1
|
Gernez E, Bennis A, Diesnis R, Niguet JP, Grzych G. Awareness of health care related to nitrous oxide abuse for diagnosis, treatment and follow-up. Ir J Med Sci 2023; 192:3087-3089. [PMID: 36867372 DOI: 10.1007/s11845-023-03327-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 02/23/2023] [Indexed: 03/04/2023]
Affiliation(s)
- Emeline Gernez
- CHU de Lille-Centre de Biologie Pathologie Génétique-Service Hormonologie Métabolisme Nutrition Oncologie-1 boulevard du professeur Jules Leclercq, 59000, Lille, France
| | - Anas Bennis
- CH de Roubaix, Service de Neurologie, 59100, Roubaix, France
| | - Remy Diesnis
- CH de Roubaix, Services des Urgences Adultes-SMUR, 59100, Roubaix, France
| | - Jean Paul Niguet
- Groupement des Hôpitaux de l'Institut Catholique de Lille, Hôpital Saint Vincent de Paul, Service de Neurologie, 59000, Lille, France
| | - Guillaume Grzych
- CHU de Lille-Centre de Biologie Pathologie Génétique-Service Hormonologie Métabolisme Nutrition Oncologie-1 boulevard du professeur Jules Leclercq, 59000, Lille, France.
| |
Collapse
|
2
|
Ungvari A, Gulej R, Csik B, Mukli P, Negri S, Tarantini S, Yabluchanskiy A, Benyo Z, Csiszar A, Ungvari Z. The Role of Methionine-Rich Diet in Unhealthy Cerebrovascular and Brain Aging: Mechanisms and Implications for Cognitive Impairment. Nutrients 2023; 15:4662. [PMID: 37960316 PMCID: PMC10650229 DOI: 10.3390/nu15214662] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 10/28/2023] [Accepted: 10/31/2023] [Indexed: 11/15/2023] Open
Abstract
As aging societies in the western world face a growing prevalence of vascular cognitive impairment and Alzheimer's disease (AD), understanding their underlying causes and associated risk factors becomes increasingly critical. A salient concern in the western dietary context is the high consumption of methionine-rich foods such as red meat. The present review delves into the impact of this methionine-heavy diet and the resultant hyperhomocysteinemia on accelerated cerebrovascular and brain aging, emphasizing their potential roles in cognitive impairment. Through a comprehensive exploration of existing evidence, a link between high methionine intake and hyperhomocysteinemia and oxidative stress, mitochondrial dysfunction, inflammation, and accelerated epigenetic aging is drawn. Moreover, the microvascular determinants of cognitive deterioration, including endothelial dysfunction, reduced cerebral blood flow, microvascular rarefaction, impaired neurovascular coupling, and blood-brain barrier (BBB) disruption, are explored. The mechanisms by which excessive methionine consumption and hyperhomocysteinemia might drive cerebromicrovascular and brain aging processes are elucidated. By presenting an intricate understanding of the relationships among methionine-rich diets, hyperhomocysteinemia, cerebrovascular and brain aging, and cognitive impairment, avenues for future research and potential therapeutic interventions are suggested.
Collapse
Affiliation(s)
- Anna Ungvari
- Department of Public Health, Semmelweis University, 1089 Budapest, Hungary
| | - Rafal Gulej
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (R.G.); (B.C.); (P.M.); (S.N.); (S.T.); (A.Y.); (A.C.); (Z.U.)
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Boglarka Csik
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (R.G.); (B.C.); (P.M.); (S.N.); (S.T.); (A.Y.); (A.C.); (Z.U.)
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- International Training Program in Geroscience, Department of Public Health, Doctoral School of Basic and Translational Medicine, Semmelweis University, 1089 Budapest, Hungary
| | - Peter Mukli
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (R.G.); (B.C.); (P.M.); (S.N.); (S.T.); (A.Y.); (A.C.); (Z.U.)
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- International Training Program in Geroscience, Department of Public Health, Doctoral School of Basic and Translational Medicine, Semmelweis University, 1089 Budapest, Hungary
| | - Sharon Negri
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (R.G.); (B.C.); (P.M.); (S.N.); (S.T.); (A.Y.); (A.C.); (Z.U.)
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Stefano Tarantini
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (R.G.); (B.C.); (P.M.); (S.N.); (S.T.); (A.Y.); (A.C.); (Z.U.)
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- International Training Program in Geroscience, Department of Public Health, Doctoral School of Basic and Translational Medicine, Semmelweis University, 1089 Budapest, Hungary
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK 73104, USA
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Andriy Yabluchanskiy
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (R.G.); (B.C.); (P.M.); (S.N.); (S.T.); (A.Y.); (A.C.); (Z.U.)
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- International Training Program in Geroscience, Department of Public Health, Doctoral School of Basic and Translational Medicine, Semmelweis University, 1089 Budapest, Hungary
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK 73104, USA
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Zoltan Benyo
- Institute of Translational Medicine, Semmelweis University, 1094 Budapest, Hungary;
- Cerebrovascular and Neurocognitive Disorders Research Group, Eötvös Loránd Research Network, Semmelweis University, 1094 Budapest, Hungary
| | - Anna Csiszar
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (R.G.); (B.C.); (P.M.); (S.N.); (S.T.); (A.Y.); (A.C.); (Z.U.)
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK 73104, USA
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- International Training Program in Geroscience, Department of Translational Medicine, Doctoral School of Basic and Translational Medicine, Semmelweis University, 1089 Budapest, Hungary
| | - Zoltan Ungvari
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (R.G.); (B.C.); (P.M.); (S.N.); (S.T.); (A.Y.); (A.C.); (Z.U.)
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- International Training Program in Geroscience, Department of Public Health, Doctoral School of Basic and Translational Medicine, Semmelweis University, 1089 Budapest, Hungary
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK 73104, USA
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| |
Collapse
|
3
|
Camici M, Garcia-Gil M, Allegrini S, Pesi R, Bernardini G, Micheli V, Tozzi MG. Inborn Errors of Purine Salvage and Catabolism. Metabolites 2023; 13:787. [PMID: 37512494 PMCID: PMC10383617 DOI: 10.3390/metabo13070787] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 06/20/2023] [Accepted: 06/21/2023] [Indexed: 07/30/2023] Open
Abstract
Cellular purine nucleotides derive mainly from de novo synthesis or nucleic acid turnover and, only marginally, from dietary intake. They are subjected to catabolism, eventually forming uric acid in humans, while bases and nucleosides may be converted back to nucleotides through the salvage pathways. Inborn errors of the purine salvage pathway and catabolism have been described by several researchers and are usually referred to as rare diseases. Since purine compounds play a fundamental role, it is not surprising that their dysmetabolism is accompanied by devastating symptoms. Nevertheless, some of these manifestations are unexpected and, so far, have no explanation or therapy. Herein, we describe several known inborn errors of purine metabolism, highlighting their unexplained pathological aspects. Our intent is to offer new points of view on this topic and suggest diagnostic tools that may possibly indicate to clinicians that the inborn errors of purine metabolism may not be very rare diseases after all.
Collapse
Affiliation(s)
- Marcella Camici
- Unità di Biochimica, Dipartimento di Biologia, Università di Pisa, Via San Zeno 51, 56127 Pisa, Italy
| | - Mercedes Garcia-Gil
- Unità di Fisiologia Generale, Dipartimento di Biologia, Università di Pisa, Via San Zeno 31, 56127 Pisa, Italy
- CISUP, Centro per l'Integrazione Della Strumentazione Dell'Università di Pisa, 56127 Pisa, Italy
- Centro di Ricerca Interdipartimentale Nutrafood "Nutraceuticals and Food for Health", Università di Pisa, 56126 Pisa, Italy
| | - Simone Allegrini
- Unità di Biochimica, Dipartimento di Biologia, Università di Pisa, Via San Zeno 51, 56127 Pisa, Italy
- CISUP, Centro per l'Integrazione Della Strumentazione Dell'Università di Pisa, 56127 Pisa, Italy
- Centro di Ricerca Interdipartimentale Nutrafood "Nutraceuticals and Food for Health", Università di Pisa, 56126 Pisa, Italy
| | - Rossana Pesi
- Unità di Biochimica, Dipartimento di Biologia, Università di Pisa, Via San Zeno 51, 56127 Pisa, Italy
| | - Giulia Bernardini
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università di Siena, Via A. Moro 2, 53100 Siena, Italy
| | - Vanna Micheli
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università di Siena, Via A. Moro 2, 53100 Siena, Italy
- LND Famiglie Italiane ODV-Via Giovanetti 15-20, 16149 Genova, Italy
| | - Maria Grazia Tozzi
- Unità di Biochimica, Dipartimento di Biologia, Università di Pisa, Via San Zeno 51, 56127 Pisa, Italy
| |
Collapse
|
4
|
Gómez J, Mota-Martorell N, Jové M, Pamplona R, Barja G. Mitochondrial ROS production, oxidative stress and aging within and between species: Evidences and recent advances on this aging effector. Exp Gerontol 2023; 174:112134. [PMID: 36849000 DOI: 10.1016/j.exger.2023.112134] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 02/21/2023] [Accepted: 02/24/2023] [Indexed: 03/01/2023]
Abstract
Mitochondria play a wide diversity of roles in cell physiology and have a key functional implication in cell bioenergetics and biology of free radicals. As the main cellular source of oxygen radicals, mitochondria have been postulated as the mediators of the cellular decline associated with the biological aging. Recent evidences have shown that mitochondrial free radical production is a highly regulated mechanism contributing to the biological determination of longevity which is species-specific. This mitochondrial free radical generation rate induces a diversity of adaptive responses and derived molecular damage to cell components, highlighting mitochondrial DNA damage, with biological consequences that influence the rate of aging of a given animal species. In this review, we explore the idea that mitochondria play a fundamental role in the determination of animal longevity. Once the basic mechanisms are discerned, molecular approaches to counter aging may be designed and developed to prevent or reverse functional decline, and to modify longevity.
Collapse
Affiliation(s)
- José Gómez
- Department of Biology and Geology, Physics and Inorganic Chemistry, ESCET, Rey Juan Carlos University, E28933 Móstoles, Madrid, Spain
| | - Natàlia Mota-Martorell
- Department of Experimental Medicine, University of Lleida (UdL), Lleida Biomedical Research Institute (IRBLleida), E25198 Lleida, Spain
| | - Mariona Jové
- Department of Experimental Medicine, University of Lleida (UdL), Lleida Biomedical Research Institute (IRBLleida), E25198 Lleida, Spain
| | - Reinald Pamplona
- Department of Experimental Medicine, University of Lleida (UdL), Lleida Biomedical Research Institute (IRBLleida), E25198 Lleida, Spain.
| | - Gustavo Barja
- Department of Genetics, Physiology and Microbiology, Faculty of Biological Sciences, Complutense University of Madrid (UCM), E28040 Madrid, Spain.
| |
Collapse
|
5
|
Stange K, Schumacher T, Miersch C, Whelan R, Klünemann M, Röntgen M. Methionine Sources Differently Affect Production of Reactive Oxygen Species, Mitochondrial Bioenergetics, and Growth of Murine and Quail Myoblasts In Vitro. Curr Issues Mol Biol 2023; 45:2661-2680. [PMID: 37185698 PMCID: PMC10136669 DOI: 10.3390/cimb45040174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 03/14/2023] [Accepted: 03/21/2023] [Indexed: 05/17/2023] Open
Abstract
An optimal supply of L-methionine (L-Met) improves muscle growth, whereas over-supplementation exerts adverse effects. To understand the underlying mechanisms, this study aims at exploring effects on the growth, viability, ROS production, and mitochondrial bioenergetics of C2C12 (mouse) and QM7 (quail) myoblasts additionally supplemented (100 or 1000 µM) with L-Met, DL-methionine (DL-Met), or DL-2-hydroxy-4-(methylthio)butanoic acid (DL-HMTBA). In both cell lines, all the supplements stimulated cell growth. However, in contrast to DL-Met, 1000 µM of L-Met (C2C12 cells only) or DL-HMTBA started to retard growth. This negative effect was stronger with DL-HMTBA and was accompanied by significantly elevated levels of extracellular H2O2, an indicator for OS, in both cell types. In addition, oversupplementation with DL-HMTBA (1000 µM) induced adaptive responses in mitochondrial bioenergetics, including reductions in basal (C2C12 and QM7) and ATP-synthase-linked (C2C12) oxygen consumption, maximal respiration rate, and reserve capacity (QM7). Only QM7 cells switched to nonmitochondrial aerobic glycolysis to reduce ROS production. In conclusion, we found a general negative effect of methionine oversupplementation on cell proliferation. However, only DL-HMTBA-induced growth retardation was associated with OS and adaptive, species-specific alterations in mitochondrial functionality. OS could be better compensated by quail cells, highlighting the role of species differences in the ability to cope with methionine oversupplementation.
