1
|
Ommi NB, Mattocks DA, Kalecký K, Bottiglieri T, Nichenametla SN. Pharmacological recapitulation of the lean phenotype induced by the lifespan-extending sulfur amino acid-restricted diet. Aging (Albany NY) 2025; 17:960-981. [PMID: 40202448 PMCID: PMC12074818 DOI: 10.18632/aging.206237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Accepted: 03/20/2025] [Indexed: 04/10/2025]
Abstract
Sulfur amino acid restriction (SAAR), lowering the dietary concentration of sulfur amino acids methionine and cysteine, induces strong anti-obesity effects in rodents. Due to difficulties in formulating the SAAR diet for human consumption, its translation is challenging. Since our previous studies suggest a mechanistic role for low glutathione (GSH) in SAAR-induced anti-obesity effects, we investigated if the pharmacological lowering of GSH recapitulates the lean phenotype in mice on a sulfur amino acid-replete diet. Male obese C57BL6/NTac mice were fed high-fat diets with 0.86% methionine (CD), 0.12% methionine (SAAR), SAAR diet supplemented with a GSH biosynthetic precursor, N-acetylcysteine in water (NAC), and CD supplemented with a GSH biosynthetic inhibitor, DL-buthionine-(S, R)-sulfoximine in water (BSO). The SAAR diet lowered hepatic GSH but increased Nrf2, Phgdh, and serine. These molecular changes culminated in lower hepatic lipid droplet frequency, epididymal fat depot weights, and body fat mass; NAC reversed all these changes. BSO mice exhibited all SAAR-induced changes, with two notable differences, i.e., a smaller effect size than that of the SAAR diet and a higher predilection for molecular changes in kidneys than in the liver. Metabolomics data indicate that BSO and the SAAR diet induce similar changes in the kidney. Unaltered plasma aspartate and alanine transaminases and cystatin-C indicate that long-term continuous administration of BSO is safe. Data demonstrate that BSO recapitulates the SAAR-induced anti-obesity effects and that GSH plays a mechanistic role. BSO dose-response studies in animals and pilot studies in humans to combat obesity are highly warranted.
Collapse
Affiliation(s)
- Naidu B. Ommi
- Animal Science Laboratory, Orentreich Foundation for the Advancement of Science Inc., Cold Spring-on-Hudson, NY 10516, USA
| | - Dwight A.L. Mattocks
- Animal Science Laboratory, Orentreich Foundation for the Advancement of Science Inc., Cold Spring-on-Hudson, NY 10516, USA
| | - Karel Kalecký
- Center of Metabolomics, Institute of Metabolic Disease, Baylor Scott and White Research Institute, Dallas, TX 75204, USA
| | - Teodoro Bottiglieri
- Center of Metabolomics, Institute of Metabolic Disease, Baylor Scott and White Research Institute, Dallas, TX 75204, USA
| | - Sailendra N. Nichenametla
- Animal Science Laboratory, Orentreich Foundation for the Advancement of Science Inc., Cold Spring-on-Hudson, NY 10516, USA
| |
Collapse
|
2
|
Tore EC, Adriaans BC, Olsen T, Vinknes KJ, Kooi ME, Elshorbagy AK, Bastani NE, Dagnelie PC, Eussen SJPM, Gundersen TE, Kožich V, Refsum H, Retterstøl K, Stolt ETK, van Greevenbroek MMJ. Estimated stearoyl-CoA desaturase activity mediates the associations of total cysteine with adiposity: The Maastricht Study. J Clin Lipidol 2025; 19:348-357. [PMID: 40024839 DOI: 10.1016/j.jacl.2024.11.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 10/17/2024] [Accepted: 11/13/2024] [Indexed: 03/04/2025]
Abstract
BACKGROUND Plasma sulfur amino acids (SAAs), particularly cysteine, are associated with obesity. One proposed mechanism is the altered regulation of the stearoyl-CoA desaturase (SCD) enzyme. Changes in the SCD enzyme activity have been linked to obesity, as well as to plasma SAA concentrations. OBJECTIVE This study aimed to investigate whether estimated SCD activity mediates the associations between plasma SAAs and measures of overall adiposity and specific fat depots. METHODS We examined cross-sectional data from a subset of the Maastricht Study (n = 1129, 50.7% men, 56.7% with (pre)diabetes). Concentrations of methionine, total homocysteine, cystathionine, total cysteine (tCys), total glutathione (tGSH), and taurine were measured in fasting plasma. Outcomes included measures of overall, peripheral and central adiposity, and liver fat. SCD activity was estimated by ratios of serum fatty acids as SCD16 and SCD18 indices. The associations between plasma SAAs and measures of adiposity or liver fat were examined with multiple linear regression analysis. Multiple mediation analysis was used to investigate whether the significant associations were mediated by SCD16 and SCD18 indices. RESULTS Plasma tCys was positively associated with all adiposity measures (β ranged from 0.15 to 0.30). SCD16 significantly mediated all associations (proportion mediated ranged from 5.1% to 9.7%). Inconsistent mediation effects were found for SCD18. Despite a significant inverse association of plasma tGSH with all adiposity measures (β ranged from -0.08 to -0.16), no significant mediation effect was found. CONCLUSIONS Plasma tCys may promote excessive body fat accumulation via upregulation of SCD activity.
Collapse
Affiliation(s)
- Elena C Tore
- Department of Internal Medicine, Maastricht University, Maastricht, The Netherlands (Drs Tore, Adriaans, Dagnelie, van Greevenbroek); CARIM, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands (Drs Tore, Adriaans, Kooi, Dagnelie, Eussen, van Greevenbroek).
| | - Bregje C Adriaans
- Department of Internal Medicine, Maastricht University, Maastricht, The Netherlands (Drs Tore, Adriaans, Dagnelie, van Greevenbroek); CARIM, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands (Drs Tore, Adriaans, Kooi, Dagnelie, Eussen, van Greevenbroek)
| | - Thomas Olsen
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway (Drs Olsen, Vinknes, Bastani, Refsum, Retterstøl, Stolt)
| | - Kathrine J Vinknes
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway (Drs Olsen, Vinknes, Bastani, Refsum, Retterstøl, Stolt)
| | - M Eline Kooi
- CARIM, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands (Drs Tore, Adriaans, Kooi, Dagnelie, Eussen, van Greevenbroek); Department of Radiology & Nuclear Medicine, Maastricht University Medical Center, Maastricht, The Netherlands (Dr Kooi)
| | - Amany K Elshorbagy
- Department of Pharmacology, University of Oxford, Oxford, UK (Drs Elshorbagy, Refsum); Department of Physiology, Faculty of Medicine, University of Alexandria, Alexandria, Egypt (Dr Elshorbagy)
| | - Nasser E Bastani
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway (Drs Olsen, Vinknes, Bastani, Refsum, Retterstøl, Stolt)
| | - Pieter C Dagnelie
- Department of Internal Medicine, Maastricht University, Maastricht, The Netherlands (Drs Tore, Adriaans, Dagnelie, van Greevenbroek); CARIM, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands (Drs Tore, Adriaans, Kooi, Dagnelie, Eussen, van Greevenbroek)
| | - Simone J P M Eussen
- CARIM, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands (Drs Tore, Adriaans, Kooi, Dagnelie, Eussen, van Greevenbroek); Department of Epidemiology, Maastricht University, Maastricht, The Netherlands (Dr Eussen); CAPHRI Care and Public Health Research Institute, Maastricht University, Maastricht, The Netherlands (Dr Eussen)
| | | | - Viktor Kožich
- Department of Pediatrics and Inherited Metabolic Disorders, Charles University-First Faculty of Medicine, and General University Hospital in Prague, Prague, Czech Republic (Dr Kožich)
| | - Helga Refsum
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway (Drs Olsen, Vinknes, Bastani, Refsum, Retterstøl, Stolt); Department of Pharmacology, University of Oxford, Oxford, UK (Drs Elshorbagy, Refsum)
| | - Kjetil Retterstøl
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway (Drs Olsen, Vinknes, Bastani, Refsum, Retterstøl, Stolt); The Lipid Clinic, Oslo University Hospital, Norway (Dr Retterstøl)
| | - Emma T K Stolt
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway (Drs Olsen, Vinknes, Bastani, Refsum, Retterstøl, Stolt)
| | - Marleen M J van Greevenbroek
- Department of Internal Medicine, Maastricht University, Maastricht, The Netherlands (Drs Tore, Adriaans, Dagnelie, van Greevenbroek); CARIM, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands (Drs Tore, Adriaans, Kooi, Dagnelie, Eussen, van Greevenbroek)
| |
Collapse
|
3
|
Olsen T, Vinknes KJ, Barvíková K, Stolt E, Lee-Ødegård S, Troensegaard H, Johannessen H, Elshorbagy A, Sokolová J, Krijt J, Křížková M, Ditrói T, Nagy P, Øvrebø B, Refsum H, Thoresen M, Retterstøl K, Kožich V. Dietary sulfur amino acid restriction in humans with overweight and obesity: Evidence of an altered plasma and urine sulfurome, and a novel metabolic signature that correlates with loss of fat mass and adipose tissue gene expression. Redox Biol 2024; 73:103192. [PMID: 38776754 PMCID: PMC11163171 DOI: 10.1016/j.redox.2024.103192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 05/03/2024] [Accepted: 05/12/2024] [Indexed: 05/25/2024] Open
Abstract
BACKGROUND In animals, dietary sulfur amino acid restriction (SAAR) improves metabolic health, possibly mediated by altering sulfur amino acid metabolism and enhanced anti-obesogenic processes in adipose tissue. AIM To assess the effects of SAAR over time on the plasma and urine SAA-related metabolites (sulfurome) in humans with overweight and obesity, and explore whether such changes were associated with body weight, body fat and adipose tissue gene expression. METHODS Fifty-nine subjects were randomly allocated to SAAR (∼2 g SAA, n = 31) or a control diet (∼5.6 g SAA, n = 28) consisting of plant-based whole-foods and supplemented with capsules to titrate contents of SAA. Sulfurome metabolites in plasma and urine at baseline, 4 and 8 weeks were measured using HPLC and LC-MS/MS. mRNA-sequencing of subcutaneous white adipose tissue (scWAT) was performed to assess changes in gene expression. Data were analyzed with mixed model regression. Principal component analyses (PCA) were performed on the sulfurome data to identify potential signatures characterizing the response to SAAR. RESULTS SAAR led to marked decrease of the main urinary excretion product sulfate (p < 0.001) and plasma and/or 24-h urine concentrations of cystathionine, sulfite, thiosulfate, H2S, hypotaurine and taurine. PCA revealed a distinct metabolic signature related to decreased transsulfuration and H2S catabolism that predicted greater weight loss and android fat mass loss in SAAR vs. controls (all pinteraction < 0.05). This signature correlated positively with scWAT expression of genes in the tricarboxylic acid cycle, electron transport and β-oxidation (FDR = 0.02). CONCLUSION SAAR leads to distinct alterations of the plasma and urine sulfurome in humans, and predicted increased loss of weight and android fat mass, and adipose tissue lipolytic gene expression in scWAT. Our data suggest that SAA are linked to obesogenic processes and that SAAR may be useful for obesity and related disorders. TRIAL IDENTIFIER: https://clinicaltrials.gov/study/NCT04701346.
Collapse
Affiliation(s)
- Thomas Olsen
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine University of Oslo, Postboks 1046 Blindern, 0317 Oslo, Norway.
| | - Kathrine J Vinknes
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine University of Oslo, Postboks 1046 Blindern, 0317 Oslo, Norway
| | - Kristýna Barvíková
- Department of Pediatrics and Inherited Metabolic Disorders, Charles University, First Faculty of Medicine, and General University Hospital, Ke Karlovu 2, 128 00 Prague, Czech Republic
| | - Emma Stolt
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine University of Oslo, Postboks 1046 Blindern, 0317 Oslo, Norway
| | - Sindre Lee-Ødegård
- Department of Endocrinology, Morbid Obesity and Preventive Medicine, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Postboks 4959 Nydalen, OUS HF Aker sykehus, 0424 Oslo, Norway
| | - Hannibal Troensegaard
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine University of Oslo, Postboks 1046 Blindern, 0317 Oslo, Norway
| | - Hanna Johannessen
- Department of Pathology, Oslo University Hospital, Rikshospitalet, Postboks 45980 Nydalen, OUS HF Rikshospitalet, 0424 Oslo, Norway
| | - Amany Elshorbagy
- Department of Physiology, Faculty of Medicine, University of Alexandria, Chamblion street, Qesm Al Attarin, Alexandria 5372066, Egypt; Department of Pharmacology, University of Oxford, Mansfield Rd, Oxford OX1 3QT, UK
| | - Jitka Sokolová
- Department of Pediatrics and Inherited Metabolic Disorders, Charles University, First Faculty of Medicine, and General University Hospital, Ke Karlovu 2, 128 00 Prague, Czech Republic
| | - Jakub Krijt
- Department of Pediatrics and Inherited Metabolic Disorders, Charles University, First Faculty of Medicine, and General University Hospital, Ke Karlovu 2, 128 00 Prague, Czech Republic
| | - Michaela Křížková
- Department of Pediatrics and Inherited Metabolic Disorders, Charles University, First Faculty of Medicine, and General University Hospital, Ke Karlovu 2, 128 00 Prague, Czech Republic
| | - Tamás Ditrói
- Department of Molecular Immunology and Toxicology and the National Tumor Biology Laboratory, National Institute of Oncology, Ráth György u. 7-9, 1122 Budapest, Hungary
| | - Péter Nagy
- Department of Molecular Immunology and Toxicology and the National Tumor Biology Laboratory, National Institute of Oncology, Ráth György u. 7-9, 1122 Budapest, Hungary; Department of Anatomy and Histology, HUN-REN-UVMB Laboratory of Redox Biology Research Group, University of Veterinary Medicine, 1078 Budapest, Hungary; Chemistry Institute, University of Debrecen, 4012 Debrecen, Hungary
| | - Bente Øvrebø
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine University of Oslo, Postboks 1046 Blindern, 0317 Oslo, Norway; Department of Food Safety, Norwegian Institute of Public Health, Postboks 222 Skøyen, 0213 Oslo, Norway
| | - Helga Refsum
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine University of Oslo, Postboks 1046 Blindern, 0317 Oslo, Norway; Department of Pharmacology, University of Oxford, Mansfield Rd, Oxford OX1 3QT, UK
| | - Magne Thoresen
- Department of Biostatistics, Institute of Basic Medical Sciences, University of Oslo, Postboks 1122 Blindern, 0317 Oslo, Norway
| | - Kjetil Retterstøl
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine University of Oslo, Postboks 1046 Blindern, 0317 Oslo, Norway; The Lipid Clinic, Department of Endocrinology, Morbid Obesity and Preventive Medicine, Oslo University Hospital, Postboks 4959 Nydalen, OUS HF Aker sykehus, 0424 Oslo, Norway
| | - Viktor Kožich
- Department of Pediatrics and Inherited Metabolic Disorders, Charles University, First Faculty of Medicine, and General University Hospital, Ke Karlovu 2, 128 00 Prague, Czech Republic.
| |
Collapse
|
4
|
Olsen T, Stolt E, Øvrebø B, Elshorbagy A, Tore EC, Lee-Ødegård S, Troensegaard H, Johannessen H, Doeland B, Vo AAD, Dahl AF, Svendsen K, Thoresen M, Refsum H, Rising R, Barvíková K, van Greevenbroek M, Kožich V, Retterstøl K, Vinknes KJ. Dietary sulfur amino acid restriction in humans with overweight and obesity: a translational randomized controlled trial. J Transl Med 2024; 22:40. [PMID: 38195568 PMCID: PMC10775517 DOI: 10.1186/s12967-023-04833-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 12/26/2023] [Indexed: 01/11/2024] Open
Abstract
BACKGROUND Dietary sulfur amino acid restriction (SAAR) improves metabolic health in animals. In this study, we investigated the effect of dietary SAAR on body weight, body composition, resting metabolic rate, gene expression profiles in white adipose tissue (WAT), and an extensive blood biomarker profile in humans with overweight or obesity. METHODS N = 59 participants with overweight or obesity (73% women) were randomized stratified by sex to an 8-week plant-based dietary intervention low (~ 2 g/day, SAAR) or high (~ 5.6 g/day, control group) in sulfur amino acids. The diets were provided in full to the participants, and both investigators and participants were blinded to the intervention. Outcome analyses were performed using linear mixed model regression adjusted for baseline values of the outcome and sex. RESULTS SAAR led to a ~ 20% greater weight loss compared to controls (β 95% CI - 1.14 (- 2.04, - 0.25) kg, p = 0.013). Despite greater weight loss, resting metabolic rate remained similar between groups. Furthermore, SAAR decreased serum leptin, and increased ketone bodies compared to controls. In WAT, 20 genes were upregulated whereas 24 genes were downregulated (FDR < 5%) in the SAAR group compared to controls. Generally applicable gene set enrichment analyses revealed that processes associated with ribosomes were upregulated, whereas processes related to structural components were downregulated. CONCLUSION Our study shows that SAAR leads to greater weight loss, decreased leptin and increased ketone bodies compared to controls. Further research on SAAR is needed to investigate the therapeutic potential for metabolic conditions in humans. TRIAL REGISTRATION ClinicalTrials.gov identifier: NCT04701346, registered Jan 8th 2021, https://www. CLINICALTRIALS gov/study/NCT04701346.
