1
|
Huang C, Liu D, Yang S, Huang Y, Wei X, Zhang P, Lin J, Xu B, Liu Y, Guo D, Li Y, Li J, Zhang H. Effect of time-restricted eating regimen on weight loss is mediated by gut microbiome. iScience 2024; 27:110202. [PMID: 38993674 PMCID: PMC11238135 DOI: 10.1016/j.isci.2024.110202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 04/16/2024] [Accepted: 06/01/2024] [Indexed: 07/13/2024] Open
Abstract
Time-restricted eating (TRE) is a promising obesity management strategy, but weight-loss efficacy varies among participants, and the underlying mechanism is unclear. The study aimed to investigate the role of gut microbiota in weight-loss response during long-term TRE intervention. We analyzed data from 51 obese adults in a 12-month TRE program, categorizing them into distinct weight loss groups (DG) and moderate weight loss groups (MG) based on their TRE responses. Shotgun metagenomic sequencing analysis revealed a significant increase in species closely associated with weight loss effectiveness and metabolic parameter changes in the DG group. Pathways related to fatty acid biosynthesis, glycogen biosynthesis, and nucleotide metabolism were reduced in the DG group and enhanced in the MG group. Next, we identified nine specific species at baseline that contributed better responses to TRE intervention and significant weight loss. Collectively, gut microbiota contributes to responsiveness heterogeneity in TRE and can predict weight-loss effectiveness.
Collapse
Affiliation(s)
- Chensihan Huang
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Deying Liu
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Shunyu Yang
- Department of Nutrition, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Yan Huang
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Xueyun Wei
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Peizhen Zhang
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Jiayang Lin
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Bingyan Xu
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Yating Liu
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Dan Guo
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Yafeng Li
- Department of Nephrology, Shanxi Provincial People’s Hospital (Fifth Hospital) of Shanxi Medical University, Taiyuan 030000, Shanxi, China
- Core Laboratory, Shanxi Provincial People’s Hospital (Fifth Hospital) of Shanxi Medical University; Taiyuan, China
- Academy of Microbial Ecology, Shanxi Medical University, Taiyuan 030000, Shanxi, China
| | - Jin Li
- Department of Endocrinology and Metabolism, Shan Xi Medical University Second Hospital, Shan Xi Medical University, Taiyuan 030000, Shanxi, China
| | - Huijie Zhang
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong, China
| |
Collapse
|
2
|
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
|
3
|
Olsen T, Elshorbagy A, Stolt E, Åsberg A, Zaré HK, Bastani NE, Refsum H, Retterstøl K, Vinknes KJ. Acute effects of oral mesna administration on the full amino acid profile and 3-methylhistidine: secondary results from the CYLOB dose-finding study. Amino Acids 2024; 56:39. [PMID: 38844567 PMCID: PMC11156715 DOI: 10.1007/s00726-024-03398-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 05/22/2024] [Indexed: 06/09/2024]
Abstract
Plasma total cysteine (tCys) is strongly associated with fat mass in humans. Mesna lowers plasma tCys in a dose-dependent manner, but it is not known whether it interferes with metabolism of other amino acids or protein. In this Phase-1 study, we show that a single dose of mesna administered at 400, 800, 1200 or 1600 mg to 6-7 individuals per dose only slightly affects amino acid profiles, with increases in plasma valine across dose levels. There were no effects of mesna on 3-methylhistidine, a marker of protein breakdown.
Collapse
Affiliation(s)
- Thomas Olsen
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway.
| | - Amany Elshorbagy
- Department of Pharmacology, University of Oxford, Oxford, UK
- Department of Physiology, Faculty of Medicine, University of Alexandria, Alexandria, Egypt
| | - Emma Stolt
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Anders Åsberg
- Department of Transplantation Medicine, Oslo University Hospital, Oslo, Norway
- Department of Pharmacy, University of Oslo, Oslo, Norway
| | - Hasse K Zaré
- Department of Pharmacology, Oslo University Hospital, Oslo, Norway
| | - Nasser E Bastani
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Helga Refsum
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway
- Department of Physiology, Faculty of Medicine, University of Alexandria, Alexandria, Egypt
| | - Kjetil Retterstøl
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway
- Department of Endocrinology, The Lipid Clinic, Morbid Obesity and Preventive Medicine, Oslo University Hospital, Oslo, Norway
| | - Kathrine J Vinknes
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
4
|
Lee DJ, Kang AN, Lee J, Kwak MJ, Mun D, Lee D, Oh S, Kim Y. Molecular characterization of Fusarium venenatum-based microbial protein in animal models of obesity using multi-omics analysis. Commun Biol 2024; 7:133. [PMID: 38278957 PMCID: PMC10817893 DOI: 10.1038/s42003-024-05791-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 01/08/2024] [Indexed: 01/28/2024] Open
Abstract
Microbial protein, produced by fermentation of Fusarium venenatum is a promising candidate alternative protein source. Previous study has demonstrated its ability to improve hyperlipidemia in rats, yet the related mechanism remains unclear. In this study, we aimed to evaluate the potential of F. venenatum as an alternative protein source and its impact on lipid metabolism using multi-omics analysis. Initial experiments with Caenorhabditis elegans revealed that F. venenatum enhanced longevity, improved immune responses, and reduced lipid metabolism by downregulating fat synthesis-related genes. Subsequently, we conducted experiments with mice on a high-fat diet to confirm the anti-obesity effects of F. venenatum. The groups fed F. venenatum showed improved lipid profiles and reduced hepatic fat accumulation. Furthermore, fecal metabolomic analysis showed higher excretion of primary bile acid and cholesterol in the groups fed F. venenatum which might lead to a decrease in lipid digestion and hepatic fat accumulation. Collectively, this series of experiments revealed the potential of F. venenatum as a sustainable alternative protein and its application as an anti-obesity supplement.
Collapse
Affiliation(s)
- Daniel Junpyo Lee
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Science, Seoul National University, Seoul, 08826, Korea
| | - An Na Kang
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Science, Seoul National University, Seoul, 08826, Korea
| | - Junbeom Lee
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Science, Seoul National University, Seoul, 08826, Korea
| | - Min-Jin Kwak
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Science, Seoul National University, Seoul, 08826, Korea
| | - Daye Mun
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Science, Seoul National University, Seoul, 08826, Korea
| | - Daseul Lee
- Agricultural Microbiology Division, Department of Agricultural Biology, National Institute of Agricultural Sciences, Wanju-gun, 55365, Korea
| | - Sangnam Oh
- Department of Functional Food and Biotechnology, Jeonju University, Jeonju, 55069, Korea.
| | - Younghoon Kim
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Science, Seoul National University, Seoul, 08826, Korea.
| |
Collapse
|
5
|
Kim H, Gu L, Yeo H, Choi U, Lee CR, Yu H, Koo S. Rapid Assembly of Pyrrole-Ligated 1,3,4-Oxadiazoles and Excellent Antibacterial Activity of Iodophenol Substituents. Molecules 2023; 28:molecules28083638. [PMID: 37110872 PMCID: PMC10142073 DOI: 10.3390/molecules28083638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 04/17/2023] [Accepted: 04/18/2023] [Indexed: 04/29/2023] Open
Abstract
Pyrrole-ligated 1,3,4-oxadiazole is a very important pharmacophore which exhibits broad therapeutic effects such as anti-tuberculosis, anti-epileptic, anti-HIV, anti-cancer, anti-inflammatory, antioxidant, and antibacterial activities. A one-pot Maillard reaction between D-Ribose and an L-amino methyl ester in DMSO with oxalic acid at 2.5 atm and 80 °C expeditiously produced pyrrole-2-carbaldehyde platform chemicals in reasonable yields, which were utilized for the synthesis of pyrrole-ligated 1,3,4-oxadiazoles. Benzohydrazide reacted with the formyl group of the pyrrole platforms to provide the corresponding imine intermediates, which underwent I2-mediated oxidative cyclization to the pyrrole-ligated 1,3,4-oxadiazole skeleton. The structure and activity relationship (SAR) of the target compounds with varying alkyl or aryl substituents of the amino acids and electron-withdrawing or electron-donating substituents on the phenyl ring of benzohydrazide were evaluated for antibacterial activity against Escherichia coli, Staphylococcus aureus, and Acinetobacter baumannii as representative Gram(-) and Gram(+) bacteria. Branched alkyl groups from the amino acid showed better antibacterial activities. Absolutely superior activities were observed for 5f-1 with an iodophenol substituent against A. baumannii (MIC < 2 μg/mL), a bacterial pathogen that displays a high resistance to commonly used antibiotics.
Collapse
Affiliation(s)
- Hyein Kim
- Department of Chemistry, Myongji University, Myongji-Ro 116, Cheoin-Gu, Yongin 17058, Gyeonggi-Do, Republic of Korea
| | - Lina Gu
- Department of Chemistry, Myongji University, Myongji-Ro 116, Cheoin-Gu, Yongin 17058, Gyeonggi-Do, Republic of Korea
- School of Pharmacy, East China University of Science and Technology, Meilong Road 130, Shanghai 200237, China
| | - Huisu Yeo
- Department of Chemistry, Myongji University, Myongji-Ro 116, Cheoin-Gu, Yongin 17058, Gyeonggi-Do, Republic of Korea
| | - Umji Choi
- Department of Biological Sciences and Bioinformatics, Myongji University, Myongji-Ro 116, Cheoin-Gu, Yongin 17058, Gyeonggi-Do, Republic of Korea
| | - Chang-Ro Lee
- Department of Biological Sciences and Bioinformatics, Myongji University, Myongji-Ro 116, Cheoin-Gu, Yongin 17058, Gyeonggi-Do, Republic of Korea
| | - Haiyang Yu
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
| | - Sangho Koo
- Department of Chemistry, Myongji University, Myongji-Ro 116, Cheoin-Gu, Yongin 17058, Gyeonggi-Do, Republic of Korea
- School of Pharmacy, East China University of Science and Technology, Meilong Road 130, Shanghai 200237, China
| |
Collapse
|
6
|
Chang R, Zhang Y, Sun J, Xu K, Li C, Zhang J, Mei W, Zhang H, Zhang J. Maternal pre-pregnancy body mass index and offspring with overweight/obesity at preschool age: The possible role of epigenome-wide DNA methylation changes in cord blood. Pediatr Obes 2023; 18:e12969. [PMID: 36102013 DOI: 10.1111/ijpo.12969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 07/20/2022] [Accepted: 07/25/2022] [Indexed: 01/09/2023]
Abstract
BACKGROUND Epigenome-wide association studies have identified some DNA methylation sites associated with body mass index (BMI) or obesity. Studies in the Asian population are lacking. OBJECTIVE To examine the association of cord blood genome-wide DNA methylation (GWDm) changes with maternal pre-pregnancy BMI and children's BMI-z score at preschool age. Additionally, we also explored the genome-wide differentially methylated regions and differentially methylated probes between preschoolers with overweight/obesity and normal-weight counterparts. METHODS This two-stage study design included (1) a GWDm analysis of 30 mother-child pairs from 633 participants of the Zhuhai birth cohort with data on newborn cord blood, maternal pre-pregnancy BMI, and children's BMI at 3 years of age; and (2) a targeted validation analysis of the cord blood of ten children with overweight/obesity and ten matched controls to validate the CpG sites. RESULTS In the first stage, no significant CpG sites were found to be associated with children's BMI-z score at preschool age after FDR correction with the p-values of the CpG sites in FOXN3 (cg23501836) and ZNF264 (cg27437574) being close to 1 × 10-6 . In the second stage, a significant difference of CpG sites in AHRR (chr5:355067-355068) and FOXN3 (chr14: 89630264-89630272 and chr14: 89630387-89630388) was found between the ten children with overweight/obesity and ten controls (p < 0.05). The CpG sites in FOXN3 (chr14:89630264-89630272 and chr14:89630295-89630296) and ZNF264 (chr19: 57703104-57703107 and chr19: 57703301-57703307) were associated with children's BMI-z score; and the CpG sites in FOXN3 (chr14: 89630264-89630272 and chr14: 89630387-89630388) were associated with maternal pre-pregnancy BMI. CONCLUSIONS DNA methylation in FOXN3 and AHRR is associated with overweight/obesity in preschool-aged children, and the methylation in FOXN3 and ZNF264 might be associated with children's BMI-z score. FOXN3 methylation may be associated with maternal pre-pregnancy BMI, suggesting its potential role in the children's BMI-z score or overweight/obesity. Our results provide novel insights into the mechanisms of children's obesity.