Collapse
Affiliation(s)
- Katja Stange
- Institute of Muscle Biology and Growth, Research Institute for Farm Animal Biology (FBN), Wilhelm-Stahl-Allee 2, 18196 Dummerstorf, Germany
| | - Toni Schumacher
- Institute of Muscle Biology and Growth, Research Institute for Farm Animal Biology (FBN), Wilhelm-Stahl-Allee 2, 18196 Dummerstorf, Germany
| | - Claudia Miersch
- Institute of Muscle Biology and Growth, Research Institute for Farm Animal Biology (FBN), Wilhelm-Stahl-Allee 2, 18196 Dummerstorf, Germany
- Nutritional Physiology and Dietetics, International University of Applied Sciences (IU), Juri-Gagarin-Ring 152, 99084 Erfurt, Germany
| | - Rose Whelan
- Evonik Operations GmbH, Rodenbacher Chaussee 4, 63457 Hanau, Germany
| | - Martina Klünemann
- Evonik Operations GmbH, Rodenbacher Chaussee 4, 63457 Hanau, Germany
| | - Monika Röntgen
- Institute of Muscle Biology and Growth, Research Institute for Farm Animal Biology (FBN), Wilhelm-Stahl-Allee 2, 18196 Dummerstorf, Germany
| |
Collapse
|
6
|
Gernez E, Deheul S, Tard C, Joncquel M, Douillard C, Grzych G. Plasma Methionine and Clinical Severity in Nitrous Oxide Consumption. TOXICS 2022; 11:12. [PMID: 36668738 PMCID: PMC9866764 DOI: 10.3390/toxics11010012] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 12/15/2022] [Accepted: 12/20/2022] [Indexed: 06/17/2023]
Abstract
In the last few years, there has been an increase in the recreational use of nitrous oxide (N2O), which can lead to neurological symptoms such as sensory or motor disorders. The literature links these symptoms to a functional inactivation of vitamin B12 by oxidation of its cobalt ion, which prevents the vitamin B12 from acting as a cofactor for methionine synthase. Thus, demyelination related to methionine deficiency could be responsible for the neurological disorders associated with N2O consumption, including the combined sclerosis of the spinal cord. We aimed to study the correlation between the plasma methionine levels and clinical severity observed in N2O users. We retrospectively collected clinical and biological data from 93 patients who chronically consumed N2O. The patients were divided into four groups based of the severity of their clinical symptoms (based on their Peripheral Neuropathy Disability (PND) score). The plasma amino acids measurement, including methionine, were performed systematically by liquid chromatography coupled with mass spectrometry. Plasma methionine is significantly correlated with the clinical severity (Spearman coefficient: −0.42; p-value < 10−5), however, the average methionine level in the four groups is within the physiological values (N: 16−23 µmol/L). There is a significant inverse correlation between plasma methionine and homocysteine (Spearman coefficient: −0.57; p-value < 10−9), which confirms the action of nitrous oxide on the methionine synthase. A decrease in plasma methionine cannot be imputed as the only mechanism involved in the pathophysiology of the neurological disorders in nitrous oxide consumption. In addition, there are few therapeutic indications for the use of methionine. Thus, we should be careful concerning the potential use of methionine in nitrous oxide consumption. As a consequence, other pathophysiological mechanisms probably need to be identified in order to find potential therapeutic targets.
Collapse
Affiliation(s)
- Emeline Gernez
- CHU de Lille, Service Hormonologie Métabolisme Nutrition Oncologie, F-59000 Lille, France
| | - Sylvie Deheul
- CHU de Lille, Centre d’Addictovigilance des Hauts-de-France, Service de Pharmacologie, F-59000 Lille, France
| | - Céline Tard
- CHU de Lille, Clinique de Neurologie, F-59000 Lille, France
| | - Marie Joncquel
- CHU de Lille, Service Hormonologie Métabolisme Nutrition Oncologie, F-59000 Lille, France
| | - Claire Douillard
- CHU de Lille, Centre de Référence des Maladies Héréditaires du Métabolisme, F-59000 Lille, France
| | - Guillaume Grzych
- CHU de Lille, Service Hormonologie Métabolisme Nutrition Oncologie, F-59000 Lille, France
| |
Collapse
|
7
|
High dietary methionine intake may contribute to the risk of nonalcoholic fatty liver disease by inhibiting hepatic H2S production. Food Res Int 2022; 158:111507. [DOI: 10.1016/j.foodres.2022.111507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Revised: 06/09/2022] [Accepted: 06/10/2022] [Indexed: 12/06/2022]
|
8
|
Wang D, Ye J, Shi R, Zhao B, Liu Z, Lin W, Liu X. Dietary protein and amino acid restriction: Roles in metabolic health and aging-related diseases. Free Radic Biol Med 2022; 178:226-242. [PMID: 34890767 DOI: 10.1016/j.freeradbiomed.2021.12.009] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 12/03/2021] [Accepted: 12/04/2021] [Indexed: 12/13/2022]
Abstract
The prevalence of obesity is a worldwide phenomenon in all age groups and is associated with aging-related diseases such as type 2 diabetes, as well metabolic and cardiovascular diseases. The use of dietary restriction (DR) while avoiding malnutrition has many profound beneficial effects on aging and metabolic health, and dietary protein or specific amino acid (AA) restrictions, rather than overall calorie intake, are considered to play key roles in the effects of DR on host health. Whereas comprehensive reviews of the underlying mechanisms are limited, protein restriction and methionine (Met) restriction improve metabolic health and aging-related neurodegenerative diseases, and may be associated with FGF21, mTOR and autophagy, improved mitochondrial function and oxidative stress. Circulating branched-chain amino acids (BCAAs) are inversely correlated with metabolic health, and BCAAs and leucine (Leu) restriction promote metabolic homeostasis in rodents. Although tryptophan (Trp) restriction extends the lifespan of rodents, the Trp-restricted diet is reported to increase inflammation in aged mice, while severe Trp restriction has side effects such as anorexia. Furthermore, inadequate protein intake in the elderly increases the risk of muscle-centric health. Therefore, the restriction of specific AAs may be an effective and executable dietary manipulation for metabolic and aging-related health in humans, which warrants further investigation to elucidate the underlying mechanisms.
Collapse
Affiliation(s)
- Danna Wang
- College of Food Science and Engineering, Northwest A&F University, Yangling, China
| | - Jin Ye
- College of Food Science and Engineering, Northwest A&F University, Yangling, China
| | - Renjie Shi
- College of Food Science and Engineering, Northwest A&F University, Yangling, China
| | - Beita Zhao
- College of Food Science and Engineering, Northwest A&F University, Yangling, China
| | - Zhigang Liu
- College of Food Science and Engineering, Northwest A&F University, Yangling, China
| | - Wei Lin
- Department of Neurosurgery, Xijing Institute of Clinical Neuroscience, Xijing Hospital, Air Force Medical University, Xi'an, Shanxi, China.
| | - Xuebo Liu
- College of Food Science and Engineering, Northwest A&F University, Yangling, China.
| |
Collapse
|
9
|
Ewing LE, Skinner CM, Pathak R, Yee EU, Krager K, Gurley PC, Melnyk S, Boerma M, Hauer-Jensen M, Koturbash I. Dietary Methionine Supplementation Exacerbates Gastrointestinal Toxicity in a Mouse Model of Abdominal Irradiation. Int J Radiat Oncol Biol Phys 2021; 109:581-593. [PMID: 33002540 PMCID: PMC7855316 DOI: 10.1016/j.ijrobp.2020.09.042] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 09/10/2020] [Accepted: 09/21/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND AND PURPOSE Identification of appropriate dietary strategies for prevention of weight and muscle loss in cancer patients is crucial for successful treatment and prolonged patient survival. High-protein oral nutritional supplements decrease mortality and improve indices of nutritional status in cancer patients; however, high-protein diets are often rich in methionine, and experimental evidence indicates that a methionine-supplemented diet (MSD) exacerbates gastrointestinal toxicity after total body irradiation. Here, we sought to investigate whether MSD can exacerbate gastrointestinal toxicity after local abdominal irradiation, an exposure regimen more relevant to clinical settings. MATERIALS AND METHODS Male CBA/CaJ mice fed either a methionine-adequate diet or MSD (6.5 mg methionine/kg diet vs 19.5 mg/kg) received localized abdominal X-irradiation (220 kV, 13 mA) using the Small Animal Radiation Research Platform, and tissues were harvested 4, 7, and 10 days after irradiation. RESULTS MSD exacerbated gastrointestinal toxicity after local abdominal irradiation with 12.5 Gy. This was evident as impaired nutrient absorption was paralleled by reduced body weight recovery. Mechanistically, significant shifts in the gut ecology, evident as decreased microbiome diversity, and substantially increased bacterial species that belong to the genus Bacteroides triggered proinflammatory responses. The latter were evident as increases in circulating neutrophils with corresponding decreases in lymphocytes and associated molecular alterations, exhibited as increases in mRNA levels of proinflammatory genes Icam1, Casp1, Cd14, and Myd88. Altered expression of the tight junction-related proteins Cldn2, Cldn5, and Cldn6 indicated a possible increase in intestinal permeability and bacterial translocation to the liver. CONCLUSIONS We report that dietary supplementation with methionine exacerbates gastrointestinal syndrome in locally irradiated mice. This study demonstrates the important roles registered dieticians should play in clinical oncology and further underlines the necessity of preclinical and clinical investigations in the role of diet in the success of cancer therapy.
Collapse
Affiliation(s)
- Laura E Ewing
- Department of Environmental and Occupational Health, University of Arkansas for Medical Sciences, Little Rock, Arkansas; Department of Pharmacology and Toxicology, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Charles M Skinner
- Department of Environmental and Occupational Health, University of Arkansas for Medical Sciences, Little Rock, Arkansas; Center for Dietary Supplements Research, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Rupak Pathak
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Eric U Yee
- Department of Pathology, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Kim Krager
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Patrick C Gurley
- Department of Environmental and Occupational Health, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Stepan Melnyk
- Arkansas Children's Research Institute, Little Rock, Arkansas
| | - Marjan Boerma
- Center for Dietary Supplements Research, University of Arkansas for Medical Sciences, Little Rock, Arkansas; Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Martin Hauer-Jensen
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Igor Koturbash
- Department of Environmental and Occupational Health, University of Arkansas for Medical Sciences, Little Rock, Arkansas; Center for Dietary Supplements Research, University of Arkansas for Medical Sciences, Little Rock, Arkansas.
| |
Collapse
|
10
|
Pamplona R, Jové M, Mota-Martorell N, Barja G. Is the NDUFV2 subunit of the hydrophilic complex I domain a key determinant of animal longevity? FEBS J 2021; 288:6652-6673. [PMID: 33455045 DOI: 10.1111/febs.15714] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 12/02/2020] [Accepted: 01/14/2021] [Indexed: 12/18/2022]
Abstract
Complex I, a component of the electron transport chain, plays a central functional role in cell bioenergetics and the biology of free radicals. The structural and functional N module of complex I is one of the main sites of the generation of free radicals. The NDUFV2 subunit/N1a cluster is a component of this module. Furthermore, the rate of free radical production is linked to animal longevity. In this review, we explore the hypothesis that NDUFV2 is the only conserved core subunit designed with a regulatory function to ensure correct electron transfer and free radical production, that low gene expression and protein abundance of the NDUFV2 subunit is an evolutionary adaptation needed to achieve a longevity phenotype, and that these features are determinants of the lower free radical generation at the mitochondrial level and a slower rate of aging of long-lived animals.
Collapse
Affiliation(s)
- Reinald Pamplona
- Department of Experimental Medicine, University of Lleida-Lleida Biomedical Research Institute (UdL-IRBLleida), Lleida, Spain
| | - Mariona Jové
- Department of Experimental Medicine, University of Lleida-Lleida Biomedical Research Institute (UdL-IRBLleida), Lleida, Spain
| | - Natalia Mota-Martorell
- Department of Experimental Medicine, University of Lleida-Lleida Biomedical Research Institute (UdL-IRBLleida), Lleida, Spain
| | - Gustavo Barja
- Department of Genetics, Physiology and Microbiology, Complutense University of Madrid, Madrid, Spain
| |
Collapse
|
11
|
Becker PH, Demir Z, Mozer Glassberg Y, Sevin C, Habes D, Imbard A, Mussini C, Rozenfeld Bar Lev M, Davit-Spraul A, Benoist JF, Thérond P, Slama A, Jacquemin E, Gonzales E, Gaignard P. Adenosine kinase deficiency: Three new cases and diagnostic value of hypermethioninemia. Mol Genet Metab 2021; 132:38-43. [PMID: 33309011 DOI: 10.1016/j.ymgme.2020.11.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 11/22/2020] [Accepted: 11/22/2020] [Indexed: 12/13/2022]
Abstract
Adenosine kinase (ADK) deficiency is characterized by liver disease, dysmorphic features, epilepsy and developmental delay. This defect disrupts the adenosine/AMP futile cycle and interferes with the upstream methionine cycle. We report the clinical, histological and biochemical courses of three ADK children carrying two new mutations and presenting with neonatal cholestasis and neurological disorders. One of them died of liver failure whereas the other two recovered from their liver damage. As the phenotype was consistent with a mitochondrial disorder, we studied liver mitochondrial respiratory chain activities in two patients and revealed a combined defect of several complexes. In addition, we retrospectively analyzed methionine plasma concentration, a hallmark of ADK deficiency, in a cohort of children and showed that methionine level in patients with ADK deficiency was strongly increased compared with patients with other liver diseases. ADK deficiency is a cause of neonatal or early infantile liver disease that may mimic primary mitochondrial disorders. In this context, an elevation of methionine plasma levels over twice the upper limit should not be considered as a nonspecific finding. ADK deficiency induced-liver dysfunction is most often transient, but could be life-threatening.
