/ Angiology, Herz- und Diabeteszentrum NRW, Ruhr-Universität Bochum, Bad Oeynhausen, Germany
- Agnes Wittenborg Institute for Translational Cardiovascular Research (AWIHK), Herz- und Diabeteszentrum NRW, Ruhr-Universität Bochum, Bad Oeynhausen, Germany
| | - Elisa Theres Vogt
- Diabetescenter, Herz- und Diabeteszentrum NRW, Ruhr-Universität Bochum, Bad Oeynhausen, Germany
| | - Bernd Stratmann
- Diabetescenter, Herz- und Diabeteszentrum NRW, Ruhr-Universität Bochum, Bad Oeynhausen, Germany
| | - Jurek Hense
- Institute of Pharmaceutical and Medicinal Chemistry, University of Münster, Münster, Germany
| | - Simon Lüdtke
- Institute of Pharmaceutical and Medicinal Chemistry, University of Münster, Münster, Germany
| | - Martina Düfer
- Institute of Pharmaceutical and Medicinal Chemistry, University of Münster, Münster, Germany
| | - Elena Tolstik
- Leibniz-Institut für Analytische Wissenschaften-ISAS e.V., Dortmund, Germany
| | - Johann Dierks
- Leibniz-Institut für Analytische Wissenschaften-ISAS e.V., Dortmund, Germany
| | - Kristina Lorenz
- Leibniz-Institut für Analytische Wissenschaften-ISAS e.V., Dortmund, Germany
- Institute of Pharmacology and Toxicology, University of Würzburg, Würzburg, Germany
| | - Tamino Huxohl
- Institute for Radiology, Nuclear Medicine and Molecular Imaging, Herz- und Diabeteszentrum NRW, Ruhr-Universität Bochum, Bad Oeynhausen, Germany
| | - Jan-Christian Reil
- Clinic for General and Interventional Cardiology/ Angiology, Herz- und Diabeteszentrum NRW, Ruhr-Universität Bochum, Bad Oeynhausen, Germany
| | - Vasco Sequeira
- Department of Translational Science Universitätsklinikum, DZHI, Würzburg, Germany
| | - Francisco Jose Schopfer
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Bruce A Freeman
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Volker Rudolph
- Clinic for General and Interventional Cardiology/ Angiology, Herz- und Diabeteszentrum NRW, Ruhr-Universität Bochum, Bad Oeynhausen, Germany
- Agnes Wittenborg Institute for Translational Cardiovascular Research (AWIHK), Herz- und Diabeteszentrum NRW, Ruhr-Universität Bochum, Bad Oeynhausen, Germany
| | - Uwe Schlomann
- Clinic for General and Interventional Cardiology/ Angiology, Herz- und Diabeteszentrum NRW, Ruhr-Universität Bochum, Bad Oeynhausen, Germany
- Agnes Wittenborg Institute for Translational Cardiovascular Research (AWIHK), Herz- und Diabeteszentrum NRW, Ruhr-Universität Bochum, Bad Oeynhausen, Germany
| | - Anna Klinke
- Clinic for General and Interventional Cardiology/ Angiology, Herz- und Diabeteszentrum NRW, Ruhr-Universität Bochum, Bad Oeynhausen, Germany.
- Agnes Wittenborg Institute for Translational Cardiovascular Research (AWIHK), Herz- und Diabeteszentrum NRW, Ruhr-Universität Bochum, Bad Oeynhausen, Germany.
| |
Collapse
|
2
|
Wessendorf-Rodriguez K, Ruchhoeft ML, Ashley EL, Galvez HM, Murray CW, Huang Y, McGregor GH, Kambhampati S, Shaw RJ, Metallo CM. Modeling compound lipid homeostasis using stable isotope tracing. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.16.618599. [PMID: 39463985 PMCID: PMC11507872 DOI: 10.1101/2024.10.16.618599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
Lipids represent the most diverse pool of metabolites found in cells, facilitating compartmentation, signaling, and other functions. Dysregulation of lipid metabolism is linked to disease states such as cancer and neurodegeneration. However, limited tools are available for quantifying metabolic fluxes across the lipidome. To directly measure reaction fluxes encompassing compound lipid homeostasis, we applied stable isotope tracing, liquid chromatography-high-resolution mass spectrometry, and network-based isotopologue modeling to non-small cell lung cancer (NSCLC) models. Compound lipid metabolic flux analysis (CL-MFA) enables the concurrent quantitation of fatty acid synthesis, elongation, headgroup assembly, and salvage reactions within virtually any biological system. Here, we resolve liver kinase B1 (LKB1)-mediated regulation of sphingolipid recycling in NSCLC cells and precision-cut lung slice cultures. We also demonstrate that widely used tissue culture conditions drive cells to upregulate fatty acid synthase flux to supraphysiological levels. Finally, we identify previously uncharacterized isozyme specificity of ceramide synthase inhibitors. These results highlight the ability of CL-MFA to quantify lipid cycling in biological systems to discover biological function and elucidate molecular mechanisms in membrane lipid metabolism.
Collapse
|
3
|
Jin Y, Liu Q, Wang Y, Wang B, An J, Chen Q, Wang T, Shang J. Propylthiouracil Induced Rat Model Reflects Heterogeneity Observed in Clinically Non-Obese Subjects with Nonalcoholic Fatty Liver Disease. Int J Mol Sci 2024; 25:10764. [PMID: 39409093 PMCID: PMC11477315 DOI: 10.3390/ijms251910764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 09/18/2024] [Accepted: 09/29/2024] [Indexed: 10/20/2024] Open
Abstract
The prevalence of nonalcoholic fatty liver disease (NAFLD) is increasing, affecting up to 30% of the population, with approximately 20% of cases occurring in non-obese individuals. The recent shift to the term metabolic dysfunction-associated steatosis liver disease (MASLD) highlights the disease's heterogeneity. However, there are no well-established animal models replicating non-obese NAFLD (NO-NAFLD). This study aimed to evaluate the relevance of the high-fat diet (HFD) combined with the propylthiouracil (PTU)-induced rat model in mimicking the histopathology and pathophysiology of NO-NAFLD. We first analyzed metabolic and clinical parameters between NO-NAFLD patients (Average BMI = 21.96 kg/m2) and obese NAFLD patients (Average BMI = 29.7 kg/m2). NO-NAFLD patients exhibited significantly higher levels of carnitines, phospholipids, and triglycerides. In the animal model, we examined serum lipid profiles, liver inflammation, histology, and transcriptomics. Hepatic steatosis in the HFD+PTU model at week 4 was comparable to that of the HFD model at week 8. The HFD+PTU model showed higher levels of carnitines, phospholipids, and triglycerides, supporting its relevance for NO-NAFLD. Additionally, the downregulation of lipid synthesis-related genes indicated differences in lipid accumulation between the two models. Overall, the HFD+PTU-induced rat model is a promising tool for studying the molecular mechanisms of NO-NAFLD.
Collapse
Affiliation(s)
- Yu Jin
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, China
- School of Pharmacy, University of Wisconsin Madison, Madison, WI 53705, USA
| | - Qiuyan Liu
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Yuqin Wang
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Bing Wang
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Jing An
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Qimeng Chen
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Tao Wang
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Jing Shang
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, China
- State Key Laboratory of Nat Mural Medicines, China Pharmaceutical University, Nanjing 210009, China
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, China Pharmaceutical University, Nanjing 210009, China
| |
Collapse
|
4
|
Słyk Ż, Stachowiak N, Małecki M. Recombinant Adeno-Associated Virus Vectors for Gene Therapy of the Central Nervous System: Delivery Routes and Clinical Aspects. Biomedicines 2024; 12:1523. [PMID: 39062095 PMCID: PMC11274884 DOI: 10.3390/biomedicines12071523] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 06/23/2024] [Accepted: 07/03/2024] [Indexed: 07/28/2024] Open
Abstract
The Central Nervous System (CNS) is vulnerable to a range of diseases, including neurodegenerative and oncological conditions, which present significant treatment challenges. The blood-brain barrier (BBB) restricts molecule penetration, complicating the achievement of therapeutic concentrations in the CNS following systemic administration. Gene therapy using recombinant adeno-associated virus (rAAV) vectors emerges as a promising strategy for treating CNS diseases, demonstrated by the registration of six gene therapy products in the past six years and 87 ongoing clinical trials. This review explores the implementation of rAAV vectors in CNS disease treatment, emphasizing AAV biology and vector engineering. Various administration methods-such as intravenous, intrathecal, and intraparenchymal routes-and experimental approaches like intranasal and intramuscular administration are evaluated, discussing their advantages and limitations in different CNS contexts. Additionally, the review underscores the importance of optimizing therapeutic efficacy through the pharmacokinetics (PK) and pharmacodynamics (PD) of rAAV vectors. A comprehensive analysis of clinical trials reveals successes and challenges, including barriers to commercialization. This review provides insights into therapeutic strategies using rAAV vectors in neurological diseases and identifies areas requiring further research, particularly in optimizing rAAV PK/PD.
Collapse
Affiliation(s)
- Żaneta Słyk
- Department of Applied Pharmacy, Faculty of Pharmacy, Medical University of Warsaw, 02-091 Warsaw, Poland
- Laboratory of Gene Therapy, Faculty of Pharmacy, Medical University of Warsaw, 02-091 Warsaw, Poland
| | - Natalia Stachowiak
- Department of Applied Pharmacy, Faculty of Pharmacy, Medical University of Warsaw, 02-091 Warsaw, Poland
| | - Maciej Małecki
- Department of Applied Pharmacy, Faculty of Pharmacy, Medical University of Warsaw, 02-091 Warsaw, Poland
- Laboratory of Gene Therapy, Faculty of Pharmacy, Medical University of Warsaw, 02-091 Warsaw, Poland
| |
Collapse
|
5
|
Alanazi IM, Alzahrani AR, Alsaad MA, Moqeem AL, Hamdi AM, Taher MM, Watson DG, Helen Grant M. The effect of mephedrone on human neuroblastoma and astrocytoma cells. Saudi Pharm J 2024; 32:102011. [PMID: 38454918 PMCID: PMC10918268 DOI: 10.1016/j.jsps.2024.102011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/09/2024] Open
Abstract
Mephedrone is an illegal drug that is used recreationally. Few studies have been conducted to investigate the mechanisms by which mephedrone is harming cells. In this research, we investigated the effect of mephedrone using toxicology coupled with LC-MS/MS based metabolomics in the two CNS derived cell lines. Methods of assessment such as neutral red (NR) assay, dimethylthiazolyl diphenyltetrazolium bromide (MTT), lactose dehydrogenase (LDH) measurement, and morphology were performed to identify the effect on cell viability and to identify the best concentration to be used in a metabolomics study. A concentration of 100 μM of mephedrone was used in the metabolomic experiment because at this concentration mephedrone had induced several intracellular changes. Although there no clear indicators of cellular damage caused by mephedrone. In astrocytes there was a clear indication that cell membrane function might be impaired by depletion of ether lipids.