Collapse
Affiliation(s)
- Ruixia Chang
- Department of Maternal and Child Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuanyuan Zhang
- Department of Maternal and Child Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiahong Sun
- Department of Maternal and Child Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ke Xu
- Department of Maternal and Child Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chunan Li
- Department of Maternal and Child Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jingli Zhang
- Traditional Chinese Medicine Hospital, Zhuhai, Guangdong, China
| | - Wenhua Mei
- Zhuhai Center for Disease Control and Prevention, Zhuhai, Guangdong, China
| | - Hongzhong Zhang
- Zhuhai Women and Children's Hospital, Zhuhai, Guangdong, China
| | - Jianduan Zhang
- Department of Maternal and Child Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
7
|
Arora D, Arora P, Goyal P, Paliwal P, Sardana K. A prospective real-world study evaluating serum levels of arginine and cysteine in women with chronic telogen effluvium: do we really need blanket prescription of amino acid supplementation? Clin Exp Dermatol 2022; 47:2006-2011. [PMID: 35844071 DOI: 10.1111/ced.15333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/11/2022] [Indexed: 01/26/2023]
Abstract
Amino acids form a major component of hair fibres and are prescribed routinely in the form of nutritional supplements in patients with chronic telogen effluvium (CTE). Such a practice is based on assumption of a nutritional deficiency state in such patients. In this prospective study, we evaluated the serum levels of cysteine and arginine in 30 women with CTE and in healthy controls. We found no significant difference between the two groups in terms of serum arginine levels. Cysteine levels were higher in patients with CTE (P < 0.001). No correlation was found between levels of serum amino acids (cysteine and arginine) and either diet type (vegetarian or not) or body mass index, and no significant correlation between levels of the two amino acids and severity of disease. Our work suggests that arginine and cysteine deficiency is not present in women with CTE. Supplementation is unlikely to be of any benefit in nutrient-replete populations and only adds to the cost of therapy.
Collapse
Affiliation(s)
- Damini Arora
- Departments of Dermatology, Dr Ram Manohar Lohia Hospital and Atal Bihari Vajpayee Institute of Medical Sciences, New Delhi, India
| | - Pooja Arora
- Departments of Dermatology, Dr Ram Manohar Lohia Hospital and Atal Bihari Vajpayee Institute of Medical Sciences, New Delhi, India
| | - Parul Goyal
- Departments of Biochemistry, Dr Ram Manohar Lohia Hospital and Atal Bihari Vajpayee Institute of Medical Sciences, New Delhi, India
| | - Purnima Paliwal
- Departments of Pathology, Dr Ram Manohar Lohia Hospital and Atal Bihari Vajpayee Institute of Medical Sciences, New Delhi, India
| | - Kabir Sardana
- Departments of Dermatology, Dr Ram Manohar Lohia Hospital and Atal Bihari Vajpayee Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
8
|
Li S, Luo X, Liao Z, Xu H, Liang M, Mai K, Zhang Y. Additional supplementation of sulfur-containing amino acids in the diets improves the intestinal health of turbot fed high-lipid diets. FISH & SHELLFISH IMMUNOLOGY 2022; 130:368-379. [PMID: 36115604 DOI: 10.1016/j.fsi.2022.09.015] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 08/29/2022] [Accepted: 09/10/2022] [Indexed: 06/15/2023]
Abstract
An eight-week feeding trial was conducted to investigate the effects of diets supplemented with three sulfur-containing amino acids (SAA), namely, methionine, cysteine, and taurine, on the intestinal health status of juvenile turbot (Scophthalmus maximus) fed high-lipid diets. Four diets were formulated, namely, a high-lipid control diet (16% lipid, HL) and three SAA-supplemented diets, which were formulated by supplementing 1.5% methionine (HLM), 1.5% cysteine (HLC), and 1.5% taurine (HLT) into the HL control diet, respectively. Each diet was assigned to triplicate tanks, and each tank was stocked with 30 juvenile fish (appr. initial weight, 8 g). The histological and morphometric results showed that dietary SAA supplementation obviously improved the intestinal morphology and integrity, in particular as reflected by higher height of microvilli and mucosal folds. Dietary SAA supplementation, in particular cysteine, up-regulated the gene expression of mucin-2 and tight junction proteins (ZO-1, Tricellilun and JAM). Dietary SAA supplementation remarkably down-regulated the gene expression of apoptosis-related factors such as p38, JNK, and Bax, expression of pro-inflammatory factors (e.g., NF-κB, AP-1 IL-1β, IL-8, and TNF-α). SAA supplementation resulted in higher antioxidative abilities in the intestine. Additionally, dietary SAA supplementation largely altered the communities of intestinal microbiota. Compared with the HL group, higher relative abundance of potential beneficial bacteria, and lower relative abundance of opportunistic pathogens were observed in SAA-supplemented groups. Dietary taurine supplementation significantly increased the relative abundance of Ligilactobacillus (in particular Lactobacillus murinus) and Limosilactobacillus (especially Lactobacillus reuteri). In conclusion, dietary sulfur-containing amino acids supplementation have promising potential in ameliorating the intestinal inflammation of turbot fed high-lipid diets. Especially dietary cysteine and taurine supplementation have more positive effects on the communities of the intestinal microbiota of turbot.
Collapse
Affiliation(s)
- Sihui Li
- Key Laboratory of Aquaculture Nutrition and Feed, Ministry of Agriculture, The Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, 5 Yushan Road, Qingdao, 266003, China
| | - Xing Luo
- Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, 106 Nanjing Road, Qingdao, 266071, China
| | - Zhangbin Liao
- Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, 106 Nanjing Road, Qingdao, 266071, China
| | - Houguo Xu
- Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, 106 Nanjing Road, Qingdao, 266071, China; Qingdao National Laboratory for Marine Science and Technology, 1 Wenhai Road, Qingdao, 266237, China.
| | - Mengqing Liang
- Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, 106 Nanjing Road, Qingdao, 266071, China; Qingdao National Laboratory for Marine Science and Technology, 1 Wenhai Road, Qingdao, 266237, China
| | - Kangsen Mai
- Key Laboratory of Aquaculture Nutrition and Feed, Ministry of Agriculture, The Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, 5 Yushan Road, Qingdao, 266003, China; Qingdao National Laboratory for Marine Science and Technology, 1 Wenhai Road, Qingdao, 266237, China
| | - Yanjiao Zhang
- Key Laboratory of Aquaculture Nutrition and Feed, Ministry of Agriculture, The Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, 5 Yushan Road, Qingdao, 266003, China; Qingdao National Laboratory for Marine Science and Technology, 1 Wenhai Road, Qingdao, 266237, China.
| |
Collapse
|
9
|
Lee-Ødegård S, Olsen T, Norheim F, Drevon CA, Birkeland KI. Potential Mechanisms for How Long-Term Physical Activity May Reduce Insulin Resistance. Metabolites 2022; 12:metabo12030208. [PMID: 35323652 PMCID: PMC8950317 DOI: 10.3390/metabo12030208] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 02/20/2022] [Accepted: 02/22/2022] [Indexed: 02/06/2023] Open
Abstract
Insulin became available for the treatment of patients with diabetes 100 years ago, and soon thereafter it became evident that the biological response to its actions differed markedly between individuals. This prompted extensive research into insulin action and resistance (IR), resulting in the universally agreed fact that IR is a core finding in patients with type 2 diabetes mellitus (T2DM). T2DM is the most prevalent form of diabetes, reaching epidemic proportions worldwide. Physical activity (PA) has the potential of improving IR and is, therefore, a cornerstone in the prevention and treatment of T2DM. Whereas most research has focused on the acute effects of PA, less is known about the effects of long-term PA on IR. Here, we describe a model of potential mechanisms behind reduced IR after long-term PA to guide further mechanistic investigations and to tailor PA interventions in the therapy of T2DM. The development of such interventions requires knowledge of normal glucose metabolism, and we briefly summarize an integrated physiological perspective on IR. We then describe the effects of long-term PA on signaling molecules involved in cellular responses to insulin, tissue-specific functions, and whole-body IR.
Collapse
Affiliation(s)
- Sindre Lee-Ødegård
- Department of Clinical Medicine, Faculty of Medicine, University of Oslo, 0372 Oslo, Norway;
| | - Thomas Olsen
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, 0372 Oslo, Norway; (T.O.); (F.N.); (C.A.D.)
| | - Frode Norheim
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, 0372 Oslo, Norway; (T.O.); (F.N.); (C.A.D.)
| | - Christian Andre Drevon
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, 0372 Oslo, Norway; (T.O.); (F.N.); (C.A.D.)
- Vitas Ltd. Analytical Services, Oslo Science Park, 0349 Oslo, Norway
| | - Kåre Inge Birkeland
- Department of Clinical Medicine, Faculty of Medicine, University of Oslo, 0372 Oslo, Norway;
- Correspondence:
| |
Collapse
|
10
|
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]
|
11
|
Gender-Specific Behaviour in Obesity Stages I-II: Imbalance of Aminothiol Status and Adipomyokine Profile in Subjects with Different Insulin Resistance Severity. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:9713582. [PMID: 34868459 PMCID: PMC8635872 DOI: 10.1155/2021/9713582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 10/11/2021] [Accepted: 11/11/2021] [Indexed: 12/03/2022]
Abstract
The hyperproduction of oxidative stress and inflammatory biomarkers, which is paralleled by decreased levels of antioxidant and anti-inflammatory mediators, is part of cellular mechanisms that contribute to the disruption of metabolic homeostasis in obesity. Whether gender-specific alterations and gender-restricted associations in these biomarkers underlie the increased cardiometabolic risk in men compared to women is unclear. We enrolled 31 women and 29 men, aged ≥50 and ≤70 years and with body mass index ≥ 30 and <40 kg/m2. We assessed the concentrations of aminothiols (cysteine, homocysteine, and glutathione), expression of oxidant/antioxidant balance, adipomyokines (leptin, adiponectin, myostatin, and interleukin-6), markers of chronic inflammation, and vitamin D, an index of nutritional state, in plasma and serum samples by using HPLC, ELISA, and chemiluminescent immunoassay methods. We measured insulin resistance (IR) by the homeostasis model assessment (HOMA) index. Despite comparable levels of visceral adiposity, IR, and a similar dietary regimen, men showed, with respect to women, higher oxidant concentrations and lower antioxidant levels, which paralleled IR severity. Myostatin levels correlated with prooxidant aminothiols among men only. Gender-specific alterations in aminothiol status and adipomyokine profile and the gender-restricted association between these biomarkers and metabolic derangement are consistent with an increased cardiometabolic risk in men compared to age-matched women with stage I-II obesity. Strict control of redox and inflammatory status, even addressing gender-specific nutritional targets, may be useful to prevent obesity-related metabolic alterations and comorbidities.