Collapse
Affiliation(s)
- Pierre-Hadrien Becker
- Department of Biochemistry, Reference Center for Mitochondrial Disease, FILNEMUS, Bicêtre University Hospital, University of Paris-Saclay, Assistance Publique-Hôpitaux de Paris, Le Kremlin-Bicêtre, France; Lip(Sys)2, University of Paris-Saclay, Chatenay-Malabry, France
| | - Zeynep Demir
- Pediatric Hepatology and Pediatric Liver Transplantation Unit, Reference Center for Mitochondrial Disease, FILNEMUS, Bicêtre University Hospital, University of Paris-Saclay, Assistance Publique-Hôpitaux de Paris, Le Kremlin-Bicêtre, France
| | - Yael Mozer Glassberg
- Institute of Gastroenterology, Nutrition, and Liver Disease, Schneider Children's Medical Center of Israel, Petah Tikva, Israel
| | - Caroline Sevin
- Pediatric Neurology Unit, Reference Center for Mitochondrial Disease, FILNEMUS, Bicêtre University Hospital, University of Paris-Saclay, Assistance Publique-Hôpitaux de Paris, Le Kremlin-Bicêtre, France; INSERM U1127, Institut du Cerveau et de la Moelle épinière (ICM), Pitié-Salpêtrière Hospital, Paris, France
| | - Dalila Habes
- Pediatric Hepatology and Pediatric Liver Transplantation Unit, Reference Center for Mitochondrial Disease, FILNEMUS, Bicêtre University Hospital, University of Paris-Saclay, Assistance Publique-Hôpitaux de Paris, Le Kremlin-Bicêtre, France
| | - Apolline Imbard
- Lip(Sys)2, University of Paris-Saclay, Chatenay-Malabry, France; Department of Metabolic Biochemistry, Necker Enfants Malades Hospital, University of Paris-Descartes, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Charlotte Mussini
- Department of Pathology, Bicêtre University Hospital, University of Paris-Saclay, Assistance Publique-Hôpitaux de Paris, Le Kremlin-Bicêtre, France
| | - Michal Rozenfeld Bar Lev
- Institute of Gastroenterology, Nutrition, and Liver Disease, Schneider Children's Medical Center of Israel, Petah Tikva, Israel
| | - Anne Davit-Spraul
- Department of Biochemistry, Reference Center for Mitochondrial Disease, FILNEMUS, Bicêtre University Hospital, University of Paris-Saclay, Assistance Publique-Hôpitaux de Paris, Le Kremlin-Bicêtre, France; Inserm U1193, Hepatinov, University Paris-Saclay, Orsay, France
| | - Jean-François Benoist
- Lip(Sys)2, University of Paris-Saclay, Chatenay-Malabry, France; Department of Metabolic Biochemistry, Necker Enfants Malades Hospital, University of Paris-Descartes, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Patrice Thérond
- Department of Biochemistry, Reference Center for Mitochondrial Disease, FILNEMUS, Bicêtre University Hospital, University of Paris-Saclay, Assistance Publique-Hôpitaux de Paris, Le Kremlin-Bicêtre, France; Lip(Sys)2, University of Paris-Saclay, Chatenay-Malabry, France
| | - Abdelhamid Slama
- Department of Biochemistry, Reference Center for Mitochondrial Disease, FILNEMUS, Bicêtre University Hospital, University of Paris-Saclay, Assistance Publique-Hôpitaux de Paris, Le Kremlin-Bicêtre, France
| | - Emmanuel Jacquemin
- Pediatric Hepatology and Pediatric Liver Transplantation Unit, Reference Center for Mitochondrial Disease, FILNEMUS, Bicêtre University Hospital, University of Paris-Saclay, Assistance Publique-Hôpitaux de Paris, Le Kremlin-Bicêtre, France; Inserm U1193, Hepatinov, University Paris-Saclay, Orsay, France
| | - Emmanuel Gonzales
- Pediatric Hepatology and Pediatric Liver Transplantation Unit, Reference Center for Mitochondrial Disease, FILNEMUS, Bicêtre University Hospital, University of Paris-Saclay, Assistance Publique-Hôpitaux de Paris, Le Kremlin-Bicêtre, France; Inserm U1193, Hepatinov, University Paris-Saclay, Orsay, France
| | - Pauline Gaignard
- Department of Biochemistry, Reference Center for Mitochondrial Disease, FILNEMUS, Bicêtre University Hospital, University of Paris-Saclay, Assistance Publique-Hôpitaux de Paris, Le Kremlin-Bicêtre, France; Lip(Sys)2, University of Paris-Saclay, Chatenay-Malabry, France.
| |
Collapse
|
12
|
The Advanced Lipoxidation End-Product Malondialdehyde-Lysine in Aging and Longevity. Antioxidants (Basel) 2020; 9:antiox9111132. [PMID: 33203089 PMCID: PMC7696601 DOI: 10.3390/antiox9111132] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Revised: 11/09/2020] [Accepted: 11/12/2020] [Indexed: 01/03/2023] Open
Abstract
The nonenzymatic adduction of malondialdehyde (MDA) to the protein amino groups leads to the formation of malondialdehyde-lysine (MDALys). The degree of unsaturation of biological membranes and the intracellular oxidative conditions are the main factors that modulate MDALys formation. The low concentration of this modification in the different cellular components, found in a wide diversity of tissues and animal species, is indicative of the presence of a complex network of cellular protection mechanisms that avoid its cytotoxic effects. In this review, we will focus on the chemistry of this lipoxidation-derived protein modification, the specificity of MDALys formation in proteins, the methodology used for its detection and quantification, the MDA-lipoxidized proteome, the metabolism of MDA-modified proteins, and the detrimental effects of this protein modification. We also propose that MDALys is an indicator of the rate of aging based on findings which demonstrate that (i) MDALys accumulates in tissues with age, (ii) the lower the concentration of MDALys the greater the longevity of the animal species, and (iii) its concentration is attenuated by anti-aging nutritional and pharmacological interventions.
Collapse
|
13
|
Ahmad MI, Ijaz MU, Hussain M, Haq IU, Zhao D, Li C. High-Fat Proteins Drive Dynamic Changes in Gut Microbiota, Hepatic Metabolome, and Endotoxemia-TLR-4-NFκB-Mediated Inflammation in Mice. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2020; 68:11710-11725. [PMID: 33034193 DOI: 10.1021/acs.jafc.0c02570] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
The responses of gut microbiota to dietary proteins have been studied previously. However, the effects of dietary proteins supplemented with a high-fat diet (HFD) on the metabolite biomarkers associated with non-alcoholic fatty liver disease (NAFLD) are not well understood. To understand the underlying mechanisms, C57BL/6J mice were fed with either a low-fat diet with casein (LFC) or an HFD with casein (HFC), fish (HFF), or mutton proteins (HFM), and their cecal microbiota and liver metabolites were analyzed. At the phylum level, the HFD group had a relatively higher abundance of Firmicutes compared to the LFC-diet group. At the genus level, the HFF-diet group had the highest abundance of Lactobacillus and Akkermansia compared to the HFC- and HFM-diet groups. Furthermore, mice fed with the HFF diet had significantly reduced levels of hepatic metabolites involved in oxidative stress and bile acid metabolism. Thus, meat proteins in HFD interact in the host to create distinct responses in the gut microbiota and its metabolites.
Collapse
Affiliation(s)
- Muhammad Ijaz Ahmad
- Key Laboratory of Meat Processing and Quality Control, MOE; Key Laboratory of Meat Processing, MARA; Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control; College of Food Science and Technology, Nanjing Agricultural University, 210095 Nanjing, China
| | - Muhammad Umair Ijaz
- Key Laboratory of Meat Processing and Quality Control, MOE; Key Laboratory of Meat Processing, MARA; Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control; College of Food Science and Technology, Nanjing Agricultural University, 210095 Nanjing, China
| | - Muzhair Hussain
- Key Laboratory of Meat Processing and Quality Control, MOE; Key Laboratory of Meat Processing, MARA; Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control; College of Food Science and Technology, Nanjing Agricultural University, 210095 Nanjing, China
| | - Ijaz Ul Haq
- Key Laboratory of Meat Processing and Quality Control, MOE; Key Laboratory of Meat Processing, MARA; Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control; College of Food Science and Technology, Nanjing Agricultural University, 210095 Nanjing, China
| | - Di Zhao
- Key Laboratory of Meat Processing and Quality Control, MOE; Key Laboratory of Meat Processing, MARA; Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control; College of Food Science and Technology, Nanjing Agricultural University, 210095 Nanjing, China
| | - Chunbao Li
- Key Laboratory of Meat Processing and Quality Control, MOE; Key Laboratory of Meat Processing, MARA; Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control; College of Food Science and Technology, Nanjing Agricultural University, 210095 Nanjing, China
| |
Collapse
|
14
|
Affiliation(s)
- Michael Hellwig
- Professur für LebensmittechemieTechnische Universität Dresden D-01062 Dresden Deutschland
| |
Collapse
|
15
|
Abstract
Oxidation is one of the deterioration reactions of proteins in food, the importance of which is comparable to others such as Maillard, lipation, or protein-phenol reactions. While research on protein oxidation has led to a precise understanding of the processes and consequences in physiological systems, knowledge about the specific effects of protein oxidation in food or the role of "oxidized" dietary protein for the human body is comparatively scarce. Food protein oxidation can occur during the whole processing axis, from primary production to intestinal digestion. The present review summarizes the current knowledge and mechanisms of food protein oxidation from a chemical, technological, and nutritional-physiological viewpoint and gives a comprehensive classification of the individual reactions. Different analytical approaches are compared, and the relationship between oxidation of food proteins and oxidative stress in vivo is critically evaluated.
Collapse
Affiliation(s)
- Michael Hellwig
- Chair of Food Chemistry, Technische Universität Dresden, D-01062, Dresden, Germany
| |
Collapse
|
16
|
Liang D, Ladva CN, Golan R, Yu T, Walker DI, Sarnat SE, Greenwald R, Uppal K, Tran V, Jones DP, Russell AG, Sarnat JA. Perturbations of the arginine metabolome following exposures to traffic-related air pollution in a panel of commuters with and without asthma. ENVIRONMENT INTERNATIONAL 2019; 127:503-513. [PMID: 30981021 PMCID: PMC6513706 DOI: 10.1016/j.envint.2019.04.003] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Revised: 03/30/2019] [Accepted: 04/01/2019] [Indexed: 05/05/2023]
Abstract
BACKGROUND Mechanisms underlying the effects of traffic-related air pollution on people with asthma remain largely unknown, despite the abundance of observational and controlled studies reporting associations between traffic sources and asthma exacerbation and hospitalizations. OBJECTIVES To identify molecular pathways perturbed following traffic pollution exposures, we analyzed data as part of the Atlanta Commuters Exposure (ACE-2) study, a crossover panel of commuters with and without asthma. METHODS We measured 27 air pollutants and conducted high-resolution metabolomics profiling on blood samples from 45 commuters before and after each exposure session. We evaluated metabolite and metabolic pathway perturbations using an untargeted metabolome-wide association study framework with pathway analyses and chemical annotation. RESULTS Most of the measured pollutants were elevated in highway commutes (p < 0.05). From both negative and positive ionization modes, 17,586 and 9087 metabolic features were extracted from plasma, respectively. 494 and 220 unique features were associated with at least 3 of the 27 exposures, respectively (p < 0.05), after controlling confounders and false discovery rates. Pathway analysis indicated alteration of several inflammatory and oxidative stress related metabolic pathways, including leukotriene, vitamin E, cytochrome P450, and tryptophan metabolism. We identified and annotated 45 unique metabolites enriched in these pathways, including arginine, histidine, and methionine. Most of these metabolites were not only associated with multiple pollutants, but also differentially expressed between participants with and without asthma. The analysis indicated that these metabolites collectively participated in an interrelated molecular network centering on arginine metabolism, underlying the impact of traffic-related pollutants on individuals with asthma. CONCLUSIONS We detected numerous significant metabolic perturbations associated with in-vehicle exposures during commuting and validated metabolites that were closely linked to several inflammatory and redox pathways, elucidating the potential molecular mechanisms of traffic-related air pollution toxicity. These results support future studies of metabolic markers of traffic exposures and the corresponding molecular mechanisms.