Collapse
Affiliation(s)
- Ibrahim M. Alanazi
- Department of Pharmacology and Toxicology, College of Medicine, Umm Al-Qura University, Al-Abidiyah, P.O.Box 13578, Makkah 21955, Saudi Arabia
| | - Abdullah R. Alzahrani
- Department of Pharmacology and Toxicology, College of Medicine, Umm Al-Qura University, Al-Abidiyah, P.O.Box 13578, Makkah 21955, Saudi Arabia
| | - Mohammad A. Alsaad
- Department of Parasitology, College of Medicine, Umm AL Qura University, Al-Abidiyah, P.O.Box 13578, Makkah 21955, Saudi Arabia
| | - Abdulaziz L. Moqeem
- Home Health Care,Directorate of Health Affairs Jeddah, Ministry of Health, P.O.Box11176, Ryiadh, Saudi Arabia
| | - Abdulmohsen M. Hamdi
- Home Health Care,Directorate of Health Affairs Jeddah, Ministry of Health, P.O.Box11176, Ryiadh, Saudi Arabia
| | - Mohiuddin M. Taher
- Science and Technology Unit, Deanship of Scientific Research, and Department of Medical Genetics, College of Medicine, Umm-Al-Qura University, Makkah, Saudi Arabia
| | - David G. Watson
- Strathclyde Institute of Pharmacy & Biomedical Sciences, University of Strathclyde, Glasgow G4 0RE, UK
| | - M. Helen Grant
- Department of Biomedical Engineering, University of Strathclyde, Glasgow G4 0NW, UK
| |
Collapse
|
6
|
de Deus IJ, Martins-Silva AF, Fagundes MMDA, Paula-Gomes S, Silva FGDE, da Cruz LL, de Abreu ARR, de Queiroz KB. Role of NLRP3 inflammasome and oxidative stress in hepatic insulin resistance and the ameliorative effect of phytochemical intervention. Front Pharmacol 2023; 14:1188829. [PMID: 37456758 PMCID: PMC10347376 DOI: 10.3389/fphar.2023.1188829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 06/19/2023] [Indexed: 07/18/2023] Open
Abstract
NLRP3 inflammasome has a key role in chronic low-grade metabolic inflammation, and its excessive activation may contribute to the beginning and progression of several diseases, including hepatic insulin resistance (hIR). Thus, this review aims to highlight the role of NLRP3 inflammasome and oxidative stress in the development of hIR and evidence related to phytochemical intervention in this context. In this review, we will address the hIR pathogenesis related to reactive oxygen species (ROS) production mechanisms, involving oxidized mitochondrial DNA (ox-mtDNA) and thioredoxin interacting protein (TXNIP) induction in the NLRP3 inflammasome activation. Moreover, we discuss the inhibitory effect of bioactive compounds on the insulin signaling pathway, and the role of microRNAs (miRNAs) in the phytochemical target mechanism in ameliorating hIR. Although most of the research in the field has been focused on evaluating the inhibitory effect of phytochemicals on the NLRP3 inflammasome pathway, further investigation and clinical studies are required to provide insights into the mechanisms of action, and, thus, encourage the use of these bioactive compounds as an additional therapeutic strategy to improve hIR and correlated conditions.
Collapse
Affiliation(s)
- Isabela Jesus de Deus
- Laboratório de Nutrição Experimental, Programa de Pós-Graduação em Saúde e Nutrição, Escola de Nutrição, Universidade Federal de Ouro Preto, Ouro Preto, Brazil
| | - Ana Flávia Martins-Silva
- Laboratório de Nutrição Experimental, Programa de Pós-Graduação em Saúde e Nutrição, Escola de Nutrição, Universidade Federal de Ouro Preto, Ouro Preto, Brazil
| | - Miliane Martins de Andrade Fagundes
- Laboratório de Nutrição Experimental, Programa de Pós-Graduação em Saúde e Nutrição, Escola de Nutrição, Universidade Federal de Ouro Preto, Ouro Preto, Brazil
- Departamento de Alimentos, Programa de Pós-Graduação em Saúde e Nutrição, Escola de Nutrição, Universidade Federal de Ouro Preto, Ouro Preto, Brazil
| | - Sílvia Paula-Gomes
- Laboratório de Bioquímica e Biologia Molecular, Programa de Pós-graduação em Ciências Biológicas, Universidade Federal de Ouro Preto, Ouro Preto, Brazil
| | - Fernanda Guimarães Drummond e Silva
- Departamento de Alimentos, Programa de Pós-Graduação em Saúde e Nutrição, Escola de Nutrição, Universidade Federal de Ouro Preto, Ouro Preto, Brazil
| | | | - Aline Rezende Ribeiro de Abreu
- Laboratório de Nutrição Experimental, Programa de Pós-Graduação em Saúde e Nutrição, Escola de Nutrição, Universidade Federal de Ouro Preto, Ouro Preto, Brazil
| | - Karina Barbosa de Queiroz
- Laboratório de Nutrição Experimental, Programa de Pós-Graduação em Saúde e Nutrição, Escola de Nutrição, Universidade Federal de Ouro Preto, Ouro Preto, Brazil
- Departamento de Alimentos, Programa de Pós-Graduação em Saúde e Nutrição, Escola de Nutrição, Universidade Federal de Ouro Preto, Ouro Preto, Brazil
| |
Collapse
|
7
|
Austin GO, Tomas A. Variation in responses to incretin therapy: Modifiable and non-modifiable factors. Front Mol Biosci 2023; 10:1170181. [PMID: 37091864 PMCID: PMC10119428 DOI: 10.3389/fmolb.2023.1170181] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 03/28/2023] [Indexed: 04/25/2023] Open
Abstract
Type 2 diabetes (T2D) and obesity have reached epidemic proportions. Incretin therapy is the second line of treatment for T2D, improving both blood glucose regulation and weight loss. Glucagon-like peptide-1 (GLP-1) and glucose-stimulated insulinotropic polypeptide (GIP) are the incretin hormones that provide the foundations for these drugs. While these therapies have been highly effective for some, the results are variable. Incretin therapies target the class B G protein-coupled receptors GLP-1R and GIPR, expressed mainly in the pancreas and the hypothalamus, while some therapeutical approaches include additional targeting of the related glucagon receptor (GCGR) in the liver. The proper functioning of these receptors is crucial for incretin therapy success and here we review several mechanisms at the cellular and molecular level that influence an individual's response to incretin therapy.
Collapse
Affiliation(s)
| | - Alejandra Tomas
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| |
Collapse
|
8
|
Yang F, Chen G. The nutritional functions of dietary sphingomyelin and its applications in food. Front Nutr 2022; 9:1002574. [PMID: 36337644 PMCID: PMC9626766 DOI: 10.3389/fnut.2022.1002574] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 09/26/2022] [Indexed: 11/13/2022] Open
Abstract
Sphingolipids are common structural components of cell membranes and are crucial for cell functions in physiological and pathophysiological conditions. Sphingomyelin and its metabolites, such as sphingoid bases, ceramide, ceramide-1-phosphate, and sphingosine-1-phosphate, play signaling roles in the regulation of human health. The diverse structures of sphingolipids elicit various functions in cellular membranes and signal transduction, which may affect cell growth, differentiation, apoptosis, and maintain biological activities. As nutrients, dietary sphingomyelin and its metabolites have wide applications in the food and pharmaceutical industry. In this review, we summarized the distribution, classifications, structures, digestion, absorption and metabolic pathways of sphingolipids, and discussed the nutritional functioning of sphingomyelin in chronic metabolic diseases. The possible implications of dietary sphingomyelin in the modern food preparations including dairy products and infant formula, skin improvement, delivery system and oil organogels are also evaluated. The production of endogenous sphingomyelin is linked to pathological changes in obesity, diabetes, and atherosclerosis. However, dietary supplementations of sphingomyelin and its metabolites have been shown to maintain cholesterol homeostasis and lipid metabolism, and to prevent or treat these diseases. This seemly paradoxical phenomenon shows that dietary sphingomyelin and its metabolites are candidates for food additives and functional food development for the prevention and treatment of chronic metabolic diseases in humans.
Collapse
Affiliation(s)
- Fang Yang
- School of Laboratory Medicine, Hubei University of Chinese Medicine, Wuhan, China
| | - Guoxun Chen
- Department of Nutrition, The University of Tennessee, Knoxville, TN, United States
| |
Collapse
|
9
|
Obesity Hinders the Protective Effect of Selenite Supplementation on Insulin Signaling. Antioxidants (Basel) 2022; 11:antiox11050862. [PMID: 35624726 PMCID: PMC9138114 DOI: 10.3390/antiox11050862] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 04/22/2022] [Accepted: 04/26/2022] [Indexed: 12/11/2022] Open
Abstract
The intake of high-fat diets (HFDs) containing large amounts of saturated long-chain fatty acids leads to obesity, oxidative stress, inflammation, and insulin resistance. The trace element selenium, as a crucial part of antioxidative selenoproteins, can protect against the development of diet-induced insulin resistance in white adipose tissue (WAT) by increasing glutathione peroxidase 3 (GPx3) and insulin receptor (IR) expression. Whether selenite (Se) can attenuate insulin resistance in established lipotoxic and obese conditions is unclear. We confirm that GPX3 mRNA expression in adipose tissue correlates with BMI in humans. Cultivating 3T3-L1 pre-adipocytes in palmitate-containing medium followed by Se treatment attenuates insulin resistance with enhanced GPx3 and IR expression and adipocyte differentiation. However, feeding obese mice a selenium-enriched high-fat diet (SRHFD) only resulted in a modest increase in overall selenoprotein gene expression in WAT in mice with unaltered body weight development, glucose tolerance, and insulin resistance. While Se supplementation improved adipocyte morphology, it did not alter WAT insulin sensitivity. However, mice fed a SRHFD exhibited increased insulin content in the pancreas. Overall, while selenite protects against palmitate-induced insulin resistance in vitro, obesity impedes the effect of selenite on insulin action and adipose tissue metabolism in vivo.
Collapse
|
10
|
Komninos D, Ramos L, van der Heijden GW, Morrison MC, Kleemann R, van Herwaarden AE, Kiliaan AJ, Arnoldussen IAC. High fat diet-induced obesity prolongs critical stages of the spermatogenic cycle in a Ldlr -/-.Leiden mouse model. Sci Rep 2022; 12:430. [PMID: 35017550 PMCID: PMC8752771 DOI: 10.1038/s41598-021-04069-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 12/10/2021] [Indexed: 02/07/2023] Open
Abstract
Obesity can disturb spermatogenesis and subsequently affect male fertility and reproduction. In our study, we aim to elucidate at which cellular level of adult spermatogenesis the detrimental effects of obesity manifest. We induced high fat diet (HFD) obesity in low-density lipoprotein receptor knock-out Leiden (Ldlr−/−.Leiden) mice, and studied the morphological structure of the testes and histologically examined the proportion of Sertoli cells, spermatocytes and spermatids in the seminiferous tubules. We examined sperm DNA damage and chromatin condensation and measured plasma levels of leptin, testosterone, cholesterol and triglycerides. HFD-induced obesity caused high plasma leptin and abnormal testosterone levels and induced an aberrant intra-tubular organisation (ITO) which is associated with an altered spermatids/spermatocytes ratio (2:1 instead of 3:1). Mice fed a HFD had a higher level of tubules in stages VII + VIII in the spermatogenic cycle. The stages VII + VII indicate crucial processes in spermatogenic development like initiation of meiosis, initiation of spermatid elongation, and release of fully matured spermatids. In conclusion, HFD-induced obese Ldlr−/−.Leiden mice develop an aberrant ITO and alterations in the spermatogenic cycle in crucial stages (stages VII and VII). Thereby, our findings stress the importance of lifestyle guidelines in infertility treatments.