Collapse
|
12
|
Krijt J, Sokolová J, Šilhavý J, Mlejnek P, Kubovčiak J, Liška F, Malínská H, Hüttl M, Marková I, Křížková M, Stipanuk MH, Křížek T, Ditroi T, Nagy P, Kožich V, Pravenec M. High cysteine diet reduces insulin resistance in SHR-CRP rats. Physiol Res 2021; 70:687-700. [PMID: 34505526 PMCID: PMC8820534 DOI: 10.33549/physiolres.934736] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 06/18/2021] [Indexed: 01/08/2023] Open
Abstract
Increased plasma total cysteine (tCys) has been associated with obesity and metabolic syndrome in human and some animal studies but the underlying mechanisms remain unclear. In this study, we aimed at evaluating the effects of high cysteine diet administered to SHR-CRP transgenic rats, a model of metabolic syndrome and inflammation. SHR-CRP rats were fed either standard (3.2 g cystine/kg diet) or high cysteine diet (HCD, enriched with additional 4 g L-cysteine/kg diet). After 4 weeks, urine, plasma and tissue samples were collected and parameters of metabolic syndrome, sulfur metabolites and hepatic gene expression were evaluated. Rats on HCD exhibited similar body weights and weights of fat depots, reduced levels of serum insulin, and reduced oxidative stress in the liver. The HCD did not change concentrations of tCys in tissues and body fluids while taurine in tissues and body fluids, and urinary sulfate were significantly increased. In contrast, betaine levels were significantly reduced possibly compensating for taurine elevation. In summary, increased Cys intake did not induce obesity while it ameliorated insulin resistance in the SHR-CRP rats, possibly due to beneficial effects of accumulating taurine.
Collapse
Affiliation(s)
- J Krijt
- Laboratory of Genetics of Model Diseases, Institute of Physiology of the Czech Academy of Sciences, Praha 4, Czech Republic. and Department of Pediatrics and Inherited Metabolic Disorders, Charles University-First Faculty of Medicine and General University Hospital in Prague, Praha 2, Czech Republic.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
D-ribose-L-cysteine prevents oxidative stress and cardiometabolic syndrome in high fructose high fat diet fed rats. Biomed Pharmacother 2021; 142:112017. [PMID: 34399203 DOI: 10.1016/j.biopha.2021.112017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 07/19/2021] [Accepted: 08/05/2021] [Indexed: 12/14/2022] Open
Abstract
Cardiometabolic syndrome has been linked with dietary modification. Therefore, we investigated the effects of D-ribose-L-cysteine (DRLC) in rats fed with high fructose high fat (HFHF) diet. Twenty rats (n = 5), divided into 4 groups were concurrently exposed to HFHF and/or DRLC (250 mg/kg, p.o) during the 8 weeks study. The result showed that compared to control group, HFHF group had significant impairment in lipid and glucose homeostasis, increased cardiac xanthine oxidase, systolic blood pressure, heart rate, %body weight change and fluid intake. Also, there were significant reductions in HDL-C, cardiac (GPX, NO&GGT), feed intake and relative heart weight in the latter, relative to the former. However, there were no significant differences in most of the observed physical and biochemical changes in HFHF + DRLC group compared to control. DRLC alone did not disrupt the level of biomarkers. Conclusively, DRLC prevented the manifestation of oxidative stress and cardiometabolic syndrome in HFHF-diet fed rats.
Collapse
|
14
|
Alqudah A, Wedyan M, Qnais E, Jawarneh H, McClements L. Plasma Amino Acids Metabolomics' Important in Glucose Management in Type 2 Diabetes. Front Pharmacol 2021; 12:695418. [PMID: 34335259 PMCID: PMC8320811 DOI: 10.3389/fphar.2021.695418] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 06/30/2021] [Indexed: 12/24/2022] Open
Abstract
The perturbation in plasma free amino acid metabolome has been observed previously in diabetes mellitus, and is associated with insulin resistance as well as the onset of cardiovascular disease in this population. In this study, we investigated, for the first time, changes in the amino acid profile in a group of people with and without type 2 diabetes (T2D) with normal BMI, from Jordan, who were only managed on metformin. Twenty one amino acids were evaluated in plasma samples from 124 people with T2D and 67 healthy controls, matched for age, gender and BMI, using amino acids analyser. Total amino acids, essential amino acids, non-essential amino acids and semi-essential amino acids were similar in T2D compared to healthy controls. Plasma concentrations of four essential amino acids were increased in the presence of T2D (Leucine, p < 0.01, Lysine, p < 0.001, Phenylalanine, p < 0.01, Tryptophan, p < 0.05). On the other hand, in relation to non-essential amino acids, Alanine and Serine were reduced in T2D (p < 0.01, p < 0.001, respectively), whereas Aspartate and Glutamate were increased in T2D compared to healthy controls (p < 0.001, p < 0.01, respectively). A semi-essential amino acid, Cystine, was also increased in T2D compared to healthy controls (p < 0.01). Citrulline, a metabolic indicator amino acid, demonstrated lower plasma concentration in T2D compared to healthy controls (p < 0.01). These amino acids were also correlated with fasting blood glucose and HbA1c (p < 0.05). Glutamate, glycine and arginine were correlated with the duration of metformin treatment (p < 0.05). No amino acid was correlated with lipid profiles. Disturbances in the metabolism of these amino acids are closely implicated in the pathogenesis of T2D and associated cardiovascular disease. Therefore, these perturbed amino acids could be explored as therapeutic targets to improve T2D management and prevent associated cardiovascular complications.
Collapse
Affiliation(s)
- Abdelrahim Alqudah
- Department of Clinical Pharmacy and Pharmacy Practice, Faculty of Pharmaceutical Sciences, The Hashemite University, Zarqa, Jordan
| | - Mohammed Wedyan
- Department of Biology and Biotechnology, Faculty of Science, The Hashemite University, Zarqa, Jordan
| | - Esam Qnais
- Department of Biology and Biotechnology, Faculty of Science, The Hashemite University, Zarqa, Jordan
| | - Hassan Jawarneh
- Department of Biology and Biotechnology, Faculty of Science, The Hashemite University, Zarqa, Jordan
| | - Lana McClements
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW, Australia
| |
Collapse
|
15
|
Liu HM, Liu LM, Zhang Y, Cong X, Wu LL, Xiang RL. Integrated analysis of lncRNA and mRNA expression profiles in the submandibular glands of DIO mice. Oral Dis 2021; 28:1846-1860. [PMID: 34192397 DOI: 10.1111/odi.13952] [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: 04/01/2021] [Revised: 06/17/2021] [Accepted: 06/26/2021] [Indexed: 11/29/2022]
Abstract
OBJECTIVE Obesity contributes to the dysfunction of salivary gland. To explore the specific underlying mechanism for obesity-induced hyposalivation, a model for high-fat diet-induced obese (DIO) mice were constructed to analyze long non-coding RNAs (lncRNAs) and messenger RNAs (mRNAs) expression profiles. METHODS The DIO group and control group were fed a diet containing 60 kcal% fat and a normal chow diet for 16 weeks respectively. Microarray analyses were performed to detect the expression profiles of lncRNA and mRNA in submandibular gland tissues from control group mice and DIO mice. Gene ontology, kyoto encyclopedia of genes and genomes, protein-protein interaction, coding-non-coding gene co-expression, transcription factors and competing endogenous RNA analyses were performed to examine the function of differentially expressed genes. RESULTS Microarray analyses identified that 624 lncRNAs, along with 297 mRNAs were differentially expressed. Bioinformatic analyses revealed that "complement and coagulation cascades," "glutathione metabolism," "cysteine and methionine metabolism," and "estrogen signaling pathway" were significantly associated with candidate lncRNAs. Transcription factors analysis on candidate lncRNAs revealed several genes such as tribbles pseudokinase 3 may play regulatory roles. CONCLUSIONS Our results revealed the expression profiles of lncRNAs and mRNAs and provided new insights into the mechanism of obesity-induced hyposalivation using bioinformatic analyses.
Collapse
Affiliation(s)
- Hui-Min Liu
- Department of Physiology and Pathophysiology, Peking University School of Basic Medical Sciences, Beijing, China
| | - Li-Mei Liu
- Department of Physiology and Pathophysiology, Peking University School of Basic Medical Sciences, Beijing, China
| | - Yan Zhang
- Department of Physiology and Pathophysiology, Peking University School of Basic Medical Sciences, Beijing, China
| | - Xin Cong
- Department of Physiology and Pathophysiology, Peking University School of Basic Medical Sciences, Beijing, China
| | - Li-Ling Wu
- Department of Physiology and Pathophysiology, Peking University School of Basic Medical Sciences, Beijing, China
| | - Ruo-Lan Xiang
- Department of Physiology and Pathophysiology, Peking University School of Basic Medical Sciences, Beijing, China
| |
Collapse
|
16
|
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]
|
17
|
Kožich V, Sokolová J, Morris AAM, Pavlíková M, Gleich F, Kölker S, Krijt J, Dionisi‐Vici C, Baumgartner MR, Blom HJ, Huemer M. Cystathionine β-synthase deficiency in the E-HOD registry-part I: pyridoxine responsiveness as a determinant of biochemical and clinical phenotype at diagnosis. J Inherit Metab Dis 2021; 44:677-692. [PMID: 33295057 PMCID: PMC8247016 DOI: 10.1002/jimd.12338] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 11/27/2020] [Accepted: 12/07/2020] [Indexed: 11/23/2022]
Abstract
Cystathionine β-synthase (CBS) deficiency has a wide clinical spectrum, ranging from neurodevelopmental problems, lens dislocation and marfanoid features in early childhood to adult onset disease with predominantly thromboembolic complications. We have analysed clinical and laboratory data at the time of diagnosis in 328 patients with CBS deficiency from the E-HOD (European network and registry for Homocystinurias and methylation Defects) registry. We developed comprehensive criteria to classify patients into four groups of pyridoxine responsivity: non-responders (NR), partial, full and extreme responders (PR, FR and ER, respectively). All groups showed overlapping concentrations of plasma total homocysteine while pyridoxine responsiveness inversely correlated with plasma/serum methionine concentrations. The FR and ER groups had a later age of onset and diagnosis and a longer diagnostic delay than NR and PR patients. Lens dislocation was common in all groups except ER but the age of dislocation increased with increasing responsiveness. Developmental delay was commonest in the NR group while no ER patient had cognitive impairment. Thromboembolism was the commonest presenting feature in ER patients, whereas it was least likely at presentation in the NR group. This probably is due to the differences in ages at presentation: all groups had a similar number of thromboembolic events per 1000 patient-years. Clinical severity of CBS deficiency depends on the degree of pyridoxine responsiveness. Therefore, a standardised pyridoxine-responsiveness test in newly diagnosed patients and a critical review of previous assessments is indispensable to ensure adequate therapy and to prevent or reduce long-term complications.