Collapse
Affiliation(s)
- Donghai Liang
- Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, USA.
| | - Chandresh N Ladva
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, USA
| | - Rachel Golan
- Department of Public Health, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Tianwei Yu
- Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, USA
| | - Douglas I Walker
- Department of Environmental Medicine & Public Health, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Stefanie E Sarnat
- Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, USA
| | - Roby Greenwald
- Division of Environmental Health, Georgia State University School of Public Health, Atlanta, USA
| | - Karan Uppal
- Clinical Biomarkers Laboratory, Division of Pulmonary, Allergy, and Critical Care Medicine, School of Medicine, Emory University, Atlanta, USA
| | - ViLinh Tran
- Clinical Biomarkers Laboratory, Division of Pulmonary, Allergy, and Critical Care Medicine, School of Medicine, Emory University, Atlanta, USA
| | - Dean P Jones
- Clinical Biomarkers Laboratory, Division of Pulmonary, Allergy, and Critical Care Medicine, School of Medicine, Emory University, Atlanta, USA
| | - Armistead G Russell
- School of Civil and Environmental Engineering, Georgia Institute of Technology, Atlanta, USA
| | - Jeremy A Sarnat
- Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, USA
| |
Collapse
|
17
|
Nuru M, Muradashvili N, Kalani A, Lominadze D, Tyagi N. High methionine, low folate and low vitamin B6/B12 (HM-LF-LV) diet causes neurodegeneration and subsequent short-term memory loss. Metab Brain Dis 2018; 33:1923-1934. [PMID: 30094804 PMCID: PMC6712979 DOI: 10.1007/s11011-018-0298-z] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Accepted: 07/26/2018] [Indexed: 01/03/2023]
Abstract
Methionine is an essential amino acid found in rich quantities in average American diet such as meats, fish and eggs. Excessive consumption of such food often exceeds the normal requirement of the methionine in our body; which found to be related to the development of neurodegenerative disorders. However, the mechanistic pathways of methionine's influence on the brain are unclear. The present study is focus on the effects of high methionine, low folate and low vitamin B6/B12 (HM-LF-LV) diet on the dysfunction of neuronal and vascular specific markers in the brain. C57BL6/J male mice (8-10 week old) were fed with HM-LF-LV diet for a 6 week period. Cognitive function of mice was determine by measuring short-term memory using a Novel Object Recognition test (NORT). Neuronal dysfunction were evaluate by measuring the levels of Neuronal nuclear antigen (NeuN), Neuron-specific-enolase (NSE) and Fluoro-jade C(FJC) fluorescence; while cerebrovascular disruption were evaluate by assessing levels of endothelial junction proteins Vascular Endothelial-Cadherin (VE-Cadherin) and Claudin-5 in harvested brain tissue. Cerebrovascular permeability was assess by evaluating microvascular leakage of fluorescently labeled albumin in vivo. Endothelial and Neuronal Nitric Oxide Synthase (eNOS, nNOS) regulation and vascular inflammation (ICAM: intercellular adhesion molecules) were also evaluate in brain tissue. All assessments were conduct at weekly intervals throughout the study duration. NORT showed a significant temporal decrease in short-term memory of mice fed on HM-LF-LV diet for 6 weeks compared to the wild-type control group. Our experimental data showed that neuronal dysfunction (decreased NeuN levels and increased FJC positive neurons in brain) was more prominent in HM-LF-LV diet fed mice compared to normal diet fed control mice. In experimental mice, cerebrovascular disruption was found to be elevated as evident from increased pial venular permeability (microvascular leakage) and decreased in VE-Cadherin expression compared to control. Slight decrease in nNOS and increase in eNOS in experimental mice suggest a trend towards the decrease in potential for neuronal development due to the long-term HM-LF-LV diet fed. Collectively, our results suggest that a diet containing high methionine, low folate and low vitamin B6/B12 results in increased neuronal degeneration and vascular dysfunction, leading to short-term memory loss. Interestingly, significant neuronal damage precedes vascular dysfunction.
Collapse
Affiliation(s)
- Mohammed Nuru
- Department of Physiology, University of Louisville School of Medicine, Louisville, KY, 40202, USA
| | - Nino Muradashvili
- Department of Physiology, University of Louisville School of Medicine, Louisville, KY, 40202, USA
| | - Anuradha Kalani
- Department of Cardiology, University of Louisville School of Medicine, Louisville, KY, 40202, USA
| | - David Lominadze
- Department of Physiology, University of Louisville School of Medicine, Louisville, KY, 40202, USA
| | - Neetu Tyagi
- Department of Physiology, University of Louisville School of Medicine, Louisville, KY, 40202, USA.
- Department of Physiology, Health Science Center, A-1201, University of Louisville, Louisville, KY, 40202, USA.
| |
Collapse
|
18
|
Wang SY, Cai GY, Chen XM. Energy restriction in renal protection. Br J Nutr 2018; 120:1149-1158. [PMID: 30401006 PMCID: PMC6316363 DOI: 10.1017/s0007114518002684] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2017] [Revised: 07/22/2018] [Accepted: 08/20/2018] [Indexed: 02/06/2023]
Abstract
Energy restriction (ER) has been widely studied as a novel intervention, and its ability to prolong life has been fully demonstrated. For example, ER can significantly extend the lifespans of model flies, worms, rodents and other mammals. The role of ER in renal protection has also been elucidated. In preclinical studies, adjusting total energy intake or consumption of specific nutrients has prophylactic or therapeutic effects on ageing-related kidney disease and acute and chronic kidney injury. Amino acid restriction has gradually attracted attention. ER mimetics have also been studied in depth. The protective mechanisms of ER and ER mimetics for renal injury include increasing AMP-activated protein kinase and sirtuin type 1 (Sirt1) levels and autophagy and reducing mammalian target of rapamycin, inflammation and oxidative stress. However, the renal protective effect of ER has mostly been investigated in rodent models, and the role of ER in patients cannot be determined due to the lack of large randomised controlled trials. To protect the kidney, the mechanism of ER must be thoroughly researched, and more accurate diet or drug interventions need to be identified.
Collapse
Affiliation(s)
| | - Guang-Yan Cai
- State Key Laboratory of Kidney Diseases, Department of Nephrology, National Clinical Research Center for Kidney Diseases, Chinese PLA Institute of Nephrology, Chinese PLA General Hospital, Beijing 100853, People’s Republic of China
| | | |
Collapse
|
19
|
Soda K. Polyamine Metabolism and Gene Methylation in Conjunction with One-Carbon Metabolism. Int J Mol Sci 2018; 19:E3106. [PMID: 30309036 PMCID: PMC6213949 DOI: 10.3390/ijms19103106] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 10/01/2018] [Accepted: 10/05/2018] [Indexed: 02/07/2023] Open
Abstract
Recent investigations have revealed that changes in DNA methylation status play an important role in aging-associated pathologies and lifespan. The methylation of DNA is regulated by DNA methyltransferases (DNMT1, DNMT3a, and DNMT3b) in the presence of S-adenosylmethionine (SAM), which serves as a methyl group donor. Increased availability of SAM enhances DNMT activity, while its metabolites, S-adenosyl-l-homocysteine (SAH) and decarboxylated S-adenosylmethionine (dcSAM), act to inhibit DNMT activity. SAH, which is converted from SAM by adding a methyl group to cytosine residues in DNA, is an intermediate precursor of homocysteine. dcSAM, converted from SAM by the enzymatic activity of adenosylmethionine decarboxylase, provides an aminopropyl group to synthesize the polyamines spermine and spermidine. Increased homocysteine levels are a significant risk factor for the development of a wide range of conditions, including cardiovascular diseases. However, successful homocysteine-lowering treatment by vitamins (B6, B12, and folate) failed to improve these conditions. Long-term increased polyamine intake elevated blood spermine levels and inhibited aging-associated pathologies in mice and humans. Spermine reversed changes (increased dcSAM, decreased DNMT activity, aberrant DNA methylation, and proinflammatory status) induced by the inhibition of ornithine decarboxylase. The relation between polyamine metabolism, one-carbon metabolism, DNA methylation, and the biological mechanism of spermine-induced lifespan extension is discussed.
Collapse
Affiliation(s)
- Kuniyasu Soda
- Cardiovascular Research Institute, Saitama Medical Center, Jichi Medical University, 1-847 Amanuma, Omiya, Saitama-city, Saitama Prefecture 330-8503, Japan.
| |
Collapse
|
20
|
Sahebi Ala F, Hassanabadi A, Golian A. Effects of dietary supplemental methionine source and betaine replacement on the growth performance and activity of mitochondrial respiratory chain enzymes in normal and heat-stressed broiler chickens. J Anim Physiol Anim Nutr (Berl) 2018; 103:87-99. [PMID: 30298681 DOI: 10.1111/jpn.13005] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Revised: 07/29/2018] [Accepted: 09/13/2018] [Indexed: 12/19/2022]
Abstract
This study aimed to evaluate the effects of dietary supplemental methionine (Met) source and betaine (Bet) replacement for Met on performance and activity of mitochondrial respiratory chain enzymes (MRCEs) in normal and heat-stressed broiler chickens. Total of 1,200-day-old Ross 308 chicks were allocated to two houses, each consisted of 12 treatments, five replicates of 10 birds each with 2 × 2×3 × 2 (temperature × Met source × Met level × Bet, respectively) split-plot factorial arrangement. Met level in the basal diets was 70% requirements (Req) that was increased to the requirement or 130% by supplemental dl- or l-Met. Bet was or was not substituted at the rate of 30% supplemental dl- or l-Met. Feed conversion ratio (FCR) in chicks fed 70% l-Met was lower than those fed 70% dl-Met diet during 1-10 days (p = 0.04). Broilers fed diets containing requirement or 130% Met, regardless of its source, showed higher weight gain (WG) than those received 70% Met diet during 11-42 days (p < 0.001). Feed intake (FI) of broilers fed 130% Met diet was decreased compared to other two groups during 11-42 days (p < 0.05). One hundred thirty percent Met requirement diet resulted in lower FCR comparing to other two groups during 11-42 days (p < 0.001). Heat-stressed birds grew less than those under normal condition (p < 0.05). Broilers fed Req Met diet under normal temperature exhibited higher activities of complexes (Cox) I and III (p < 0.05). Cox I activity in heat-stressed birds fed Bet + diet was similar to those fed Bet-diet under normal temperature (p = 0.046). It is concluded that performance and the activities of Cox I and III were increased as the level of Met increased. Bet replacement for 30% supplemental Met resulted in similar consequences comparing to non-Bet replacement diets on performance, but increased the activity of Cox III. l-Met was effective than dl-Met at the cellular level. High ambient temperature depressed performance and MRCE activity.
Collapse
Affiliation(s)
- Fatemeh Sahebi Ala
- Department of Animal Science, Faculty of Agriculture, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Ahmad Hassanabadi
- Department of Animal Science, Faculty of Agriculture, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Abolghasem Golian
- Department of Animal Science, Faculty of Agriculture, Ferdowsi University of Mashhad, Mashhad, Iran
| |
Collapse
|
21
|
Václav R, Valera F. Host preference of a haematophagous avian ectoparasite: a micronutrient supplementation experiment to test an evolutionary trade-off. Biol J Linn Soc Lond 2018. [DOI: 10.1093/biolinnean/bly089] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Affiliation(s)
- Radovan Václav
- Institute of Zoology, Slovak Academy of Sciences, Dúbravská cesta, Bratislava, Slovakia
| | - Francisco Valera
- Estación Experimental de Zonas Áridas (EEZA-CSIC), Ctra. de Sacramento s/n, La Cañada de S. Urbano, Almería, Spain
| |
Collapse
|
22
|
Koturbash I. 2017 Michael Fry Award Lecture When DNA is Actually Not a Target: Radiation Epigenetics as a Tool to Understand and Control Cellular Response to Ionizing Radiation. Radiat Res 2018; 190:5-11. [PMID: 29697303 PMCID: PMC6036898 DOI: 10.1667/rr15027.1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Aside from the generally accepted potential to cause DNA damage, it is becoming increasingly recognized that ionizing radiation has the capability to target the cellular epigenome. Epigenetics unifies the chemical marks and molecules that collectively facilitate the proper reading of genetic material. Among the epigenetic mechanisms of regulation, methylation of DNA is known to be the key player in the postirradiation response by controlling the expression of genetic information and activity of transposable elements. Radiation-induced alterations to DNA methylation may lead to cellular epigenetic reprogramming that, in turn, can substantially compromise the genomic integrity and has been proposed as one of the mechanisms of radiation-induced carcinogenesis. DNA methylation is strongly dependent on the one-carbon metabolism. This metabolic pathway is central to the support of DNA methylation by means of providing the donor of methyl groups, as well as for the synthesis of amino acids. To better understand the mechanisms of radiation-induced health effects, we study how exposure to radiation affects DNA methylation and one-carbon metabolism. Also, a tight interaction that exists between DNA methylation and one-carbon metabolism allows us to simultaneously manipulate both cellular epigenetic and metabolic profiles to modulate the normal and cancerous tissue response to radiotherapy.