Collapse
Affiliation(s)
- Thomas Olsen
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway.
| | - Emma Stolt
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Bente Øvrebø
- Department of Food Safety, Norwegian Institute of Public Health, Oslo, Norway
| | - Amany Elshorbagy
- Department of Physiology, Faculty of Medicine, University of Alexandria, Alexandria, Egypt
- Department of Pharmacology, University of Oxford, Oxford, UK
| | - Elena C Tore
- Department of Internal Medicine and CARIM School of Cardiovascular Diseases, Maastricht University, Maastricht, The Netherlands
| | - Sindre Lee-Ødegård
- Department of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Hannibal Troensegaard
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Hanna Johannessen
- Department of Paedriatic Surgery, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Beate Doeland
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Anna A D Vo
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Anja F Dahl
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Karianne Svendsen
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
- The Lipid Clinic, Department of Endocrinology, Morbid Obesity and Preventive Medicine, Oslo University Hospital, Oslo, Norway
| | - Magne Thoresen
- Department of Biostatistics, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Helga Refsum
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
- Department of Pharmacology, University of Oxford, Oxford, UK
| | | | - Kristýna Barvíková
- Department of Pediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Marleen van Greevenbroek
- Department of Internal Medicine and CARIM School of Cardiovascular Diseases, Maastricht University, Maastricht, The Netherlands
| | - Viktor Kožich
- Department of Pediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Kjetil Retterstøl
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
- The Lipid Clinic, Department of Endocrinology, Morbid Obesity and Preventive Medicine, Oslo University Hospital, Oslo, Norway
| | - Kathrine J Vinknes
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| |
Collapse
|
5
|
Vinknes KJ, Olsen T, Zaré HK, Bastani NE, Stolt E, Dahl AF, Cox RD, Refsum H, Retterstøl K, Åsberg A, Elshorbagy A. Cysteine-lowering treatment with mesna against obesity: Proof of concept and results from a human phase I, dose-finding study. Diabetes Obes Metab 2023; 25:3161-3170. [PMID: 37435697 PMCID: PMC11497255 DOI: 10.1111/dom.15210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 06/14/2023] [Accepted: 06/21/2023] [Indexed: 07/13/2023]
Abstract
AIM To investigate whether mesna-sodium-2-mercaptoethane sulfonate) can reduce diet-induced fat gain in mice, and to assess the safety of single ascending mesna doses in humans to find the dose associated with lowering of plasma tCys by at least 30%. METHODS C3H/HeH mice were shifted to a high-fat diet ± mesna in drinking water; body composition was measured at weeks 0, 2 and 4. In an open, phase I, single ascending dose study, oral mesna (400, 800, 1200, 1600 mg) was administered to 17 men with overweight or obesity. Mesna and tCys concentrations were measured repeatedly for a duration of 48 hours postdosing in plasma, as well as in 24-hour urine. RESULTS Compared with controls, mesna-treated mice had lower tCys and lower estimated mean fat mass gain from baseline (week 2: 4.54 ± 0.40 vs. 6.52 ± 0.36 g; week 4: 6.95 ± 0.35 vs. 8.19 ± 0.34 g; Poverall = .002), but similar lean mass gain. In men with overweight, mesna doses of 400-1600 mg showed dose linearity and were well tolerated. Mesna doses of 800 mg or higher decreased plasma tCys by 30% or more at nadir (4h post-dosing). With increasing mesna dose, tCys AUC0-12h decreased (Ptrend < .001), and urine tCys excretion increased (Ptrend = .004). CONCLUSIONS Mesna reduces diet-induced fat gain in mice. In men with overweight, single oral doses of mesna (800-1600 mg) were well tolerated and lowered plasma tCys efficiently. The effect of sustained tCys-lowering by repeated mesna administration on weight loss in humans deserves investigation.
Collapse
Affiliation(s)
- Kathrine J. Vinknes
- Department of Nutrition, Institute of Basic Medical SciencesUniversity of OsloOsloNorway
| | - Thomas Olsen
- Department of Nutrition, Institute of Basic Medical SciencesUniversity of OsloOsloNorway
| | | | - Nasser E. Bastani
- Department of Nutrition, Institute of Basic Medical SciencesUniversity of OsloOsloNorway
| | - Emma Stolt
- Department of Nutrition, Institute of Basic Medical SciencesUniversity of OsloOsloNorway
| | - Anja F. Dahl
- Department of Nutrition, Institute of Basic Medical SciencesUniversity of OsloOsloNorway
| | - Roger D. Cox
- MRC Harwell InstituteMammalian Genetics UnitHarwell CampusOxfordUK
| | - Helga Refsum
- Department of Nutrition, Institute of Basic Medical SciencesUniversity of OsloOsloNorway
- Department of PharmacologyUniversity of OxfordOxfordUK
| | - Kjetil Retterstøl
- Department of Nutrition, Institute of Basic Medical SciencesUniversity of OsloOsloNorway
- The Lipid Clinic, Department of Endocrinology, Morbid Obesity and Preventive MedicineOslo University HospitalOsloNorway
| | - Anders Åsberg
- Department of Transplantation MedicineOslo University HospitalOsloNorway
- Department of PharmacyUniversity of OsloOsloNorway
| | - Amany Elshorbagy
- Department of PharmacologyUniversity of OxfordOxfordUK
- Department of Physiology, Faculty of MedicineUniversity of AlexandriaAlexandriaEgypt
| |
Collapse
|
6
|
Tore EC, Eussen SJPM, Bastani NE, Dagnelie PC, Elshorbagy AK, Grootswagers P, Kožich V, Olsen T, Refsum H, Retterstøl K, Stehouwer CDA, Stolt ETK, Vinknes KJ, van Greevenbroek MMJ. The Associations of Habitual Intake of Sulfur Amino Acids, Proteins and Diet Quality with Plasma Sulfur Amino Acid Concentrations: The Maastricht Study. J Nutr 2023; 153:2027-2040. [PMID: 37164267 DOI: 10.1016/j.tjnut.2023.05.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 04/12/2023] [Accepted: 05/04/2023] [Indexed: 05/12/2023] Open
Abstract
BACKGROUND Plasma sulfur amino acids (SAAs), i.e., methionine, total cysteine (tCys), total homocysteine (tHcy), cystathionine, total glutathione (tGSH), and taurine, are potential risk factors for obesity and cardiometabolic disorders. However, except for plasma tHcy, little is known about how dietary intake modifies plasma SAA concentrations. OBJECTIVE To investigate whether the intake of SAAs and proteins or diet quality is associated with plasma SAAs. METHODS Data from a cross-sectional subset of The Maastricht Study (n = 1145, 50.5% men, 61 interquartile range: [55, 66] y, 22.5% with prediabetes and 34.3% with type 2 diabetes) were investigated. Dietary intake was assessed using a validated food frequency questionnaire. The intake of SAAs (total, methionine, and cysteine) and proteins (total, animal, and plant) was estimated from the Dutch and Danish food composition tables. Diet quality was assessed using the Dutch Healthy Diet Index, the Mediterranean Diet Score, and the Dietary Approaches to Stop Hypertension score. Fasting plasma SAAs were measured by liquid chromatography (LC) tandem mass spectrometry (MS) (LC/MS-MS). Associations were investigated with multiple linear regressions with tertiles of dietary intake measures (main exposures) and z-standardized plasma SAAs (outcomes). RESULTS Intake of total SAAs and total proteins was positively associated with plasma tCys and cystathionine. Associations were stronger in women and in those with normal body weight. Higher intake of cysteine and plant proteins was associated with lower plasma tHcy and higher cystathionine. Higher methionine intake was associated with lower plasma tGSH, whereas cysteine intake was positively associated with tGSH. Higher intake of methionine and animal proteins was associated with higher plasma taurine. Better diet quality was consistently related to lower plasma tHcy concentrations, but it was not associated with the other SAAs. CONCLUSION Targeted dietary modifications might be effective in modifying plasma concentrations of tCys, tHcy, and cystathionine, which have been associated with obesity and cardiometabolic disorders.
Collapse
Affiliation(s)
- Elena C Tore
- Department of Internal Medicine, Maastricht University, Maastricht, the Netherlands; CARIM School for Cardiovascular Disease, Maastricht University, Maastricht, the Netherlands.
| | - Simone J P M Eussen
- CARIM School for Cardiovascular Disease, Maastricht University, Maastricht, the Netherlands; Department of Epidemiology, Maastricht University, Maastricht, the Netherlands; CAPHRI Care and Public Health Research Institute, Maastricht University, Maastricht, the Netherlands
| | - Nasser E Bastani
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Pieter C Dagnelie
- Department of Internal Medicine, Maastricht University, Maastricht, the Netherlands; CARIM School for Cardiovascular Disease, Maastricht University, Maastricht, the Netherlands
| | - Amany K Elshorbagy
- Department of Pharmacology, University of Oxford, Oxford, United Kingdom; Department of Physiology, Faculty of Medicine, University of Alexandria, Alexandria, Egypt
| | - Pol Grootswagers
- Division of Human Nutrition and Health, Wageningen University, Wageningen, the Netherlands
| | - Viktor Kožich
- Department of Pediatrics and Inherited Metabolic Disorders, Charles University-First Faculty of Medicine, and General University Hospital in Prague, Czech Republic
| | - Thomas Olsen
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Helga Refsum
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway; Department of Pharmacology, University of Oxford, Oxford, United Kingdom
| | - Kjetil Retterstøl
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Coen DA Stehouwer
- Department of Internal Medicine, Maastricht University, Maastricht, the Netherlands; CARIM School for Cardiovascular Disease, Maastricht University, Maastricht, the Netherlands
| | - Emma T K Stolt
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Kathrine J Vinknes
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Marleen M J van Greevenbroek
- Department of Internal Medicine, Maastricht University, Maastricht, the Netherlands; CARIM School for Cardiovascular Disease, Maastricht University, Maastricht, the Netherlands
| |
Collapse
|
7
|
Elshorbagy A, Bastani NE, Lee-Ødegård S, Øvrebø B, Haj-Yasein N, Svendsen K, Turner C, Refsum H, Vinknes KJ, Olsen T. The association of fasting plasma thiol fractions with body fat compartments, biomarker profile, and adipose tissue gene expression. Amino Acids 2023; 55:313-323. [PMID: 36542145 PMCID: PMC10038976 DOI: 10.1007/s00726-022-03229-2] [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: 07/15/2022] [Accepted: 12/17/2022] [Indexed: 12/24/2022]
Abstract
People with high plasma total cysteine (tCys) have higher fat mass and higher concentrations of the atherogenic apolipoprotein B (apoB). The disulfide form, cystine, enhanced human adipogenesis and correlated with total fat mass in a Middle-Eastern cohort. In 35 European adults with overweight (88.6% women) and with dual-X-ray absorptiometry measurements of regional fat, we investigated how cystine compared to other free disulfides in their association with total regional adiposity, plasma lipid and glucose biomarkers, and adipose tissue lipid enzyme mRNA (n = 19). Most total plasma homocysteine (tHcy) (78%) was protein-bound; 63% of total glutathione (tGSH) was reduced. tCys was 49% protein-bound, 30% mixed-disulfide, 15% cystine, and 6% reduced. Controlling for age and lean mass, cystine and total free cysteine were the fractions most strongly associated with android and total fat: 1% higher cystine predicted 1.97% higher android fat mass (95% CI 0.64, 3.31) and 1.25% (0.65, 2.98) higher total fat mass (both p = 0.005). A positive association between tCys and apoB (β: 0.64%; 95% CI 0.17, 1.12%, p = 0.009) was apparently driven by free cysteine and cystine; cystine was also inversely associated with the HDL-associated apolipoprotein A1 (β: -0.57%; 95% CI -0.96, -0.17%, p = 0.007). No independent positive associations with adiposity were noted for tGSH or tHcy fractions. Plasma cystine correlated with CPT1a mRNA (Spearman's r = 0.68, p = 0.001). In conclusion, plasma cystine-but not homocysteine or glutathione disulfides-is associated with android adiposity and an atherogenic plasma apolipoprotein profile. The role of cystine in human adiposity and cardiometabolic risk deserves investigation. ClinicalTrials.gov identifiers: NCT02647970 and NCT03629392.
Collapse
Affiliation(s)
- Amany Elshorbagy
- Department of Pharmacology, University of Oxford, Oxford, UK
- Department of Physiology, Faculty of Medicine, University of Alexandria, Alexandria, Egypt
| | - Nasser E Bastani
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Blindern, Postboks 1046, Oslo, Norway
| | - Sindre Lee-Ødegård
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Blindern, Postboks 1046, Oslo, Norway
| | - Bente Øvrebø
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Blindern, Postboks 1046, Oslo, Norway
| | - Nadia Haj-Yasein
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Blindern, Postboks 1046, Oslo, Norway
| | - Karianne Svendsen
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Blindern, Postboks 1046, Oslo, Norway
- The Cancer Registry of Norway, Oslo University Hospital, Oslo, Norway
| | - Cheryl Turner
- Department of Pharmacology, University of Oxford, Oxford, UK
| | - Helga Refsum
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Blindern, Postboks 1046, Oslo, Norway
| | - Kathrine J Vinknes
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Blindern, Postboks 1046, Oslo, Norway
| | - Thomas Olsen
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Blindern, Postboks 1046, Oslo, Norway.
| |
Collapse
|
8
|
Metabolomics Reveal the Regulatory Effect of Polysaccharides from Fermented Barley Bran Extract on Lipid Accumulation in HepG2 Cells. Metabolites 2023; 13:metabo13020223. [PMID: 36837842 PMCID: PMC9962758 DOI: 10.3390/metabo13020223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Revised: 01/31/2023] [Accepted: 02/01/2023] [Indexed: 02/05/2023] Open
Abstract
Barley bran has potential bioactivities due to its high content of polyphenols and dietary fiber, etc. Fermentation has been considered as an effective way to promote the functional activity of food raw materials. In this study, polysaccharides from barley bran extract fermented by Lactiplantibacillus plantarum dy-1 (FBBE-PS) were analyzed, and its effects on lipid accumulation and oxidative stress in high-fat HepG2 cells induced by sodium oleate were evaluated. The results showed that the molecular weight decreased and monosaccharide composition of polysaccharides changed significantly after fermentation. In addition, 50 μg/mL FBBE-PS could reduce the triglyceride (TG) content and reaction oxygen species (ROS) level in high-fat HepG2 cells by 21.62% and 30.01%, respectively, while increasing the activities of superoxide dismutase (SOD) and catalase (CAT) represented by 64.87% and 22.93%, respectively. RT-qPCR analysis revealed that FBBE-PS could up-regulate the lipid metabolism-related genes such as ppar-α, acox-1 and cpt-1α, and oxidation-related genes such as nrf2, ho-1, nqo-1, sod1, cat, etc. The metabolomics analysis indicated that FBBE-PS could alleviate lipid deposition by inhibiting the biosynthesis of unsaturated fatty acids, which is consistent with the downregulation of scd-1 expression. It is demonstrated that fermentation can alter the properties and physiological activities of polysaccharides in barley bran, and FBBE-PS exhibited an alleviating effect on lipid deposition and oxidative stress in high-fat cells.