Collapse
Affiliation(s)
- D Komninos
- Department of Obstetrics and Gynaecology, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA, Nijmegen, The Netherlands
| | - L Ramos
- Department of Obstetrics and Gynaecology, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA, Nijmegen, The Netherlands
| | - G W van der Heijden
- Department of Obstetrics and Gynaecology, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA, Nijmegen, The Netherlands
| | - M C Morrison
- Department of Metabolic Health Research, Netherlands Organisation for Applied Scientific Research (TNO), Zernikedreef 9, 2333 CK, Leiden, The Netherlands.,Department of Human and Animal Physiology, Wageningen University, De Elst 1, 6708 WD, Wageningen, The Netherlands
| | - R Kleemann
- Department of Metabolic Health Research, Netherlands Organisation for Applied Scientific Research (TNO), Zernikedreef 9, 2333 CK, Leiden, The Netherlands
| | - A E van Herwaarden
- Department of Laboratory Medicine, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA, Nijmegen, The Netherlands
| | - A J Kiliaan
- Department of Medical Imaging, Anatomy, Donders Institute for Brain, Cognition and Behaviour, Preclinical Imaging Center PRIME, Radboud University Medical Center, Geert Grooteplein Noord 21, 6525 EZ, Nijmegen, The Netherlands.
| | - I A C Arnoldussen
- Department of Medical Imaging, Anatomy, Donders Institute for Brain, Cognition and Behaviour, Preclinical Imaging Center PRIME, Radboud University Medical Center, Geert Grooteplein Noord 21, 6525 EZ, Nijmegen, The Netherlands
| |
Collapse
|
11
|
Wang S, Wei Y, Hu C, Liu F. Proteomic analysis reveals proteins and pathways associated with declined testosterone production in male obese mice after chronic high-altitude exposure. Front Endocrinol (Lausanne) 2022; 13:1046901. [PMID: 36531490 PMCID: PMC9748565 DOI: 10.3389/fendo.2022.1046901] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Accepted: 11/10/2022] [Indexed: 12/05/2022] Open
Abstract
OBJECTIVE Obesity is common in highland areas owing to lifestyle alterations. There are pieces of evidence to suggest that both obesity and hypoxia may promote oxidative stress, leading to hypogonadism in males. These findings indicate an increased risk of hypogonadism in obese males following hypoxia exposure. However, the mechanisms underlying the disease process remain unclear. The current study aims to explore the mechanism of testosterone production dysfunction in obese male mice exposed to a chronic high-altitude hypoxia environment. METHODS An obese male mouse model was generated by inducing obesity in mice via a high-fat diet for 14 weeks, and the obese mice were then exposed to a high-altitude hypoxia environment for 24 days. Sera and testicular tissues were collected to detect serum lipids, sex hormone level, and testicular oxidative stress indicators. Morphological examination was performed to assess pathological alterations in testicular tissues and suborganelles in leydig cells. Proteomic alterations in testicular tissues were investigated using quantitative proteomics in Obese/Control and Obese-Hypoxia/Obese groups. RESULTS The results showed that chronic high-altitude hypoxia exposure aggravated low testosterone production in obese male mice accompanied by increased testicular oxidative stress and histological damages. In total, 363 and 242 differentially expressed proteins (DEPs) were identified in the two comparison groups, Obese/Control and Obese-Hypoxia/Obese, respectively. Functional enrichment analysis demonstrated that several significant functional terms and pathways related to testosterone production were altered in the two comparison groups. These included cholesterol metabolism, steroid hormone biosynthesis, peroxisome proliferator-activated receptor (PPAR) signaling pathway, oxidative stress responses, as well as retinol metabolism. Finally, 10 representative DEPs were selected for parallel reaction monitoring verification. Among them, StAR, DHCR7, NSDHL, CYP51A1, FDPS, FDX1, CYP11A1, ALDH1A1, and GPX3 were confirmed to be downregulated in the two groups. CONCLUSIONS Chronic hypoxia exposure could exacerbate low testosterone production in obese male mice by influencing the expression of key proteins involved in steroid hormone biosynthesis, cholesterol biosynthesis, oxidative stress responses and retinol metabolism.
Collapse
Affiliation(s)
- Shuqiong Wang
- Research Center for High Altitude Medicine, Qinghai University, Xining, China
- Key Laboratory of High Altitude Medicine, Ministry of Education, Xining, China
- Key Laboratory of Application and Foundation for High Altitude Medicine Research in Qinghai Province, Qinghai-Utah Joint Research Key Lab for High Altitude Medicine, Xining, China
- Department of Endocrinology, Qinghai Provincial People’s Hospital, Xining, China
| | - Youwen Wei
- Department of Plague Prevention and Control, Qinghai Institute for Endemic Disease Prevention and Control, Xining, China
| | - Caiyan Hu
- Department of Laboratory Medicine, Baoding First Central Hospital, Baoding, China
| | - Fang Liu
- Department of Biochemistry, Medical College, Qinghai University, Xining, China
- *Correspondence: Fang Liu,
| |
Collapse
|
12
|
Sphingolipids in foodstuff: Compositions, distribution, digestion, metabolism and health effects - A comprehensive review. Food Res Int 2021; 147:110566. [PMID: 34399542 DOI: 10.1016/j.foodres.2021.110566] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 06/25/2021] [Accepted: 06/27/2021] [Indexed: 12/26/2022]
Abstract
Sphingolipids (SLs) are common in all eukaryotes, prokaryotes, and viruses, and played a vital role in human health. They are involved in physiological processes, including intracellular transport, cell division, and signal transduction. However, there are limited reviews on dietary effects on endogenous SLs metabolism and further on human health. Various dietary conditions, including the SLs-enriched diet, high-fat diet, and vitamins, can change the level of endogenous SLs metabolites and even affect human health. This review systematically summarizes the main known SLs in foods concerning their variety and contents, as well as their isolation and identification approaches. Moreover, the present review discusses the role of dietary (particularly SLs-enriched diet, high-fat diet, and vitamins) in endogenous SLs metabolism, highlighting how exogenous SLs are digested and absorbed. The role of SLs family in the pathogenesis of diseases, including cancers, neurological disorders, infectious and inflammatory diseases, and cardiovascular diseases, and in recently coronavirus disease-19 outbreak was also discussed. In the post-epidemic era, we believe that the concern for health and the need for plant-based products will increase. Therefore, a need for research on the absorption and metabolism pathway of SLs (especially plant-derived SLs) and their bioavailability is necessary. Moreover, the effects of storage treatment and processing on the content and composition of SLs in food are worth exploring. Further studies should also be conducted on the dose-response of SLs on human health to support the development of SLs supplements. More importantly, new approaches, such as, making SLs based hydrogels can effectively achieve sustained release and targeted therapies.
Collapse
|
13
|
Changes in Key Mitochondrial Lipids Accompany Mitochondrial Dysfunction and Oxidative Stress in NAFLD. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:9986299. [PMID: 34257827 PMCID: PMC8257344 DOI: 10.1155/2021/9986299] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Accepted: 05/06/2021] [Indexed: 12/30/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a dysmetabolic hepatic damage of increasing severity: simple fat accumulation (steatosis), nonalcoholic steatohepatitis (NASH), and hepatic fibrosis. Oxidative stress is considered an important factor in producing hepatocyte injury associated with NAFLD progression. Studies also suggest a link between the accumulation of specific hepatic lipid species, mitochondrial dysfunction, and the progression of NAFLD. However, it is unclear whether mitochondrial lipid modifications are involved in NAFLD progression. To gain insight into the relationship between mitochondrial lipids and disease progression through different stages of NAFLD, we performed lipidomic analyses on mouse livers at different stages of western diet-induced NAFLD, with or without hepatic fibrosis. After organelle separation, we studied separately the mitochondrial and the “nonmitochondrial” hepatic lipidomes. We identified 719 lipid species from 16 lipid families. Remarkably, the western diet triggered time-dependent changes in the mitochondrial lipidome, whereas the “nonmitochondrial” lipidome showed little difference with levels of hepatic steatosis or the presence of fibrosis. In mitochondria, the changes in the lipidome preceded hepatic fibrosis. In particular, two critical phospholipids, phosphatidic acid (PA) and cardiolipin (CL), displayed opposite responses in mitochondria. Decrease in CL and increase in PA were concurrent with an increase of coenzyme Q. Electron paramagnetic resonance spectroscopy superoxide spin trapping and Cu2+ measurement showed the progressive increase in oxidative stress in the liver. Overall, these results suggest mitochondrial lipid modifications could act as an early event in mitochondrial dysfunction and NAFLD progression.
Collapse
|
14
|
Rodrigues RR, Gurung M, Li Z, García-Jaramillo M, Greer R, Gaulke C, Bauchinger F, You H, Pederson JW, Vasquez-Perez S, White KD, Frink B, Philmus B, Jump DB, Trinchieri G, Berry D, Sharpton TJ, Dzutsev A, Morgun A, Shulzhenko N. Transkingdom interactions between Lactobacilli and hepatic mitochondria attenuate western diet-induced diabetes. Nat Commun 2021; 12:101. [PMID: 33397942 PMCID: PMC7782853 DOI: 10.1038/s41467-020-20313-x] [Citation(s) in RCA: 91] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Accepted: 11/09/2020] [Indexed: 02/06/2023] Open
Abstract
Western diet (WD) is one of the major culprits of metabolic disease including type 2 diabetes (T2D) with gut microbiota playing an important role in modulating effects of the diet. Herein, we use a data-driven approach (Transkingdom Network analysis) to model host-microbiome interactions under WD to infer which members of microbiota contribute to the altered host metabolism. Interrogation of this network pointed to taxa with potential beneficial or harmful effects on host's metabolism. We then validate the functional role of the predicted bacteria in regulating metabolism and show that they act via different host pathways. Our gene expression and electron microscopy studies show that two species from Lactobacillus genus act upon mitochondria in the liver leading to the improvement of lipid metabolism. Metabolomics analyses revealed that reduced glutathione may mediate these effects. Our study identifies potential probiotic strains for T2D and provides important insights into mechanisms of their action.