Collapse
Affiliation(s)
- Igor Koturbash
- Department of Environmental and Occupational Health, Fay W. Boozman College of Public Health, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| |
Collapse
|
23
|
Miousse IR, Pathak R, Garg S, Skinner CM, Melnyk S, Pavliv O, Hendrickson H, Landes RD, Lumen A, Tackett AJ, Deutz NE, Hauer-Jensen M, Koturbash I. Short-term dietary methionine supplementation affects one-carbon metabolism and DNA methylation in the mouse gut and leads to altered microbiome profiles, barrier function, gene expression and histomorphology. GENES & NUTRITION 2017; 12:22. [PMID: 28904640 PMCID: PMC5588631 DOI: 10.1186/s12263-017-0576-0] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Accepted: 08/28/2017] [Indexed: 12/24/2022]
Abstract
BACKGROUND Methionine, a central molecule in one-carbon metabolism, is an essential amino acid required for normal growth and development. Despite its importance to biological systems, methionine is toxic when administered at supra-physiological levels. The aim of this study was to investigate the effects of short-term methionine dietary modulation on the proximal jejunum, the section of the gut specifically responsible for amino acid absorption, in a mouse model. Eight-week-old CBA/J male mice were fed methionine-adequate (MAD; 6.5 g/kg) or methionine-supplemented (MSD; 19.5 g/kg) diets for 3.5 or 6 days (average food intake 100 g/kg body weight). The study design was developed in order to address the short-term effects of the methionine supplementation that corresponds to methionine dietary intake in Western populations. Biochemical indices in the blood as well as metabolic, epigenetic, transcriptomic, metagenomic, and histomorphological parameters in the gut were evaluated. RESULTS By day 6, feeding mice with MSD (protein intake <10% different from MAD) resulted in increased plasma (2.3-fold; p < 0.054), but decreased proximal jejunum methionine concentrations (2.2-fold; p < 0.05) independently of the expression of neutral amino acid transporters. MSD has also caused small bowel bacteria colonization, increased the abundance of pathogenic bacterial species Burkholderiales and decreased the gene expression of the intestinal transmembrane proteins-Cldn8 (0.18-fold, p < 0.05), Cldn9 (0.24-fold, p < 0.01) and Cldn10 (0.05-fold, p < 0.05). Feeding MSD led to substantial histomorphological alterations in the proximal jejunum exhibited as a trend towards decreased plasma citrulline concentrations (1.8-fold, p < 0.07), as well as loss of crypt depth (by 28%, p < 0.05) and mucosal surface (by 20%, p < 0.001). CONCLUSIONS Together, these changes indicate that short-term feeding of MSD substantially alters the normal gut physiology. These effects may contribute to the pathogenesis of intestinal inflammatory diseases and/or sensitize the gut to exposure to other stressors.
Collapse
Affiliation(s)
- Isabelle R. Miousse
- Department of Environmental and Occupational Health, University of Arkansas for Medical Sciences, 4301 W. Markham Str., Slot 820-11, Little Rock, AR 72205-7199 USA
| | - Rupak Pathak
- Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205 USA
| | - Sarita Garg
- Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205 USA
| | - Charles M. Skinner
- Department of Environmental and Occupational Health, University of Arkansas for Medical Sciences, 4301 W. Markham Str., Slot 820-11, Little Rock, AR 72205-7199 USA
| | - Stepan Melnyk
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72205 USA
| | - Oleksandra Pavliv
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72205 USA
| | - Howard Hendrickson
- Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205 USA
| | - Reid D. Landes
- Department of Biostatistics, University of Arkansas for Medical Sciences, Little Rock, AR 72205 USA
| | - Annie Lumen
- Division of Biochemical Toxicology, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR USA
| | - Alan J. Tackett
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72205 USA
- Department of Biochemistry, University of Arkansas for Medical Sciences, Little Rock, AR USA
| | - Nicolaas E.P. Deutz
- Department of Health and Kinesiology, Center for Translational Research on Aging and Longevity, Texas A&M University, College Station, TX USA
| | - Martin Hauer-Jensen
- Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205 USA
| | - Igor Koturbash
- Department of Environmental and Occupational Health, University of Arkansas for Medical Sciences, 4301 W. Markham Str., Slot 820-11, Little Rock, AR 72205-7199 USA
| |
Collapse
|
24
|
Mastrototaro L, Sponder G, Saremi B, Aschenbach JR. Gastrointestinal methionine shuttle: Priority handling of precious goods. IUBMB Life 2016; 68:924-934. [PMID: 27753190 DOI: 10.1002/iub.1571] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Accepted: 09/22/2016] [Indexed: 01/05/2023]
Affiliation(s)
- Lucia Mastrototaro
- Institute of Veterinary Physiology, Department of Veterinary Medicine, Free University of Berlin; Berlin Germany
| | - Gerhard Sponder
- Institute of Veterinary Physiology, Department of Veterinary Medicine, Free University of Berlin; Berlin Germany
| | - Behnam Saremi
- Evonik Nutrition & Care GmbH; Animal Nutrition-Animal Nutrition Services; Hanau Germany
| | - Jörg R. Aschenbach
- Institute of Veterinary Physiology, Department of Veterinary Medicine, Free University of Berlin; Berlin Germany
| |
Collapse
|
25
|
Mechanistic basis of hypermethioninemia. Amino Acids 2016; 48:2479-2489. [PMID: 27465642 DOI: 10.1007/s00726-016-2302-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Accepted: 07/19/2016] [Indexed: 02/07/2023]
Abstract
Hypermethioninemia is a condition defined as elevated plasma methionine levels and may be a consequence of different conditions that include non-genetic and genetic causes. In severe cases, hypermethioninemia may lead to development of neurological and hepatic impairments, but mechanisms are still not well elucidated. Therefore, this review aims to reunite the knowledge acquired about the methionine-induced brain and liver toxicity focusing on the results obtained by studies from patients, in vitro experiments, and in vivo animal models. In general, some studies have shown that methionine decreases Na+,K+-ATPase activity, induces oxidative stress, increases acetylcholinesterase activity, and leads to dendritic spine downregulation in brain. Concerning to liver, hypermethioninemia seems to provoke changes in cell morphology, lipid accumulation, oxidative stress, inflammation, and ATP depletion. It is possible to infer that oxidative damage is one of the most important mechanisms responsible for methionine toxicity, since different studies showed that this amino acid induces oxidative stress in brain and liver tissues. Besides, reactive oxygen species may mediate other alterations induced by methionine, such as the reduction in brain Na+,K+-ATPase activity, and liver inflammation.
Collapse
|
26
|
Karunasinghe N, Zhu S, Ferguson LR. Benefits of Selenium Supplementation on Leukocyte DNA Integrity Interact with Dietary Micronutrients: A Short Communication. Nutrients 2016; 8:E249. [PMID: 27128937 PMCID: PMC4882662 DOI: 10.3390/nu8050249] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Revised: 04/19/2016] [Accepted: 04/21/2016] [Indexed: 01/05/2023] Open
Abstract
A male cohort from New Zealand has previously shown variability in Selenium (Se) supplementation effects on measured biomarkers. The current analysis is to understand the reasons for variability of the H₂O₂-induced DNA damage recorded after Se supplementation. We have looked at the variation of demographic, lifestyle, medication, genetic and dietary factors and biomarkers measured at baseline and post-supplementation in these two extreme subgroups A and B. Group A showed increased H₂O₂-induced DNA damage and group B showed decreased damage after Se supplementation. We have also considered correlations of biomarkers and dietary factors in the complete dataset. The glutathione peroxidase (GPx) activity and DNA damage were significantly lower at post-supplementation in Group B compared to Group A. Post-supplementation, Group B showed a significant reduction in the GPx activity, while Group A showed a significant increase in DNA damage compared to baseline levels. Dietary methionine intake was significantly higher and folate intake was significantly lower in Group B compared to Group A. Se supplementation significantly increased the caspase-cleaved keratin 18 levels in both groups, indicating increased apoptotic potential of this supplement. Parameter correlation with the complete dataset showed dietary methionine to have a significant negative correlation with H₂O₂-induced DNA damage post-supplementation. The data suggest that Se supplementation is beneficial for the leukocyte DNA integrity only in interaction with the dietary methionine and folate intake.
Collapse
Affiliation(s)
- Nishi Karunasinghe
- Auckland Cancer Society Research Centre, Faculty of Medical and Health Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand.
| | - Shuotun Zhu
- Discipline of Nutrition, Faculty of Medical and Health Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand.
| | - Lynnette R Ferguson
- Auckland Cancer Society Research Centre, Faculty of Medical and Health Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand.
- Discipline of Nutrition, Faculty of Medical and Health Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand.
| |
Collapse
|
27
|
Chaturvedi P, Kamat PK, Kalani A, Familtseva A, Tyagi SC. High Methionine Diet Poses Cardiac Threat: A Molecular Insight. J Cell Physiol 2016; 231:1554-61. [PMID: 26565991 DOI: 10.1002/jcp.25247] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Accepted: 11/10/2015] [Indexed: 12/21/2022]
Abstract
High methionine diet (HMD) for example red meat which includes lamb, beef, pork can pose cardiac threat and vascular dysfunction but the mechanisms are unclear. We hypothesize that a diet rich in methionine can malfunction the cardiovascular system in three ways: (1) by augmenting oxidative stress; (2) by inflammatory manifestations; and (3) by matrix/vascular remodeling. To test this hypothesis we used four groups of mice: (1) WT; (2) WT + methionine; (3) CBS(+/-) ; (4) CBS(+/-) +methionine. We observed high oxidative stress in mice fed with methionine which was even higher in CBS(+/-) and CBS(+/-) +methionine. Higher oxidative stress was indicated by high levels of SOD-1 in methionine fed mouse hearts whereas IL-1β, IL-6, TNFα, and TLR4 showed high inflammatory manifestations. The upregulated levels of eNOS/iNOS and upregulated levels of MMP2/MMP9 along with high collagen deposition indicated vascular and matrix remodeling in methionine fed mouse. We evaluated the cardiac function which was dysregulated in the mice fed with HMD. These mice had decreased ejection fraction and left ventricular dysfunction which subsequently leads to adverse cardiac remodeling. In conclusion, our study clearly shows that HMD poses a cardiac threat by increasing oxidative stress, inflammatory manifestations, matrix/vascular remodeling, and decreased cardiac function.
Collapse
Affiliation(s)
- Pankaj Chaturvedi
- Department of Physiology, School of Medicine, University of Louisville, Louisville, Kentucky
| | - Pradip K Kamat
- Department of Anesthesiology, University of Florida, Gainesville, Florida
| | - Anuradha Kalani
- Department of Physiology, School of Medicine, University of Louisville, Louisville, Kentucky
| | - Anastasia Familtseva
- Department of Physiology, School of Medicine, University of Louisville, Louisville, Kentucky
| | - Suresh C Tyagi
- Department of Physiology, School of Medicine, University of Louisville, Louisville, Kentucky
| |
Collapse
|
28
|
Stender S, Chakrabarti RS, Xing C, Gotway G, Cohen JC, Hobbs HH. Adult-onset liver disease and hepatocellular carcinoma in S-adenosylhomocysteine hydrolase deficiency. Mol Genet Metab 2015; 116:269-74. [PMID: 26527160 PMCID: PMC4733618 DOI: 10.1016/j.ymgme.2015.10.009] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Revised: 10/23/2015] [Accepted: 10/23/2015] [Indexed: 10/22/2022]
Abstract
BACKGROUND The etiology of liver disease remains elusive in some adults presenting with severe hepatic dysfunction. METHODS AND RESULTS Here we describe a woman of Pakistani descent who had elevated aminotransferases at age 23. She developed muscle weakness in her mid-20s, and was diagnosed with hepatocellular carcinoma at age 29. She died without a diagnosis at age 32 after having a liver transplant. Exome sequencing revealed that she was homozygous for a missense mutation (R49H) in AHCY, the gene encoding S-adenosylhomocysteine (SAH) hydrolase. SAH hydrolase catalyzes the final step in conversion of methionine to homocysteine and inactivating mutations in this enzyme cause a rare autosomal recessive disorder, SAH hydrolase deficiency, that typically presents in infancy. An asymptomatic 7-year old son of the proband is also homozygous for the AHCY-R49H mutation and has elevated serum aminotransferase levels, as well as markedly elevated serum levels of SAH, S-adenosylmethionine (SAM), and methionine, which are hallmarks of SAH hydrolase deficiency. CONCLUSION This report reveals several new aspects of SAH hydrolase deficiency. Affected women with SAH hydrolase deficiency can give birth to healthy children. SAH hydrolase deficiency can remain asymptomatic in childhood, and the disorder can be associated with early onset hepatocellular carcinoma. The measurement of serum amino acids should be considered in patients with liver disease or hepatocellular carcinoma of unknown etiology.