Collapse
|
9
|
Associations between plasma sulfur amino acids and specific fat depots in two independent cohorts: CODAM and The Maastricht Study. Eur J Nutr 2023; 62:891-904. [PMID: 36322288 PMCID: PMC9941263 DOI: 10.1007/s00394-022-03041-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 10/20/2022] [Indexed: 02/23/2023]
Abstract
PURPOSE Sulfur amino acids (SAAs) have been associated with obesity and obesity-related metabolic diseases. We investigated whether plasma SAAs (methionine, total cysteine (tCys), total homocysteine, cystathionine and total glutathione) are related to specific fat depots. METHODS We examined cross-sectional subsets from the CODAM cohort (n = 470, 61.3% men, median [IQR]: 67 [61, 71] years) and The Maastricht Study (DMS; n = 371, 53.4% men, 63 [55, 68] years), enriched with (pre)diabetic individuals. SAAs were measured in fasting EDTA plasma with LC-MS/MS. Outcomes comprised BMI, skinfolds, waist circumference (WC), dual-energy X-ray absorptiometry (DXA, DMS), body composition, abdominal subcutaneous and visceral adipose tissues (CODAM: ultrasound, DMS: MRI) and liver fat (estimated, in CODAM, or MRI-derived, in DMS, liver fat percentage and fatty liver disease). Associations were examined with linear or logistic regressions adjusted for relevant confounders with z-standardized primary exposures and outcomes. RESULTS Methionine was associated with all measures of liver fat, e.g., fatty liver disease [CODAM: OR = 1.49 (95% CI 1.19, 1.88); DMS: OR = 1.51 (1.09, 2.14)], but not with other fat depots. tCys was associated with overall obesity, e.g., BMI [CODAM: β = 0.19 (0.09, 0.28); DMS: β = 0.24 (0.14, 0.34)]; peripheral adiposity, e.g., biceps and triceps skinfolds [CODAM: β = 0.15 (0.08, 0.23); DMS: β = 0.20 (0.12, 0.29)]; and central adiposity, e.g., WC [CODAM: β = 0.16 (0.08, 0.25); DMS: β = 0.17 (0.08, 0.27)]. Associations of tCys with VAT and liver fat were inconsistent. Other SAAs were not associated with body fat. CONCLUSION Plasma concentrations of methionine and tCys showed distinct associations with different fat depots, with similar strengths in the two cohorts.
Collapse
|
10
|
Meguid NA, Hemimi M, Ghozlan SAS, Kandeel WA, Hashish AF, Gouda AS, Nazim WS, Mohamed MF. Differential expression of cystathionine beta synthase in adolescents with Down syndrome: impact on adiposity. J Diabetes Metab Disord 2022; 21:1491-1497. [PMID: 36404855 PMCID: PMC9672282 DOI: 10.1007/s40200-022-01087-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 10/05/2021] [Accepted: 07/01/2022] [Indexed: 11/29/2022]
Abstract
Purpose Obesity is more prevalent among people with Down Syndrome (DS) compared to general population. In this pilot study, we investigated the effect of cystathionine beta-synthase (CBS) overdosage on the regulation of transsulfuration pathway and the obesity phenotype in fifty adolescents (25 obese/overweight and 25 lean) with trisomy 21. Methods The transcriptional levels of CBS in leukocytes and its translational levels in plasma were quantified using real time polymerase chain reaction and enzyme-linked immunosorbent assay respectively. Meanwhile, ultra performance liquid chromatography tandem mass spectrometry was used to determine the plasma concentrations of methionine, homocysteine, cystathionine and cysteine. Fasting plasma lipid profiles were assessed by colorimetric assays. The anthropometric measurements and indices of all subjects were recorded. Results Both DS groups had comparable levels of CBS transcripts (p = 0.2734). The plasma levels of the enzyme were significantly higher in the lean DS cases (p = 0.0174) compared to the obese/overweight participants. Total cholesterol, triglycerides, high-density lipoprotein, low-density lipoprotein, methionine, homocysteine, cystathionine and cysteine showed similar plasma levels in both groups. However, the plasma cysteine levels exceeded the normal range in all DS cases. We reported a statistically significant inverse association between CBS enzyme levels and weight (r= - 0.3498, p = 0.0128), hip circumference (r= - 0.3584, p = 0.0106), body mass index (r= - 0.3719, p = 0.0078) and body adiposity index (r= - 0.3183, p = 0.0243). Conclusions Our data suggests that the high concentrations of CBS enzyme together with cysteine modulate the DS obesity presumably through increased hydrogen sulfide production which has recently showed anti-adiposity effects.
Collapse
Affiliation(s)
- Nagwa A. Meguid
- Department of Research on Children with Special Needs, Medical Research and Clinical Studies Institute, National Research Centre, Cairo, Egypt
- CONEM Egypt Child Brain Research Group, National Research Centre, Cairo, Egypt
| | - Maha Hemimi
- Department of Research on Children with Special Needs, Medical Research and Clinical Studies Institute, National Research Centre, Cairo, Egypt
| | - Said A. S. Ghozlan
- Department of Chemistry, Faculty of Science, Cairo University, Giza, Egypt
| | - Wafaa A. Kandeel
- Department of Biological Anthropology, Medical Research and Clinical Studies Institute, National Research Centre, Cairo, Egypt
- Theodor Bilharz Research Institute, Giza, Egypt
| | - Adel F. Hashish
- Department of Research on Children with Special Needs, Medical Research and Clinical Studies Institute, National Research Centre, Cairo, Egypt
| | - Amr S. Gouda
- Department of Biochemical Genetics, Human Genetics and Genome Research Institute, National Research Centre, Cairo, Egypt
| | - Walaa S. Nazim
- Department of Biochemical Genetics, Human Genetics and Genome Research Institute, National Research Centre, Cairo, Egypt
| | - Magda F. Mohamed
- Department of Chemistry (Biochemistry Branch), Faculty of Science, Cairo University, Giza, Egypt
- Department of Chemistry, College of Science and Arts at Khulais, University of Jeddah, Jeddah, Saudi Arabia
| |
Collapse
|
11
|
Su Y, Elshorbagy A, Turner C, Refsum H, Kwok T. The Association of Circulating Amino Acids and Dietary Inflammatory Potential with Muscle Health in Chinese Community-Dwelling Older People. Nutrients 2022; 14:nu14122471. [PMID: 35745201 PMCID: PMC9229609 DOI: 10.3390/nu14122471] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 06/09/2022] [Accepted: 06/10/2022] [Indexed: 02/06/2023] Open
Abstract
Amino acids (AAs) and dietary inflammatory potential play essential roles in muscle health. We examined the associations of dietary inflammatory index (DII) of habitual diet with serum AA profile, and ascertained if the associations between DII and muscle outcomes were mediated by serum AAs, in 2994 older Chinese community-dwelling men and women (mean age 72 years) in Hong Kong. Higher serum branched chain AAs (BCAAs), aromatic AAs and total glutathione (tGSH) were generally associated with better muscle status at baseline. A more pro-inflammatory diet, correlating with higher serum total homocysteine and cystathionine, was directly (90.2%) and indirectly (9.8%) through lower tGSH associated with 4-year decline in hand grip strength in men. Higher tGSH was associated with favorable 4-year changes in hand grip strength, gait speed and time needed for 5-time chair stands in men and 4-year change in muscle mass in women. Higher leucine and isoleucine were associated with decreased risk of sarcopenia in men; the associations were abolished after adjustment for BMI. In older men, perturbations in serum sulfur AAs metabolism may be biomarkers of DII related adverse muscle status, while the lower risk of sarcopenia with higher BCAAs may partly be due to preserved BMI.
Collapse
Affiliation(s)
- Yi Su
- Key Laboratory of Molecular Epidemiology of Hunan Province, School of Medicine, Hunan Normal University, Changsha 410013, China;
| | - Amany Elshorbagy
- Department of Physiology, Faculty of Medicine, University of Alexandria, Alexandria 21526, Egypt;
- Department of Pharmacology, University of Oxford, Oxford OX1 2JD, UK;
| | - Cheryl Turner
- Department of Pharmacology, University of Oxford, Oxford OX1 2JD, UK;
| | - Helga Refsum
- Institute of Basic Medical Sciences, Department of Nutrition, University of Oslo, 0316 Oslo, Norway;
| | - Timothy Kwok
- Department of Medicine and Therapeutics, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong 999077, China
- Jockey Club Centre for Osteoporosis Care and Control, The Chinese University of Hong Kong, Hong Kong 999077, China
- Correspondence: ; Tel.: +852-2632-3128; Fax: +852-2637-3852
| |
Collapse
|
12
|
The association of serum sulfur amino acids and related metabolites with incident diabetes: a prospective cohort study. Eur J Nutr 2022; 61:3161-3173. [PMID: 35415822 DOI: 10.1007/s00394-022-02872-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 03/08/2022] [Indexed: 12/20/2022]
Abstract
AIM Plasma total cysteine (tCys) is associated with fat mass and insulin resistance, whereas taurine is inversely related to diabetes risk. We investigated the association of serum sulfur amino acids (SAAs) and related amino acids (AAs) with incident diabetes. METHODS Serum AAs were measured at baseline in 2997 subjects aged ≥ 65 years. Diabetes was recorded at baseline and after 4 years. Logistic regression evaluated the association of SAAs [methionine, total homocysteine (tHcy), cystathionine, tCys, and taurine] and related metabolites [serine, total glutathione (tGSH), glutamine, and glutamic acid] with diabetes risk. RESULTS Among 2564 subjects without diabetes at baseline, 4.6% developed diabetes. Each SD increment in serum tCys was associated with a 68% higher risk (95% CI 1.27, 2.23) of diabetes [OR for upper vs. lower quartile 2.87 (1.39, 5.91)], after full adjustments (age, sex, other AAs, adiposity, eGFR, physical activity, blood pressure, diet and medication); equivalent ORs for cystathionine were 1.33 (1.08, 1.64) and 1.68 (0.85, 3.29). Subjects who were simultaneously in the upper tertiles of both cystathionine and tCys had a fivefold risk [OR = 5.04 (1.55, 16.32)] of diabetes compared with those in the lowest tertiles. Higher serine was independently associated with a lower risk of developing diabetes [fully adjusted OR per SD = 0.68 (0.54, 0.86)]. Glutamic acid and glutamine showed positive and negative associations, respectively, with incident diabetes in age- and sex-adjusted analysis, but only the glutamic acid association was independent of other confounders [fully adjusted OR per SD = 1.95 (1.19, 3.21); for upper quartile = 7.94 (3.04, 20.75)]. tGSH was inversely related to diabetes after adjusting for age and sex, but not other confounders. No consistent associations were observed for methionine, tHcy or taurine. CONCLUSION Specific SAAs and related metabolites show strong and independent associations with incident diabetes. This suggests that perturbations in the SAA metabolic pathway may be an early marker for diabetes risk.
Collapse
|
13
|
Rafiee Z, García-Serrano AM, Duarte JMN. Taurine Supplementation as a Neuroprotective Strategy upon Brain Dysfunction in Metabolic Syndrome and Diabetes. Nutrients 2022; 14:1292. [PMID: 35334949 PMCID: PMC8952284 DOI: 10.3390/nu14061292] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Revised: 03/14/2022] [Accepted: 03/15/2022] [Indexed: 02/07/2023] Open
Abstract
Obesity, type 2 diabetes, and their associated comorbidities impact brain metabolism and function and constitute risk factors for cognitive impairment. Alterations to taurine homeostasis can impact a number of biological processes, such as osmolarity control, calcium homeostasis, and inhibitory neurotransmission, and have been reported in both metabolic and neurodegenerative disorders. Models of neurodegenerative disorders show reduced brain taurine concentrations. On the other hand, models of insulin-dependent diabetes, insulin resistance, and diet-induced obesity display taurine accumulation in the hippocampus. Given the possible cytoprotective actions of taurine, such cerebral accumulation of taurine might constitute a compensatory mechanism that attempts to prevent neurodegeneration. The present article provides an overview of brain taurine homeostasis and reviews the mechanisms by which taurine can afford neuroprotection in individuals with obesity and diabetes. We conclude that further research is needed for understanding taurine homeostasis in metabolic disorders with an impact on brain function.
Collapse
Affiliation(s)
- Zeinab Rafiee
- Department of Experimental Medical Science, Faculty of Medicine, Lund University, 22100 Lund, Sweden; (Z.R.); (A.M.G.-S.)
- Wallenberg Centre for Molecular Medicine, Lund University, 22100 Lund, Sweden
| | - Alba M. García-Serrano
- Department of Experimental Medical Science, Faculty of Medicine, Lund University, 22100 Lund, Sweden; (Z.R.); (A.M.G.-S.)
- Wallenberg Centre for Molecular Medicine, Lund University, 22100 Lund, Sweden
| | - João M. N. Duarte
- Department of Experimental Medical Science, Faculty of Medicine, Lund University, 22100 Lund, Sweden; (Z.R.); (A.M.G.-S.)
- Wallenberg Centre for Molecular Medicine, Lund University, 22100 Lund, Sweden
| |
Collapse
|
14
|
Pavão ML, Ferin R, Lima A, Baptista J. Cysteine and related aminothiols in cardiovascular disease, obesity and insulin resistance. Adv Clin Chem 2022; 109:75-127. [DOI: 10.1016/bs.acc.2022.03.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
15
|
Chang L, Lin F, Cheng K, Li J, Sun X, Figeys D, Jiang J, Ye Y, Liu J. A simultaneous identification and quantification strategy for determination of sulfhydryl-containing metabolites in normal- and high-fat diet hamsters using stable isotope labeling combined with LC-MS. Anal Chim Acta 2021; 1184:339016. [PMID: 34625243 DOI: 10.1016/j.aca.2021.339016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 07/25/2021] [Accepted: 08/30/2021] [Indexed: 10/20/2022]
Abstract
Sulfur-containing metabolites are related to several physiologic disorders and metabolic diseases. In this study, a simultaneous identification and quantification strategy in one batch for determination of sulfhydryl-containing metabolites was developed using stable isotope labeling combined with liquid chromatography-tandem mass spectrometry (SIL-LC-MS). In the proposed method, a pair of isotope labeling reagents, D0/D5-N-ethylmaleimide (D0/D5-NEM), was used to derivatize sulfhydryl-containing metabolites in blood and plasma of normal- and high-fat-diet (NFD and HFD) hamsters for reduced (-SH) and total (-SH, -S-S-, S-glutathionylated proteins) analysis. Quality control (QC) samples and test samples were prepared for LC-MS analysis. First, both QC samples and stable isotope labeled internal standards were used to monitor the status of the instrument and ensure the reliability of the analysis. Subsequently, an inhouse database containing 45 sulfhydryl-containing metabolites was established by MS1 based on QC samples. Then, qualitatively differential sulfhydryl-containing metabolites were found by MS2 between the NFD and HFD hamsters of the test samples, including 3 in reduced and 8 in total analysis of blood samples, and 2 in reduced and 2 in total analysis of plasma samples. Next, in quantitative analysis, satisfied linearities for 6 sulfhydryl-containing metabolites were obtained with the correlation coefficient (R2) > 0.99 and absolute quantification was carried out. The results showed that glutathione and cysteine have different concentrations in blood and plasma of hamsters. Finally, the correlation of sulfhydryl-containing metabolites with blood lipid and oxidative stress levels was determined, which provided insight into the hyperlipidemia-related oxidative stress. Taken together, the developed method of simultaneous identification with the inhouse database and MS2 and quantification with standards in one batch provides a promising strategy for the analysis of sulfhydryl-containing metabolites in biological samples, which may promote the in-depth investigation on sulfhydryl-containing metabolites and related diseases.