Collapse
Affiliation(s)
| | - Manoj Gurung
- Veterinary Medicine, Oregon State University, Corvallis, OR, USA
| | - Zhipeng Li
- Veterinary Medicine, Oregon State University, Corvallis, OR, USA
| | | | - Renee Greer
- Veterinary Medicine, Oregon State University, Corvallis, OR, USA
| | | | - Franziska Bauchinger
- Department of Microbiology and Ecosystem Science, University of Vienna, Vienna, Austria
| | - Hyekyoung You
- Veterinary Medicine, Oregon State University, Corvallis, OR, USA
| | - Jacob W Pederson
- Veterinary Medicine, Oregon State University, Corvallis, OR, USA
| | | | - Kimberly D White
- Veterinary Medicine, Oregon State University, Corvallis, OR, USA
| | - Briana Frink
- Veterinary Medicine, Oregon State University, Corvallis, OR, USA
| | - Benjamin Philmus
- College of Pharmacy, Oregon State University, Corvallis, OR, USA
| | - Donald B Jump
- College of Public Health and Human Sciences, Oregon State University, Corvallis, OR, USA
| | - Giorgio Trinchieri
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - David Berry
- Department of Microbiology and Ecosystem Science, University of Vienna, Vienna, Austria
| | | | - Amiran Dzutsev
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Andrey Morgun
- College of Pharmacy, Oregon State University, Corvallis, OR, USA.
| | | |
Collapse
|
15
|
Kurhaluk N, Bojková B, Kajo K, Macháleková K, Kisková T. Addition of palm olein to lard-supplemented diet indicates myocardial dysfunction and augments oxidative stress by authophagy-lysosome pathway in rats. J Anim Physiol Anim Nutr (Berl) 2020; 105:587-598. [PMID: 33314355 DOI: 10.1111/jpn.13477] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 05/13/2020] [Accepted: 06/13/2020] [Indexed: 12/18/2022]
Abstract
This study evaluated a prolonged effect of palm oil addition to lard-supplemented diet (PLD) on the oxidative status, lysosomal enzyme activities, markers of hepatotoxicity and basic lipid profile in female rats. Female Sprague-Dawley rats received PLD (10% of total fat: 7.5% from palm oil and 2.5% from lard), and the control group received lard-supplemented diet (2.5% fat) from 28 days of age for 14 weeks. Histopathological evaluation of the liver from animals fed the PLD showed slight steatosis and signs of mild chronic inflammation. Reduction of extramedullary hematopoiesis and an increased ratio of red/white pulp were observed in the spleen. PLD induced oxidative stress (evaluated in the liver, heart, spleen, muscle and kidney) evidenced by an increase in conjugated dienes and malondialdehyde in all tissues except the muscle; protein carbonyl derivatives were increased as well. The changes in the antioxidant enzyme activities in the evaluated tissues were ambiguous except for the prominent increase in the heart. Lysosomal enzyme activities showed a tendency to increase in the heart and kidney and to decrease in the muscle and spleen. The De Ritis ratio, which is a biomarker of hepatotoxicity, was higher in the heart from animals fed the PLD. The palm oil addition to the lard-supplemented diet-induced prominent oxidative stress, particularly in myocardial tissue with involvement of the authophagy-lysosome pathway.
Collapse
Affiliation(s)
- Natalia Kurhaluk
- Department of Physiology, Institute of Biology and Earth Science, Pomeranian University in Slupsk, Słupsk, Poland
| | - Bianka Bojková
- Department of Animal Physiology, Institute of Biology and Ecology, Faculty of Science, P.J. Šafárik University in Košice, Košice, Slovakia
| | - Karol Kajo
- Department of Pathology, St. Elizabeth Cancer Institute Hospital, Bratislava, Slovakia.,Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Katarína Macháleková
- Department of Pathology, St. Elizabeth Cancer Institute Hospital, Bratislava, Slovakia
| | - Terézia Kisková
- Department of Animal Physiology, Institute of Biology and Ecology, Faculty of Science, P.J. Šafárik University in Košice, Košice, Slovakia
| |
Collapse
|
16
|
Weber M, Mera P, Casas J, Salvador J, Rodríguez A, Alonso S, Sebastián D, Soler-Vázquez MC, Montironi C, Recalde S, Fucho R, Calderón-Domínguez M, Mir JF, Bartrons R, Escola-Gil JC, Sánchez-Infantes D, Zorzano A, Llorente-Cortes V, Casals N, Valentí V, Frühbeck G, Herrero L, Serra D. Liver CPT1A gene therapy reduces diet-induced hepatic steatosis in mice and highlights potential lipid biomarkers for human NAFLD. FASEB J 2020; 34:11816-11837. [PMID: 32666604 DOI: 10.1096/fj.202000678r] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Revised: 06/12/2020] [Accepted: 06/19/2020] [Indexed: 12/25/2022]
Abstract
The prevalence of nonalcoholic fatty liver disease (NAFLD) has increased drastically due to the global obesity pandemic but at present there are no approved therapies. Here, we aimed to revert high-fat diet (HFD)-induced obesity and NAFLD in mice by enhancing liver fatty acid oxidation (FAO). Moreover, we searched for potential new lipid biomarkers for monitoring liver steatosis in humans. We used adeno-associated virus (AAV) to deliver a permanently active mutant form of human carnitine palmitoyltransferase 1A (hCPT1AM), the key enzyme in FAO, in the liver of a mouse model of HFD-induced obesity and NAFLD. Expression of hCPT1AM enhanced hepatic FAO and autophagy, reduced liver steatosis, and improved glucose homeostasis. Lipidomic analysis in mice and humans before and after therapeutic interventions, such as hepatic AAV9-hCPT1AM administration and RYGB surgery, respectively, led to the identification of specific triacylglyceride (TAG) specie (C50:1) as a potential biomarker to monitor NAFFLD disease. To sum up, here we show for the first time that liver hCPT1AM gene therapy in a mouse model of established obesity, diabetes, and NAFLD can reduce HFD-induced derangements. Moreover, our study highlights TAG (C50:1) as a potential noninvasive biomarker that might be useful to monitor NAFLD in mice and humans.
Collapse
Affiliation(s)
- Minéia Weber
- Department of Biochemistry and Physiology, School of Pharmacy and Food Sciences, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, Barcelona, Spain
| | - Paula Mera
- Department of Biochemistry and Physiology, School of Pharmacy and Food Sciences, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Josefina Casas
- Research Unit on BioActive Molecules, Department of Biological Chemistry, Institute of Advanced Chemistry of Catalonia (IQAC)/CSIC, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Instituto de Salud Carlos III, Madrid, Spain
| | - Javier Salvador
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain.,Department of Endocrinology & Nutrition, Clínica Universidad de Navarra, Pamplona, Spain
| | - Amaia Rodríguez
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain.,Metabolic Research Laboratory, Clínica Universidad de Navarra, IdiSNA, Pamplona, Spain
| | - Sergio Alonso
- Cancer Genetics and Epigenetics Group, Program of Predictive and Personalized Medicine of Cancer, Germans Trias i Pujol Research Institute (IGTP-PMPPC), Campus Can Ruti, Barcelona, Spain
| | - David Sebastián
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology, Departament de Bioquímica i Biomedicina Molecular, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
| | - M Carmen Soler-Vázquez
- Department of Biochemistry and Physiology, School of Pharmacy and Food Sciences, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, Barcelona, Spain
| | - Carla Montironi
- Pathology Department, Hospital Clinic de Barcelona, Barcelona, Spain.,Liver Cancer Translational Research Laboratory, Liver Unit, IDIBAPS-Hospital Clínic, Universitat de Barcelona, Barcelona, Spain
| | - Sandra Recalde
- Department of Biochemistry and Physiology, School of Pharmacy and Food Sciences, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, Barcelona, Spain
| | - Raquel Fucho
- Department of Biochemistry and Physiology, School of Pharmacy and Food Sciences, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, Barcelona, Spain
| | - María Calderón-Domínguez
- Department of Biochemistry and Physiology, School of Pharmacy and Food Sciences, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, Barcelona, Spain
| | - Joan Francesc Mir
- Department of Biochemistry and Physiology, School of Pharmacy and Food Sciences, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, Barcelona, Spain
| | - Ramon Bartrons
- Departament de Ciències Fisiològiques, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Spain
| | - Joan Carles Escola-Gil
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain.,IIB Sant Pau, Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - David Sánchez-Infantes
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain.,Germans Trias i Pujol Research Institute (IGTP-PMPPC), Campus Can Ruti, Barcelona, Spain
| | - Antonio Zorzano
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology, Departament de Bioquímica i Biomedicina Molecular, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
| | - Vicenta Llorente-Cortes
- Institute of Biomedical Research of Barcelona (IIBB), Spanish National Research Council (CSIC), Biomedical Research Institute Sant Pau (IIB Sant Pau), Barcelona, Spain.,CIBERCV, Institute of Health Carlos III, Madrid, Spain.,Cardiovascular Research Center, CSIC-ICCC, Barcelona, Spain
| | - Núria Casals
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain.,Basic Sciences Department, Faculty of Medicine and Health Sciences, Universitat Internacional de Catalunya, Sant Cugat del Vallès, Spain
| | - Víctor Valentí
- Metabolic Research Laboratory, Clínica Universidad de Navarra, IdiSNA, Pamplona, Spain.,Department of Surgery, Clínica Universidad de Navarra, Pamplona, Spain
| | - Gema Frühbeck
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain.,Metabolic Research Laboratory, Clínica Universidad de Navarra, IdiSNA, Pamplona, Spain
| | - Laura Herrero
- Department of Biochemistry and Physiology, School of Pharmacy and Food Sciences, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Dolors Serra
- Department of Biochemistry and Physiology, School of Pharmacy and Food Sciences, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
17
|
Seifi K, Rezaei M, Yansari AT, Zamiri MJ, Riazi GH, Heidari R. Short chain fatty acids may improve hepatic mitochondrial energy efficiency in heat stressed-broilers. J Therm Biol 2020; 89:102520. [PMID: 32364974 DOI: 10.1016/j.jtherbio.2020.102520] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2019] [Revised: 12/16/2019] [Accepted: 01/10/2020] [Indexed: 01/10/2023]
Abstract
The present study was conducted to investigate the effects of four dietary fat types and two environmental temperatures on the hepatic mitochondrial energetic in male broilers exposed to heat stress. The birds were kept in two separate rooms at 24 °C or 36 °C from 32 to 42 d of age with four experimental groups in each room. The birds fed on the diets supplemented containing rich sources of long-chain saturated fatty acids (beef tallow), middle-length-chain saturated FA (coconut oil), monounsaturated FA (olive oil), or polyunsaturated FA (soybean oil) for ten days. At 36 °C, the highest body weight and lowest feed conversion ratio were recorded in the birds fed on the diets supplemented with coconut oil or beef tallow. Temperature and fat type significantly affected the activities of the mitochondrial electron transport chain complexes (P < 0.01). There was a significant interaction between the temperature and fat type (P < 0.01). Generally, electron transport chain complexes I-V enzymatic activities were decreased at 36 °C. The coconut oil-fed birds showed the highest complex I activity at both temperatures. The beef tallow-fed broilers showed the lowest complex II activity at 24 °C. In birds exposed to 36 °C, complex II activity was higher for birds fed saturated coconut oil or beef tallow than those feeding the unsaturated olive oil or soybean oil-supplemented diets. At 24 °C, the highest and lowest complex III activities were recorded for the coconut oil- and beef tallow-supplemented diets, respectively. At 36 °C, the activity of complex III was coconut oil > beef tallow > olive oil > soybean oil. At 24 °C, complex IV activity was highest in coconut oil- or soybean oil-fed broilers; and at 36 °C, complex IV showed the lowest activity in soybean oil-fed birds. The highest complex IV activity was observed in coconut oil-fed chickens followed by olive oil-fed and beef tallow-fed birds, respectively. At 24 or 36 °C, the highest and lowest complex V activity was observed in coconut oil-fed and soybean oil-fed chickens, respectively. ATP concentration and mitochondrial membrane potential were in the order of coconut oil > beef tallow > olive oil > soybean oil at both temperatures. Temperature and fat type significantly affected the avANT mRNA concentration. Exposure of broilers to 36 °C generally decreased the mRNA expression of avANT, with beef tallow- or coconut oil-supplemented birds showing a lower avANT mRNA expression than those receiving olive oil- or soybean oil-supplemented diets. These findings provide further information on the use of fat sources in the diet of heat stressed-broilers.