Collapse
Affiliation(s)
- Viktor Kožich
- Department of Pediatrics and Inherited Metabolic DisordersCharles University‐First Faculty of Medicine and General University Hospital in PraguePragueCzech Republic
| | - Jitka Sokolová
- Department of Pediatrics and Inherited Metabolic DisordersCharles University‐First Faculty of Medicine and General University Hospital in PraguePragueCzech Republic
| | - Andrew A. M. Morris
- Manchester Centre for Genomic MedicineManchester University Hospitals NHS TrustManchesterUK
| | - Markéta Pavlíková
- Department of Probability and Mathematical StatisticsCharles University‐Faculty of Mathematics and PhysicsPragueCzech Republic
| | - Florian Gleich
- Division of Neuropaediatrics and Metabolic Medicine, Centre for Paediatric and Adolescent MedicineUniversity HospitalHeidelbergGermany
| | - Stefan Kölker
- Division of Neuropaediatrics and Metabolic Medicine, Centre for Paediatric and Adolescent MedicineUniversity HospitalHeidelbergGermany
| | - Jakub Krijt
- Department of Pediatrics and Inherited Metabolic DisordersCharles University‐First Faculty of Medicine and General University Hospital in PraguePragueCzech Republic
| | - Carlo Dionisi‐Vici
- Division of MetabolismBambino Gesù Children's Research Hospital, IRCCSRomeItaly
| | - Matthias R. Baumgartner
- Division of Metabolism and Children's Research CenterUniversity Children's HospitalZurichSwitzerland
- University of ZürichZürichSwitzerland
| | - Henk J. Blom
- Department of Clinical Genetics, Center for Lysosomal and Metabolic DiseasesErasmus Medical CenterRotterdamNetherlands
| | - Martina Huemer
- Division of Metabolism and Children's Research CenterUniversity Children's HospitalZurichSwitzerland
- Department of PediatricsLandeskrankenhaus BregenzBregenzAustria
| | | |
Collapse
|
18
|
Bala CG, Rusu A, Ciobanu D, Bucsa C, Roman G. Amino Acid Signature of Oxidative Stress in Patients with Type 2 Diabetes: Targeted Exploratory Metabolomic Research. Antioxidants (Basel) 2021; 10:610. [PMID: 33921149 PMCID: PMC8071553 DOI: 10.3390/antiox10040610] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 04/11/2021] [Accepted: 04/12/2021] [Indexed: 02/06/2023] Open
Abstract
Oxidative stress plays a key role in the development of chronic diabetes-related complications. Previous metabolomic studies showed a positive association of diabetes and insulin resistance with branched-chain amino acids (AAs) and aromatic AAs. The purpose of this research is to identify distinct metabolic changes associated with increased oxidative stress, as assessed by nitrotyrosine levels, in type 2 diabetes (T2DM). Serum samples of 80 patients with insulin-treated T2DM are analyzed by AA-targeted metabolomics using ultrahigh-performance liquid chromatography/mass spectrometry. Patients are divided into two groups based on their nitrotyrosine levels: the highest level of oxidative stress (Q4 nitrotyrosine) and lower levels (Q1-Q3 nitrotyrosine). The identification of biomarkers is performed in MetaboAnalyst version 5.0 using a t-test corrected for false discovery rate, unsupervised principal component analysis and supervised partial least-squares discriminant analysis (PLS-DA). Four AAs have significantly different levels between the groups for highest and lower oxidative stress. Cysteine, phenylalanine and tyrosine are substantially increased while citrulline is decreased (p-value <0.05 and variable importance in the projection [VIP] >1). Corresponding pathways that might be disrupted in patients with high oxidative stress are phenylalanine, tyrosine and tryptophan biosynthesis, arginine biosynthesis, phenylalanine metabolism, cysteine and methionine metabolism and tyrosine metabolism.
Collapse
Affiliation(s)
- Cornelia G. Bala
- Department of Diabetes and Nutrition Diseases, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400006 Cluj-Napoca, Romania; (C.G.B.); (D.C.); (G.R.)
| | - Adriana Rusu
- Department of Diabetes and Nutrition Diseases, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400006 Cluj-Napoca, Romania; (C.G.B.); (D.C.); (G.R.)
| | - Dana Ciobanu
- Department of Diabetes and Nutrition Diseases, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400006 Cluj-Napoca, Romania; (C.G.B.); (D.C.); (G.R.)
| | - Camelia Bucsa
- Drug Information Research Centre, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania;
| | - Gabriela Roman
- Department of Diabetes and Nutrition Diseases, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400006 Cluj-Napoca, Romania; (C.G.B.); (D.C.); (G.R.)
| |
Collapse
|
19
|
Stolt E, Olsen T, Elshorbagy A, Kožich V, van Greevenbroek M, Øvrebø B, Thoresen M, Refsum H, Retterstøl K, Vinknes KJ. Sulfur amino acid restriction, energy metabolism and obesity: a study protocol of an 8-week randomized controlled dietary intervention with whole foods and amino acid supplements. J Transl Med 2021; 19:153. [PMID: 33858441 PMCID: PMC8051033 DOI: 10.1186/s12967-021-02824-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Accepted: 04/09/2021] [Indexed: 01/21/2023] Open
Abstract
Background Dietary sulfur amino acid (SAA) restriction is an established animal model for increasing lifespan and improving metabolic health. Data from human studies are limited. In the study outlined in this protocol, we will evaluate if dietary SAA restriction can reduce body weight and improve resting energy expenditure (REE) and parameters related to metabolic health. Method/design Men and women (calculated sample size = 60), aged 18–45 years, with body mass index of 27–35 kg/m2 will be included in a double-blind 8-week dietary intervention study. The participants will be randomized in a 1:1 manner to a diet with either low or high SAA. Both groups will receive an equal base diet consisting of low-SAA plant-based whole foods and an amino acid supplement free of SAA. Contrasting SAA contents will be achieved using capsules with or without methionine and cysteine (SAAhigh, total diet SAA ~ 50–60 mg/kg body weight/day; SAAlow, total diet SAA ~ 15–25 mg/kg body weight/day). The primary outcome is body weight change. Data and material collection will also include body composition (dual X-ray absorptiometry), resting energy expenditure (whole-room indirect calorimetry) and samples of blood, urine, feces and adipose tissue at baseline, at 4 weeks and at study completion. Measures will be taken to promote and monitor diet adherence. Data will be analyzed using linear mixed model regression to account for the repeated measures design and within-subject correlation. Discussion The strength of this study is the randomized double-blind design. A limitation is the restrictive nature of the diet which may lead to poor compliance. If this study reveals a beneficial effect of the SAAlow diet on body composition and metabolic health, it opens up for new strategies for prevention and treatment of overweight, obesity and its associated disorders. Trial registration ClinicalTrials.gov: NCT04701346, Registration date: January 8th, 2021 Supplementary Information The online version contains supplementary material available at 10.1186/s12967-021-02824-3.
Collapse
Affiliation(s)
- Emma Stolt
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Sognsvannveien 9, 0372, Oslo, Norway
| | - Thomas Olsen
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Sognsvannveien 9, 0372, Oslo, Norway.
| | - Amany Elshorbagy
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Sognsvannveien 9, 0372, Oslo, Norway.,Department of Pharmacology, University of Oxford, Oxford, UK.,Department of Physiology, Faculty of Medicine, University of Alexandria, Alexandria, Egypt
| | - Viktor Kožich
- 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
| | - Bente Øvrebø
- Department of Sports Science and Physical Education, Faculty of Health and Sport Sciences, University of Agder, Kristiansand, 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, Sognsvannveien 9, 0372, Oslo, Norway
| | - Kjetil Retterstøl
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Sognsvannveien 9, 0372, Oslo, Norway.,The Lipid Clinic, Oslo University Hospital, Oslo, Norway
| | - Kathrine J Vinknes
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Sognsvannveien 9, 0372, Oslo, Norway
| |
Collapse
|
20
|
Ren H, Liu TC, Lu Y, Zhang K, Xu Y, Zhou P, Tang X. A comparison study of the influence of milk protein versus whey protein in high-protein diets on adiposity in rats. Food Funct 2021; 12:1008-1019. [PMID: 33502407 DOI: 10.1039/d0fo01960g] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
High-protein diets are known to reduce weight and fat deposition. However, there have been only a few studies on the efficacy of different types of high-protein diets in preventing obesity. Therefore, the emphasis of this study lies in comparing the efficacy of two high-protein diets (milk protein and whey protein) in preventing obesity and exploring specific mechanisms. Eighty Sprague Dawley rats were divided into two groups and fed with milk protein concentrate (MPC) and whey protein concentrate (WPC) for 12 weeks. Each group was divided into four levels: two low-fat regimens with either low or high protein content (L-14%, L-40%) and two high-fat regimens with either low or high protein content (H-14%, H-40%). The studies we have performed showed that rats treated with MPC at the 40% protein level had significantly reduced body weight, fat weight and fat ratio gain induced by a high-fat diet, while the protein level in the WPC group had no effect on body weight or body fat in rats fed with a high-fat diet. What is more, rats fed with MPC at the H-40% energy level showed a significant decrease in plasma triglyceride, total cholesterol and low-density lipoprotein cholesterol levels and a significant increase in plasma high-density lipoprotein cholesterol levels compared with the H-14% energy level group. In contrast, in the WPC groups, increasing the protein content in high-fat diets had no significant influence on plasma lipid levels. The results of the amino acid composition of the two proteins and plasma showed that the MPC diet of 40% protein level increased the transsulfuration pathway in rats, thereby increasing the level of H2S. This research work has shown that not all types of high-protein diets can effectively prevent obesity induced by high-fat diets, as effectiveness depends on the amino acid composition of the protein.
Collapse
Affiliation(s)
- Haoyi Ren
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, Jiangsu, China.
| | | | | | | | | | | | | |
Collapse
|
21
|
Olsen T, Øvrebø B, Turner C, Bastani NE, Refsum H, Vinknes KJ. Effects of short-term methionine and cysteine restriction and enrichment with polyunsaturated fatty acids on oral glucose tolerance, plasma amino acids, fatty acids, lactate and pyruvate: results from a pilot study. BMC Res Notes 2021; 14:43. [PMID: 33531059 PMCID: PMC7852127 DOI: 10.1186/s13104-021-05463-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 01/22/2021] [Indexed: 11/22/2022] Open
Abstract
Objective In this 7-day pilot study we randomized healthy, normal-weight men and women to either a dietary intervention with methionine and cysteine restriction enriched in PUFA (Met/Cyslow + PUFA, n = 7) or with high contents of methionine, cysteine and SFA (Met/Cyshigh + SFA, n = 7). The objective was to describe the short-term responses in oral glucose tolerance, amino acid profile, total fatty acid profile, pyruvate and lactate following a Met/Cyslow + PUFA diet vs. Met/Cyshigh + SFA. Results The diet groups consisted of five women and two men, aged 20–38 years. After the 7-d intervention median pre- and post-oral glucose tolerance test (OGTT) glucose concentrations were 5 mmol/L and 4 mmol/L respectively in the Met/Cyslow + PUFA group. In the Met/Cyshigh + SFA group, median pre- and post-OGTT glucose concentrations were 4.8 mmol/L and 4.65 mmol/L after the 7-d intervention. The responses in the amino acid profiles were similar in both groups during the intervention with the exception of serine. Fatty acids decreased from baseline to day 7 in both groups. Plasma lactate and pyruvate were similar for both groups with an increase to day 3 before approaching baseline values at day 7. Trial registration ClinicalTrials.gov: NCT02647970, registration date: January 6th 2016.