Collapse
Affiliation(s)
- Lu Chang
- Key Laboratory of Systems Bioengineering (Ministry of Education), School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300350, PR China; Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, PR China
| | - Feifei Lin
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China; Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, PR China
| | - Kai Cheng
- SIMM-University of Ottawa Joint Research Center in Systems and Personalized Pharmacology and Ottawa Institute of Systems Biology and Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, Ontario, K1H 8M5, Canada
| | - Jiaomeng Li
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, PR China
| | - Xiaochu Sun
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, PR China
| | - Daniel Figeys
- SIMM-University of Ottawa Joint Research Center in Systems and Personalized Pharmacology and Ottawa Institute of Systems Biology and Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, Ontario, K1H 8M5, Canada
| | - Jianlan Jiang
- Key Laboratory of Systems Bioengineering (Ministry of Education), School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300350, PR China.
| | - Yang Ye
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China; Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, PR China.
| | - Jia Liu
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, PR China; School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310058, PR China.
| |
Collapse
|
16
|
Demerdash HM. Weight regain after bariatric surgery: Promoters and potential predictors. World J Meta-Anal 2021; 9:438-454. [DOI: 10.13105/wjma.v9.i5.438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 06/07/2021] [Accepted: 09/17/2021] [Indexed: 02/06/2023] Open
|
17
|
Elkafrawy H, Mehanna R, Ali F, Barghash A, Dessouky I, Jernerén F, Turner C, Refsum H, Elshorbagy A. Extracellular cystine influences human preadipocyte differentiation and correlates with fat mass in healthy adults. Amino Acids 2021; 53:1623-1634. [PMID: 34519922 PMCID: PMC8521515 DOI: 10.1007/s00726-021-03071-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 08/19/2021] [Indexed: 02/08/2023]
Abstract
Plasma cysteine is associated with human obesity, but it is unknown whether this is mediated by reduced, disulfide (cystine and mixed-disulfides) or protein-bound (bCys) fractions. We investigated which cysteine fractions are associated with adiposity in vivo and if a relevant fraction influences human adipogenesis in vitro. In the current study, plasma cysteine fractions were correlated with body fat mass in 35 adults. Strong positive correlations with fat mass were observed for cystine and mixed disulfides (r ≥ 0.61, P < 0.001), but not the quantitatively major form, bCys. Primary human preadipocytes were differentiated in media containing cystine concentrations varying from 10-50 μM, a range similar to that in plasma. Increasing extracellular cystine (10-50 μM) enhanced mRNA expression of PPARG2 (to sixfold), PPARG1, PLIN1, SCD1 and CDO1 (P = 0.042- < 0.001). Adipocyte lipid accumulation and lipid-droplet size showed dose-dependent increases from lowest to highest cystine concentrations (P < 0.001), and the malonedialdehyde/total antioxidant capacity increased, suggesting increased oxidative stress. In conclusion, increased cystine concentrations, within the physiological range, are positively associated with both fat mass in healthy adults and human adipogenic differentiation in vitro. The potential role of cystine as a modifiable factor regulating human adipocyte turnover and metabolism deserves further study.
Collapse
Affiliation(s)
- Hagar Elkafrawy
- Department of Medical Biochemistry, Faculty of Medicine, Alexandria University, Alexandria, Egypt
- Center of Excellence for Research in Regenerative Medicine and Applications (CERRMA), Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Radwa Mehanna
- Center of Excellence for Research in Regenerative Medicine and Applications (CERRMA), Faculty of Medicine, Alexandria University, Alexandria, Egypt
- Department of Physiology, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Fayrouz Ali
- Department of Medical Biochemistry, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Ayman Barghash
- Department of Medical Biochemistry, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Iman Dessouky
- Department of Medical Biochemistry, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Fredrik Jernerén
- Department of Pharmacology, University of Oxford, Oxford, UK
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | - Cheryl Turner
- Department of Pharmacology, University of Oxford, Oxford, UK
| | - Helga Refsum
- Department of Pharmacology, University of Oxford, Oxford, UK
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Amany Elshorbagy
- Department of Physiology, Faculty of Medicine, Alexandria University, Alexandria, Egypt.
- Department of Pharmacology, University of Oxford, Oxford, UK.
| |
Collapse
|
18
|
Redox Imbalance and Methylation Disturbances in Early Childhood Obesity. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:2207125. [PMID: 34457110 PMCID: PMC8387800 DOI: 10.1155/2021/2207125] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Revised: 07/13/2021] [Accepted: 08/02/2021] [Indexed: 11/29/2022]
Abstract
Obesity is increasing worldwide in prepubertal children, reducing the age of onset of associated comorbidities, including type 2 diabetes. Sulfur-containing amino acids, methionine, cysteine, and their derivatives play important roles in the transmethylation and transsulfuration pathways. Dysregulation of these pathways leads to alterations in the cellular methylation patterns and an imbalanced redox state. Therefore, we tested the hypothesis that one-carbon metabolism is already dysregulated in prepubertal children with obesity. Peripheral blood was collected from 64 children, and the plasma metabolites from transmethylation and transsulfuration pathways were quantified by HPLC. The cohort was stratified by BMI z-scores and HOMA-IR indices into healthy lean (HL), healthy obese (HO), and unhealthy obese (UHO). Fasting insulin levels were higher in the HO group compared to the HL, while the UHO had the highest. All groups presented normal fasting glycemia. Furthermore, high-density lipoprotein (HDL) was lower while triglycerides and lactate levels were higher in the UHO compared to HO subjects. S-adenosylhomocysteine (SAH) and total homocysteine levels were increased in the HO group compared to HL. Additionally, glutathione metabolism was also altered. Free cystine and oxidized glutathione (GSSG) were increased in the HO as compared to HL subjects. Importantly, the adipocyte secretory function was already compromised at this young age. Elevated circulating leptin and decreased adiponectin levels were observed in the UHO as compared to the HO subjects. Some of these alterations were concomitant with alterations in the DNA methylation patterns in the obese group, independent of the impaired insulin levels. In conclusion, our study informs on novel and important metabolic alterations in the transmethylation and the transsulfuration pathways in the early stages of obesity. Moreover, the altered secretory function of the adipocyte very early in life may be relevant in identifying early metabolic markers of disease that may inform on the increased risk for specific future comorbidities in this population.
Collapse
|
19
|
de Oliveira Leite L, Costa Dias Pitangueira J, Ferreira Damascena N, Ribas de Farias Costa P. Homocysteine levels and cardiovascular risk factors in children and adolescents: systematic review and meta-analysis. Nutr Rev 2021; 79:1067-1078. [PMID: 33351941 DOI: 10.1093/nutrit/nuaa116] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
CONTEXT Studies have indicated that homocysteine levels are nontraditional markers for cardiovascular disease. The onset of atherosclerotic disease begins in childhood and adolescence; thus, prevention of its risk factors should occur early. OBJECTIVE This systematic review and meta-analysis was conducted to summarize the association between high homocysteine levels and traditional cardiovascular risk factors in children and adolescents. DATA SOURCES This systematic review and meta-analysis were developed according to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses and the protocol was submitted to PROSPERO. Only observational studies in children and adolescents with homocysteine levels as an exposure variable and cardiovascular risk factors as outcome variables were included and searched in the following electronic bibliographic databases: PubMed/MEDLINE, Web of Science, Embase, Latin American and Caribbean Literature in Health Sciences, Ovid and Scopus. DATA EXTRACTION Two authors independently extracted data from eligible studies. The methodological quality of the studies was assessed using the Newcastle-Ottawa scale. DATA ANALYSIS Seven studies were included in the systematic review; they were published from 1999 to 2017, predominantly were of a cross-sectional design, and mainly evaluated adolescents. In the meta-analysis (n = 6), cross-sectional studies (n = 3) identified that high homocysteine levels were positive and weakly correlated with overweight in children and adolescents (odds ratio, 1.08; 95%CI, 1.04-1.11). CONCLUSION High homocysteine levels were weakly associated with overweight in children and adolescents in the reviewed cross-sectional studies. However, for the other traditional cardiovascular risk factors, the findings, although important, were inconclusive. Additional robust longitudinal studies are recommended to be conducted to better identify these associations. SYSTEMATIC REVIEW REGISTRATION PROSPERO registration no. CRD42018086252.
Collapse
|
20
|
Maternal cysteine intake influenced oxidative status and lipid-related gut microbiota and plasma metabolomics in male suckling piglets. Anim Feed Sci Technol 2021. [DOI: 10.1016/j.anifeedsci.2021.114947] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
21
|
Comas F, Moreno-Navarrete JM. The Impact of H 2S on Obesity-Associated Metabolic Disturbances. Antioxidants (Basel) 2021; 10:antiox10050633. [PMID: 33919190 PMCID: PMC8143163 DOI: 10.3390/antiox10050633] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 04/18/2021] [Accepted: 04/19/2021] [Indexed: 12/20/2022] Open
Abstract
Over the last several decades, hydrogen sulfide (H2S) has gained attention as a new signaling molecule, with extensive physiological and pathophysiological roles in human disorders affecting vascular biology, immune functions, cellular survival, metabolism, longevity, development, and stress resistance. Apart from its known functions in oxidative stress and inflammation, new evidence has emerged revealing that H2S carries out physiological functions by targeting proteins, enzymes, and transcription factors through a post-translational modification known as persulfidation. This review article provides a critical overview of the current state of the literature addressing the role of H2S in obesity-associated metabolic disturbances, with particular emphasis on its mechanisms of action in obesity, diabetes, non-alcoholic fatty liver disease (NAFLD), and cardiovascular diseases.
Collapse
Affiliation(s)
- Ferran Comas
- Department of Diabetes, Endocrinology and Nutrition, Institut d’Investigació Biomèdica de Girona (IdIBGi), CIBEROBN (CB06/03/010) and Instituto de Salud Carlos III (ISCIII), 17007 Girona, Spain;
| | - José María Moreno-Navarrete
- Department of Diabetes, Endocrinology and Nutrition, Institut d’Investigació Biomèdica de Girona (IdIBGi), CIBEROBN (CB06/03/010) and Instituto de Salud Carlos III (ISCIII), 17007 Girona, Spain;
- Department of Medical Sciences, Universitat de Girona, 17003 Girona, Spain
- Correspondence: ; Tel.: +(34)-872-98-70-87
| |
Collapse
|
22
|
Transsulfuration metabolites and the association with incident atrial fibrillation – An observational cohort study among Norwegian patients with stable angina pectoris. Int J Cardiol 2020; 317:75-80. [DOI: 10.1016/j.ijcard.2020.05.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 04/16/2020] [Accepted: 05/04/2020] [Indexed: 11/19/2022]
|
23
|
Olsen T, Turner C, Øvrebø B, Bastani NE, Refsum H, Vinknes KJ. Postprandial effects of a meal low in sulfur amino acids and high in polyunsaturated fatty acids compared to a meal high in sulfur amino acids and saturated fatty acids on stearoyl CoA-desaturase indices and plasma sulfur amino acids: a pilot study. BMC Res Notes 2020; 13:379. [PMID: 32778150 PMCID: PMC7419218 DOI: 10.1186/s13104-020-05222-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 08/04/2020] [Indexed: 01/01/2023] Open
Abstract
Objective The sulfur amino acid (SAA) cysteine is positively related, whereas polyunsaturated fatty acids (PUFAs) are inversely related to activity of the lipogenic enzyme stearoyl-CoA desaturase (SCD). High SCD activity promotes obesity in animals, and plasma activity indices positively associates with fat mass in humans. SCD may thus be a target for dietary intervention with SAA restriction and PUFA enrichment with unknown potential benefits for body composition. We randomized ten healthy individuals to a meal restricted in SAAs and enriched with PUFAs (Cys/Metlow + PUFA) (n = 5) or a meal enriched in SAA and saturated fatty acids (Cys/Methigh + SFA) (n = 5). We measured plasma SCD activity indices (SCD16 and SCD18) and SAAs response hourly from baseline and up to 4 h postprandial. Results SCD16 was unchanged whereas SCD18 tended to increase in the Cys/Metlow + PUFA compared to the Cys/Methigh + SFA group (ptime*group interaction = 0.08). Plasma concentrations of total cysteine fractions including free and reduced cysteine decreased in the Cys/Metlow + PUFA compared to the Cys/Methigh + SFA group (both ptime*group interaction < 0.001). In conclusion, a meal low in SAA but high in PUFAs reduced plasma cysteine fractions but not SCD activity indices. This pilot study can be useful for the design and diet composition of future dietary interventions that targets SCD and SAA. Trial registration ClinicalTrials.gov: NCT02647970, registration date: 6 January 2016
Collapse
Affiliation(s)
- Thomas Olsen
- Department of Nutrition, Institute of Medical Biosciences, University of Oslo, 0372, Oslo, Norway. .,Institute of Medical Biosciences, Domus Medica, Sognsvannsveien 9, 0372, Oslo, Norway.
| | - Cheryl Turner
- Department of Pharmacology, University of Oxford, Oxford, OX1 3QT, UK
| | - Bente Øvrebø
- Department of Nutrition, Institute of Medical Biosciences, University of Oslo, 0372, Oslo, Norway.,Øvrebø Nutrition, 0550, Oslo, Norway
| | - Nasser E Bastani
- Department of Nutrition, Institute of Medical Biosciences, University of Oslo, 0372, Oslo, Norway
| | - Helga Refsum
- Department of Nutrition, Institute of Medical Biosciences, University of Oslo, 0372, Oslo, Norway
| | - Kathrine J Vinknes
- Department of Nutrition, Institute of Medical Biosciences, University of Oslo, 0372, Oslo, Norway
| |
Collapse
|
24
|
Elshorbagy AK, Graham I, Refsum H. Body mass index determines the response of plasma sulfur amino acids to methionine loading. Biochimie 2020; 173:107-113. [DOI: 10.1016/j.biochi.2020.03.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Accepted: 03/02/2020] [Indexed: 01/02/2023]
|
25
|
Olsen T, Øvrebø B, Haj-Yasein N, Lee S, Svendsen K, Hjorth M, Bastani NE, Norheim F, Drevon CA, Refsum H, Vinknes KJ. Effects of dietary methionine and cysteine restriction on plasma biomarkers, serum fibroblast growth factor 21, and adipose tissue gene expression in women with overweight or obesity: a double-blind randomized controlled pilot study. J Transl Med 2020; 18:122. [PMID: 32160926 PMCID: PMC7065370 DOI: 10.1186/s12967-020-02288-x] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Accepted: 02/27/2020] [Indexed: 12/15/2022] Open
Abstract
Background Dietary restriction of methionine and cysteine is a well-described model that improves metabolic health in rodents. To investigate the translational potential in humans, we evaluated the effects of dietary methionine and cysteine restriction on cardiometabolic risk factors, plasma and urinary amino acid profile, serum fibroblast growth factor 21 (FGF21), and subcutaneous adipose tissue gene expression in women with overweight and obesity in a double-blind randomized controlled pilot study. Methods Twenty women with overweight or obesity were allocated to a diet low (Met/Cys-low, n = 7), medium (Met/Cys-medium, n = 7) or high (Met/Cys-high, n = 6) in methionine and cysteine for 7 days. The diets differed only by methionine and cysteine content. Blood and urine were collected at day 0, 1, 3 and 7 and subcutaneous adipose tissue biopsies were taken at day 0 and 7. Results Plasma methionine and cystathionine and urinary total cysteine decreased, whereas FGF21 increased in the Met/Cys-low vs. Met/Cys-high group. The Met/Cys-low group had increased mRNA expression of lipogenic genes in adipose tissue including DGAT1. When we excluded one participant with high fasting insulin at baseline, the Met/Cys-low group showed increased expression of ACAC, DGAT1, and tendencies for increased expression of FASN and SCD1 compared to the Met/Cys-high group. The participants reported satisfactory compliance and that the diets were moderately easy to follow. Conclusions Our data suggest that dietary methionine and cysteine restriction may have beneficial effects on circulating biomarkers, including FGF21, and influence subcutaneous adipose tissue gene expression. These results will aid in the design and implementation of future large-scale dietary interventions with methionine and cysteine restriction. Trial registration ClinicalTrials.gov Identifier: NCT03629392, registration date: 14/08/2018 https://clinicaltrials.gov/ct2/show/NCT03629392.