Collapse
Affiliation(s)
- Kazem Seifi
- Department of Animal Science, Faculty of Animal Science and Fisheries, Sari Agricultural Sciences and Natural Sciences University, Sari, Iran.
| | - Mansour Rezaei
- Department of Animal Science, Faculty of Animal Science and Fisheries, Sari Agricultural Sciences and Natural Sciences University, Sari, Iran
| | - Asad Teimouri Yansari
- Department of Animal Science, Faculty of Animal Science and Fisheries, Sari Agricultural Sciences and Natural Sciences University, Sari, Iran
| | - Mohammad Javad Zamiri
- Department of Animal Science, Faculty of Agriculture, Shiraz University, Shiraz, Iran
| | | | - Reza Heidari
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
18
|
Zhang WQ, Zhao TT, Gui DK, Gao CL, Gu JL, Gan WJ, Huang W, Xu Y, Zhou H, Chen WN, Liu ZL, Xu YH. Sodium Butyrate Improves Liver Glycogen Metabolism in Type 2 Diabetes Mellitus. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2019; 67:7694-7705. [PMID: 31250637 DOI: 10.1021/acs.jafc.9b02083] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Liver plays a central role in modulating blood glucose level. Our most recent findings suggested that supplementation with microbiota metabolite sodium butyrate (NaB) could ameliorate progression of type 2 diabetes mellitus (T2DM) and decrease blood HbA1c in db/db mice. To further investigate the role of butyrate in homeostasis of blood glucose and glycogen metabolism, we carried out the present study. In db/db mice, we found significant hypertrophy and steatosis in hepatic lobules accompanied by reduced glycogen storage, and expression of GPR43 was significantly decreased by 59.38 ± 3.33%; NaB administration significantly increased NaB receptor G-protein coupled receptor 43 (GPR43) level and increased glycogen storage in both mice and HepG2 cells. Glucose transporter 2 (GLUT2) and sodium-glucose cotransporter 1 (SGLT1) on cell membrane were upregulated by NaB. The activation of intracellular signaling Protein kinase B (PKB), also known as AKT, was inhibited while glycogen synthase kinase 3 (GSK3) was activated by NaB in both in vivo and in vitro studies. The present study demonstrated that microbiota metabolite NaB possessed beneficial effects on preserving blood glucose homeostasis by promoting glycogen metabolism in liver cells, and the GPR43-AKT-GSK3 signaling pathway should contribute to this effect.
Collapse
Affiliation(s)
- Wen-Qian Zhang
- Faculty of Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicine , Macau University of Science and Technology , Avenida Wai Long , Taipa , Macao 999078 , People's Republic of China
| | - Ting-Ting Zhao
- Faculty of Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicine , Macau University of Science and Technology , Avenida Wai Long , Taipa , Macao 999078 , People's Republic of China
| | - Ding-Kun Gui
- Department of Nephrology , Shanghai Jiaotong University Affiliated Sixth People's Hospital , Shanghai 200080 , People's Republic of China
| | - Chen-Lin Gao
- Faculty of Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicine , Macau University of Science and Technology , Avenida Wai Long , Taipa , Macao 999078 , People's Republic of China
- Department of Endocrinology , Affiliated Hospital of Southwest Medical University , Luzhou , Sichuan 646000 , People's Republic of China
| | - Jun-Ling Gu
- Faculty of Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicine , Macau University of Science and Technology , Avenida Wai Long , Taipa , Macao 999078 , People's Republic of China
| | - Wen-Jun Gan
- Faculty of Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicine , Macau University of Science and Technology , Avenida Wai Long , Taipa , Macao 999078 , People's Republic of China
| | - Wei Huang
- Department of Endocrinology , Affiliated Hospital of Southwest Medical University , Luzhou , Sichuan 646000 , People's Republic of China
| | - Yong Xu
- Department of Endocrinology , Affiliated Hospital of Southwest Medical University , Luzhou , Sichuan 646000 , People's Republic of China
| | - Hua Zhou
- Faculty of Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicine , Macau University of Science and Technology , Avenida Wai Long , Taipa , Macao 999078 , People's Republic of China
- Macau Institute for Applied Research in Medicine and Health , Avenida Wai Long , Taipa , Macao 999078 , People's Republic of China
| | - Wei-Ni Chen
- Department of Endocrinology , Zhuhai Hospital of Integrated Traditional Chinese and Western Medicine , Zhuhai 519000 , People's Republic of China
| | - Zhi-Long Liu
- Department of Endocrinology , Zhuhai Hospital of Integrated Traditional Chinese and Western Medicine , Zhuhai 519000 , People's Republic of China
| | - You-Hua Xu
- Faculty of Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicine , Macau University of Science and Technology , Avenida Wai Long , Taipa , Macao 999078 , People's Republic of China
| |
Collapse
|
19
|
Zhang C, Wang K, Yang L, Liu R, Chu Y, Qin X, Yang P, Yu H. Lipid metabolism in inflammation-related diseases. Analyst 2019; 143:4526-4536. [PMID: 30128447 DOI: 10.1039/c8an01046c] [Citation(s) in RCA: 127] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
There are thousands of lipid species existing in cells, which belong to eight different categories. Lipids are the essential building blocks of cells. Recent studies have started to unveil the important functions of lipids in regulating cell metabolism. However, we are still at a very early stage in fully understanding the physiological and pathological functions of lipids. The application of lipidomics for studying lipid metabolism can provide a direct readout of the cellular status and broadens our understanding of the mechanisms that underpin metabolic disease states. This review provides an introduction to lipid metabolism and its role in modulating homeostasis and immunity. We also describe representative applications of lipidomics for studying lipid metabolism in inflammation-related diseases.
Collapse
Affiliation(s)
- Cuiping Zhang
- Key Laboratory of Medical Epigenetics and Metabolism, Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China.
| | | | | | | | | | | | | | | |
Collapse
|
20
|
Junghans L, Teleki A, Wijaya AW, Becker M, Schweikert M, Takors R. From nutritional wealth to autophagy: In vivo metabolic dynamics in the cytosol, mitochondrion and shuttles of IgG producing CHO cells. Metab Eng 2019; 54:145-159. [PMID: 30930288 DOI: 10.1016/j.ymben.2019.02.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2018] [Revised: 01/27/2019] [Accepted: 02/27/2019] [Indexed: 10/27/2022]
Abstract
To fulfil the optimization needs of current biopharmaceutical processes the knowledge how to improve cell specific productivities is of outmost importance. This requires a detailed understanding of cellular metabolism on a subcellular level inside compartments such as cytosol and mitochondrion. Using IgG1 producing Chinese hamster ovary (CHO) cells, a pioneering protocol for compartment-specific metabolome analysis was applied. Various production-like growth conditions ranging from ample glucose and amino acid supply via moderate to severe nitrogen limitation were investigated in batch cultures. The combined application of quantitative metabolite pool analysis, 13C tracer studies and non-stationary flux calculations revealed that Pyr/H+ symport (MPC1/2) bore the bulk of the mitochondrial transport under ample nutrient supply. Glutamine limitation induced the concerted adaptation of the bidirectional Mal/aKG (OGC) and the Mal/HPO42- antiporter (DIC), even installing completely reversed shuttle fluxes. As a result, NADPH and ATP formation were adjusted to cellular needs unraveling the key role of cytosolic malic enzyme for NADPH production. Highest cell specific IgG1 productivities were closely correlated to a strong mitochondrial malate export according to the anabolic demands. The requirement to install proper NADPH supply for optimizing the production of monoclonal antibodies is clearly outlined. Interestingly, it was observed that mitochondrial citric acid cycle activity was always maintained enabling constant cytosolic adenylate energy charges at physiological levels, even under autophagy conditions.
Collapse
Affiliation(s)
- Lisa Junghans
- Institute of Biochemical Engineering, University of Stuttgart, Allmandring 31, 70569, Stuttgart, Germany
| | - Attila Teleki
- Institute of Biochemical Engineering, University of Stuttgart, Allmandring 31, 70569, Stuttgart, Germany
| | - Andy Wiranata Wijaya
- Institute of Biochemical Engineering, University of Stuttgart, Allmandring 31, 70569, Stuttgart, Germany
| | - Max Becker
- Institute of Biochemical Engineering, University of Stuttgart, Allmandring 31, 70569, Stuttgart, Germany
| | - Michael Schweikert
- Institute of Biomaterials and Biomolecular Systems, Department of Biobased Materials, University of Stuttgart, Pfaffenwaldring 57, 70569, Stuttgart, Germany
| | - Ralf Takors
- Institute of Biochemical Engineering, University of Stuttgart, Allmandring 31, 70569, Stuttgart, Germany.
| |
Collapse
|
21
|
Tomášová P, Čermáková M, Pelantová H, Vecka M, Kratochvílová H, Lipš M, Lindner J, Šedivá B, Haluzík M, Kuzma M. Minor lipids profiling in subcutaneous and epicardial fat tissue using LC/MS with an optimized preanalytical phase. J Chromatogr B Analyt Technol Biomed Life Sci 2019; 1113:50-59. [PMID: 30897405 DOI: 10.1016/j.jchromb.2019.03.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Revised: 03/05/2019] [Accepted: 03/06/2019] [Indexed: 12/25/2022]
Abstract
Analysis of bioactive lipids in adipose tissue could lead to better understanding of the pathogenesis of obesity and its complications. However, current MS methods are limited by a high content of triacylglycerols (TAGs), which markedly surpasses the amount of other lipids and suppresses their ionization. The aim of our study was thus to optimize the preanalytical phase of lipid analysis in adipose tissue, focusing in particular on less-abundant lipids. Next, the optimized method was used to describe the differences between epicardial and subcutaneous adipose tissues obtained from patients undergoing cardiac surgery. Lipids were extracted using a modified Folch method with subsequent detachment of TAGs by thin layer chromatography (TLC). The extracts with/without TAGs were analyzed by tandem LC/MS. The repeatability of the presented method expressed by the median of the coefficients of variation was 12/5% for analysis with/without TAGs separation, respectively. The difference in the relative abundance of TAGs gained with/without TLC was, on average, 19% and did not reach significance (p value > 0.05) for any identified TAG. The novel preanalytical step allowed us to detect 37 lipids, which could not have been detected without TAG separation, because their signal to noise ratio is <5 in current methods of untargeted lipidomics. These lipids belong predominately to ceramides, glycerophosphatidylserines, glycerophosphatidylinsitols, sphingomyelins, glycerophosphatidylcholines, glycerophosphatidylethanolamines, diacylglycerols. The two adipose tissue depots differed mainly in the following lipid classes: glycerophosphatidylcholines, glycerophosphatidylinositols, glycerophosphatidylethanolamine, and sphingomyelins. Moreover, other major lipids showed distinctly different distributions between the two adipose tissues. Among these, the changes in TAGs were the most striking, which correspond to previously published data describing the differences between omental and subcutaneous adipose tissue. Implementation of the TLC step for the elimination of TAGs was crucial for enhancing the MS detection limit of minor lipids in adipose tissue. The differences between the overall lipid profiles of subcutaneous and epicardial tissue reflect their different functions arising from their location.