Collapse
Affiliation(s)
- Thomas Olsen
- Department of Nutrition, Institute of Medical Biosciences, Domus Medica, University of Oslo, Sognsvannsveien 9, 0372, Oslo, Norway.
| | - Bente Øvrebø
- Department of Nutrition, Institute of Medical Biosciences, Domus Medica, University of Oslo, Sognsvannsveien 9, 0372, Oslo, Norway.,Department of Sport Science and Physical Education, University of Agder, 4604, Kristiansand, Norway
| | - Cheryl Turner
- Department of Pharmacology, University of Oxford, Oxford, OX1 3QT, UK
| | - Nasser E Bastani
- Department of Nutrition, Institute of Medical Biosciences, Domus Medica, University of Oslo, Sognsvannsveien 9, 0372, Oslo, Norway
| | - Helga Refsum
- Department of Nutrition, Institute of Medical Biosciences, Domus Medica, University of Oslo, Sognsvannsveien 9, 0372, Oslo, Norway
| | - Kathrine J Vinknes
- Department of Nutrition, Institute of Medical Biosciences, Domus Medica, University of Oslo, Sognsvannsveien 9, 0372, Oslo, Norway
| |
Collapse
|
22
|
Puckett DL, Alquraishi M, Chowanadisai W, Bettaieb A. The Role of PKM2 in Metabolic Reprogramming: Insights into the Regulatory Roles of Non-Coding RNAs. Int J Mol Sci 2021; 22:1171. [PMID: 33503959 PMCID: PMC7865720 DOI: 10.3390/ijms22031171] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 01/13/2021] [Accepted: 01/14/2021] [Indexed: 01/17/2023] Open
Abstract
Pyruvate kinase is a key regulator in glycolysis through the conversion of phosphoenolpyruvate (PEP) into pyruvate. Pyruvate kinase exists in various isoforms that can exhibit diverse biological functions and outcomes. The pyruvate kinase isoenzyme type M2 (PKM2) controls cell progression and survival through the regulation of key signaling pathways. In cancer cells, the dimer form of PKM2 predominates and plays an integral role in cancer metabolism. This predominance of the inactive dimeric form promotes the accumulation of phosphometabolites, allowing cancer cells to engage in high levels of synthetic processing to enhance their proliferative capacity. PKM2 has been recognized for its role in regulating gene expression and transcription factors critical for health and disease. This role enables PKM2 to exert profound regulatory effects that promote cancer cell metabolism, proliferation, and migration. In addition to its role in cancer, PKM2 regulates aspects essential to cellular homeostasis in non-cancer tissues and, in some cases, promotes tissue-specific pathways in health and diseases. In pursuit of understanding the diverse tissue-specific roles of PKM2, investigations targeting tissues such as the kidney, liver, adipose, and pancreas have been conducted. Findings from these studies enhance our understanding of PKM2 functions in various diseases beyond cancer. Therefore, there is substantial interest in PKM2 modulation as a potential therapeutic target for the treatment of multiple conditions. Indeed, a vast plethora of research has focused on identifying therapeutic strategies for targeting PKM2. Recently, targeting PKM2 through its regulatory microRNAs, long non-coding RNAs (lncRNAs), and circular RNAs (circRNAs) has gathered increasing interest. Thus, the goal of this review is to highlight recent advancements in PKM2 research, with a focus on PKM2 regulatory microRNAs and lncRNAs and their subsequent physiological significance.
Collapse
Affiliation(s)
- Dexter L. Puckett
- Department of Nutrition, University of Tennessee Knoxville, Knoxville, TN 37996, USA; (D.L.P.); (M.A.)
| | - Mohammed Alquraishi
- Department of Nutrition, University of Tennessee Knoxville, Knoxville, TN 37996, USA; (D.L.P.); (M.A.)
| | - Winyoo Chowanadisai
- Department of Nutrition, Oklahoma State University, Stillwater, OK 74078, USA;
| | - Ahmed Bettaieb
- Department of Nutrition, University of Tennessee Knoxville, Knoxville, TN 37996, USA; (D.L.P.); (M.A.)
| |
Collapse
|
23
|
Rodgers M, Migdal AL, Rodríguez TG, Chen ZZ, Nath AK, Gerszten RE, Kasid N, Toschi E, Tripaldi J, Heineman B, Phan M, Ngo L, Maratos-Flier E, Dushay J. Weight Loss Outcomes Among Early High Responders to Exenatide Treatment: A Randomized, Placebo Controlled Study in Overweight and Obese Women. Front Endocrinol (Lausanne) 2021; 12:742873. [PMID: 34867786 PMCID: PMC8635796 DOI: 10.3389/fendo.2021.742873] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 10/18/2021] [Indexed: 01/04/2023] Open
Abstract
OBJECTIVE As there is significant heterogeneity in the weight loss response to pharmacotherapy, one of the most important clinical questions in obesity medicine is how to predict an individual's response to pharmacotherapy. The present study examines patterns of weight loss among overweight and obese women who demonstrated early robust response to twice daily exenatide treatment compared to those treated with hypocaloric diet and matched placebo injections. METHODS We randomized 182 women (BMI 25-48 kg/m2) to treatment with exenatide alone or matched placebo injections plus hypocaloric diet. In both treatment groups, women who demonstrated ≥ 5% weight loss at 12 weeks were characterized as high responders and those who lost ≥10% of body weight were classified as super responders. Our primary outcome was long-term change in body weight among early high responders to either treatment. An exploratory metabolomic analysis was also performed. RESULTS We observed individual variability in weight loss with both exenatide and hypocaloric diet plus placebo injections. There was a trend toward a higher percentage of subjects who achieved ≥ 5% weight loss with exenatide compared to diet (56% of those treated with exenatide, 76% of those treated with diet, p = 0.05) but no significant difference in those who achieved ≥ 10% weight loss (23% of individuals treated with exenatide and 36% of those treated with diet, p = 0.55). In both treatment groups, higher weight loss at 3 months of treatment predicted super responder status (diet p=0.0098, exenatide p=0.0080). Both treatment groups also demonstrated similar peak weight loss during the study period. We observed lower cysteine concentrations in the exenatide responder group (0.81 vs 0.48 p < 0.0001) and a trend toward higher levels of serotonin, aminoisobutyric acid, anandamide, and sarcosine in the exenatide super responder group. CONCLUSION In a population of early high responders, longer term weight loss with exenatide treatment is similar to that achieved with a hypocaloric diet. CLINICAL TRIAL REGISTRATION www.clinicaltrialsgov, identifier NCT01590433.
Collapse
|
24
|
Wu ZE, Fraser K, Kruger MC, Sequeira IR, Yip W, Lu LW, Plank LD, Murphy R, Cooper GJS, Martin JC, Hollingsworth KG, Poppitt SD. Untargeted metabolomics reveals plasma metabolites predictive of ectopic fat in pancreas and liver as assessed by magnetic resonance imaging: the TOFI_Asia study. Int J Obes (Lond) 2021; 45:1844-1854. [PMID: 33994541 PMCID: PMC8310794 DOI: 10.1038/s41366-021-00854-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 04/10/2021] [Accepted: 04/30/2021] [Indexed: 02/04/2023]
Abstract
BACKGROUND Excess visceral obesity and ectopic organ fat is associated with increased risk of cardiometabolic disease. However, circulating markers for early detection of ectopic fat, particularly pancreas and liver, are lacking. METHODS Lipid storage in pancreas, liver, abdominal subcutaneous adipose tissue (SAT) and visceral adipose tissue (VAT) from 68 healthy or pre-diabetic Caucasian and Chinese women enroled in the TOFI_Asia study was assessed by magnetic resonance imaging/spectroscopy (MRI/S). Plasma metabolites were measured with untargeted liquid chromatography-mass spectroscopy (LC-MS). Multivariate partial least squares (PLS) regression identified metabolites predictive of VAT/SAT and ectopic fat; univariate linear regression adjusting for potential covariates identified individual metabolites associated with VAT/SAT and ectopic fat; linear regression adjusted for ethnicity identified clinical and anthropometric correlates for each fat depot. RESULTS PLS identified 56, 64 and 31 metabolites which jointly predicted pancreatic fat (R2Y = 0.81, Q2 = 0.69), liver fat (RY2 = 0.8, Q2 = 0.66) and VAT/SAT ((R2Y = 0.7, Q2 = 0.62)) respectively. Among the PLS-identified metabolites, none of them remained significantly associated with pancreatic fat after adjusting for all covariates. Dihydrosphingomyelin (dhSM(d36:0)), 3 phosphatidylethanolamines, 5 diacylglycerols (DG) and 40 triacylglycerols (TG) were associated with liver fat independent of covariates. Three DGs and 12 TGs were associated with VAT/SAT independent of covariates. Notably, comparison with clinical correlates showed better predictivity of ectopic fat by these PLS-identified plasma metabolite markers. CONCLUSIONS Untargeted metabolomics identified candidate markers of visceral and ectopic fat that improved fat level prediction over clinical markers. Several plasma metabolites were associated with level of liver fat and VAT/SAT ratio independent of age, total and visceral adiposity, whereas pancreatic fat deposition was only associated with increased sulfolithocholic acid independent of adiposity-related parameters, but not age.