Collapse
Affiliation(s)
- Thomas Olsen
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Postboks 1046, Blindern, 0317, Oslo, Norway.
| | - Bente Øvrebø
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Postboks 1046, Blindern, 0317, Oslo, Norway
| | - Nadia Haj-Yasein
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Postboks 1046, Blindern, 0317, Oslo, Norway
| | - Sindre Lee
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Postboks 1046, Blindern, 0317, Oslo, Norway
| | - Karianne Svendsen
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Postboks 1046, Blindern, 0317, Oslo, Norway.,The Lipid Clinic, Department of Endocrinology, Morbid Obesity and Preventive Medicine, Oslo University Hospital, OUS HF Aker Sykehus, Postboks 4959, Nydalen, 0424, Oslo, Norway
| | - Marit Hjorth
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Postboks 1046, Blindern, 0317, Oslo, Norway
| | - Nasser E Bastani
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Postboks 1046, Blindern, 0317, Oslo, Norway
| | - Frode Norheim
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Postboks 1046, Blindern, 0317, Oslo, Norway
| | - Christian A Drevon
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Postboks 1046, Blindern, 0317, Oslo, Norway
| | - Helga Refsum
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Postboks 1046, Blindern, 0317, Oslo, Norway
| | - Kathrine J Vinknes
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Postboks 1046, Blindern, 0317, Oslo, Norway
| |
Collapse
|
26
|
Padoin S, de Freitas VH, Cleto DAM, Zeffa AC, Nakamura FY, Andrello AC, de Paula Ramos S. Effects of Futsal Demands on Serum and Salivary Levels of Trace Elements and Minerals Detected by Total Reflection X-Ray Fluorescence. Biol Trace Elem Res 2020; 193:73-80. [PMID: 30924068 DOI: 10.1007/s12011-019-01697-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2018] [Accepted: 02/10/2019] [Indexed: 12/13/2022]
Abstract
The aim of this study was to monitor the circulating and salivary ion concentrations by total reflection X-ray fluorescence (TXRF) in futsal players submitted to the futsal-specific intermittent shuttle protocol (FISP). TXRF may allow identification of changes in ion concentrations induced by physical efforts. Saliva and blood samples of 13 male futsal players were collected before (Pre) and after (Post) the FISP. Salivary and plasma ion levels were detected by TXRF, and differences from Pre to Post (paired t test or Wilcoxon test) and correlations between both biological fluids were determined (P < 0.05). All saliva samples presented phosphorus (P), sulfur (S), chlorine (Cl), potassium (K), calcium (Ca), iron (Fe), zinc (Zn), bromine (Br), and rubidium (Rb). S, Cl, Ca, Fe, Cu, Zn, Br, and Rb were detected in all blood samples. K, Cu, Br, and Rb presented reduced secretion rate from Pre to Post samples (P < 0.05). The salivary concentrations of K (r = - 0.53) and Zn (r = 0.54) were correlated with plasmatic concentrations. After FISP, salivary secretion of S (r = - 0.76), Cl (r = - 0.64), P (r = - 0.67), Mn (r = - 0.74), and Zn (r = 0.69) were correlated with plasma levels. We concluded that TXRF may be used to monitor salivary (P, S, Cl, K, Ca, Fe, Zn, Br, and Rb) and circulating (S, Cl, Ca, Fe, Cu, Zn, Br, and Rb) levels of several elements in futsal athletes. However, an acute bout of futsal-specific physical effort did not significantly imbalance ion concentrations in saliva or plasma.
Collapse
Affiliation(s)
- Susana Padoin
- Master Student in Physical Education, State University of Londrina, Londrina, Brazil
| | | | | | - Aline Campos Zeffa
- Master Student in Odontology, State University of Londrina, Londrina, Brazil
| | - Fábio Yuzo Nakamura
- Department of Medicine and Aging Sciences, University "G. d'Annunzio" Chieti-Pescara, Chieti, Italy
| | | | - Solange de Paula Ramos
- Study Group of Tissue Regeneration, Adaptation and Repair. Center of Biological Sciences, State University of Londrina, Londrina, Brazil.
| |
Collapse
|
27
|
Su Y, Elshorbagy A, Turner C, Refsum H, Chan R, Kwok T. Circulating amino acids are associated with bone mineral density decline and ten-year major osteoporotic fracture risk in older community-dwelling adults. Bone 2019; 129:115082. [PMID: 31622772 PMCID: PMC6925590 DOI: 10.1016/j.bone.2019.115082] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 09/17/2019] [Accepted: 09/26/2019] [Indexed: 11/18/2022]
Abstract
With aging, poor bone mineral density (BMD) and accelerated decrease in BMD are strong risk factors for fracture. Reports of the associations of dietary protein intake with bone strength are inconsistent, possibly owing to differences in protein sources and amino acid (AA) composition. We examined the associations of serum AA with 4-year hip BMD loss and subsequent fracture risk within 10 years in older community-dwelling adults, and further addressed whether lifestyle, dietary protein intake and its source, and body composition would affect the associations. In 1424 men and 1573 women (mean age 72 years), using binary logistic regression, higher serum valine, leucine, isoleucine and tryptophan concentrations were associated (or approaching a borderline significance in case of the last three ones) with less hip BMD decline (defined as BMD loss ≥ 2.8 times the precision error of the BMD measurement at femoral neck) in 4 years later, with the OR (95%CI) /SD of AA increase, ranging from 0.83 (0.75, 0.91) to 0.92 (0.87, 0.98) after multiple adjustments for baseline age, gender, BMI, BMD, estimated glomerular filtration rate (eGFR), dietary protein intake (animal- and plant-derived protein intakes), calcium intake, established lifestyles (physical activity level, smoking and alcohol drinking status), osteoporosis medications, and changes of body fat and lean muscle mass. Higher serum total homocysteine (tHcy) concentration was independently associated with BMD decline 4 years later (OR (95%CI) /SD of 1.16 (1.05, 1.27)). Using multivariate Cox regression, higher serum tryptophan concentration potentially predicted low risk of incident major osteoporotic fractures (MOFs) (HR/SD (95%CI)=0.86 (0.75, 0.98)) after multiple adjustments. Higher serum tHcy was associated with MOFs (HR/SD (95%CI)=1.29 (1.12, 1.50)) risk after multiple adjustments in men. These findings suggest that a specific AA profile correlates with greater BMD and lower subsequent fracture risk, independent of diet and lifestyle factors.
Collapse
Affiliation(s)
- Yi Su
- Department of Medicine and Therapeutics, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Amany Elshorbagy
- Department of Physiology, Faculty of Medicine, University of Alexandria, Alexandria, Egypt
| | - Cheryl Turner
- Department of Pharmacology, University of Oxford, Oxford, United Kingdom
| | - Helga Refsum
- Institute of Basic Medical Sciences, Department of Nutrition, University of Oslo, Oslo, Norway
| | - Ruth Chan
- Department of Medicine and Therapeutics, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Timothy Kwok
- Department of Medicine and Therapeutics, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China; Jockey Club Centre for Osteoporosis Care and Control, The Chinese University of Hong Kong, Hong Kong, China.
| |
Collapse
|
28
|
Belikova J, Lizogub V, Kuzminets A, Lavrenchuk I. Normalization of heart rate variability with taurine and meldonium complex in post-infarction patients with type 2 diabetes mellitus. J Med Life 2019; 12:290-295. [PMID: 31666833 PMCID: PMC6814880 DOI: 10.25122/jml-2019-0052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
The purpose of this study is to scrutiny the Dynamics of heart rate variability (HRV) in patients with PICS with 2nd type DM against the background of Taurine (TN) and meldonium (ME). The results of the investigations prove the decrease of the oxidative stress, which is basis of DACN, under the influence of sulfur-containing amino acid taurine (TN), and meldonium (ME) — a competitive inhibitor of gamma-butyrobetaine hydroxylase. Biochemical mechanisms of synergistic action of ME and TN are also described. The results of the studies of 98 patients with PICS and concomitant 2nd type diabetes mellitus were analyzed. They were distributed by simple randomization method into two groups, comparable according to age and sex: the main group (MG) (n = 68): and group of comparison (GoC) (n = 30). HRV was evaluated twice daily at the Cardiosense HMEGG system: at baseline and after 12 weeks of treatment. For the assessment of HRV the frequency and spectral parameters were used. While evaluating the different methods of treatment, their influence on the range of spectral and time indices of HRV was determined (p = 0.001 by the criterion of Kruskall-Wallis). It was learned that the combined application of ME and TN gives a statistically significant (p <0.01) increase of SDNN, HF at night, pNN — on 50% by day (p <0.01, p <0.001 and p <0.01 respectively), and statistically significant decrease in LF at night, compared to GHG.
Collapse
Affiliation(s)
- Juliia Belikova
- Department of Internal Medicine No 4, Bogomolets National Medical University, Kyiv, Ukraine
| | - Victor Lizogub
- Department of Internal Medicine No 4, Bogomolets National Medical University, Kyiv, Ukraine
| | - Andrii Kuzminets
- Department of the Therapy, Infectious Disease and Dermatology Postgraduate Education, Bogomolets National Medical University, Kyiv, Ukraine
| | - Iryna Lavrenchuk
- Department of the Therapy, Infectious Disease and Dermatology Postgraduate Education, Bogomolets National Medical University, Kyiv, Ukraine
| |
Collapse
|
29
|
Dhar I, Lysne V, Svingen GFT, Ueland PM, Gregory JF, Bønaa KH, Nygård OK. Elevated plasma cystathionine is associated with increased risk of mortality among patients with suspected or established coronary heart disease. Am J Clin Nutr 2019; 109:1546-1554. [PMID: 31005968 DOI: 10.1093/ajcn/nqy391] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Accepted: 12/26/2018] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Elevated circulating cystathionine levels are related to atherosclerotic cardiovascular disease, a leading cause of death globally. OBJECTIVE We investigated whether plasma cystathionine was associated with mortality in patients with suspected or established coronary heart disease (CHD). METHODS Data from 2 independent cohorts of patients with suspected stable angina pectoris (SAP) (3033 patients; median 10.7 y follow-up; 648 deaths) or acute myocardial infarction (AMI) (3670 patients; median 7.0 y follow-up; 758 deaths) were included. Hazard ratios with 95% CIs per SD increment of log-transformed cystathionine were calculated using Cox regression modeling. Endpoint data was obtained from a national health registry. RESULTS Among patients with SAP, there was a positive association between plasma cystathionine and death (age- and sex-adjusted HRs [95% CI] per SD: 1.23 [1.14, 1.32], 1.29 [1.16, 1.44], and 1.17 [1.05, 1.29] for total, cardiovascular, and noncardiovascular mortality, respectively). Corresponding risk estimates were 1.28 (1.19, 1.37) for all-cause, 1.33 (1.22, 1.45) for cardiovascular, and 1.19 (1.06, 1.34) for noncardiovascular death among AMI patients. In both cohorts, estimates were slightly attenuated after multivariate adjustments for established CHD risk factors. Subgroup analyses showed that the relation between cystathionine and all-cause mortality in SAP patients was stronger among nonsmokers and those with lower plasma concentration of pyridoxal-5'-phosphate (P-interaction ≤ 0.01 for both). CONCLUSIONS Elevated plasma cystathionine is associated with both cardiovascular and noncardiovascular mortality among patients with suspected or established CHD. The joint risk associations of high plasma cystathionine with lifestyle factors and impaired vitamin B-6 status on mortality need further investigation. This trial was registered at clinicaltrials.gov as NCT00354081 and NCT00266487.
Collapse
Affiliation(s)
- Indu Dhar
- Department of Clinical Science, KG Jebsen Centre for Diabetes Research, University of Bergen, Bergen, Norway.,KG Jebsen Centre for Diabetes Research, University of Bergen, Bergen, Norway
| | - Vegard Lysne
- Department of Clinical Science, KG Jebsen Centre for Diabetes Research, University of Bergen, Bergen, Norway
| | - Gard F T Svingen
- Department of Heart Disease, Haukeland University Hospital, Bergen, Norway
| | - Per M Ueland
- Department of Clinical Science, KG Jebsen Centre for Diabetes Research, University of Bergen, Bergen, Norway.,Bevital AS, Bergen, Norway
| | - Jesse F Gregory
- Food Science and Human Nutrition Department, University of Florida, Gainesville, FL
| | - Kaare H Bønaa
- Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Ottar K Nygård
- Department of Clinical Science, KG Jebsen Centre for Diabetes Research, University of Bergen, Bergen, Norway.,KG Jebsen Centre for Diabetes Research, University of Bergen, Bergen, Norway.,Department of Heart Disease, Haukeland University Hospital, Bergen, Norway
| |
Collapse
|
30
|
Olsen T, Øvrebø B, Turner C, Bastani NE, Refsum H, Vinknes KJ. Combining Dietary Sulfur Amino Acid Restriction with Polyunsaturated Fatty Acid Intake in Humans: A Randomized Controlled Pilot Trial. Nutrients 2018; 10:nu10121822. [PMID: 30477080 PMCID: PMC6315936 DOI: 10.3390/nu10121822] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Revised: 11/19/2018] [Accepted: 11/20/2018] [Indexed: 01/09/2023] Open
Abstract
Dietary and plasma total cysteine (tCys) have been associated with adiposity, possibly through interaction with stearoyl–CoA desaturase (SCD), which is an enzyme that is involved in fatty acid and energy metabolism. We evaluated the effect of a dietary intervention with low cysteine and methionine and high polyunsaturated fatty acids (PUFAs) on plasma and urinary sulfur amino acids and SCD activity indices. Fourteen normal-weight healthy subjects were randomized to a seven-day diet low in cysteine and methionine and high in PUFAs (Cys/Metlow + PUFA), or high in saturated fatty acids (SFA), cysteine, and methionine (Cys/Methigh + SFA). Compared with the Cys/Methigh + SFA group, plasma methionine and cystathionine decreased (p-values < 0.05), whereas cystine tended to increase (p = 0.06) in the Cys/Metlow + PUFA group. Plasma total cysteine (tCys) was not significantly different between the groups. Urinary cysteine and taurine decreased in the Cys/Metlow + PUFA group compared with the Cys/Methigh + SFA group (p-values < 0.05). Plasma SCD-activity indices were not different between the groups, but the change in cystine correlated with the SCD-16 index in the Cys/Metlow + PUFA group. A diet low in methionine and cysteine decreased plasma methionine and urinary cysteine and taurine. Plasma tCys was unchanged, suggesting that compensatory mechanisms are activated during methionine and cysteine restriction to maintain plasma tCys.
Collapse
Affiliation(s)
- Thomas Olsen
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, 0372 Oslo, Norway.
| | - Bente Øvrebø
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, 0372 Oslo, Norway.
- Øvrebø Nutrition, 0550 Oslo, Norway.
| | - Cheryl Turner
- Department of Pharmacology, University of Oxford, Oxford OX1 3QT, UK.
| | - Nasser E Bastani
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, 0372 Oslo, Norway.
| | - Helga Refsum
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, 0372 Oslo, Norway.
| | - Kathrine J Vinknes
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, 0372 Oslo, Norway.
| |
Collapse
|
31
|
Liu H, Wang W, Zhang C, Xu C, Duan H, Tian X, Zhang D. Heritability and Genome-Wide Association Study of Plasma Cholesterol in Chinese Adult Twins. Front Endocrinol (Lausanne) 2018; 9:677. [PMID: 30498476 PMCID: PMC6249314 DOI: 10.3389/fendo.2018.00677] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Accepted: 10/29/2018] [Indexed: 12/14/2022] Open
Abstract
Dyslipidemia represents a strong and independent risk factor for cardiovascular disease. Plasma cholesterol, such as total cholesterol (TC), low density lipoprotein cholesterol (LDL-C), and high density lipoprotein cholesterol (HDL-C), is the common indicator of diagnosing dyslipidemia. Here based on 382 Chinese twin pairs, we explored the magnitude of genetic impact on TC, HDL-C, LDL-C variation and further searched for genetic susceptibility loci for them using genome-wide association study (GWAS). The ACE model was the best fit model with additive genetic parameter (A) accounting for 26.6%, common or shared environmental parameter (C) accounting for 47.8%, unique/non-shared environmental parameter (E) accounting for 25.6% for the variance in HDL-C. The AE model was the best fit model for TC (A: 61.4%; E: 38.6%) and LDL-C (A: 65.5%; E: 34.5%). While no SNPs reached the genome-wide significance level (P < 5 × 10-8), 8, 14, 9 SNPs exceeded the suggestive significance level (P < 1 × 10-5) for TC, HDL-C, LDL-C, respectively. The promising genetic regions for TC, HDL-C, LDL-C were on chromosome 11 around rs7107698, chromosome 5 around rs12518218, chromosome 2 around rs10490120, respectively. Gene-based analysis found 1038, 1033 and 1090 genes nominally associated with TC, HDL-C, LDL-C (P < 0.05), especially FAF1, KLKB1 for TC, KLKB1 for HDL-C, and NTRK1, FAF1, SNTB2 for LDL-C, respectively. The number of common related genes among TC, HDL-C and LDL-C was 71, including FAF1, KLKB1, etc. Pathway enrichment analysis discovered known related pathways-zinc transporters, metal ion SLC transporters for TC, cell adhesion molecules CAMs, IL-6 signaling for HDL, FC epsilon RI signaling pathway, NFAT pathway for LDL, respectively. In conclusion, the TC and LDL-C level is moderately heritable and the HDL-C level is lowly heritable in Chinese population. The genomic loci, functional genes and pathways are identified to account for the heritability of plasma cholesterol level. Our findings provide important insights into plasma cholesterol molecular physiology and expect future research to replicate and validate our results.