Collapse
Affiliation(s)
- Stefan Stender
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX, United States; McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, TX, United States.
| | - Rima S Chakrabarti
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX, United States; Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX, United States.
| | - Chao Xing
- McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, TX, United States.
| | - Garrett Gotway
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, United States.
| | - Jonathan C Cohen
- McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, TX, United States.
| | - Helen H Hobbs
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX, United States; McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, TX, United States; Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX, United States.
| |
Collapse
|
29
|
Lai YS, Chen WC, Kuo TC, Ho CT, Kuo CH, Tseng YJ, Lu KH, Lin SH, Panyod S, Sheen LY. Mass-Spectrometry-Based Serum Metabolomics of a C57BL/6J Mouse Model of High-Fat-Diet-Induced Non-alcoholic Fatty Liver Disease Development. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2015; 63:7873-7884. [PMID: 26262841 DOI: 10.1021/acs.jafc.5b02830] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Obesity, dyslipidemia, insulin resistance, oxidative stress, and inflammation are key clinical risk factors for the progression of non-alcoholic fatty liver disease (NAFLD). Currently, there is no comprehensive metabolic profile of a well-established animal model that effectively mimics the etiology and pathogenesis of NAFLD in humans. Here, we report the pathophysiological and metabolomic changes associated with NAFLD development in a C57BL/6J mouse model in which NAFLD was induced by feeding a high-fat diet (HFD) for 4, 8, 12, and 16 weeks. Serum metabolomic analysis was conducted using ultrahigh-performance liquid chromatography coupled with quadrupole time-of-flight mass spectrometry (UHPLC-QTOF-MS) and gas chromatography-mass spectrometry (GC-MS) to establish a metabolomic profile. Analysis of the metabolomic profile in combination with principal component analysis revealed marked differences in metabolites between the control and HFD group depending upon NAFLD severity. A total of 30 potential biomarkers were strongly associated with the development of NAFLD. Among these, 11 metabolites were mainly related to carbohydrate metabolism, hepatic biotransformation, collagen synthesis, and gut microbial metabolism, which are characteristics of obesity, as well as significantly increased serum glucose, total cholesterol, and hepatic triglyceride levels during the onset of NAFLD (4 weeks). At 8 weeks, 5 additional metabolites that are chiefly involved in perturbation of lipid metabolism and insulin secretion were found to be associated with hyperinsulinemia, hyperlipidemia, and hepatic steatosis in the mid-term of NAFLD progression. At the end of 12 and 16 weeks, 14 additional metabolites were predominantly correlated to abnormal bile acid synthesis, oxidative stress, and inflammation, representing hepatic inflammatory infiltration during NAFLD development. These results provide potential biomarkers for early risk assessment of NAFLD and further insights into NAFLD development.
Collapse
Affiliation(s)
| | | | | | - Chi-Tang Ho
- Department of Food Science, Rutgers University , New Brunswick, New Jersey 08901, United States
| | | | | | | | | | | | | |
Collapse
|
30
|
Barja G. The mitochondrial free radical theory of aging. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2015; 127:1-27. [PMID: 25149212 DOI: 10.1016/b978-0-12-394625-6.00001-5] [Citation(s) in RCA: 132] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The mitochondrial free radical theory of aging is reviewed. Only two parameters currently correlate with species longevity in the right sense: the mitochondrial rate of reactive oxygen species (mitROS) production and the degree of fatty acid unsaturation of tissue membranes. Both are low in long-lived animals. In addition, the best-known manipulation that extends longevity, dietary restriction, also decreases the rate of mitROS production and oxidative damage to mtDNA. The same occurs during protein restriction as well as during methionine restriction. These two manipulations also increase maximum longevity in rodents. The decrease in mitROS generation and oxidative stress that takes place in caloric restriction seems to be due to restriction of a single dietary substance: methionine. The information available supports a mitochondrial free radical theory of aging focused on low generation of endogenous damage and low sensitivity of membranes to oxidation in long-lived animals.
Collapse
Affiliation(s)
- Gustavo Barja
- Department of Animal Physiology II, Faculty of Biological Sciences, Complutense University, Madrid Spain
| |
Collapse
|
31
|
Badás EP, Martínez J, Rivero de Aguilar Cachafeiro J, Miranda F, Figuerola J, Merino S. Ageing and reproduction: antioxidant supplementation alleviates telomere loss in wild birds. J Evol Biol 2015; 28:896-905. [PMID: 25758014 DOI: 10.1111/jeb.12615] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Revised: 02/26/2015] [Accepted: 03/09/2015] [Indexed: 01/10/2023]
Abstract
Reproduction is inherently costly. Environmental stressors, such as infection and limited food resources, can compromise investment at each breeding attempt. For example, recent data on captive birds showed that increased reproductive effort accelerates ageing. However, the effects of nutritional status and infection on ageing remain unknown. Telomeres function as protective caps at the ends of eukaryotic chromosomes, and changes in telomere length is a commonly used proxy for ageing. To partially address the mechanisms of ageing following reproduction, we supplemented, medicated or administered a combined treatment to wild blue tits (Cyanistes caeruleus) breeding in central Spain during 2012. The nutritional supplement consisted of two different antioxidants, whereas the medication was an antimalarial treatment against blood parasites. We evaluated the effect of these manipulations on reproductive success and parasite loads in the first breeding season, and on changes in telomere length between two consecutive breeding seasons. Supplemented birds showed no reduction in blood parasite infections in 2012, although they exhibited higher body mass and fledging success. The antimalarial drugs reduced infections by several parasite species, but this had no effect on fitness parameters. In the following season, telomeres from supplemented birds had shortened less. Altogether, we found that supplementation with antioxidants provided fitness benefits in the short term and reduced telomere loss a year following treatment. Our results provide indirect empirical support for accelerated telomere loss as a cost of reproduction.
Collapse
Affiliation(s)
- E P Badás
- Department of Evolutionary Ecology, National Museum of Natural Sciences, Madrid, Spain
| | | | | | | | | | | |
Collapse
|
32
|
Dietary L-methionine restriction decreases oxidative stress in porcine liver mitochondria. Exp Gerontol 2015; 65:35-41. [PMID: 25765145 DOI: 10.1016/j.exger.2015.03.004] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2014] [Revised: 03/06/2015] [Accepted: 03/07/2015] [Indexed: 11/23/2022]
Abstract
Dietary methionine restriction (MetR) has been reported to improve hepatocyte function in mammals. However, the underlying mechanisms remain largely unknown. This study was conducted with a swine model to test the hypothesis that MetR decreases generation of reactive oxygen species (ROS) and attenuates oxidative damage in hepatic mitochondria. Twenty-four 35-day old pigs were fed a control diet or a Met-restricted diet for two weeks. Liver mitochondria were isolated to determine: 8-oxodG in mitochondrial DNA, oxidative-derived proteins markers, including glutamic semialdehyde (GSA), aminoadipic semialdehydes (AASA), carboxyethyl-lysine (CEL), carboxymethyl-lysine (CML), and malondialdehyde lysine (MDAL), mitochondrial H2O2 generation rate; rates of oxygen consumption; free radical leak (FRL); anti-oxidative capacity, electron transport complex activity; and protein abundances of respiratory chain complex subunits (NDUFA9, SDHA, Core 2, and Cox 1), manganese superoxide dismutase (MnSOD), and apoptosis-inducing factor (AIF). Compared with the control, MetR decreased mitochondrial 8-oxodG content, H2O2 generation, FRL (P<0.05), and increased rates of oxygen consumption. Abundances of markers for protein oxidative damage, including GSA, AASA, CEL, and CML, were decreased (P<0.05) by 40%, 30%, 32%, and 28%, respectively, compared with the control. Western blot analysis revealed that MetR decreased (P<0.05) the protein abundances of complex subunits, NDUFA9 and AIF without affecting expression of SDHA, Core 2, Cox 1 or MnSOD. The complex I activity (P<0.05) were lowered in MetR group as compared with that of control. Collectively, our findings indicate that dietary MetR decreases mitochondrial ROS generation primarily via inhibiting complex I activity and ROS generation rather than augmenting anti-oxidative capacity, thereby ameliorating oxidative damage to hepatic mitochondrial DNA and proteins.
Collapse
|
33
|
Brown-Borg HM, Rakoczy SG, Wonderlich JA, Rojanathammanee L, Kopchick JJ, Armstrong V, Raasakka D. Growth hormone signaling is necessary for lifespan extension by dietary methionine. Aging Cell 2014; 13:1019-27. [PMID: 25234161 PMCID: PMC4244257 DOI: 10.1111/acel.12269] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/02/2014] [Indexed: 11/29/2022] Open
Abstract
Growth hormone significantly impacts lifespan in mammals. Mouse longevity is extended when growth hormone (GH) signaling is interrupted but markedly shortened with high-plasma hormone levels. Methionine metabolism is enhanced in growth hormone deficiency, for example, in the Ames dwarf, but suppressed in GH transgenic mice. Methionine intake affects also lifespan, and thus, GH mutant mice and respective wild-type littermates were fed 0.16%, 0.43%, or 1.3% methionine to evaluate the interaction between hormone status and methionine. All wild-type and GH transgenic mice lived longer when fed 0.16% methionine but not when fed higher levels. In contrast, animals without growth hormone signaling due to hormone deficiency or resistance did not respond to altered levels of methionine in terms of lifespan, body weight, or food consumption. Taken together, our results suggest that the presence of growth hormone is necessary to sense dietary methionine changes, thus strongly linking growth and lifespan to amino acid availability.
Collapse
Affiliation(s)
- Holly M Brown-Borg
- Department of Basic Sciences, University of North Dakota School of Medicine and Health SciencesGrand Forks, ND, 58203, USA
| | - Sharlene G Rakoczy
- Department of Basic Sciences, University of North Dakota School of Medicine and Health SciencesGrand Forks, ND, 58203, USA
| | - Joseph A Wonderlich
- Department of Basic Sciences, University of North Dakota School of Medicine and Health SciencesGrand Forks, ND, 58203, USA
| | - Lalida Rojanathammanee
- Department of Basic Sciences, University of North Dakota School of Medicine and Health SciencesGrand Forks, ND, 58203, USA
| | - John J Kopchick
- Department of Biomedical Sciences Heritage College of Osteopathic Medicine, Edison Biotechnology Institute, Ohio UniversityAthens, OH, 45701, USA
| | - Vanessa Armstrong
- Department of Basic Sciences, University of North Dakota School of Medicine and Health SciencesGrand Forks, ND, 58203, USA
| | - Debbie Raasakka
- Department of Basic Sciences, University of North Dakota School of Medicine and Health SciencesGrand Forks, ND, 58203, USA
| |
Collapse
|
34
|
Schwarzer M, Osterholt M, Lunkenbein A, Schrepper A, Amorim P, Doenst T. Mitochondrial reactive oxygen species production and respiratory complex activity in rats with pressure overload-induced heart failure. J Physiol 2014; 592:3767-82. [PMID: 24951621 DOI: 10.1113/jphysiol.2014.274704] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
We investigated the impact of cardiac reactive oxygen species (ROS) during the development of pressure overload-induced heart failure. We used our previously described rat model where transverse aortic constriction (TAC) induces compensated hypertrophy after 2 weeks, heart failure with preserved ejection fraction at 6 and 10 weeks, and heart failure with systolic dysfunction after 20 weeks. We measured mitochondrial ROS production rates, ROS damage and assessed the therapeutic potential of in vivo antioxidant therapies. In compensated hypertrophy (2 weeks of TAC) ROS production rates were normal at both mitochondrial ROS production sites (complexes I and III). Complex I ROS production rates increased with the appearance of diastolic dysfunction (6 weeks of TAC) and remained high thereafter. Surprisingly, maximal ROS production at complex III peaked at 6 weeks of pressure overload. Mitochondrial respiratory capacity (state 3 respiration) was elevated 2 and 6 weeks after TAC, decreased after this point and was significantly impaired at 20 weeks, when contractile function was also impaired and ROS damage was found with increased hydroxynonenal. Treatment with the ROS scavenger α-phenyl-N-tert-butyl nitrone or the uncoupling agent dinitrophenol significantly reduced ROS production rates at 6 weeks. Despite the decline in ROS production capacity, no differences in contractile function between treated and untreated animals were observed. Increased ROS production occurs early in the development of heart failure with a peak at the onset of diastolic dysfunction. However, ROS production may not be related to the onset of contractile dysfunction.