Collapse
Affiliation(s)
- Zhanxuan E. Wu
- grid.417738.e0000 0001 2110 5328Food Nutrition & Health, Food and Bio-based Products, AgResearch Limited, Palmerston North, New Zealand ,grid.148374.d0000 0001 0696 9806School of Health Sciences, Massey University, Palmerston North, New Zealand ,High-Value Nutrition National Science Challenge, Auckland, New Zealand
| | - Karl Fraser
- grid.417738.e0000 0001 2110 5328Food Nutrition & Health, Food and Bio-based Products, AgResearch Limited, Palmerston North, New Zealand ,High-Value Nutrition National Science Challenge, Auckland, New Zealand ,grid.148374.d0000 0001 0696 9806Riddet Institute, Massey University, Palmerston North, New Zealand
| | - Marlena C. Kruger
- grid.148374.d0000 0001 0696 9806School of Health Sciences, Massey University, Palmerston North, New Zealand ,grid.148374.d0000 0001 0696 9806Riddet Institute, Massey University, Palmerston North, New Zealand
| | - Ivana R. Sequeira
- High-Value Nutrition National Science Challenge, Auckland, New Zealand ,grid.9654.e0000 0004 0372 3343Human Nutrition Unit, School of Biological Sciences, University of Auckland, Auckland, New Zealand
| | - Wilson Yip
- High-Value Nutrition National Science Challenge, Auckland, New Zealand ,grid.9654.e0000 0004 0372 3343Human Nutrition Unit, School of Biological Sciences, University of Auckland, Auckland, New Zealand
| | - Louise W. Lu
- High-Value Nutrition National Science Challenge, Auckland, New Zealand ,grid.9654.e0000 0004 0372 3343Human Nutrition Unit, School of Biological Sciences, University of Auckland, Auckland, New Zealand
| | - Lindsay D. Plank
- grid.9654.e0000 0004 0372 3343Department of Surgery, University of Auckland, Auckland, New Zealand
| | - Rinki Murphy
- High-Value Nutrition National Science Challenge, Auckland, New Zealand ,grid.9654.e0000 0004 0372 3343Department of Medicine, University of Auckland, Auckland, New Zealand
| | - Garth J. S. Cooper
- grid.9654.e0000 0004 0372 3343Department of Medicine, University of Auckland, Auckland, New Zealand ,grid.9654.e0000 0004 0372 3343School of Biological Sciences University of Auckland, Auckland, New Zealand ,grid.5379.80000000121662407Centre for Advanced Discovery and Experimental Therapeutics, School of Medical Sciences, University of Manchester, Manchester, UK
| | - Jean-Charles Martin
- grid.5399.60000 0001 2176 4817Aix-Marseille University, INSERM, INRAe, C2VN, BioMeT, Marseille, France
| | - Kieren G. Hollingsworth
- grid.1006.70000 0001 0462 7212Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Sally D. Poppitt
- High-Value Nutrition National Science Challenge, Auckland, New Zealand ,grid.148374.d0000 0001 0696 9806Riddet Institute, Massey University, Palmerston North, New Zealand ,grid.9654.e0000 0004 0372 3343Human Nutrition Unit, School of Biological Sciences, University of Auckland, Auckland, New Zealand ,grid.9654.e0000 0004 0372 3343Department of Medicine, University of Auckland, Auckland, New Zealand ,grid.9654.e0000 0004 0372 3343School of Biological Sciences University of Auckland, Auckland, New Zealand
| |
Collapse
|
25
|
Pecze L, Randi EB, Szabo C. Meta-analysis of metabolites involved in bioenergetic pathways reveals a pseudohypoxic state in Down syndrome. Mol Med 2020; 26:102. [PMID: 33167881 PMCID: PMC7653803 DOI: 10.1186/s10020-020-00225-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 10/12/2020] [Indexed: 12/14/2022] Open
Abstract
Clinical observations and preclinical studies both suggest that Down syndrome (DS) may be associated with significant metabolic and bioenergetic alterations. However, the relevant scientific literature has not yet been systematically reviewed. The aim of the current study was to conduct a meta-analysis of metabolites involved in bioenergetics pathways in DS to conclusively determine the difference between DS and control subjects. We discuss these findings and their potential relevance in the context of pathogenesis and experimental therapy of DS. Articles published before July 1, 2020, were identified by using the search terms “Down syndrome” and “metabolite name” or “trisomy 21” and “metabolite name”. Moreover, DS-related metabolomics studies and bioenergetics literature were also reviewed. 41 published reports and associated databases were identified, from which the descriptive information and the relevant metabolomic parameters were extracted and analyzed. Mixed effect model revealed the following changes in DS: significantly decreased ATP, CoQ10, homocysteine, serine, arginine and tyrosine; slightly decreased ADP; significantly increased uric acid, succinate, lactate and cysteine; slightly increased phosphate, pyruvate and citrate. However, the concentrations of AMP, 2,3-diphosphoglycerate, glucose, and glutamine were comparable in the DS vs. control populations. We conclude that cells of subjects with DS are in a pseudo-hypoxic state: the cellular metabolic and bio-energetic mechanisms exhibit pathophysiological alterations that resemble the cellular responses associated with hypoxia, even though the supply of the cells with oxygen is not disrupted. This fundamental alteration may be, at least in part, responsible for a variety of functional deficits associated with DS, including reduced exercise difference, impaired neurocognitive status and neurodegeneration.
Collapse
Affiliation(s)
- Laszlo Pecze
- Chair of Pharmacology, Section of Medicine, University of Fribourg, Fribourg, Switzerland
| | - Elisa B Randi
- Chair of Pharmacology, Section of Medicine, University of Fribourg, Fribourg, Switzerland
| | - Csaba Szabo
- Chair of Pharmacology, Section of Medicine, University of Fribourg, Fribourg, Switzerland.
| |
Collapse
|
26
|
Schwinger C, Chowdhury R, Sharma S, Bhandari N, Taneja S, Ueland PM, Strand TA. Association of Plasma Total Cysteine and Anthropometric Status in 6-30 Months Old Indian Children. Nutrients 2020; 12:nu12103146. [PMID: 33076294 PMCID: PMC7602373 DOI: 10.3390/nu12103146] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 10/09/2020] [Accepted: 10/10/2020] [Indexed: 12/21/2022] Open
Abstract
High-quality protein has been associated with child growth; however, the role of the amino acid cysteine remains unclear. The aim was to measure the extent to which plasma total cysteine (tCys) concentration is associated with anthropometric status in children aged 6–30 months living in New Delhi, India. The study was a prospective cohort study including 2102 children. We calculated Z-scores for height-for-age (HAZ), weight-for-height (WHZ), or weight-for-age (WAZ) according to the WHO Child Growth Standards. We used multiple regression models to estimate the association between tCys and the anthropometric indices. A high proportion of the children were categorized as malnourished at enrolment; 41% were stunted (HAZ ≤ −2), 19% were wasted (WHZ ≤ −2) and 42% underweight (WAZ ≤ −2). Plasma total cysteine (tCys) was significantly associated with HAZ, WHZ and WAZ after adjusting for relevant confounders (p < 0.001). Low tCys (≤25th percentile) was associated with a decrease of 0.28 Z-scores for HAZ, 0.10 Z-scores for WHZ, and 0.21 Z-scores for WAZ compared to being >25th percentile. In young Indian children from low-to-middle socioeconomic neighborhoods, a low plasma total cysteine concentration was associated with an increased risk of poor anthropometric status.
Collapse
Affiliation(s)
- Catherine Schwinger
- Centre for Intervention Science in Maternal and Child Health, Centre for International Health, Department of Global Public Health and Primary Care, University of Bergen, Catherine Schwinger, Årstadveien 21, 5009 Bergen, Norway; (R.C.); (N.B.); (T.A.S.)
- Correspondence: ; Tel.: +47-5558-9733
| | - Ranadip Chowdhury
- Centre for Intervention Science in Maternal and Child Health, Centre for International Health, Department of Global Public Health and Primary Care, University of Bergen, Catherine Schwinger, Årstadveien 21, 5009 Bergen, Norway; (R.C.); (N.B.); (T.A.S.)
- Society for Applied Studies, New Delhi 110016, India;
| | - Shakun Sharma
- Department of Child Health, Institute of Medicine, Tribuhvan University, Kathmandu 44613, Nepal;
| | - Nita Bhandari
- Centre for Intervention Science in Maternal and Child Health, Centre for International Health, Department of Global Public Health and Primary Care, University of Bergen, Catherine Schwinger, Årstadveien 21, 5009 Bergen, Norway; (R.C.); (N.B.); (T.A.S.)
- Society for Applied Studies, New Delhi 110016, India;
| | - Sunita Taneja
- Society for Applied Studies, New Delhi 110016, India;
| | - Per M. Ueland
- Department of Clinical Science, University of Bergen,5020 Bergen, Norway;
| | - Tor A. Strand
- Centre for Intervention Science in Maternal and Child Health, Centre for International Health, Department of Global Public Health and Primary Care, University of Bergen, Catherine Schwinger, Årstadveien 21, 5009 Bergen, Norway; (R.C.); (N.B.); (T.A.S.)
- Department of Research, Innlandet Hospital Trust, 2618 Lillehammer, Norway
| |
Collapse
|
27
|
Stabler SP. Alterations in Sulfur Amino Acids as Biomarkers of Disease. J Nutr 2020; 150:2532S-2537S. [PMID: 33000156 DOI: 10.1093/jn/nxaa118] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 03/31/2020] [Accepted: 04/02/2020] [Indexed: 01/30/2023] Open
Abstract
Homocysteine (Hcy) is methylated by methionine synthase to form methionine with methyl-cobalamin as a cofactor. The reaction demethylates 5-methyltetrahydrofolate to tetrahydrofolate, which is required for DNA and RNA synthesis. Deficiency of either of the cobalamin (Cbl) and/or folate cofactors results in elevated Hcy and megaloblastic anemia. Elevated Hcy is a sensitive biomarker of Cbl and/or folate status and more specific than serum vitamin assays. Elevated Hcy normalizes when the correct vitamin is given. Elevated Hcy is associated with alcohol use disorder and drugs that target folate or Cbl metabolism, and is a risk factor for thrombotic vascular disease. Elevated methionine and cystathionine are associated with liver disease. Elevated Hcy, cystathionine, and cysteine, but not methionine, are common in patients with chronic renal failure. Higher cysteine predicts obesity and future weight gain. Serum S-adenosylhomocysteine (AdoHcy) is elevated in Cbl deficiency and chronic renal failure. Drugs that require methylation for catabolism may deplete liver S-adenosylmethionine and raise AdoHcy and Hcy. Deficiency of Cbl or folate or perturbations of their metabolism cause major changes in sulfur amino acids.
Collapse
Affiliation(s)
- Sally P Stabler
- Division of Hematology, Department of Medicine, University of Colorado Anschutz Medical Campus, Denver, CO, USA
| |
Collapse
|
28
|
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
|
29
|
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
|
30
|
Cirulli F, Musillo C, Berry A. Maternal Obesity as a Risk Factor for Brain Development and Mental Health in the Offspring. Neuroscience 2020; 447:122-135. [PMID: 32032668 DOI: 10.1016/j.neuroscience.2020.01.023] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 01/10/2020] [Accepted: 01/14/2020] [Indexed: 12/30/2022]
Abstract
Maternal obesity plays a key role in the health trajectory of the offspring. Although research on this topic has largely focused on the potential of this condition to increase the risk for child obesity, it is becoming more and more evident that it can also significantly impact cognitive function and mental health. The mechanisms underlying these effects are starting to be elucidated and point to the placenta as a critical organ that may mediate changes in the response to stress, immune function and oxidative stress. Long-term effects of maternal obesity may rely upon epigenetic changes in selected genes that are involved in metabolic and trophic regulations of the brain. More recent evidence also indicates the gut microbiota as a potential mediator of these effects. Overall, understanding cause-effect relationships can allow the development of preventive measures that could rely upon dietary changes in the mother and the offspring. Addressing diets appears more feasible than developing new pharmacological targets and has the potential to affect the multiple interconnected physiological pathways engaged by these complex regulations, allowing prevention of both metabolic and mental disorders.
Collapse
Affiliation(s)
- Francesca Cirulli
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, Rome, Italy.
| | - Chiara Musillo
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, Rome, Italy
| | - Alessandra Berry
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, Rome, Italy
| |
Collapse
|
31
|
Xie X, Huo F, Yue Y, Chao J, Yin C. NEM assisted real-time fluorescence detection of Cys in cytoplasm and mice imaging by a Coumarin probe containing carboxyl group. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2020; 225:117517. [PMID: 31521001 DOI: 10.1016/j.saa.2019.117517] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 07/29/2019] [Accepted: 09/04/2019] [Indexed: 06/10/2023]
Abstract
Alterations of the homeostasis balance of cysteine (Cys) are associated with a variety of diseases and cellular functions, and therefore, Cys dynamic real-time living cell intracellular imaging and quantification are important for understanding the pathophysiological processes. Thus, Cys probe that can permeate high efficiently is the first one to be affected. In fact, it is difficult for organic molecular probes to infiltrate cells because of the unique structure of the cell membrane. In this work, we found that probe containing-carboxyl just stagnated in cytomembrane due to carboxyl of probe and amino group of membrane protein forming peptide chains, nevertheless, the addition of NEM, improved membrane permeability by NEM reacting with sulfhydryl of membrane protein, which made probe permeate high efficiently and sequentially real-time detect the Cys in cytoplasm. It is the first time noted that NEM can regulate Cys probe containing-carboxyl for high efficient detection in cytoplasm. Additionally, probe was successfully applied to image Cys in mouse.