Collapse
Affiliation(s)
- Hui Liu
- Department of Epidemiology and Health Statistics, Public Health College, Qingdao University, Qingdao, China
| | - Weijing Wang
- Department of Epidemiology and Health Statistics, Public Health College, Qingdao University, Qingdao, China
| | - Caixia Zhang
- Department of Epidemiology and Health Statistics, Public Health College, Qingdao University, Qingdao, China
| | - Chunsheng Xu
- Qingdao Municipal Centre for Disease Control and Prevention, Qingdao, China
| | - Haiping Duan
- Qingdao Municipal Centre for Disease Control and Prevention, Qingdao, China
| | - Xiaocao Tian
- Qingdao Municipal Centre for Disease Control and Prevention, Qingdao, China
| | - Dongfeng Zhang
- Department of Epidemiology and Health Statistics, Public Health College, Qingdao University, Qingdao, China
| |
Collapse
|
32
|
Dhar I, Svingen GF, Ueland PM, Lysne V, Svenningsson MM, Tell GS, Nygård OK. Plasma Cystathionine and Risk of Incident Stroke in Patients With Suspected Stable Angina Pectoris. J Am Heart Assoc 2018; 7:e008824. [PMID: 30371177 PMCID: PMC6201441 DOI: 10.1161/jaha.118.008824] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Accepted: 07/20/2018] [Indexed: 12/14/2022]
Abstract
Background Cystathionine is an intermediate product in the transsulfuration pathway and formed during the B6-dependent conversion of methionine to cysteine. Elevated plasma cystathionine has been related to atherosclerosis, which is a major etiological factor for ischemic stroke. However, the role of cystathionine in stroke development is unknown. Therefore, we prospectively assessed the association of circulating levels of cystathionine with risk of total and ischemic stroke. Methods and Results Two-thousand thirty-six patients (64% men; median age, 62 years) undergoing coronary angiography for suspected stable angina pectoris were included. Stroke cases were identified by linkage to the CVDNOR (Cardiovascular Disease in Norway) project. Hazard ratios with confidence intervals (95% confidence interval) were estimated by using Cox-regression analyses. During 7.3 years of median follow-up, 124 (6.1%) incident strokes were ascertained, which comprised 100 cases of ischemic stroke. There was a positive association of plasma cystathionine with risk of total stroke and ischemic stroke. Comparing the fourth versus the first cystathionine quartiles, age- and sex-adjusted hazard ratios (95% confidence interval) were 2.11 (1.19-3.75) and 2.56 (1.31-4.99) for total and ischemic stroke, respectively. Additional adjustment for major stroke risk factors only slightly attenuated the associations, which tended to be stronger in patients without previous or existing atrial fibrillation at baseline (hazard ratio [95% confidence interval], 2.43 [1.27-4.65] and 2.88 [1.39-5.98] for total and ischemic stroke, respectively). Conclusions In patients with suspected stable angina pectoris, plasma cystathionine was independently related to increased risk of total stroke and, in particular, ischemic stroke. Clinical Trial Registration URL : http://www.clinicaltrials.gov . Unique identifier: NCT 00354081.
Collapse
Affiliation(s)
- Indu Dhar
- Department of Clinical ScienceUniversity of BergenNorway
- KG Jebsen Centre for Diabetes ResearchUniversity of BergenNorway
| | - Gard F.T. Svingen
- Department of Heart DiseaseHaukeland University HospitalBergenNorway
| | - Per M. Ueland
- Department of Clinical ScienceUniversity of BergenNorway
- Bevital ASBergenNorway
| | - Vegard Lysne
- Department of Clinical ScienceUniversity of BergenNorway
| | | | - Grethe S. Tell
- Department of Global Public Health and Primary CareUniversity of BergenNorway
- Norwegian Institute of Public HealthBergenNorway
| | - Ottar K. Nygård
- Department of Clinical ScienceUniversity of BergenNorway
- KG Jebsen Centre for Diabetes ResearchUniversity of BergenNorway
- Department of Heart DiseaseHaukeland University HospitalBergenNorway
| |
Collapse
|
33
|
Li YC, Li YZ, Li R, Lan L, Li CL, Huang M, Shi D, Feng RN, Sun CH. Dietary Sulfur-Containing Amino Acids Are Associated with Higher Prevalence of Overweight/Obesity in Northern Chinese Adults, an Internet-Based Cross-Sectional Study. ANNALS OF NUTRITION AND METABOLISM 2018; 73:44-53. [PMID: 29879713 DOI: 10.1159/000490194] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Accepted: 05/14/2018] [Indexed: 01/19/2023]
Abstract
BACKGROUND/AIMS Elevation of plasma sulfur-containing amino acids (SAAs) is generally associated with higher body mass index (BMI) and unfavorable lipid profiles. It is not known how dietary SAAs relate to these associations in humans. METHODS A convenient tool named internet-based dietary questionnaire for Chinese (IDQC) was used to estimate dietary SAAs intake. A total of 936 participants were randomly recruited and asked to complete the IDQC. Furthermore, 90 subjects were randomly selected to perform a subgroup study. The associations between dietary SAAs and prevalence of obesity, lipid profiles, and status of insulin resistance (IR), inflammation and oxidative stress were assessed. RESULTS Dietary total SAAs and cysteine of overweight/obese participants were significantly higher. Dietary total SAAs and cysteine were positively associated with BMI and waist circumference. Higher dietary total SAAs were associated with higher prevalence of overweight/obesity. Higher dietary total SAAs and cysteine also associated with higher serum triglyceride (total cholesterol), low density lipoprotein, fasting blood glucose, 2 h-postprandial glucose, and homeostasis model assessment of IR. In the subgroup study, positive associations between dietary SAAs and inflammation biomarkers were also observed. CONCLUSIONS Dietary SAAs are associated with higher prevalence of overweight/obesity, unfavorable lipid profiles and status of IR, and inflammation.
Collapse
Affiliation(s)
- Yan-Chuan Li
- Department of Nutrition and Food Hygiene, School of Public Health, Harbin Medical University, Harbin, China.,Department of Nutrition and Food Hygiene, School of Public Health, Hainan Medical University, Haikou, China
| | - Yu-Zheng Li
- Department of Nutrition and Food Hygiene, School of Public Health, Harbin Medical University, Harbin, China
| | - Rui Li
- Department of Food and School Hygiene, Dalian Center for Disease Control and Prevention, Dalian, China
| | - Li Lan
- Department of Chronic Disease Prevention and Control, Harbin Center for Disease Control and Prevention, Harbin, China
| | - Chun-Long Li
- Department of General Surgery, the Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Min Huang
- Department of Nutrition and Food Hygiene, School of Public Health, Harbin Medical University, Harbin, China
| | - Dan Shi
- Department of Nutrition and Food Hygiene, School of Public Health, Harbin Medical University, Harbin, China
| | - Ren-Nan Feng
- Department of Nutrition and Food Hygiene, School of Public Health, Harbin Medical University, Harbin, China
| | - Chang-Hao Sun
- Department of Nutrition and Food Hygiene, School of Public Health, Harbin Medical University, Harbin, China
| |
Collapse
|
34
|
Dhar I, Svingen GFT, Pedersen ER, DeRatt B, Ulvik A, Strand E, Ueland PM, Bønaa KH, Gregory JF, Nygård OK. Plasma cystathionine and risk of acute myocardial infarction among patients with coronary heart disease: Results from two independent cohorts. Int J Cardiol 2018; 266:24-30. [PMID: 29728335 DOI: 10.1016/j.ijcard.2018.04.083] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Revised: 04/06/2018] [Accepted: 04/18/2018] [Indexed: 01/02/2023]
Abstract
BACKGROUND Cystathionine is a thio-ether and a metabolite formed from homocysteine during transsulfuration. Elevated plasma cystathionine levels are reported in patients with cardiovascular disease; however prospective relationships with acute myocardial infarction (AMI) are unknown. We investigated associations between plasma cystathionine and AMI among patients with suspected and/or verified coronary heart disease (CHD). METHODS Subjects from two independent cohort studies, the Western Norway Coronary Angiography Cohort (WECAC) (3033 patients with stable angina pectoris; 263 events within 4.8 years of median follow-up) and the Norwegian Vitamin Trial (NORVIT) (3670 patients with AMI; 683 events within 3.2 years of median follow-up) were included. RESULTS In both cohorts, plasma cystathionine was associated with several traditional CHD risk factors (P < 0.001). Comparing the cystathionine quartile 4 to 1, age and gender adjusted hazard ratios (95% confidence intervals) for AMI were 2.08 (1.43-3.03) and 1.41 (1.12-1.76) in WECAC and NORVIT, respectively. Additional adjustment for traditional risk factors slightly attenuated the risk estimates, which were generally stronger in both cohorts among non-smokers, patients with higher age, and lower BMI or PLP status (P-interaction ≤ 0.04). Risk associations also tended to be stronger in patients not treated with B-vitamins. Additionally, in a subset of 80 WECAC patients, plasma cystathionine associated strongly negatively with glutathione, an important antioxidant and positively with lanthionine, a marker of H2S production (P < 0.001). CONCLUSIONS Plasma cystathionine is associated with increased risk of AMI among patients with either suspected or verified coronary heart disease, and is possibly related to altered redox homeostasis.
Collapse
Affiliation(s)
- Indu Dhar
- Department of Clinical Science, University of Bergen, Bergen, Norway; KG Jebsen Centre for Diabetes Research, University of Bergen, Bergen, Norway.
| | - Gard F T Svingen
- Department of Heart Disease, Haukeland University Hospital, Bergen, Norway
| | - Eva R Pedersen
- Department of Heart Disease, Haukeland University Hospital, Bergen, Norway
| | - Barbara DeRatt
- Food Science and Human Nutrition Department, University of Florida, Gainesville, FL
| | | | - Elin Strand
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Per M Ueland
- Department of Clinical Science, University of Bergen, Bergen, Norway; Bevital AS, Bergen, Norway
| | - Kaare H Bønaa
- Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Jesse F Gregory
- Food Science and Human Nutrition Department, University of Florida, Gainesville, FL
| | - Ottar K Nygård
- Department of Clinical Science, University of Bergen, Bergen, Norway; KG Jebsen Centre for Diabetes Research, University of Bergen, Bergen, Norway; Department of Heart Disease, Haukeland University Hospital, Bergen, Norway
| |
Collapse
|
35
|
Costa PRF, Kinra S, D'Almeida V, Assis AMO. Serum Homocysteine and Cysteine Levels and Anthropometric Changes: A Longitudinal Study among Brazilian Children and Adolescents. J Am Coll Nutr 2017; 37:80-86. [PMID: 29087241 DOI: 10.1080/07315724.2017.1360806] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
OBJECTIVE This article aimed to identify the influence of high serum homocysteine (Hcy) and cysteine (Cys) levels, alone or in conjunction, on changes in anthropometric parameters in children and adolescents over a 12-month follow-up period. METHODS This is a cohort study involving 483 boys and girls 7-15 years of age. The outcome variables were body mass index (BMI)-for-age and waist circumference (WC) and the principal exposure variables were serum Hcy and Cys levels, alone or in conjunction. A generalized estimating equation (GEE) approach was used to identify longitudinal changes in the outcome variables. RESULTS Irrespective of age, sex, socioeconomic conditions, dietary intake, or the practice of physical activity, the children and adolescents in the fifth quintile of distribution of Hcy levels had a z-score increase of 0.50 (p < 0.01) and a 3.62 cm increase (p < 0.01) in mean BMI-for-age and WC, respectively, over the period of the study. In individuals with Cys values above the fifth quintile, a z-score increase of 0.59 (p < 0.01) and a 5 cm increase (p < 0.01) were found in BMI-for-age and WC, respectively. When serum Hcy and Cys levels were both above the fifth quintile of distribution, a z-score increase of 0.87 (p < 0.01) and a 6.57 cm increase (p < 0.01) were found in mean BMI-for-age and WC, respectively, over the 12-month follow-up. CONCLUSION High serum Hcy and Cys levels contributed to an increase in BMI-for-age and WC in children and adolescents over a 12-month follow-up period, with these increases being even greater when these 2 biochemical parameters were simultaneously high.
Collapse
Affiliation(s)
- Priscila R F Costa
- a Public Health Institute (ISC) , Federal University of Bahia , Salvador-Ba , Brazil.,b Department of Nutrition Science , Federal University of Bahia , Salvador-Ba , Brazil
| | - Sanjay Kinra
- c Department of Non-communicable Disease Epidemiology , London School of Hygiene and Tropical Medicine , London , United Kingdom
| | - Vânia D'Almeida
- d Department of Psychobiology , Federal University of São Paulo , São Paulo , Brazil
| | - Ana Marlúcia O Assis
- b Department of Nutrition Science , Federal University of Bahia , Salvador-Ba , Brazil
| |
Collapse
|
36
|
Myocardial metabolic alterations in mice with diet-induced atherosclerosis: linking sulfur amino acid and lipid metabolism. Sci Rep 2017; 7:13597. [PMID: 29051579 PMCID: PMC5648757 DOI: 10.1038/s41598-017-13991-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Accepted: 10/04/2017] [Indexed: 02/08/2023] Open
Abstract
Atherosclerosis is a leading cause of cardiovascular disease (CVD), but the effect of diet on the atherosclerotic heart’s metabolism is unclear. We used an integrated metabolomics and lipidomics approach to evaluate metabolic perturbations in heart and serum from mice fed an atherogenic diet (AD) for 8, 16, and 25 weeks. Nuclear magnetic resonance (NMR)-based metabolomics revealed significant changes in sulfur amino acid (SAA) and lipid metabolism in heart from AD mice compared with heart from normal diet mice. Higher SAA levels in AD mice were quantitatively verified using liquid chromatography-mass spectrometry (LC/MS). Lipidomic profiling revealed that fatty acid and triglyceride (TG) levels in the AD group were altered depending on the degree of unsaturation. Additionally, levels of SCD1, SREBP-1, and PPARγ were reduced in AD mice after 25 weeks, while levels of reactive oxygen species were elevated. The results suggest that a long-term AD leads to SAA metabolism dysregulation and increased oxidative stress in the heart, causing SCD1 activity suppression and accumulation of toxic TGs with a low degree of unsaturation. These findings demonstrate that the SAA metabolic pathway is a promising therapeutic target for CVD treatment and that metabolomics can be used to investigate the metabolic signature of atherosclerosis.
Collapse
|
37
|
Ribas de Farias Costa P, Kinra S, D'Almeida V, Oliveira Assis AM. Serum homocysteine and cysteine levels and changes in the lipid profile of children and adolescents over a 12-month follow-up period. Clin Nutr ESPEN 2017; 21:13-19. [PMID: 30014864 DOI: 10.1016/j.clnesp.2017.07.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Revised: 02/01/2017] [Accepted: 07/11/2017] [Indexed: 10/19/2022]
Abstract
INTRODUCTION There is evidence of associations between high serum homocysteine (Hcy) and cysteine (Cys) levels and changes in the lipid profile of adults; however, little information is available with respect to these associations in the pediatric age group. OBJECTIVE To identify the effect of high baseline serum Hcy and Cys levels, alone or in conjunction, on the lipid profile of children and adolescents over a 12-month follow-up period. METHODS A cohort study involving 540 boys and girls of 7-15 years of age was conducted over 12 months. The outcome variables were the lipid indicators and the principal exposure variable was serum Hcy and Cys levels. A generalized estimating equation (GEE) approach was used to identify the associations of interest. RESULTS Irrespective of age, sex, socioeconomic status, diet or anthropometric status, when serum Hcy levels were above the 5th quintile of distribution at baseline, HDL-cholesterol decreased by a mean of 2.91 mg/dl (p < 0.01), while triglyceride levels increased by a mean of 1.58 mg/dl (p < 0.01) over the 12-month follow-up period. In individuals with high baseline Cys levels, there was a reduction of 1.95 mg/dl (p < 0.01) and an increase of 1.76 mg/dl (p < 0.01) in mean serum HDL-cholesterol and triglyceride levels, respectively, over the 12-month period. When serum Hcy and Cys levels were both above the 5th quintile of distribution at baseline, there was a reduction of 3.65 mg/dl (p < 0.01) in mean HDL-cholesterol and an increase of 3.53 mg/dl (p < 0.01) in mean triglyceride levels in the 12 months of follow-up. CONCLUSIONS High baseline serum Hcy and Cys levels resulted in a reduction in mean HDL-cholesterol levels and an increase in mean triglycerides levels over the 12 months of follow-up in children and adolescents, with these alterations being greater when these two biochemical parameters were simultaneously high at baseline.