Collapse
Affiliation(s)
- Michael Schwarzer
- Department of Cardiothoracic Surgery, Jena University Hospital Friedrich Schiller University of Jena, Jena, Germany
| | - Moritz Osterholt
- Department of Cardiothoracic Surgery, Jena University Hospital Friedrich Schiller University of Jena, Jena, Germany
| | - Anne Lunkenbein
- Department of Cardiac Surgery, University of Leipzig Heart Center, Leipzig, Germany
| | - Andrea Schrepper
- Department of Cardiothoracic Surgery, Jena University Hospital Friedrich Schiller University of Jena, Jena, Germany
| | - Paulo Amorim
- Department of Cardiothoracic Surgery, Jena University Hospital Friedrich Schiller University of Jena, Jena, Germany
| | - Torsten Doenst
- Department of Cardiothoracic Surgery, Jena University Hospital Friedrich Schiller University of Jena, Jena, Germany
| |
Collapse
|
35
|
Ruckenstuhl C, Netzberger C, Entfellner I, Carmona-Gutierrez D, Kickenweiz T, Stekovic S, Gleixner C, Schmid C, Klug L, Sorgo AG, Eisenberg T, Büttner S, Mariño G, Koziel R, Jansen-Dürr P, Fröhlich KU, Kroemer G, Madeo F. Lifespan extension by methionine restriction requires autophagy-dependent vacuolar acidification. PLoS Genet 2014; 10:e1004347. [PMID: 24785424 PMCID: PMC4006742 DOI: 10.1371/journal.pgen.1004347] [Citation(s) in RCA: 171] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2013] [Accepted: 03/19/2014] [Indexed: 11/19/2022] Open
Abstract
Reduced supply of the amino acid methionine increases longevity across species through an as yet elusive mechanism. Here, we report that methionine restriction (MetR) extends yeast chronological lifespan in an autophagy-dependent manner. Single deletion of several genes essential for autophagy (ATG5, ATG7 or ATG8) fully abolished the longevity-enhancing capacity of MetR. While pharmacological or genetic inhibition of TOR1 increased lifespan in methionine-prototroph yeast, TOR1 suppression failed to extend the longevity of methionine-restricted yeast cells. Notably, vacuole-acidity was specifically enhanced by MetR, a phenotype that essentially required autophagy. Overexpression of vacuolar ATPase components (Vma1p or Vph2p) suffices to increase chronological lifespan of methionine-prototrophic yeast. In contrast, lifespan extension upon MetR was prevented by inhibition of vacuolar acidity upon disruption of the vacuolar ATPase. In conclusion, autophagy promotes lifespan extension upon MetR and requires the subsequent stimulation of vacuolar acidification, while it is epistatic to the equally autophagy-dependent anti-aging pathway triggered by TOR1 inhibition or deletion.
Collapse
Affiliation(s)
| | | | - Iryna Entfellner
- Institute for Molecular Biosciences, University of Graz, Graz, Austria
| | | | - Thomas Kickenweiz
- Institute for Molecular Biosciences, University of Graz, Graz, Austria
| | - Slaven Stekovic
- Institute for Molecular Biosciences, University of Graz, Graz, Austria
| | | | - Christian Schmid
- Institute for Molecular Biosciences, University of Graz, Graz, Austria
| | - Lisa Klug
- Institute for Molecular Biosciences, University of Graz, Graz, Austria
| | - Alice G. Sorgo
- Institute for Molecular Biosciences, University of Graz, Graz, Austria
| | - Tobias Eisenberg
- Institute for Molecular Biosciences, University of Graz, Graz, Austria
| | - Sabrina Büttner
- Institute for Molecular Biosciences, University of Graz, Graz, Austria
| | - Guillermo Mariño
- INSERM, U848, Villejuif, France
- Institut Gustave Roussy, Villejuif, France
- Université Paris Sud, Paris 11, Villejuif, France
| | - Rafal Koziel
- Institute for Biomedical Aging Research (IBA), Austrian Academy of Sciences, Innsbruck, Austria
| | - Pidder Jansen-Dürr
- Institute for Biomedical Aging Research (IBA), Austrian Academy of Sciences, Innsbruck, Austria
| | - Kai-Uwe Fröhlich
- Institute for Molecular Biosciences, University of Graz, Graz, Austria
| | - Guido Kroemer
- INSERM, U848, Villejuif, France
- Institut Gustave Roussy, Villejuif, France
- Metabolomics Platform, Institut Gustave Roussy, Villejuif, France
- Centre de Recherche des Cordeliers, Paris, France
- Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, Paris, France
- Université Paris Descartes, Paris 5, Paris, France
| | - Frank Madeo
- Institute for Molecular Biosciences, University of Graz, Graz, Austria
| |
Collapse
|
36
|
Spencer J, Phister TG, Smart KA, Greetham D. Tolerance of pentose utilising yeast to hydrogen peroxide-induced oxidative stress. BMC Res Notes 2014; 7:151. [PMID: 24636079 PMCID: PMC4004043 DOI: 10.1186/1756-0500-7-151] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2013] [Accepted: 03/11/2014] [Indexed: 11/27/2022] Open
Abstract
Background Bioethanol fermentations follow traditional beverage fermentations where the yeast is exposed to adverse conditions such as oxidative stress. Lignocellulosic bioethanol fermentations involve the conversion of pentose and hexose sugars into ethanol. Environmental stress conditions such as osmotic stress and ethanol stress may affect the fermentation performance; however, oxidative stress as a consequence of metabolic output can also occur. However, the effect of oxidative stress on yeast with pentose utilising capabilities has yet to be investigated. Results Assaying for the effect of hydrogen peroxide-induced oxidative stress on Candida, Pichia and Scheffersomyces spp. has demonstrated that these yeast tolerate hydrogen peroxide-induced oxidative stress in a manner consistent with that demonstrated by Saccharomyces cerevisiae. Pichia guillermondii appears to be more tolerant to hydrogen peroxide-induced oxidative stress when compared to Candida shehatae, Candida succiphila or Scheffersomyces stipitis. Conclusions Sensitivity to hydrogen peroxide-induced oxidative stress increased in the presence of minimal media; however, addition of amino acids and nucleobases was observed to increase tolerance. In particular adenine increased tolerance and methionine reduced tolerance to hydrogen peroxide-induced oxidative stress.
Collapse
Affiliation(s)
| | | | | | - Darren Greetham
- School of Biosciences, University of Nottingham, Loughborough, Leics LE12 5RD, UK.
| |
Collapse
|
37
|
Maddineni S, Nichenametla S, Sinha R, Wilson RP, Richie JP. Methionine restriction affects oxidative stress and glutathione-related redox pathways in the rat. Exp Biol Med (Maywood) 2013; 238:392-9. [PMID: 23760005 DOI: 10.1177/1535370213477988] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Lifelong dietary methionine restriction (MR) is associated with increased longevity and decreased incidence of age-related disorders and diseases in rats and mice. A reduction in the levels of oxidative stress may be a contributing mechanistic factor for the beneficial effects of MR. To examine this, we determined the effects of an 80% dietary restriction of Met on different biomarkers of oxidative stress and antioxidant pathways in blood, liver, kidney and brain in the rat. Male F-344 rats were fed control (0.86% methionine) or MR (0.17% methionine) diets for up to six months. Blood and tissues were analyzed for glutathione (GSH) concentrations, related enzyme activities and biomarkers of oxidative stress. MR was associated with reductions in oxidative stress biomarkers including plasma 8-hydoxydeoxyguanosine (8-OHdG) and 8-isoprostane and erythrocyte protein-bound glutathione after one month with levels remaining low for at least six months (P < 0.05). Levels of free GSH in blood were increased after 1-6 months of MR feeding whereas liver GSH levels were reduced over this time (P < 0.05). In MR rats, GSH peroxidase activity was decreased in liver and increased in kidney compared with controls. No changes in the activities of GSH reductase in liver and kidney and superoxide dismutase in liver were observed as a result of MR feeding. Altogether, these findings indicate that oxidative stress is reduced by MR feeding in rats, but this effect cannot be explained by changes in the activity of antioxidant enzymes.
Collapse
Affiliation(s)
- Sreenivasa Maddineni
- Department of Comparative Medicine, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | | | | | | | | |
Collapse
|
38
|
Sanchez-Roman I, Barja G. Regulation of longevity and oxidative stress by nutritional interventions: role of methionine restriction. Exp Gerontol 2013; 48:1030-42. [PMID: 23454735 DOI: 10.1016/j.exger.2013.02.021] [Citation(s) in RCA: 116] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2012] [Revised: 01/17/2013] [Accepted: 02/21/2013] [Indexed: 11/25/2022]
Abstract
Comparative studies indicate that long-lived mammals have low rates of mitochondrial reactive oxygen species production (mtROSp) and oxidative damage in their mitochondrial DNA (mtDNA). Dietary restriction (DR), around 40%, extends the mean and maximum life span of a wide range of species and lowers mtROSp and oxidative damage to mtDNA, which supports the mitochondrial free radical theory of aging (MFRTA). Regarding the dietary factor responsible for the life extension effect of DR, neither carbohydrate nor lipid restriction seems to modify maximum longevity. However protein restriction (PR) and methionine restriction (at least 80% MetR) increase maximum lifespan in rats and mice. Interestingly, only 7weeks of 40% PR (at least in liver) or 40% MetR (in all the studied organs, heart, brain, liver or kidney) is enough to decrease mtROSp and oxidative damage to mtDNA in rats, whereas neither carbohydrate nor lipid restriction changes these parameters. In addition, old rats also conserve the capacity to respond to 7weeks of 40% MetR with these beneficial changes. Most importantly, 40% MetR, differing from what happens during both 40% DR and 80% MetR, does not decrease growth rate and body size of rats. All the available studies suggest that the decrease in methionine ingestion that occurs during DR is responsible for part of the aging-delaying effect of this intervention likely through the decrease of mtROSp and ensuing DNA damage that it exerts. We conclude that lowering mtROS generation is a conserved mechanism, shared by long-lived species and dietary, protein, and methionine restricted animals, that decreases damage to macromolecules situated near the complex I mtROS generator, especially mtDNA. This would decrease the accumulation rate of somatic mutations in mtDNA and maybe finally also in nuclear DNA.
Collapse
Affiliation(s)
- Ines Sanchez-Roman
- Department of Animal Physiology-II, Faculty of Biological Sciences, Complutense University of Madrid (UCM), Spain
| | | |
Collapse
|
39
|
Hensen SMM, Heldens L, van Enckevort CMW, van Genesen ST, Pruijn GJM, Lubsen NH. Activation of the antioxidant response in methionine deprived human cells results in an HSF1-independent increase in HSPA1A mRNA levels. Biochimie 2013; 95:1245-51. [PMID: 23395854 DOI: 10.1016/j.biochi.2013.01.017] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2012] [Accepted: 01/29/2013] [Indexed: 12/30/2022]
Abstract
In cells starved for leucine, lysine or glutamine heat shock factor 1 (HSF1) is inactivated and the level of the transcripts of the HSF1 target genes HSPA1A (Hsp70) and DNAJB1 (Hsp40) drops. We show here that in HEK293 cells deprived of methionine HSF1 was similarly inactivated but that the level of HSPA1A and DNAJB1 mRNA increased. This increase was also seen in cells expressing a dominant negative HSF1 mutant (HSF379 or HSF1-K80Q), confirming that the increase is HSF1 independent. The antioxidant N-acetylcysteine completely inhibited the increase in HSPA1A and DNAJB1 mRNA levels upon methionine starvation, indicating that this increase is a response to oxidative stress resulting from a lack of methionine. Cells starved for methionine contained higher levels of c-Fos and FosB mRNA, but knockdown of these transcription factors had no effect on the HSPA1A or DNAJB1 mRNA level. Knockdown of NRF2 mRNA resulted in the inhibition of the increase in the HSPA1A mRNA, but not the DNAJB1 mRNA, level in methionine starved cells. We conclude that methionine deprivation results in both the amino acid deprivation response and an antioxidant response mediated at least in part by NRF2. This antioxidant response includes an HSF1 independent increase in the levels of HSPA1A and DNAJB1 mRNA.