Collapse
Affiliation(s)
- Xixi Xie
- Department of Chemistry, Xinzhou Teachers University, Xinzhou 034000, Shanxi, China; Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Key Laboratory of Materials for Energy Conversion and Storage of Shanxi Province, Institute of Molecular Science, Shanxi University, Taiyuan 030006, China
| | - Fangjun Huo
- Research Institute of Applied Chemistry, Shanxi University, Taiyuan 030006, China
| | - Yongkang Yue
- Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Key Laboratory of Materials for Energy Conversion and Storage of Shanxi Province, Institute of Molecular Science, Shanxi University, Taiyuan 030006, China
| | - Jianbin Chao
- Research Institute of Applied Chemistry, Shanxi University, Taiyuan 030006, China
| | - Caixia Yin
- Department of Chemistry, Xinzhou Teachers University, Xinzhou 034000, Shanxi, China; Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Key Laboratory of Materials for Energy Conversion and Storage of Shanxi Province, Institute of Molecular Science, Shanxi University, Taiyuan 030006, China.
| |
Collapse
|
32
|
Hypercysteinemia, A Potential Risk Factor for Central Obesity and Related Disorders in Azores, Portugal. J Nutr Metab 2019; 2019:1826780. [PMID: 31321096 PMCID: PMC6609363 DOI: 10.1155/2019/1826780] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Accepted: 05/27/2019] [Indexed: 01/08/2023] Open
Abstract
In Azores, the standardized mortality rate for coronary artery disease (CAD) is nearly the double when compared to mainland Portugal. The aim of this study was to compare the prevalence of conventional CAD risk factors, as well as the plasma aminothiol profile (and its major determinants), between two groups of healthy subjects from Ponta Delgada (in Azores) and Lisbon (in mainland) cities, searching for precocious biomarker(s) of the disease. The study groups consisted of 101 healthy volunteers from Ponta Delgada (PDL) and 121 from Lisbon, aged 20–69 years. No differences in the prevalence of classical CAD risk factors were found between the study groups, except in physical inactivity and related central obesity, which were both higher in PDL men than in those from Lisbon. Hypercysteinemia, which seems to result from sulfur-rich amino acid diets and/or vitamin B12 malabsorption, revealed to be significantly more prevalent in PDL vs. Lisbon subjects (18% vs. 4%, P=0.001), namely, in male gender. Moreover, plasma Cys levels predicted waist circumference (β coefficient = 0.102, P=0.032) and concomitant central obesity and were also associated with insulin resistance. Nevertheless, hyperhomocysteinemia prevalence was similar in both groups, despite the fact that PDL subjects exhibited a higher rate of vitamin B12 deficiency compared to those from Lisbon (19% vs. 6%, P=0.003). Owing to the nature of this study design, a cause-effect relationship between high plasma Cys levels and central obesity or CAD risk could not be derived, but results strongly suggest that hypercysteinemia is a potential risk factor for metabolic disorders, i.e., obesity and insulin resistance, and CAD in Azores, a hypothesis that asks for confirmation through further large prospective studies.
Collapse
|
33
|
Wang P, Duan L, Liao Y. A retrievable and highly selective peptide-based fluorescent probe for detection of Cd2+ and Cys in aqueous solutions and live cells. Microchem J 2019. [DOI: 10.1016/j.microc.2019.02.004] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
34
|
Disposable electrochemical sensor based on copper-electrodeposited screen-printed gold electrode and its application in sensing l-Cysteine. Electrochim Acta 2019. [DOI: 10.1016/j.electacta.2018.08.140] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
|
35
|
Lee S, Olsen T, Vinknes KJ, Refsum H, Gulseth HL, Birkeland KI, Drevon CA. Plasma Sulphur-Containing Amino Acids, Physical Exercise and Insulin Sensitivity in Overweight Dysglycemic and Normal Weight Normoglycemic Men. Nutrients 2018; 11:nu11010010. [PMID: 30577516 PMCID: PMC6356487 DOI: 10.3390/nu11010010] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Revised: 12/19/2018] [Accepted: 12/19/2018] [Indexed: 12/30/2022] Open
Abstract
Plasma sulphur-containing amino acids and related metabolites are associated with insulin sensitivity, although the mechanisms are unclear. We examined the effect of exercise on this relationship. Dysglycemic (n = 13) and normoglycemic (n = 13) men underwent 45 min cycling before and after 12 weeks exercise intervention. We performed hyperinsulinemic euglycemic clamp, mRNA-sequencing of skeletal muscle and adipose tissue biopsies, and targeted profiling of plasma metabolites by LC-MS/MS. Insulin sensitivity increased similarly in dysglycemic and normoglycemic men after 12 weeks of exercise, in parallel to similar increases in concentration of plasma glutamine, and decreased concentrations of plasma glutamate, cysteine, taurine, and glutathione. Change in plasma concentrations of cysteine and glutathione exhibited the strongest correlations to exercise-improved insulin sensitivity, and expression of a cluster of genes essential for oxidative phosphorylation and fatty acid metabolism in both skeletal muscle and adipose tissue, as well as mitochondria-related genes such as mitofilin. Forty-five min of cycling decreased plasma concentrations of glutamine and methionine, and increased plasma concentrations of glutamate, homocysteine, cystathionine, cysteine, glutathione, and taurine. Similar acute responses were seen in both groups before and after the 12 weeks training period. Both acute and long-term exercise may influence transsulphuration and glutathione biosynthesis, linking exercise-improved insulin sensitivity to oxidative stress and mitochondrial function.
Collapse
Affiliation(s)
- Sindre Lee
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, 0317 Oslo, Norway.
- Department of Endocrinology, Morbid Obesity and Preventive Medicine, Oslo University Hospital; 0586 Oslo, Norway.
| | - Thomas Olsen
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, 0317 Oslo, Norway.
| | - Kathrine J Vinknes
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, 0317 Oslo, Norway.
| | - Helga Refsum
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, 0317 Oslo, Norway.
| | - Hanne L Gulseth
- Department of Endocrinology, Morbid Obesity and Preventive Medicine, Oslo University Hospital; 0586 Oslo, Norway.
- Department of Non-communicable Diseases, Norwegian Institute of Public Health; 0473 Oslo, Norway.
| | - Kåre I Birkeland
- Department of Endocrinology, Morbid Obesity and Preventive Medicine, Oslo University Hospital; 0586 Oslo, Norway.
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo; 0450 Oslo, Norway.
| | - Christian A Drevon
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, 0317 Oslo, Norway.
| |
Collapse
|
36
|
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: 36] [Impact Index Per Article: 5.1] [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
|
37
|
Elshorbagy AK, Samocha-Bonet D, Jernerén F, Turner C, Refsum H, Heilbronn LK. Food Overconsumption in Healthy Adults Triggers Early and Sustained Increases in Serum Branched-Chain Amino Acids and Changes in Cysteine Linked to Fat Gain. J Nutr 2018; 148:1073-1080. [PMID: 29901727 DOI: 10.1093/jn/nxy062] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Accepted: 03/06/2018] [Indexed: 01/03/2023] Open
Abstract
Background Plasma concentrations of branched-chain amino acids (BCAAs) and the sulfur-containing amino acid cysteine are associated with obesity and insulin resistance. BCAAs predict future diabetes. Objective We investigated amino acid changes during food overconsumption. Methods Forty healthy men and women with a body mass index (mean ± SEM) of 25.6 ± 0.6 were overfed by 1250 kcal/d for 28 d, increasing consumption of all macronutrients. Insulin sensitivity and body composition were assessed at baseline (day 0) and day 28. Fasting serum amino acids were measured at days 0, 3, and 28. Linear mixed-effects models evaluated the effect of time in the total group and separately in those with low and high body fat gain (below compared with at or above median fat gain, 1.95 kg). At days 0 and 28, insulin-induced suppression of serum amino acids during a hyperinsulinemic-euglycemic clamp test and, in a subset (n = 20), adipose tissue mRNA expression of selected amino acid metabolizing enzymes were assessed. Results Weight increased by 2.8 kg. High fat gainers gained 2.6 kg fat mass compared with 1.1 kg in low fat gainers. Valine and isoleucine increased at day 3 (+17% and +22%, respectively; P ≤ 0.002) and remained elevated at day 28, despite a decline in valine (P = 0.019) from day 3 values. Methionine, cystathionine, and taurine were unaffected. Serum total cysteine (tCys) transiently increased at day 3 (+11%; P = 0.022) only in high fat gainers (P-interaction = 0.043), in whom the cysteine catabolic enzyme cysteine dioxygenase (CDO1) was induced (+26%; P = 0.025) in adipose tissue (P-interaction = 0.045). Overconsumption did not alter adipose tissue mRNA expression of the BCAA-metabolizing enzymes branched-chain keto acid dehydrogenase E1α polypeptide (BCKDHA) or branched-chain amino transferase 1 (BCAT1). In the total population at day 0, insulin infusion decreased all serum amino acids (-11% to -47%; P < 0.01), except for homocysteine and tCys, which were unchanged, and glutathione, which was increased by 54%. At day 28, insulin increased tCys (+8%), and the insulin-induced suppression of taurine and phenylalanine observed at day 0, but not that of BCAAs, was significantly impaired. Conclusions These findings highlight the role of nutrient oversupply in increasing fasting BCAA concentrations in healthy adults. The link between cysteine availability, CDO1 expression, and fat gain deserves investigation. This trial was registered at www.clinicaltrials.gov as NCT00562393.
Collapse
Affiliation(s)
- Amany K Elshorbagy
- Department of Physiology, Faculty of Medicine, University of Alexandria, Alexandria, Egypt
| | - Dorit Samocha-Bonet
- Faculty of Medicine, University of New South Wales, Sydney, New South Wales, Australia.,Diabetes and Metabolism Division, Garvan Institute of Medical Research, Sydney, Australia
| | - Fredrik Jernerén
- Department of Pharmacology, University of Oxford, Oxford, United Kingdom.,Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | - Cheryl Turner
- Department of Pharmacology, University of Oxford, Oxford, United Kingdom
| | - Helga Refsum
- Department of Pharmacology, University of Oxford, Oxford, United Kingdom.,Institute of Basic Medical Sciences, Department of Nutrition, University of Oslo, Oslo, Norway
| | - Leonie K Heilbronn
- Diabetes and Metabolism Division, Garvan Institute of Medical Research, Sydney, Australia.,Discipline of Medicine, University of Adelaide, Adelaide, South Australia, Australia
| |
Collapse
|
38
|
Dong Z, Sinha R, Richie JP. Disease prevention and delayed aging by dietary sulfur amino acid restriction: translational implications. Ann N Y Acad Sci 2018; 1418:44-55. [PMID: 29399808 DOI: 10.1111/nyas.13584] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Revised: 11/21/2017] [Accepted: 11/27/2017] [Indexed: 01/01/2023]
Abstract
Sulfur amino acids (SAAs) play numerous critical roles in metabolism and overall health maintenance. Preclinical studies have demonstrated that SAA-restricted diets have many beneficial effects, including extending life span and preventing the development of a variety of diseases. Dietary sulfur amino acid restriction (SAAR) is characterized by chronic restrictions of methionine and cysteine but not calories and is associated with reductions in body weight, adiposity and oxidative stress, and metabolic changes in adipose tissue and liver resulting in enhanced insulin sensitivity and energy expenditure. SAAR-induced changes in blood biomarkers include reductions in insulin, insulin-like growth factor-1, glucose, and leptin and increases in adiponectin and fibroblast growth factor 21. On the basis of these preclinical data, SAAR may also have similar benefits in humans. While little is known of the translational significance of SAAR, its potential feasibility in humans is supported by findings of its effectiveness in rodents, even when initiated in adult animals. To date, there have been no controlled feeding studies of SAAR in humans; however, there have been numerous relevant epidemiologic and disease-based clinical investigations reported. Here, we summarize observations from these clinical investigations to provide insight into the potential effectiveness of SAAR for humans.