Collapse
Affiliation(s)
- Priscila Ribas de Farias Costa
- Public Health Institute (ISC), Federal University of Bahia. Basilio da Gama Street, Canela, 40110-040, Salvador, BA, Brazil; Department of Nutrition Science, Federal University of Bahia. Araújo Pinho Avenue, 32, 40110-090, Canela, Salvador, BA, Brazil.
| | - Sanjay Kinra
- Department of Non-communicable Disease Epidemiology, London School of Hygiene and Tropical Medicine, Keppel Street, WC1E 7HT, London, UK.
| | - Vânia D'Almeida
- Department of Psychobiology, Federal University of São Paulo, Napoleão de Barros Street, 925, Vila Clementino, 04024002, São Paulo, SP, Brazil.
| | - Ana Marlúcia Oliveira Assis
- Department of Nutrition Science, Federal University of Bahia. Araújo Pinho Avenue, 32, 40110-090, Canela, Salvador, BA, Brazil.
| |
Collapse
|
38
|
Exploring the Lean Phenotype of Glutathione-Depleted Mice: Thiol, Amino Acid and Fatty Acid Profiles. PLoS One 2016; 11:e0163214. [PMID: 27788147 PMCID: PMC5082875 DOI: 10.1371/journal.pone.0163214] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Accepted: 09/05/2016] [Indexed: 11/20/2022] Open
Abstract
BACKGROUND Although reduced glutathione (rGSH) is decreased in obese mice and humans, block of GSH synthesis by buthionine sulfoximine (BSO) results in a lean, insulin-sensitive phenotype. Data is lacking about the effect of BSO on GSH precursors, cysteine and glutamate. Plasma total cysteine (tCys) is positively associated with stearoyl-coenzyme A desaturase (SCD) activity and adiposity in humans and animal models. OBJECTIVE To explore the phenotype, amino acid and fatty acid profiles in BSO-treated mice. DESIGN Male C3H/HeH mice aged 11 weeks were fed a high-fat diet with or without BSO in drinking water (30 mmol/L) for 8 weeks. Amino acid and fatty acid changes were assessed, as well as food consumption, energy expenditure, locomotor activity, body composition and liver vacuolation (steatosis). RESULTS Despite higher food intake, BSO decreased particularly fat mass but also lean mass (both P<0.001), and prevented fatty liver vacuolation. Physical activity increased during the dark phase. BSO decreased plasma free fatty acids and enhanced insulin sensitivity. BSO did not alter liver rGSH, but decreased plasma total GSH (tGSH) and rGSH (by ~70%), and liver tGSH (by 82%). Glutamate accumulated in plasma and liver. Urine excretion of cysteine and its precursors was increased by BSO. tCys, rCys and cystine decreased in plasma (by 23-45%, P<0.001 for all), but were maintained in liver, at the expense of decreased taurine. Free and total plasma concentrations of the SCD products, oleic and palmitoleic acids were decreased (by 27-38%, P <0.001 for all). CONCLUSION Counterintuitively, block of GSH synthesis decreases circulating tCys, raising the question of whether the BSO-induced obesity-resistance is linked to cysteine depletion. Cysteine-supplementation of BSO-treated mice is warranted to dissect the effects of cysteine and GSH depletion on energy metabolism.
Collapse
|
39
|
Niewiadomski J, Zhou JQ, Roman HB, Liu X, Hirschberger LL, Locasale JW, Stipanuk MH. Effects of a block in cysteine catabolism on energy balance and fat metabolism in mice. Ann N Y Acad Sci 2016; 1363:99-115. [PMID: 26995761 DOI: 10.1111/nyas.13021] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Revised: 12/08/2015] [Accepted: 01/19/2016] [Indexed: 01/14/2023]
Abstract
To gain further insights into the effects of elevated cysteine levels on energy metabolism and the possible mechanisms underlying these effects, we conducted studies in cysteine dioxygenase (Cdo1)-null mice. Cysteine dioxygenase (CDO) catalyzes the first step of the major pathway for cysteine catabolism. When CDO is absent, tissue and plasma cysteine levels are elevated, resulting in enhanced flux of cysteine through desulfhydration reactions. When Cdo1-null mice were fed a high-fat diet, they gained more weight than their wild-type controls, regardless of whether the diet was supplemented with taurine. Cdo1-null mice had markedly lower leptin levels, higher feed intakes, and markedly higher abundance of hepatic stearoyl-CoA desaturase 1 (SCD1) compared to wild-type control mice, and these differences were not affected by the fat or taurine content of the diet. Thus, reported associations of elevated cysteine levels with greater weight gain and with elevated hepatic Scd1 expression are also seen in the Cdo1-null mouse model. Hepatic accumulation of acylcarnitines suggests impaired mitochondrial β-oxidation of fatty acids in Cdo1-null mice. The strong associations of elevated cysteine levels with excess H2 S production and impairments in energy metabolism suggest that H2 S signaling could be involved.
Collapse
Affiliation(s)
| | - James Q Zhou
- Division of Nutritional Sciences, Cornell University, Ithaca, New York
| | - Heather B Roman
- Division of Nutritional Sciences, Cornell University, Ithaca, New York
| | - Xiaojing Liu
- Division of Nutritional Sciences, Cornell University, Ithaca, New York
| | | | - Jason W Locasale
- Division of Nutritional Sciences, Cornell University, Ithaca, New York
| | - Martha H Stipanuk
- Division of Nutritional Sciences, Cornell University, Ithaca, New York
| |
Collapse
|
40
|
Glutathionyl systems and metabolic dysfunction in obesity. Nutr Rev 2015; 73:858-68. [DOI: 10.1093/nutrit/nuv042] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Accepted: 04/18/2015] [Indexed: 12/18/2022] Open
|
41
|
Xiao Y, Su X, Huang W, Zhang J, Peng C, Huang H, Wu X, Huang H, Xia M, Ling W. Role of S-adenosylhomocysteine in cardiovascular disease and its potential epigenetic mechanism. Int J Biochem Cell Biol 2015; 67:158-66. [PMID: 26117455 DOI: 10.1016/j.biocel.2015.06.015] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Revised: 06/08/2015] [Accepted: 06/16/2015] [Indexed: 12/28/2022]
Abstract
Transmethylation reactions utilize S-adenosylmethionine (SAM) as a methyl donor and are central to the regulation of many biological processes: more than fifty SAM-dependent methyltransferases methylate a broad spectrum of cellular compounds including DNA, histones, phospholipids and other small molecules. Common to all SAM-dependent transmethylation reactions is the release of the potent inhibitor S-adenosylhomocysteine (SAH) as a by-product. SAH is reversibly hydrolyzed to adenosine and homocysteine by SAH hydrolase. Hyperhomocysteinemia is an independent risk factor for cardiovascular disease. However, a major unanswered question is if homocysteine is causally involved in disease pathogenesis or simply a passive and indirect indicator of a more complex mechanism. A chronic elevation in homocysteine levels results in a parallel increase in intracellular or plasma SAH, which is a more sensitive biomarker of cardiovascular disease than homocysteine and suggests that SAH is a critical pathological factor in homocysteine-associated disorders. Previous reports indicate that supplementation with folate and B vitamins efficiently lowers homocysteine levels but not plasma SAH levels, which possibly explains the failure of homocysteine-lowering vitamins to reduce vascular events in several recent clinical intervention studies. Furthermore, more studies are focusing on the role and mechanisms of SAH in different chronic diseases related to hyperhomocysteinemia, such as cardiovascular disease, kidney disease, diabetes, and obesity. This review summarizes the current role of SAH in cardiovascular disease and its effect on several related risk factors. It also explores possible the mechanisms, such as epigenetics and oxidative stress, of SAH. This article is part of a Directed Issue entitled: Epigenetic dynamics in development and disease.
Collapse
Affiliation(s)
- Yunjun Xiao
- Department of Nutrition and Food Hygiene, Key Laboratory of Modern Toxicology of Shenzhen, Shenzhen Center for Disease Control and Prevention, Shenzhen, China.
| | - Xuefen Su
- The Jockey Club School of Public Health and Primary Care, School of Public Health, The Chinese University of Hong Kong, Hong Kong, China
| | - Wei Huang
- Department of Nutrition and Food Hygiene, Key Laboratory of Modern Toxicology of Shenzhen, Shenzhen Center for Disease Control and Prevention, Shenzhen, China
| | - Jinzhou Zhang
- Department of Nutrition and Food Hygiene, Key Laboratory of Modern Toxicology of Shenzhen, Shenzhen Center for Disease Control and Prevention, Shenzhen, China
| | - Chaoqiong Peng
- Department of Nutrition and Food Hygiene, Key Laboratory of Modern Toxicology of Shenzhen, Shenzhen Center for Disease Control and Prevention, Shenzhen, China
| | - Haixiong Huang
- Department of Nutrition and Food Hygiene, Key Laboratory of Modern Toxicology of Shenzhen, Shenzhen Center for Disease Control and Prevention, Shenzhen, China
| | - Xiaomin Wu
- Department of Nutrition and Food Hygiene, Key Laboratory of Modern Toxicology of Shenzhen, Shenzhen Center for Disease Control and Prevention, Shenzhen, China
| | - Haiyan Huang
- Department of Nutrition and Food Hygiene, Key Laboratory of Modern Toxicology of Shenzhen, Shenzhen Center for Disease Control and Prevention, Shenzhen, China
| | - Min Xia
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Wenhua Ling
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
42
|
Stearoyl-CoA Desaturase-1: Is It the Link between Sulfur Amino Acids and Lipid Metabolism? BIOLOGY 2015; 4:383-96. [PMID: 26046927 PMCID: PMC4498306 DOI: 10.3390/biology4020383] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Revised: 05/02/2015] [Accepted: 05/14/2015] [Indexed: 12/11/2022]
Abstract
An association between sulfur amino acids (methionine, cysteine, homocysteine and taurine) and lipid metabolism has been described in several experimental and population-based studies. Changes in the metabolism of these amino acids influence serum lipoprotein concentrations, although the underlying mechanisms are still poorly understood. However, recent evidence has suggested that the enzyme stearoyl-CoA desaturase-1 (SCD-1) may be the link between these two metabolic pathways. SCD-1 is a key enzyme for the synthesis of monounsaturated fatty acids. Its main substrates C16:0 and C18:0 and products palmitoleic acid (C16:1) and oleic acid (C18:1) are the most abundant fatty acids in triglycerides, cholesterol esters and membrane phospholipids. A significant suppression of SCD-1 has been observed in several animal models with disrupted sulfur amino acid metabolism, and the activity of SCD-1 is also associated with the levels of these amino acids in humans. This enzyme also appears to be involved in the etiology of metabolic syndromes because its suppression results in decreased fat deposits (regardless of food intake), improved insulin sensitivity and higher basal energy expenditure. Interestingly, this anti-obesogenic phenotype has also been described in humans and animals with sulfur amino acid disorders, which is consistent with the hypothesis that SCD-1 activity is influenced by these amino acids, in particularly cysteine, which is a strong and independent predictor of SCD-1 activity and fat storage. In this narrative review, we discuss the evidence linking sulfur amino acids, SCD-1 and lipid metabolism.
Collapse
|
43
|
Elshorbagy AK. Body composition in gene knockouts of sulfur amino acid-metabolizing enzymes. Mamm Genome 2014; 25:455-63. [PMID: 24952018 DOI: 10.1007/s00335-014-9527-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2014] [Accepted: 05/20/2014] [Indexed: 01/10/2023]
Abstract
Plasma concentrations of several amino acids are elevated in human obesity and insulin resistance, but there is no conclusive evidence on whether the amino acid alterations are causal. Dietary restriction of the essential SAA methionine (MR) in rats produces a hypermetabolic phenotype, with an integrated set of transcriptional changes in lipid enzymes in liver and adipose tissue. MR also induces an array of changes in methionine metabolites, including elevated plasma homocysteine and decreased cystathionine, cysteine, glutathione, and taurine. Several knockouts of enzymes acting downstream of methionine recapitulate the phenotypic results of MR, suggesting that the MR phenotype may be driven by changes distal to methionine. Here we review the changes in SAA and body composition in seven relevant knockout mouse models. All seven models feature decreased body weight, which in five of these have been further explored and shown to result from predominantly decreased fat mass. Common to several models is increased energy expenditure, enhanced insulin sensitivity, and protection against dietary obesity, as occurs in MR. A decrease in plasma total cysteine concentrations is also seen in most models. The lean phenotype could often be reversed by dietary supplementation of cysteine or choline, but not taurine, betaine or a H2S donor. Importantly, the plasma concentrations of both cysteine and choline are positively associated with fat mass in large populations studies, while taurine, betaine, and H2S are not. Collectively, the emerging data from dietary and knockout models are in harmony with human epidemiologic data, suggesting that the availability of key nutrients in the SAA pathway regulates fat storage pathways.
Collapse
Affiliation(s)
- Amany K Elshorbagy
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford, OX1 3QT, UK,
| |
Collapse
|
44
|
Poloni S, Leistner-Segal S, Bandeira IC, D'Almeida V, de Souza CFM, Spritzer PM, Castro K, Tonon T, Nalin T, Imbard A, Blom HJ, Schwartz IVD. Body composition in patients with classical homocystinuria: body mass relates to homocysteine and choline metabolism. Gene 2014; 546:443-7. [PMID: 24815046 DOI: 10.1016/j.gene.2014.05.015] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2013] [Revised: 04/20/2014] [Accepted: 05/05/2014] [Indexed: 01/10/2023]
Abstract
INTRODUCTION Classical homocystinuria is a rare genetic disease caused by cystathionine β-synthase deficiency, resulting in homocysteine accumulation. Growing evidence suggests that reduced fat mass in patients with classical homocystinuria may be associated with alterations in choline and homocysteine pathways. This study aimed to evaluate the body composition of patients with classical homocystinuria, identifying changes in body fat percentage and correlating findings with biochemical markers of homocysteine and choline pathways, lipoprotein levels and bone mineral density (BMD) T-scores. METHODS Nine patients with classical homocystinuria were included in the study. Levels of homocysteine, methionine, cysteine, choline, betaine, dimethylglycine and ethanolamine were determined. Body composition was assessed by bioelectrical impedance analysis (BIA) in patients and in 18 controls. Data on the last BMD measurement and lipoprotein profile were obtained from medical records. RESULTS Of 9 patients, 4 (44%) had a low body fat percentage, but no statistically significant differences were found between patients and controls. Homocysteine and methionine levels were negatively correlated with body mass index (BMI), while cysteine showed a positive correlation with BMI (p<0.05). There was a trend between total choline levels and body fat percentage (r=0.439, p=0.07). HDL cholesterol correlated with choline and ethanolamine levels (r=0.757, p=0.049; r=0.847, p=0.016, respectively), and total cholesterol also correlated with choline levels (r=0.775, p=0.041). There was no association between BMD T-scores and body composition. CONCLUSIONS These results suggest that reduced fat mass is common in patients with classical homocystinuria, and that alterations in homocysteine and choline pathways affect body mass and lipid metabolism.