Collapse
Affiliation(s)
- Petra Tomášová
- Institute of Microbiology, Czech Academy of Sciences, Vídeňská 1083, 142 20 Prague 4, Czech Republic; 4th Medical Department, First Faculty of Medicine, Charles University and General Faculty Hospital in Prague, U Nemocnice 2, 128 08 Praha 2, Czech Republic
| | - Martina Čermáková
- Institute of Microbiology, Czech Academy of Sciences, Vídeňská 1083, 142 20 Prague 4, Czech Republic; Faculty of Chemical Technology, University of Chemistry and Technology Prague, Technická 5, 166 28 Prague 6, Czech Republic
| | - Helena Pelantová
- Institute of Microbiology, Czech Academy of Sciences, Vídeňská 1083, 142 20 Prague 4, Czech Republic
| | - Marek Vecka
- 4th Medical Department, First Faculty of Medicine, Charles University and General Faculty Hospital in Prague, U Nemocnice 2, 128 08 Praha 2, Czech Republic
| | - Helena Kratochvílová
- Institute of Medical Biochemistry and Laboratory Diagnostics, First Faculty of Medicine, Charles University and General University Hospital in Prague, U Nemocnice 2, 128 08, Prague 2, Czech Republic; Centre for Experimental Medicine, Institute for Clinical and Experimental Medicine, Vídeňská 1958/9, 140 21, Prague 4, Czech Republic
| | - Michal Lipš
- Department of Anaesthesiology, Resuscitation and Intensive Care, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Jaroslav Lindner
- 2nd Department of Surgery - Department of Cardiovascular Surgery, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Blanka Šedivá
- Faculty of Applied Sciences, University of West Bohemia, Univerzitní 8, 306 14 Plzeň, Czech Republic; Institute of Microbiology, Czech Academy of Sciences, Vídeňská 1083, 142 20 Prague 4, Czech Republic
| | - Martin Haluzík
- Institute of Medical Biochemistry and Laboratory Diagnostics, First Faculty of Medicine, Charles University and General University Hospital in Prague, U Nemocnice 2, 128 08, Prague 2, Czech Republic; Centre for Experimental Medicine, Institute for Clinical and Experimental Medicine, Vídeňská 1958/9, 140 21, Prague 4, Czech Republic
| | - Marek Kuzma
- Institute of Microbiology, Czech Academy of Sciences, Vídeňská 1083, 142 20 Prague 4, Czech Republic.
| |
Collapse
|
22
|
20-Week follow-up of hepatic steatosis installation and liver mitochondrial structure and activity and their interrelation in rats fed a high-fat-high-fructose diet. Br J Nutr 2019; 119:368-380. [PMID: 29498345 DOI: 10.1017/s0007114517003713] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The incidence of obesity and its metabolic complications are rapidly increasing and become a major public health issue. This trend is associated with an increase in the prevalence of non-alcoholic fatty liver disease (NAFLD), insulin resistance and diabetes. The sequence of events leading to NAFLD progression and mitochondrial dysfunction and their interrelation remains to be elucidated. This study aimed to explore the installation and progression of NAFLD and its association with the liver mitochondrial structure and activity changes in rats fed an obesogenic diet up to 20 weeks. Male Wistar rats were fed either a standard or high-fat-high-fructose (HFHFR) diet and killed on 4, 8, 12, 16 and 20 weeks of diet intake. Rats fed the HFHFR diet developed mildly overweight, associated with increased adipose tissue weight, hepatic steatosis, hyperglycaemia and hyperinsulinaemia after 8 weeks of HFHFR diet. Hepatic steatosis and many biochemical modifications plateaued at 8-12 weeks of HFHFR diet with slight amelioration afterwards. Interestingly, several biochemical and physiological parameters of mitochondrial function, as well as its phospholipid composition, in particular cardiolipin content, were tightly related to hepatic steatosis installation. These results showed once again the interrelation between hepatic steatosis development and mitochondrial activity alterations without being able to say whether the mitochondrial alterations preceded or followed the installation/progression of hepatic steatosis. Because both hepatic steatosis and mitochondrial alterations occurred as early as 4 weeks of diet, future studies should consider these four 1st weeks to reveal the exact interconnection between these major consequences of obesogenic diet intake.
Collapse
|
23
|
Darbandi M, Darbandi S, Agarwal A, Sengupta P, Durairajanayagam D, Henkel R, Sadeghi MR. Reactive oxygen species and male reproductive hormones. Reprod Biol Endocrinol 2018; 16:87. [PMID: 30205828 PMCID: PMC6134507 DOI: 10.1186/s12958-018-0406-2] [Citation(s) in RCA: 199] [Impact Index Per Article: 28.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Accepted: 08/30/2018] [Indexed: 12/12/2022] Open
Abstract
Reports of the increasing incidence of male infertility paired with decreasing semen quality have triggered studies on the effects of lifestyle and environmental factors on the male reproductive potential. There are numerous exogenous and endogenous factors that are able to induce excessive production of reactive oxygen species (ROS) beyond that of cellular antioxidant capacity, thus causing oxidative stress. In turn, oxidative stress negatively affects male reproductive functions and may induce infertility either directly or indirectly by affecting the hypothalamus-pituitary-gonadal (HPG) axis and/or disrupting its crosstalk with other hormonal axes. This review discusses the important exogenous and endogenous factors leading to the generation of ROS in different parts of the male reproductive tract. It also highlights the negative impact of oxidative stress on the regulation and cross-talk between the reproductive hormones. It further describes the mechanism of ROS-induced derangement of male reproductive hormonal profiles that could ultimately lead to male infertility. An understanding of the disruptive effects of ROS on male reproductive hormones would encourage further investigations directed towards the prevention of ROS-mediated hormonal imbalances, which in turn could help in the management of male infertility.
Collapse
Affiliation(s)
- Mahsa Darbandi
- Reproductive Biotechnology Research Center, Avicenna Research Institute, Academic Center for Education, Culture and Research, Tehran, Iran
| | - Sara Darbandi
- Reproductive Biotechnology Research Center, Avicenna Research Institute, Academic Center for Education, Culture and Research, Tehran, Iran
| | - Ashok Agarwal
- American Center for Reproductive Medicine, Cleveland Clinic, Cleveland, Ohio 44195 USA
| | - Pallav Sengupta
- Department of Physiology, Faculty of Medicine, MAHSA University, Jalan SP2, Bandar Saujana Putra, 42610 Jenjarom, Selangor Malaysia
| | - Damayanthi Durairajanayagam
- Department of Physiology, Faculty of Medicine, Universiti Teknologi MARA, Sungai Buloh Campus, Jalan Hospital, 47000 Sungai Buloh, Selangor Malaysia
| | - Ralf Henkel
- Department of Medical Biosciences, University of the Western Cape, Bellville, Cape Town, 7535 South Africa
| | - Mohammad Reza Sadeghi
- Reproductive Immunology Research Center, Avicenna Research Institute, Academic Center for Education, Culture and Research, Tehran, Iran
| |
Collapse
|
24
|
Yan J, Yu Y, Kang JW, Tam ZY, Xu S, Fong ELS, Singh SP, Song Z, Tucker-Kellogg L, So PTC, Yu H. Development of a classification model for non-alcoholic steatohepatitis (NASH) using confocal Raman micro-spectroscopy. JOURNAL OF BIOPHOTONICS 2017; 10. [PMID: 28635128 PMCID: PMC5902180 DOI: 10.1002/jbio.201600303] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is the most common liver disorder in developed countries [1]. A subset of individuals with NAFLD progress to non-alcoholic steatohepatitis (NASH), an advanced form of NAFLD which predisposes individuals to cirrhosis, liver failure and hepatocellular carcinoma. The current gold standard for NASH diagnosis and staging is based on histological evaluation, which is largely semi-quantitative and subjective. To address the need for an automated and objective approach to NASH detection, we combined Raman micro-spectroscopy and machine learning techniques to develop a classification model based on a well-established NASH mouse model, using spectrum pre-processing, biochemical component analysis (BCA) and logistic regression. By employing a selected pool of biochemical components, we identified biochemical changes specific to NASH and show that the classification model is capable of accurately detecting NASH (AUC=0.85-0.87) in mice. The unique biochemical fingerprint generated in this study may serve as a useful criterion to be leveraged for further validation in clinical samples.
Collapse
Affiliation(s)
- Jie Yan
- Institute of Bioengineering and Nanotechnology, Agency for Science, Technology and Research (A*STAR), Singapore 138669
| | - Yang Yu
- Institute of Bioengineering and Nanotechnology, Agency for Science, Technology and Research (A*STAR), Singapore 138669
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, 2 Medical Drive, Singapore 117597
- BioSyM, Singapore-MIT Alliance for Research and Technology, Singapore 138602
| | - Jeon Woong Kang
- Laser Biomedical Research Center, George R. Harrison Spectroscopy Laboratory, Massachusetts Institute of Technology, Cambridge, MA 02139 USA
| | - Zhi Yang Tam
- BioSyM, Singapore-MIT Alliance for Research and Technology, Singapore 138602
| | - Shuoyu Xu
- InvitroCue Pte Ltd, Singapore 138667
| | - Eliza Li Shan Fong
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, 2 Medical Drive, Singapore 117597
| | - Surya Pratap Singh
- Laser Biomedical Research Center, George R. Harrison Spectroscopy Laboratory, Massachusetts Institute of Technology, Cambridge, MA 02139 USA
| | - Ziwei Song
- Institute of Bioengineering and Nanotechnology, Agency for Science, Technology and Research (A*STAR), Singapore 138669
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, 2 Medical Drive, Singapore 117597
| | - Lisa Tucker-Kellogg
- BioSyM, Singapore-MIT Alliance for Research and Technology, Singapore 138602
- Duke-NUS Graduate Medical School Singapore, National University of Singapore, Singapore 169857
| | - Peter T. C. So
- BioSyM, Singapore-MIT Alliance for Research and Technology, Singapore 138602
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139 USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139 USA
| | - Hanry Yu
- Institute of Bioengineering and Nanotechnology, Agency for Science, Technology and Research (A*STAR), Singapore 138669
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, 2 Medical Drive, Singapore 117597
- BioSyM, Singapore-MIT Alliance for Research and Technology, Singapore 138602
- Mechanobiology Institute, National University of Singapore, Singapore 117411
- Corresponding author: , Tel. No. +65 65163466, Fax No. +65 68748261
| |
Collapse
|
25
|
Ademowo OS, Dias HKI, Burton DGA, Griffiths HR. Lipid (per) oxidation in mitochondria: an emerging target in the ageing process? Biogerontology 2017; 18:859-879. [PMID: 28540446 PMCID: PMC5684309 DOI: 10.1007/s10522-017-9710-z] [Citation(s) in RCA: 130] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Accepted: 05/09/2017] [Indexed: 12/11/2022]
Abstract
Lipids are essential for physiological processes such as maintaining membrane integrity, providing a source of energy and acting as signalling molecules to control processes including cell proliferation, metabolism, inflammation and apoptosis. Disruption of lipid homeostasis can promote pathological changes that contribute towards biological ageing and age-related diseases. Several age-related diseases have been associated with altered lipid metabolism and an elevation in highly damaging lipid peroxidation products; the latter has been ascribed, at least in part, to mitochondrial dysfunction and elevated ROS formation. In addition, senescent cells, which are known to contribute significantly to age-related pathologies, are also associated with impaired mitochondrial function and changes in lipid metabolism. Therapeutic targeting of dysfunctional mitochondrial and pathological lipid metabolism is an emerging strategy for alleviating their negative impact during ageing and the progression to age-related diseases. Such therapies could include the use of drugs that prevent mitochondrial uncoupling, inhibit inflammatory lipid synthesis, modulate lipid transport or storage, reduce mitochondrial oxidative stress and eliminate senescent cells from tissues. In this review, we provide an overview of lipid structure and function, with emphasis on mitochondrial lipids and their potential for therapeutic targeting during ageing and age-related disease.