Collapse
Affiliation(s)
- Sanne M M Hensen
- Department of Biomolecular Chemistry, Radboud University Nijmegen, P.O. Box 9101, NL-6500 HB Nijmegen, The Netherlands
| | | | | | | | | | | |
Collapse
|
40
|
Abstract
One of the many functions of taurine is to protect cells against oxidation, by protecting mitochondrial integrity and respiration. Taurine metabolism has attracted much attention in fish nutrition due to the fact that as plant ingredients replace fishmeal, dietary taurine has declined. As the endogenous synthesis of taurine might be too low to protect cells against oxidative stress and apoptosis, the present study aimed to test whether taurine may protect liver cells from apoptosis. Liver cells isolated from Atlantic salmon (Salmo salar) were grown in media supplemented with a physiological concentration of taurine (25 (se0·5) mm) or without any taurine supplementation (14 (se3) μm) for 3 d. To increase oxidation in the mitochondria and maximise any cellular response of taurine supplementation, 100 μm-CdCl2was added or not added to the cells at day 3. At day 4, cells were harvested and assessed for viability. As expected, the addition of CdCl2decreased cell viability without showing any interaction with taurine supplementation. Cells grown in the taurine-supplemented media had lower protein abundance of active caspase-3. In addition, the protein abundance of phosphorylated mitogen-activating phosphokinase (P-p63, P-p42/44 and P-p38) as well as cytochrome P450 were reduced when taurine was added to the media. Cells grown without taurine supplementation had a more condensed chromatin and more smeared DNA, also pointing to a higher apoptosis in these cells. In conclusion, taurine attenuated apoptosis in primary liver cells isolated from Atlantic salmon, and as such, taurine may be conditionally indispensable in Atlantic salmon.
Collapse
|
41
|
Effects of aging and methionine restriction applied at old age on ROS generation and oxidative damage in rat liver mitochondria. Biogerontology 2012; 13:399-411. [PMID: 22580750 DOI: 10.1007/s10522-012-9384-5] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2012] [Accepted: 04/20/2012] [Indexed: 01/08/2023]
Abstract
It is known that a global decrease in food ingestion (dietary restriction, DR) lowers mitochondrial ROS generation (mitROS) and oxidative stress in young immature rats. This seems to be caused by the decreased methionine ingestion of DR animals. This is interesting since isocaloric methionine restriction in the diet (MetR) also increases, like DR, rodent maximum longevity. However, it is not known if old rats maintain the capacity to lower mitROS generation and oxidative stress in response to MetR similarly to young immature animals, and whether MetR implemented at old age can reverse aging-related variations in oxidative stress. In this investigation the effects of aging and 7 weeks of MetR were investigated in liver mitochondria of Wistar rats. MetR implemented at old age decreased mitROS generation, percent free radical leak at the respiratory chain and mtDNA oxidative damage without changing oxygen consumption. Protein oxidation, lipoxidation and glycoxidation increased with age, and MetR in old rats partially or totally reversed these age-related increases. Aging increased the amount of SIRT1, and MetR decreased SIRT1 and TFAM and increased complex IV. No changes were observed in the protein amounts of PGC1, Nrf2, MnSOD, AIF, complexes I, II and III, and in the extent of genomic DNA methylation. In conclusion, treating old rats with isocaloric short-term MetR lowers mitROS production and free radical leak and oxidative damage to mtDNA, and reverses aging-related increases in protein modification. Aged rats maintain the capacity to lower mitochondrial ROS generation and oxidative stress in response to a short-term exposure to restriction of a single dietary substance: methionine.
Collapse
|
42
|
Does the oxidative stress theory of aging explain longevity differences in birds? I. Mitochondrial ROS production. Exp Gerontol 2011; 47:203-10. [PMID: 22123429 DOI: 10.1016/j.exger.2011.11.006] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2011] [Revised: 10/08/2011] [Accepted: 11/14/2011] [Indexed: 11/21/2022]
Abstract
Mitochondrial reactive oxygen species (ROS) production rates are reported to be inversely related to maximum lifespan potential (MLSP) in mammals and also to be higher in short-living mammals compared to short-living birds. The mammal-bird comparison, however, is mainly based on studies of rats and pigeons. To date, there has been no systematic examination of ROS production in birds that differ in MLSP. Here we report a comparison of mitochondrial ROS production in two short-living (quails) and three long-living bird species (parrots) that exhibit, on average, a 5-fold longevity difference. Mitochondrial ROS production was determined both in isolated mitochondria (heart, skeletal muscle and liver) as traditionally done and also in intact erythrocytes. In all four tissues, mitochondrial ROS production was similar in quails and parrots and showed no correspondence with known longevity differences. The lack of a consistent difference between quails and parrots was not due to differences in mitochondrial content as ROS production in relation to oxygen consumption (determined as the free radical leak) showed a similar pattern. These findings cast doubt on the robustness of the oxidative stress theory of aging.
Collapse
|
43
|
Sanchez-Roman I, Gomez A, Gomez J, Suarez H, Sanchez C, Naudi A, Ayala V, Portero-Otin M, Lopez-Torres M, Pamplona R, Barja G. Forty percent methionine restriction lowers DNA methylation, complex I ROS generation, and oxidative damage to mtDNA and mitochondrial proteins in rat heart. J Bioenerg Biomembr 2011; 43:699-708. [PMID: 22006472 DOI: 10.1007/s10863-011-9389-9] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2011] [Accepted: 09/29/2011] [Indexed: 01/09/2023]
Abstract
Methionine dietary restriction (MetR), like dietary restriction (DR), increases rodent maximum longevity. However, the mechanism responsible for the retardation of aging with MetR is still not entirely known. As DR decreases oxidative damage and mitochondrial free radical production, it is plausible to hypothesize that a decrease in oxidative stress is the mechanism for longevity extension with MetR. In the present investigation male Wistar rats were subjected to isocaloric 40% MetR during 7 weeks. It was found that 40% MetR decreases heart mitochondrial ROS production at complex I during forward electron flow, lowers oxidative damage to mitochondrial DNA and proteins, and decreases the degree of methylation of genomic DNA. No significant changes occurred for mitochondrial oxygen consumption, the amounts of the four respiratory complexes (I to IV), and the mitochondrial protein apoptosis-inducing factor (AIF). These results indicate that methionine can be the dietary factor responsible for the decrease in mitochondrial ROS generation and oxidative stress, and likely for part of the increase in longevity, that takes place during DR. They also highlight some of the mechanisms involved in the generation of these beneficial effects.
Collapse
Affiliation(s)
- Ines Sanchez-Roman
- Department of Animal Physiology-II, Faculty of Biological Sciences, Complutense University of Madrid (UCM), Madrid, Spain
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Spasojević I, Bogdanović Pristov J, Vujisić L, Spasić M. The reaction of methionine with hydroxyl radical: reactive intermediates and methanethiol production. Amino Acids 2011; 42:2439-45. [DOI: 10.1007/s00726-011-1049-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2011] [Accepted: 07/22/2011] [Indexed: 01/10/2023]
|
45
|
Krzywanski DM, Moellering DR, Fetterman JL, Dunham-Snary KJ, Sammy MJ, Ballinger SW. The mitochondrial paradigm for cardiovascular disease susceptibility and cellular function: a complementary concept to Mendelian genetics. J Transl Med 2011; 91:1122-35. [PMID: 21647091 PMCID: PMC3654682 DOI: 10.1038/labinvest.2011.95] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
While there is general agreement that cardiovascular disease (CVD) development is influenced by a combination of genetic, environmental, and behavioral contributors, the actual mechanistic basis of how these factors initiate or promote CVD development in some individuals while others with identical risk profiles do not, is not clearly understood. This review considers the potential role for mitochondrial genetics and function in determining CVD susceptibility from the standpoint that the original features that molded cellular function were based upon mitochondrial-nuclear relationships established millions of years ago and were likely refined during prehistoric environmental selection events that today, are largely absent. Consequently, contemporary risk factors that influence our susceptibility to a variety of age-related diseases, including CVD were probably not part of the dynamics that defined the processes of mitochondrial-nuclear interaction, and thus, cell function. In this regard, the selective conditions that contributed to cellular functionality and evolution should be given more consideration when interpreting and designing experimental data and strategies. Finally, future studies that probe beyond epidemiologic associations are required. These studies will serve as the initial steps for addressing the provocative concept that contemporary human disease susceptibility is the result of selection events for mitochondrial function that increased chances for prehistoric human survival and reproductive success.
Collapse
Affiliation(s)
- David M Krzywanski
- Division of Molecular and Cellular Pathology, Department of Pathology, The University of Alabama at Birmingham, Birmingham, AL, USA
| | | | | | | | | | | |
Collapse
|
46
|
Gomez J, Sanchez-Roman I, Gomez A, Sanchez C, Suarez H, Lopez-Torres M, Barja G. Methionine and homocysteine modulate the rate of ROS generation of isolated mitochondria in vitro. J Bioenerg Biomembr 2011; 43:377-86. [PMID: 21748404 DOI: 10.1007/s10863-011-9368-1] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2011] [Accepted: 06/05/2011] [Indexed: 11/28/2022]
Abstract
Dietary methionine restriction and supplementation in mammals have beneficial (antiaging) and detrimental effects respectively, which have been related to chronic modifications in the rate of mitochondrial ROS generation. However it is not known if methionine or its metabolites can have, in addition, direct effects on the rate of mitochondrial ROS production. This is studied here for the methionine cycle metabolites S-adenosylmethionine (SAM), S-adenosylhomocysteine (SAH), homocysteine and methionine itself in isolated rat liver, kidney, heart, and brain mitochondria. The results show that methionine increases ROS production in liver and kidney mitochondria, homocysteine increases it in kidney and decreases it in the other three organs, and SAM and SAH have no effects. The variations in ROS production are localized at complexes I or III. These changes add to previously described chronic effects of methionine restriction and supplementation in vivo.
Collapse
Affiliation(s)
- Jose Gomez
- Department of Animal Physiology II, Faculty of Biological Sciences, Complutense University, Madrid, Spain
| | | | | | | | | | | | | |
Collapse
|
47
|
|
48
|
Matsui S, Tsujimoto Y, Ozawa T, Matsushima K. Antioxidant effects of antioxidant biofactor on reactive oxygen species in human gingival fibroblasts. J Clin Biochem Nutr 2011; 48:209-13. [PMID: 21562640 PMCID: PMC3082075 DOI: 10.3164/jcbn.10-85] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2010] [Accepted: 08/22/2010] [Indexed: 11/22/2022] Open
Abstract
The purpose of this study was to investigate the effects of antioxidant biofactor (AOB) on reactive oxygen species (ROS). Generation of superoxide radical (O2•−) and hydroxyl radical (•OH) was determined using an electron spin resonance (ESR) spin-trapping method. AOB was added at different concentrations to these free radical generating systems. The generation of both O2•− and •OH was scavenged by the addition of AOB in a dose-dependent manner. These results indicate that AOB has strong antioxidant properties against these radicals. We further investigated the anti-oxidative effect of AOB on human gingival fibroblasts (HGFs). HGFs were treated for 3 h with α-MEM containing a combination of AOB and H2O2 (AOB + H2O2 group), containing H2O2 (H2O2 group), or containing AOB alone (AOB group). Non-stimulated HGFs were used as a control group. The number of surviving cells was in the order of the AOB group > control group > AOB + H2O2 group > H2O2 group. The level of expression of type I collagen mRNA and production of collagen were also in the order of the AOB group > control group > AOB + H2O2 group > H2O2 group. In conclusion, our results suggest that AOB may protect HGFs against oxidative stress by reducing stress-induced ROS.
Collapse
Affiliation(s)
- Satoshi Matsui
- Department of Endodontics, Nihon University School of Dentistry at Matsudo, 870-1, Sakaecho, Nishi-2, Matsudo, Chiba 271-8587, Japan
| | | | | | | |
Collapse
|
49
|
Lee BC, Gladyshev VN. The biological significance of methionine sulfoxide stereochemistry. Free Radic Biol Med 2011; 50:221-7. [PMID: 21075204 PMCID: PMC3311537 DOI: 10.1016/j.freeradbiomed.2010.11.008] [Citation(s) in RCA: 118] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2010] [Revised: 11/04/2010] [Accepted: 11/05/2010] [Indexed: 12/22/2022]
Abstract
Methionine can be oxidized by reactive oxygen species to a mixture of two diastereomers, methionine-S-sulfoxide and methionine-R-sulfoxide. Both free amino acid and protein-based forms of methionine-S-sulfoxide are stereospecifically reduced by MsrA, whereas the reduction of methionine-R-sulfoxide requires two enzymes, MsrB and fRMsr, which act on its protein-based and free amino acid forms, respectively. However, mammals lack fRMsr and are characterized by deficiency in the reduction of free methionine-R-sulfoxide. The biological significance of such biased reduction of methionine sulfoxide has not been fully explored. MsrA and MsrB activities decrease during aging, leading to accumulation of protein-based and free amino acid forms of methionine sulfoxide. Since methionine is an indispensible amino acid in human nutrition and a key metabolite in sulfur, methylation, and transsulfuration pathways, the consequences of accumulation of its oxidized forms require further studies. Finally, in addition to methionine, methylsulfinyl groups are present in various drugs and natural compounds, and their differential reduction by Msrs may have important therapeutic implications.
Collapse
Affiliation(s)
- Byung Cheon Lee
- Division of Genetics, Department of Medicine, Brigham & Women's Hospital and Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | | |
Collapse
|
50
|
|