Collapse
Affiliation(s)
- Soraia Poloni
- Post-Graduation Program in Genetics and Molecular Biology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil; BRAIN Laboratory (Basic Research and Advanced Investigations in Neurosciences), Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil.
| | - Sandra Leistner-Segal
- BRAIN Laboratory (Basic Research and Advanced Investigations in Neurosciences), Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil; Medical Genetics Service, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Isabel Cristina Bandeira
- BRAIN Laboratory (Basic Research and Advanced Investigations in Neurosciences), Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Vânia D'Almeida
- Reference Center for Inborn Errors of Metabolism, Universidade Federal de São Paulo, São Paulo, Brazil
| | | | - Poli Mara Spritzer
- Gynecological Endocrinology Unit, Division of Endocrinology, Hospital de Clinicas de Porto Alegre, Department of Physiology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Kamila Castro
- Food and Nutrition Research Center, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Tássia Tonon
- Post-Graduation Program in Genetics and Molecular Biology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Tatiéle Nalin
- Post-Graduation Program in Genetics and Molecular Biology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Apolline Imbard
- Biochemistry-Hormonology Laboratory, Robert Debré Hospital, APHP, Paris, France; Laboratory of Clinical Biochemistry and Metabolism, Department of General Pediatrics, University Medical Centre Freiburg, Freiburg, Germany
| | - Henk J Blom
- Laboratory of Clinical Biochemistry and Metabolism, Department of General Pediatrics, University Medical Centre Freiburg, Freiburg, Germany
| | - Ida V D Schwartz
- Post-Graduation Program in Genetics and Molecular Biology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil; BRAIN Laboratory (Basic Research and Advanced Investigations in Neurosciences), Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil; Medical Genetics Service, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| |
Collapse
|
45
|
Bel-Serrat S, Mouratidou T, Huybrechts I, Cuenca-García M, Manios Y, Gómez-Martínez S, Molnár D, Kafatos A, Gottrand F, Widhalm K, Sjöström M, Wästlund A, Stehle P, Azzini E, Vyncke K, González-Gross M, Moreno LA. The role of dietary fat on the association between dietary amino acids and serum lipid profile in European adolescents participating in the HELENA Study. Eur J Clin Nutr 2014; 68:464-73. [PMID: 24495993 DOI: 10.1038/ejcn.2013.284] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2013] [Revised: 11/21/2013] [Accepted: 12/05/2013] [Indexed: 01/04/2023]
Abstract
BACKGROUND/OBJECTIVES The objective of this study was to examine the relationship between amino acid (AA) intake and serum lipid profile in European adolescents from eight European cities participating in the cross-sectional (2006-2007) HELENA (Healthy Lifestyle in Europe by Nutrition in Adolescence) study, and to assess whether this association was independent of total fat intake. SUBJECTS/METHODS Diet, skinfold thickness, triglycerides (TG), total cholesterol (TC), high-density lipoprotein cholesterol (HDL-c), TC/HDL-c ratio, low-density lipoprotein cholesterol (LDL-c), apolipoprotein B (Apo B), apolipoprotein A1 (Apo A1) and Apo B/Apo A1 ratio were measured in 454 12.5- to 17.5-year-old adolescents (44% boys). Intake was assessed via two non-consecutive 24-h dietary recalls. Data on maternal education and sedentary behaviors were obtained via questionnaires. Physical activity was objectively measured by accelerometry. RESULTS Alanine, arginine, asparaginic acid, glycine, histidine, lysine and serine intakes were inversely associated with serum TG concentrations in both boys and girls. Intake of other AA like alanine and/or arginine was also inversely associated with serum TC, LDL-c and Apo B/Apo A1 ratio only in girls. An inverse association was observed between intakes of alanine, isoleucine, leucine, methionine, serine, tryptophan, tyrosine and valine and TC/HDL-c ratio among female adolescents. Similar results were found in males for serine and tryptophan intakes. It is noteworthy, however, that associations were no longer significant in both genders when total fat intake was considered as a confounding factor. CONCLUSIONS In this sample of adolescents, the association between AA intakes and serum lipid profile did not persist when dietary fat was considered. Therefore, dietary interventions and health promotion activities should focus on fat intake to improve lipid profile and potentially prevent cardiovascular disease.
Collapse
Affiliation(s)
- S Bel-Serrat
- 8217;Growth, Exercise, Nutrition and Development' (GENUD) Research Group, Department of Physiatry and Nursery, Faculty of Health Sciences, University of Zaragoza, Zaragoza, Spain
| | - T Mouratidou
- 8217;Growth, Exercise, Nutrition and Development' (GENUD) Research Group, Department of Physiatry and Nursery, Faculty of Health Sciences, University of Zaragoza, Zaragoza, Spain
| | - I Huybrechts
- 1] Department of Public Health, Faculty of Medicine and Health Sciences, University Hospital, Ghent University, Ghent, Belgium [2] International Agency for Research on Cancer (IARC), Dietary Exposure Assessment Group (DEX), Lyon, France
| | - M Cuenca-García
- Department of Physiology, Medicine School, Granada University, Granada, Spain
| | - Y Manios
- Department of Nutrition and Dietetics, Harokopio University, Athens, Greece
| | - S Gómez-Martínez
- Immunonutrition Research Group, Department of Metabolism and Nutrition, Institute of Food Science, Technology and Nutrition (ICTAN), Spanish National Research Council (CSIC), Madrid, Spain
| | - D Molnár
- Department of Pediatrics, University of Pécs, Pécs-József, Hungary
| | - A Kafatos
- Department of Social Medicine, Preventive Medicine and Nutrition Unit, University of Crete School of Medicine, Heraklion, Greece
| | - F Gottrand
- Inserm U995, Faculty of Medicine, University Lille 2, Lille, France
| | - K Widhalm
- Department of Pediatrics, Division of Clinical Nutrition and Prevention, Medical University of Vienna, Vienna, Austria
| | - M Sjöström
- Department of Biosciences and Nutrition, Karolinska Institute, Huddinge, Sweden
| | - A Wästlund
- Department of Biosciences and Nutrition, Karolinska Institute, Huddinge, Sweden
| | - P Stehle
- Institut für Ernährungs- und Lebensmittelwissenschaften - Humanernährung, Rheinische Friedich-Wilhelms-Universität, Bonn, Germany
| | - E Azzini
- National Research Institute for Food and Nutrition, INRAN, Rome, Italy
| | - K Vyncke
- 1] Department of Public Health, Faculty of Medicine and Health Sciences, University Hospital, Ghent University, Ghent, Belgium [2] Research Foundation - Flanders (FWO), Brussels, Belgium
| | - M González-Gross
- 1] ImFine Research Group, Departamento de Salud y Rendimiento Humano, Facultad de Ciencias de la Actividad Física y del Deporte-INEF, Universidad Politécnica de Madrid, Madrid, Spain [2] CIBER: CB12/03/30038 Fisiopatología de la Obesidad y la Nutrición, CIBERobn, Instituto de Salud Carlos III (ISCIII), Spain
| | - L A Moreno
- 8217;Growth, Exercise, Nutrition and Development' (GENUD) Research Group, Department of Physiatry and Nursery, Faculty of Health Sciences, University of Zaragoza, Zaragoza, Spain
| |
Collapse
|
46
|
Samuelsson M, Skogh E, Lundberg K, Vrethem M, Öllinger K. Taurine and glutathione in plasma and cerebrospinal fluid in olanzapine treated patients with schizophrenia. Psychiatry Res 2013; 210:819-24. [PMID: 24113127 DOI: 10.1016/j.psychres.2013.09.014] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2012] [Revised: 09/04/2013] [Accepted: 09/12/2013] [Indexed: 01/22/2023]
Abstract
Oxidative stress has been implicated in the pathophysiology of schizophrenia. Taurine and glutathione (GSH) have antioxidant and central nervous system protective properties, and are proposed to be involved in the pathology of schizophrenia. The aim of this study was to compare the blood and cerebrospinal fluid (CSF) levels of taurine and GSH in patients with schizophrenia, medicated with oral olanzapine, compared with controls. In total, 37 patients with schizophrenia and 45 healthy volunteers were recruited. We found the plasma taurine levels to be elevated in patients compared with controls. No differences were, however, found between patients and controls regarding taurine in CSF or GSH concentrations in plasma and CSF. Moreover, in the patient group no correlations between taurine and GSH levels and the symptoms or function of the disorder were found. The higher levels of plasma but not CSF taurine in patients with schizophrenia treated with OLA may implicate the involvement of taurine in the pathophysiology of the disease. The absence of GSH differences both in plasma and CSF between patients and controls is interesting in the perspective of earlier research proposing a dysregulation of GSH metabolism as a vulnerability factor for the development of schizophrenia.
Collapse
Affiliation(s)
- Martin Samuelsson
- Division of Psychiatry, Department of Clinical and Experimental Medicine, Linköping University, S-58185 Linköping, Sweden.
| | | | | | | | | |
Collapse
|
47
|
Elshorbagy AK, Nijpels G, Valdivia-Garcia M, Stehouwer CDA, Ocke M, Refsum H, Dekker JM. S-adenosylmethionine is associated with fat mass and truncal adiposity in older adults. J Nutr 2013; 143:1982-8. [PMID: 24068793 DOI: 10.3945/jn.113.179192] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
S-adenosylmethionine (SAM) is synthesized from methionine, which is abundant in animal-derived protein, in an energy-consuming reaction. SAM and S-adenosylhomocysteine (SAH) correlate with body mass index (BMI). Plasma total concentration of the SAM-associated product cysteine (tCys) correlates with fat mass in humans and cysteine promotes adiposity in animals. In a cross-sectional study of 610 participants, we investigated whether SAM and SAH are associated with BMI via lean mass or fat mass and dietary protein sources as determinants of SAM and tCys concentrations. Plasma SAM was not associated with lean mass, but mean adjusted fat mass increased from 24 kg (95% CI: 22.6, 25.1) to 30 kg (95% CI: 28.7, 31.3) across SAM quartiles (P < 0.001) and trunk fat:total fat ratio increased from 0.48 to 0.52 (P < 0.001). Erythrocyte SAM was also positively associated with fat mass and trunk fat:total fat ratio. The association of SAM with fat mass was not weakened by adjustment for serum tCys, lipids, creatinine, or dietary or lifestyle confounders. Concentrations of the SAM precursor, methionine, and the SAM product, SAH, were not independently associated with adiposity. Intake of animal-derived protein was not related to serum methionine but was positively associated with plasma SAM (partial r = 0.11) and serum tCys (partial r = 0.13; P < 0.05 for both after adjustment for age, gender, and total energy intake). In conclusion, plasma SAM, but not methionine, is independently associated with fat mass and truncal adiposity, suggesting increased conversion of methionine to SAM in obese individuals. Prospective studies are needed to investigate the interactions among dietary energy and animal protein content, SAM concentrations, and change in body weight and cardiometabolic risk.
Collapse
|
48
|
Glycine restores glutathione and protects against oxidative stress in vascular tissue from sucrose-fed rats. Clin Sci (Lond) 2013; 126:19-29. [PMID: 23742196 DOI: 10.1042/cs20130164] [Citation(s) in RCA: 77] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The attenuation of oxidative stress could be an important mechanism whereby the incidence of vascular complications in the MS (metabolic syndrome) may be diminished. The present study was undertaken to investigate the mechanism by which glycine, supplemented to the diet of SF (sucrose-fed) rats, modulates glutathione biosynthesis and protects against oxidative stress and altered endothelium-dependent relaxation in isolated aorta. Glycine reduced O2•- (superoxide anion radical) release in the presence of NADPH, and decreased protein carbonyl and lipid peroxidation. This effect of glycine could be because of the increased amount of glutathione synthetase, which may be responsible for increased glutathione (GSH) content in vascular tissue from SF rats. Moreover, glycine increased the amount of Cu,Zn-SOD (copper/zinc superoxide dismutase) and eNOS (endothelial NO synthase) in aorta from SF animals. Finally, it improved the relaxation response to ACh (acetylcholine) found impaired in aortic rings from SF rats. In the presence of NAC (N-acetylcysteine), a precursor of GSH, an improved ACh-mediated aortic relaxation of aortic rings from SF rats was observed, whereas BSO (buthionine sulfoximine), an inhibitor of glutathione biosynthesis, inhibited the relaxing effect of NAC in aortas from both control and SF rats. This experiment emphasizes the role of GSH in endothelial function in SF rats. The present data suggest that glycine rectifies vascular reactivity by increasing the biosynthesis of glutathione. Glutathione protects vascular tissue against oxidative stress, and enhances the availability of NO, which exerts its relaxing effect, thus contributing to the reduction of high BP (blood pressure) in the SF rats.
Collapse
|
49
|
Vinknes KJ, Dekker JM, Drevon CA, Refsum H, Nurk E, Nijpels G, Stehouwer CDA, Teerlink T, Tell GS, Nygård O, Vollset SE, Ueland PM, Elshorbagy AK. Plasma sulfur amino acids and stearoyl-CoA desaturase activity in two Caucasian populations. Prostaglandins Leukot Essent Fatty Acids 2013; 89:297-303. [PMID: 24120123 DOI: 10.1016/j.plefa.2013.09.007] [Citation(s) in RCA: 10] [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: 07/02/2013] [Revised: 09/04/2013] [Accepted: 09/09/2013] [Indexed: 12/29/2022]
Abstract
In rats, dietary restriction of the cysteine precursor methionine suppresses hepatic stearoyl-CoA desaturase (SCD)-1 expression and activity, whereas cysteine supplementation reverses these effects. In 2 independent cohorts: Hordaland Health Study (HUSK; N=2021, aged 71-74y), Norway, and Hoorn study (N=686, aged 50-87y), Netherlands, we examined the cross-sectional associations of plasma sulfur-containing compounds (SCC; methionine, S-adenosylmethionine, S-adenosylhomocysteine, homocysteine, cystathionine, total cysteine (tCys), glutathione and cysteinylglycine) with SCD-16 index (16:1n-7/16:0), estimated from fatty acid profiles of total plasma or serum lipids. Only tCys was consistently associated with SCD-16 index after adjustments for sex and age (HUSK: partial r=0.14; Hoorn: partial r=0.11, P<0.001 for both), and after further adjustments for other SCC, body fat, diet, exercise and plasma lipids (HUSK: partial r=0.07, P=0.004; Hoorn: partial r=0.12, P=0.013). Together with animal data showing an effect of dietary cysteine on SCD1, our results suggest a role for cysteine in SCD1 regulation in humans.
Collapse
Affiliation(s)
- K J Vinknes
- Department of Nutrition, Institute of Basic Medical Science, Faculty of Medicine, University of Oslo, Post box 1046 Blindern, 0317 Oslo, Norway.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Increase or decrease hydrogen sulfide exert opposite lipolysis, but reduce global insulin resistance in high fatty diet induced obese mice. PLoS One 2013; 8:e73892. [PMID: 24058499 PMCID: PMC3772803 DOI: 10.1371/journal.pone.0073892] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2013] [Accepted: 07/26/2013] [Indexed: 11/19/2022] Open
Abstract
Objective Adipose tissue expressed endogenous cystathionine gamma lyase (CSE)/hydrogen sulfide (H2S) system. H2S precursor inhibited catecholamine stimulated lipolysis. Thus, we hypothesized that CSE/H2S system regulates lipolysis which contributed to the pathogenesis of insulin resistance. Methods We treated rat adipocyte with DL-propargylglycine (PAG, a CSE inhibitor), L-cysteine (an H2S precursor) plus pyridoxial phosphate (co-enzyme) or the H2S chronic release donor GYY4137, then the glycerol level was assayed for assessing the lipolysis. Then, the effects of PAG and GYY4137 on insulin resistance in high fatty diet (HFD) induced obese mice were investigated. Results Here, we found that PAG time-dependently increased basal or isoproterenol stimulated lipolysis. However, L-cysteine plus pyridoxial phosphate or GYY4137 significantly reduced it. PAG increased phosphorylated protein kinase A substrate, perilipin 1 and hormone sensitive lipase, but L-cysteine and GYY4137 decreased the parameters. In HFD induced obese mice, PAG increased adipose basal lipolysis, thus blunted fat mass increase, resulting in lowering insulin resistance evidenced by reduction of fasting glucose, insulin level, HOMA index, oral glucose tolerance test (OGTT) curve area and elevating the insulin tolerance test (ITT) response. GYY4137 inhibited lipolysis in vivo without increasing fat mass, but also ameliorated the insulin resistance in HFD mice. Conclusion These results implicated that inhibition endogenous CSE/H2S system in adipocytes increased lipolysis by a protein kinase A-perilipin/hormone-sensitive lipase pathway, thus blunted fat mass increase and reduced insulin resistance in obese mice; giving H2S donor decreased lipolysis, also reduced insulin resistance induced by HFD. Our data showed that increase or decrease H2S induced opposite lipolysis, but had the same effect on insulin resistance. The paradoxical regulation may be resulted from different action of H2S on metabolic and endocrine function in adipocyte.
Collapse
|