Collapse
Affiliation(s)
- O S Ademowo
- Life & Health Sciences, Aston University, Birmingham, UK
| | - H K I Dias
- Life & Health Sciences, Aston University, Birmingham, UK
| | - D G A Burton
- Life & Health Sciences, Aston University, Birmingham, UK
| | - H R Griffiths
- Life & Health Sciences, Aston University, Birmingham, UK.
- Health and Medical Sciences, University of Surrey, Guildford, UK.
| |
Collapse
|
26
|
Hung CL, Pan SH, Han CL, Chang CW, Hsu YL, Su CH, Shih SC, Lai YJ, Chiang Chiau JS, Yeh HI, Liu CY, Lee HC, Lam CS. Membrane Proteomics of Impaired Energetics and Cytoskeletal Disorganization in Elderly Diet-Induced Diabetic Mice. J Proteome Res 2017; 16:3504-3513. [DOI: 10.1021/acs.jproteome.7b00148] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Chung-Lieh Hung
- Department
of Medicine, Mackay Medical College, New Taipei City, 252, Taiwan
| | - Szu-Hua Pan
- Graduate
Institute of Medical Genomics and Proteomics, College of Medicine, National Taiwan University, Taipei, 100, Taiwan
| | | | - Ching-Wei Chang
- Department
of Medicine, Mackay Medical College, New Taipei City, 252, Taiwan
| | - Yuan-Ling Hsu
- Graduate
Institute of Medical Genomics and Proteomics, College of Medicine, National Taiwan University, Taipei, 100, Taiwan
| | | | - Shou-Chuan Shih
- Department
of Medicine, Mackay Medical College, New Taipei City, 252, Taiwan
| | - Yu-Jun Lai
- Department
of Medicine, Mackay Medical College, New Taipei City, 252, Taiwan
| | | | - Hung-I Yeh
- Department
of Medicine, Mackay Medical College, New Taipei City, 252, Taiwan
| | - Chia-Yuan Liu
- Department
of Medicine, Mackay Medical College, New Taipei City, 252, Taiwan
| | - Hung-Chang Lee
- Department
of Medicine, Mackay Medical College, New Taipei City, 252, Taiwan
- MacKay Children’s
Hospital, Taipei, 104, Taiwan
- Mackay Junior
College of Medicine, Nursing, and Management, New Taipei City, 252, Taiwan
| | - Carolyn S.P. Lam
- National Heart
Centre Singapore, 169609, Singapore
- Duke-National
University of Singapore, 169857, Singapore
| |
Collapse
|
27
|
Maternal intake of trans-unsaturated or interesterified fatty acids during pregnancy and lactation modifies mitochondrial bioenergetics in the liver of adult offspring in mice. Br J Nutr 2017; 118:41-52. [DOI: 10.1017/s0007114517001817] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
AbstractThe quality of dietary lipids in the maternal diet can programme the offspring to diseases in later life. We investigated whether the maternal intake of palm oil or interesterified fat, substitutes for trans-unsaturated fatty acids (FA), induces metabolic changes in the adult offspring. During pregnancy and lactation, C57BL/6 female mice received normolipidic diets containing partially hydrogenated vegetable fat rich in trans-unsaturated fatty acids (TG), palm oil (PG), interesterified fat (IG) or soyabean oil (CG). After weaning, male offspring from all groups received the control diet until day 110. Plasma glucose and TAG and liver FA profiles were ascertained. Liver mitochondrial function was accessed with high-resolution respirometry by measuring VO2, fluorimetry for detection of hydrogen peroxide (H2O2) production and mitochondrial Ca2+ uptake. The results showed that the IG offspring presented a 20 % increase in plasma glucose and both the IG and TG offspring presented a 2- and 1·9-fold increase in TAG, respectively, when compared with CG offspring. Liver MUFA and PUFA contents decreased in the TG and IG offspring when compared with CG offspring. Liver MUFA content also decreased in the PG offspring. These modifications in FA composition possibly affected liver mitochondrial function, as respiration was impaired in the TG offspring and H2O2 production was higher in the IG offspring. In addition, mitochondrial Ca2+ retention capacity was reduced by approximately 40 and 55 % in the TG and IG offspring, respectively. In conclusion, maternal consumption of trans-unsaturated and interesterified fat affected offspring health by compromising mitochondrial bioenergetics and lipid metabolism in the liver.
Collapse
|
28
|
The Immunosuppressant Mycophenolic Acid Alters Nucleotide and Lipid Metabolism in an Intestinal Cell Model. Sci Rep 2017; 7:45088. [PMID: 28327659 PMCID: PMC5361167 DOI: 10.1038/srep45088] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Accepted: 02/16/2017] [Indexed: 01/14/2023] Open
Abstract
The study objective was to elucidate the molecular mechanisms underlying the negative effects of mycophenolic acid (MPA) on human intestinal cells. Effects of MPA exposure and guanosine supplementation on nucleotide concentrations in LS180 cells were assessed using liquid chromatography-mass spectrometry. Proteomics analysis was carried out using stable isotope labeling by amino acids in cell culture combined with gel-based liquid chromatography-mass spectrometry and lipidome analysis using 1H nuclear magnetic resonance spectroscopy. Despite supplementation, depletion of guanosine nucleotides (p < 0.001 at 24 and 72 h; 5, 100, and 250 μM MPA) and upregulation of uridine and cytidine nucleotides (p < 0.001 at 24 h; 5 μM MPA) occurred after exposure to MPA. MPA significantly altered 35 proteins mainly related to nucleotide-dependent processes and lipid metabolism. Cross-reference with previous studies of MPA-associated protein changes widely corroborated these results, but showed differences that may be model- and/or method-dependent. MPA exposure increased intracellular concentrations of fatty acids, cholesterol, and phosphatidylcholine (p < 0.01 at 72 h; 100 μM MPA) which corresponded to the changes in lipid-metabolizing proteins. MPA affected intracellular nucleotide levels, nucleotide-dependent processes, expression of structural proteins, fatty acid and lipid metabolism in LS180 cells. These changes may compromise intestinal membrane integrity and contribute to gastrointestinal toxicity.
Collapse
|
29
|
Entezari MH, Hadi A, Kafeshani M. Effects of dietary approaches to stop hypertension diet versus usual dietary advice on glycemic indices in women at risk for cardiovascular disease; a randomized controlled clinical trial. J Renal Inj Prev 2016. [DOI: 10.15171/jrip.2017.39] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
|
30
|
Suzuki S, Sato Y, Umegaki K, Chiba T. Influence of High-fat and High-cholesterol Diet on Major CYP Activities in the Liver. YAKUGAKU ZASSHI 2016; 136:1297-305. [PMID: 27592832 DOI: 10.1248/yakushi.16-00034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
We previously reported that a high-fat and high-cholesterol (HFHC) diet for 12 weeks induced non-alcoholic steatohepatitis (NASH) and influenced major CYP subtype gene expression levels and activities in a mouse model. In the present study, we determined the effects of the HFHC diet on CYP expression levels and activities prior to the establishment of NASH. When male C57BL/6J mice were fed the HFHC or a normal chow diet (Control) ad libitum for 4 weeks, body weights were significantly lower, whereas liver weights and hepatic lipid contents were significantly higher in the HFHC group than in the Control group. Under these conditions, hepatic microsomal luciferin-H (human CYP2C9 substrate) hydroxylation activity was significantly lower in the HFHC group than in the Control group. In order to investigate drug efficacy in mice fed the HFHC diet, an intraperitoneal glucose tolerance test was conducted with or without a pretreatment with tolbutamide (a CYP2C substrate) after 4 weeks of feeding. The plasma glucose-lowering effects of tolbutamide were attenuated in the HFHC group even though luciferin-H hydroxylation activity was suppressed in this group. The reason for this discrepancy was attributed to the mRNA expression levels of Cyp2c44 being lower and those of Cyp2c29 and Cyp2c66, which are involved in the metabolism of tolbutamide, being higher in the HFHC group than in the Control group. These results indicate that the expression of Cyp2c in the liver is influenced by the HFHC diet prior to the establishment of NASH and its regulation differed among the subtypes examined.
Collapse
Affiliation(s)
- Sachina Suzuki
- Information Center, National Institute of Health and Nutrition, National Institutes of Biomedical Innovation, Health and Nutrition
| | | | | | | |
Collapse
|
31
|
Kovach CP, Al Koborssy D, Huang Z, Chelette BM, Fadool JM, Fadool DA. Mitochondrial Ultrastructure and Glucose Signaling Pathways Attributed to the Kv1.3 Ion Channel. Front Physiol 2016; 7:178. [PMID: 27242550 PMCID: PMC4871887 DOI: 10.3389/fphys.2016.00178] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Accepted: 05/04/2016] [Indexed: 12/20/2022] Open
Abstract
Gene-targeted deletion of the potassium channel Kv1.3 (Kv1.3−∕−) results in “Super-smeller” mice with a sensory phenotype that includes an increased olfactory ability linked to changes in olfactory circuitry, increased abundance of olfactory cilia, and increased expression of odorant receptors and the G-protein, Golf. Kv1.3−∕− mice also have a metabolic phenotype including lower body weight and decreased adiposity, increased total energy expenditure (TEE), increased locomotor activity, and resistance to both diet- and genetic-induced obesity. We explored two cellular aspects to elucidate the mechanism by which loss of Kv1.3 channel in the olfactory bulb (OB) may enhance glucose utilization and metabolic rate. First, using in situ hybridization we find that Kv1.3 and the insulin-dependent glucose transporter type 4 (GLUT4) are co-localized to the mitral cell layer of the OB. Disruption of Kv1.3 conduction via construction of a pore mutation (W386F Kv1.3) was sufficient to independently translocate GLUT4 to the plasma membrane in HEK 293 cells. Because olfactory sensory perception and the maintenance of action potential (AP) firing frequency by mitral cells of the OB is highly energy demanding and Kv1.3 is also expressed in mitochondria, we next explored the structure of this organelle in mitral cells. We challenged wildtype (WT) and Kv1.3−∕− male mice with a moderately high-fat diet (MHF, 31.8 % kcal fat) for 4 months and then examined OB ultrastructure using transmission electron microscopy. In WT mice, mitochondria were significantly enlarged following diet-induced obesity (DIO) and there were fewer mitochondria, likely due to mitophagy. Interestingly, mitochondria were significantly smaller in Kv1.3−∕− mice compared with that of WT mice. Similar to their metabolic resistance to DIO, the Kv1.3−∕− mice had unchanged mitochondria in terms of cross sectional area and abundance following a challenge with modified diet. We are very interested to understand how targeted disruption of the Kv1.3 channel in the OB can modify TEE. Our study demonstrates that Kv1.3 regulates mitochondrial structure and alters glucose utilization; two important metabolic changes that could drive whole system changes in metabolism initiated at the OB.
Collapse
Affiliation(s)
- Christopher P Kovach
- Program in Neuroscience, Florida State UniversityTallahassee, FL, USA; Department of Biological Science, Florida State UniversityTallahassee, FL, USA
| | - Dolly Al Koborssy
- Program in Neuroscience, Florida State University Tallahassee, FL, USA
| | - Zhenbo Huang
- Program in Neuroscience, Florida State University Tallahassee, FL, USA
| | | | - James M Fadool
- Program in Neuroscience, Florida State UniversityTallahassee, FL, USA; Department of Biological Science, Florida State UniversityTallahassee, FL, USA
| | - Debra A Fadool
- Program in Neuroscience, Florida State UniversityTallahassee, FL, USA; Department of Biological Science, Florida State UniversityTallahassee, FL, USA; Institute of Molecular Biophysics, Florida State UniversityTallahassee, FL, USA
| |
Collapse
|
32
|
Sandlers Y, Mercier K, Pathmasiri W, Carlson J, McRitchie S, Sumner S, Vernon HJ. Metabolomics Reveals New Mechanisms for Pathogenesis in Barth Syndrome and Introduces Novel Roles for Cardiolipin in Cellular Function. PLoS One 2016; 11:e0151802. [PMID: 27015085 PMCID: PMC4807847 DOI: 10.1371/journal.pone.0151802] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Accepted: 03/04/2016] [Indexed: 02/07/2023] Open
Abstract
Barth Syndrome is the only known Mendelian disorder of cardiolipin remodeling, with characteristic clinical features of cardiomyopathy, skeletal myopathy, and neutropenia. While the primary biochemical defects of reduced mature cardiolipin and increased monolysocardiolipin are well-described, much of the downstream biochemical dysregulation has not been uncovered, and biomarkers are limited. In order to further expand upon the knowledge of the biochemical abnormalities in Barth Syndrome, we analyzed metabolite profiles in plasma from a cohort of individuals with Barth Syndrome compared to age-matched controls via 1H nuclear magnetic resonance spectroscopy and liquid chromatography-mass spectrometry. A clear distinction between metabolite profiles of individuals with Barth Syndrome and controls was observed, and was defined by an array of metabolite classes including amino acids and lipids. Pathway analysis of these discriminating metabolites revealed involvement of mitochondrial and extra-mitochondrial biochemical pathways including: insulin regulation of fatty acid metabolism, lipid metabolism, biogenic amine metabolism, amino acid metabolism, endothelial nitric oxide synthase signaling, and tRNA biosynthesis. Taken together, this data indicates broad metabolic dysregulation in Barth Syndrome with wide cellular effects.
Collapse
Affiliation(s)
- Yana Sandlers
- Department of Chemistry, Cleveland State University, Cleveland, OH, United States of America
| | - Kelly Mercier
- Research Triangle International, Durham, NC, United States of America
| | - Wimal Pathmasiri
- Research Triangle International, Durham, NC, United States of America
| | - Jim Carlson
- Research Triangle International, Durham, NC, United States of America
| | - Susan McRitchie
- Research Triangle International, Durham, NC, United States of America
| | - Susan Sumner
- Research Triangle International, Durham, NC, United States of America
| | - Hilary J. Vernon
- Department of Neurogenetics, Kennedy Krieger Institute, Baltimore, MD, United States of America
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University, Baltimore, MD, United States of America
- * E-mail:
| |
Collapse
|
33
|
Altered Energetics of Exercise Explain Risk of Rhabdomyolysis in Very Long-Chain Acyl-CoA Dehydrogenase Deficiency. PLoS One 2016; 11:e0147818. [PMID: 26881790 PMCID: PMC4755596 DOI: 10.1371/journal.pone.0147818] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Accepted: 01/09/2016] [Indexed: 12/31/2022] Open
Abstract
Rhabdomyolysis is common in very long-chain acyl-CoA dehydrogenase deficiency (VLCADD) and other metabolic myopathies, but its pathogenic basis is poorly understood. Here, we show that prolonged bicycling exercise against a standardized moderate workload in VLCADD patients is associated with threefold bigger changes in phosphocreatine (PCr) and inorganic phosphate (Pi) concentrations in quadriceps muscle and twofold lower changes in plasma acetyl-carnitine levels than in healthy subjects. This result is consistent with the hypothesis that muscle ATP homeostasis during exercise is compromised in VLCADD. However, the measured rates of PCr and Pi recovery post-exercise showed that the mitochondrial capacity for ATP synthesis in VLCADD muscle was normal. Mathematical modeling of oxidative ATP metabolism in muscle composed of three different fiber types indicated that the observed altered energy balance during submaximal exercise in VLCADD patients may be explained by a slow-to-fast shift in quadriceps fiber-type composition corresponding to 30% of the slow-twitch fiber-type pool in healthy quadriceps muscle. This study demonstrates for the first time that quadriceps energy balance during exercise in VLCADD patients is altered but not because of failing mitochondrial function. Our findings provide new clues to understanding the risk of rhabdomyolysis following exercise in human VLCADD.
Collapse
|
34
|
Kakimoto PA, Kowaltowski AJ. Effects of high fat diets on rodent liver bioenergetics and oxidative imbalance. Redox Biol 2016; 8:216-25. [PMID: 26826574 PMCID: PMC4753394 DOI: 10.1016/j.redox.2016.01.009] [Citation(s) in RCA: 123] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Revised: 01/11/2016] [Accepted: 01/13/2016] [Indexed: 02/08/2023] Open
Abstract
Human metabolic diseases can be mimicked in rodents by using dietary interventions such as high fat diets (HFD). Nonalcoholic fatty liver disease (NAFLD) develops as a result of HFD and the disease may progress in a manner involving increased production of oxidants. The main intracellular source of these oxidants are mitochondria, which are also responsible for lipid metabolism and thus widely recognized as important players in the pathology and progression of steatosis. Here, we review publications that study redox and bioenergetic effects of HFD in the liver. We find that dietary composition and protocol implementations vary widely, as do the results of these dietary interventions. Overall, all HFD promote steatosis, changes in β-oxidation, generation and consequences of oxidants, while effects on body weight, insulin signaling and other bioenergetic parameters are more variable with the experimental models adopted. Our review provides a broad analysis of the bioenergetic and redox changes promoted by HFD as well as suggestions for changes and specifications in methodologies that may help explain apparent disparities in the current literature. High fat diets (HFDs) induce steatosis, even with no weight changes . HFDs activate β-oxidation. HFDs promote oxidative imbalance.
Collapse
Affiliation(s)
- Pâmela A Kakimoto
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, Brazil
| | - Alicia J Kowaltowski
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, Brazil
| |
Collapse
|
35
|
Schäfer A, Neschen S, Kahle M, Sarioglu H, Gaisbauer T, Imhof A, Adamski J, Hauck SM, Ueffing M. The Epoxyeicosatrienoic Acid Pathway Enhances Hepatic Insulin Signaling and is Repressed in Insulin-Resistant Mouse Liver. Mol Cell Proteomics 2015; 14:2764-74. [PMID: 26070664 PMCID: PMC4597150 DOI: 10.1074/mcp.m115.049064] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Indexed: 11/06/2022] Open
Abstract
Although it is widely accepted that ectopic lipid accumulation in the liver is associated with hepatic insulin resistance, the underlying molecular mechanisms have not been well characterized. Here we employed time resolved quantitative proteomic profiling of mice fed a high fat diet to determine which pathways were affected during the transition of the liver to an insulin-resistant state. We identified several metabolic pathways underlying altered protein expression. In order to test the functional impact of a critical subset of these alterations, we focused on the epoxyeicosatrienoic acid (EET) eicosanoid pathway, whose deregulation coincided with the onset of hepatic insulin resistance. These results suggested that EETs may be positive modulators of hepatic insulin signaling. Analyzing EET activity in primary hepatocytes, we found that EETs enhance insulin signaling on the level of Akt. In contrast, EETs did not influence insulin receptor or insulin receptor substrate-1 phosphorylation. This effect was mediated through the eicosanoids, as overexpression of the deregulated enzymes in absence of arachidonic acid had no impact on insulin signaling. The stimulation of insulin signaling by EETs and depression of the pathway in insulin resistant liver suggest a likely role in hepatic insulin resistance. Our findings support therapeutic potential for inhibiting EET degradation.
Collapse
Affiliation(s)
- Alexander Schäfer
- From the ‡Research Unit Protein Science, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Germany, Ingolstädter Landstr.1 8674 Neuherberg; §German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Susanne Neschen
- §German Center for Diabetes Research (DZD), Neuherberg, Germany; ¶Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Germany, Ingolstädter Landstr.1 8674 Neuherberg
| | - Melanie Kahle
- §German Center for Diabetes Research (DZD), Neuherberg, Germany; ¶Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Germany, Ingolstädter Landstr.1 8674 Neuherberg
| | - Hakan Sarioglu
- From the ‡Research Unit Protein Science, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Germany, Ingolstädter Landstr.1 8674 Neuherberg; §German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Tobias Gaisbauer
- §German Center for Diabetes Research (DZD), Neuherberg, Germany; ¶Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Germany, Ingolstädter Landstr.1 8674 Neuherberg
| | - Axel Imhof
- ‖Munich Center of Integrated Protein Science, Adolf-Butenandt Institute, Ludwig Maximilians University of Munich, Germany, Schillerstraβe 44, 80336 Munich
| | - Jerzy Adamski
- §German Center for Diabetes Research (DZD), Neuherberg, Germany; ¶Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Germany, Ingolstädter Landstr.1 8674 Neuherberg; **Institute of Experimental Genetics, Technical University Munich, Freising-Weihenstephan, Germany
| | - Stefanie M Hauck
- From the ‡Research Unit Protein Science, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Germany, Ingolstädter Landstr.1 8674 Neuherberg; §German Center for Diabetes Research (DZD), Neuherberg, Germany;
| | - Marius Ueffing
- From the ‡Research Unit Protein Science, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Germany, Ingolstädter Landstr.1 8674 Neuherberg; §German Center for Diabetes Research (DZD), Neuherberg, Germany; ‡‡Centre of Ophthalmology, Institute for Ophthalmic Research, University of Tübingen, Germany, Röntgenweg 11,72076 Tübingen
| |
Collapse
|