Collapse
Affiliation(s)
- Zhen Dong
- Department of Public Health Sciences, Pennsylvania State University College of Medicine, Hershey, Pennsylvania
| | - Raghu Sinha
- Department of Biochemistry and Molecular Biology, Pennsylvania State University College of Medicine, Hershey, Pennsylvania
| | - John P Richie
- Department of Public Health Sciences, Pennsylvania State University College of Medicine, Hershey, Pennsylvania
| |
Collapse
|
39
|
Cai J, Shi X, Wang H, Fan J, Feng Y, Lin X, Yang J, Cui Q, Tang C, Xu G, Geng B. Cystathionine γ lyase–hydrogen sulfide increases peroxisome proliferator-activated receptor γ activity by sulfhydration at C139 site thereby promoting glucose uptake and lipid storage in adipocytes. Biochim Biophys Acta Mol Cell Biol Lipids 2016; 1861:419-29. [DOI: 10.1016/j.bbalip.2016.03.001] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2015] [Revised: 02/25/2016] [Accepted: 03/01/2016] [Indexed: 10/22/2022]
|
40
|
Pravenec M, Kožich V, Krijt J, Sokolová J, Zídek V, Landa V, Mlejnek P, Šilhavý J, Šimáková M, Škop V, Trnovská J, Kazdová L, Kajiya T, Wang J, Kurtz TW. Genetic Variation in Renal Expression ofFolate Receptor 1(Folr1) Gene Predisposes Spontaneously Hypertensive Rats to Metabolic Syndrome. Hypertension 2016; 67:335-41. [DOI: 10.1161/hypertensionaha.115.06158] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Accepted: 11/18/2015] [Indexed: 01/30/2023]
Affiliation(s)
- Michal Pravenec
- From the Department of Model Diseases, Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic (M.P., V.Z., V.L., P.M., J.Š., M.Š.); Institute of Inherited Metabolic Disorders, 1st Faculty of Medicine, Charles University in Prague and General University Hospital in Prague, Prague, Czech Republic (V.K., J.K., J.Š.); Center of Experimental Medicine, Department of Metabolism and Diabetes, Institute for Clinical and Experimental Medicine, Prague, Czech Republic (V.Š., J.T., L.K.); and
| | - Viktor Kožich
- From the Department of Model Diseases, Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic (M.P., V.Z., V.L., P.M., J.Š., M.Š.); Institute of Inherited Metabolic Disorders, 1st Faculty of Medicine, Charles University in Prague and General University Hospital in Prague, Prague, Czech Republic (V.K., J.K., J.Š.); Center of Experimental Medicine, Department of Metabolism and Diabetes, Institute for Clinical and Experimental Medicine, Prague, Czech Republic (V.Š., J.T., L.K.); and
| | - Jakub Krijt
- From the Department of Model Diseases, Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic (M.P., V.Z., V.L., P.M., J.Š., M.Š.); Institute of Inherited Metabolic Disorders, 1st Faculty of Medicine, Charles University in Prague and General University Hospital in Prague, Prague, Czech Republic (V.K., J.K., J.Š.); Center of Experimental Medicine, Department of Metabolism and Diabetes, Institute for Clinical and Experimental Medicine, Prague, Czech Republic (V.Š., J.T., L.K.); and
| | - Jitka Sokolová
- From the Department of Model Diseases, Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic (M.P., V.Z., V.L., P.M., J.Š., M.Š.); Institute of Inherited Metabolic Disorders, 1st Faculty of Medicine, Charles University in Prague and General University Hospital in Prague, Prague, Czech Republic (V.K., J.K., J.Š.); Center of Experimental Medicine, Department of Metabolism and Diabetes, Institute for Clinical and Experimental Medicine, Prague, Czech Republic (V.Š., J.T., L.K.); and
| | - Václav Zídek
- From the Department of Model Diseases, Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic (M.P., V.Z., V.L., P.M., J.Š., M.Š.); Institute of Inherited Metabolic Disorders, 1st Faculty of Medicine, Charles University in Prague and General University Hospital in Prague, Prague, Czech Republic (V.K., J.K., J.Š.); Center of Experimental Medicine, Department of Metabolism and Diabetes, Institute for Clinical and Experimental Medicine, Prague, Czech Republic (V.Š., J.T., L.K.); and
| | - Vladimír Landa
- From the Department of Model Diseases, Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic (M.P., V.Z., V.L., P.M., J.Š., M.Š.); Institute of Inherited Metabolic Disorders, 1st Faculty of Medicine, Charles University in Prague and General University Hospital in Prague, Prague, Czech Republic (V.K., J.K., J.Š.); Center of Experimental Medicine, Department of Metabolism and Diabetes, Institute for Clinical and Experimental Medicine, Prague, Czech Republic (V.Š., J.T., L.K.); and
| | - Petr Mlejnek
- From the Department of Model Diseases, Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic (M.P., V.Z., V.L., P.M., J.Š., M.Š.); Institute of Inherited Metabolic Disorders, 1st Faculty of Medicine, Charles University in Prague and General University Hospital in Prague, Prague, Czech Republic (V.K., J.K., J.Š.); Center of Experimental Medicine, Department of Metabolism and Diabetes, Institute for Clinical and Experimental Medicine, Prague, Czech Republic (V.Š., J.T., L.K.); and
| | - Jan Šilhavý
- From the Department of Model Diseases, Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic (M.P., V.Z., V.L., P.M., J.Š., M.Š.); Institute of Inherited Metabolic Disorders, 1st Faculty of Medicine, Charles University in Prague and General University Hospital in Prague, Prague, Czech Republic (V.K., J.K., J.Š.); Center of Experimental Medicine, Department of Metabolism and Diabetes, Institute for Clinical and Experimental Medicine, Prague, Czech Republic (V.Š., J.T., L.K.); and
| | - Miroslava Šimáková
- From the Department of Model Diseases, Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic (M.P., V.Z., V.L., P.M., J.Š., M.Š.); Institute of Inherited Metabolic Disorders, 1st Faculty of Medicine, Charles University in Prague and General University Hospital in Prague, Prague, Czech Republic (V.K., J.K., J.Š.); Center of Experimental Medicine, Department of Metabolism and Diabetes, Institute for Clinical and Experimental Medicine, Prague, Czech Republic (V.Š., J.T., L.K.); and
| | - Vojtěch Škop
- From the Department of Model Diseases, Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic (M.P., V.Z., V.L., P.M., J.Š., M.Š.); Institute of Inherited Metabolic Disorders, 1st Faculty of Medicine, Charles University in Prague and General University Hospital in Prague, Prague, Czech Republic (V.K., J.K., J.Š.); Center of Experimental Medicine, Department of Metabolism and Diabetes, Institute for Clinical and Experimental Medicine, Prague, Czech Republic (V.Š., J.T., L.K.); and
| | - Jaroslava Trnovská
- From the Department of Model Diseases, Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic (M.P., V.Z., V.L., P.M., J.Š., M.Š.); Institute of Inherited Metabolic Disorders, 1st Faculty of Medicine, Charles University in Prague and General University Hospital in Prague, Prague, Czech Republic (V.K., J.K., J.Š.); Center of Experimental Medicine, Department of Metabolism and Diabetes, Institute for Clinical and Experimental Medicine, Prague, Czech Republic (V.Š., J.T., L.K.); and
| | - Ludmila Kazdová
- From the Department of Model Diseases, Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic (M.P., V.Z., V.L., P.M., J.Š., M.Š.); Institute of Inherited Metabolic Disorders, 1st Faculty of Medicine, Charles University in Prague and General University Hospital in Prague, Prague, Czech Republic (V.K., J.K., J.Š.); Center of Experimental Medicine, Department of Metabolism and Diabetes, Institute for Clinical and Experimental Medicine, Prague, Czech Republic (V.Š., J.T., L.K.); and
| | - Takashi Kajiya
- From the Department of Model Diseases, Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic (M.P., V.Z., V.L., P.M., J.Š., M.Š.); Institute of Inherited Metabolic Disorders, 1st Faculty of Medicine, Charles University in Prague and General University Hospital in Prague, Prague, Czech Republic (V.K., J.K., J.Š.); Center of Experimental Medicine, Department of Metabolism and Diabetes, Institute for Clinical and Experimental Medicine, Prague, Czech Republic (V.Š., J.T., L.K.); and
| | - Jiaming Wang
- From the Department of Model Diseases, Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic (M.P., V.Z., V.L., P.M., J.Š., M.Š.); Institute of Inherited Metabolic Disorders, 1st Faculty of Medicine, Charles University in Prague and General University Hospital in Prague, Prague, Czech Republic (V.K., J.K., J.Š.); Center of Experimental Medicine, Department of Metabolism and Diabetes, Institute for Clinical and Experimental Medicine, Prague, Czech Republic (V.Š., J.T., L.K.); and
| | - Theodore W. Kurtz
- From the Department of Model Diseases, Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic (M.P., V.Z., V.L., P.M., J.Š., M.Š.); Institute of Inherited Metabolic Disorders, 1st Faculty of Medicine, Charles University in Prague and General University Hospital in Prague, Prague, Czech Republic (V.K., J.K., J.Š.); Center of Experimental Medicine, Department of Metabolism and Diabetes, Institute for Clinical and Experimental Medicine, Prague, Czech Republic (V.Š., J.T., L.K.); and
| |
Collapse
|
41
|
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
|
42
|
Nakatsu D, Horiuchi Y, Kano F, Noguchi Y, Sugawara T, Takamoto I, Kubota N, Kadowaki T, Murata M. L-cysteine reversibly inhibits glucose-induced biphasic insulin secretion and ATP production by inactivating PKM2. Proc Natl Acad Sci U S A 2015; 112:E1067-76. [PMID: 25713368 PMCID: PMC4364213 DOI: 10.1073/pnas.1417197112] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Increase in the concentration of plasma L-cysteine is closely associated with defective insulin secretion from pancreatic β-cells, which results in type 2 diabetes (T2D). In this study, we investigated the effects of prolonged L-cysteine treatment on glucose-stimulated insulin secretion (GSIS) from mouse insulinoma 6 (MIN6) cells and from mouse pancreatic islets, and found that the treatment reversibly inhibited glucose-induced ATP production and resulting GSIS without affecting proinsulin and insulin synthesis. Comprehensive metabolic analyses using capillary electrophoresis time-of-flight mass spectrometry showed that prolonged L-cysteine treatment decreased the levels of pyruvate and its downstream metabolites. In addition, methyl pyruvate, a membrane-permeable form of pyruvate, rescued L-cysteine-induced inhibition of GSIS. Based on these results, we found that both in vitro and in MIN6 cells, L-cysteine specifically inhibited the activity of pyruvate kinase muscle isoform 2 (PKM2), an isoform of pyruvate kinases that catalyze the conversion of phosphoenolpyruvate to pyruvate. L-cysteine also induced PKM2 subunit dissociation (tetramers to dimers/monomers) in cells, which resulted in impaired glucose-induced ATP production for GSIS. DASA-10 (NCGC00181061, a substituted N,N'-diarylsulfonamide), a specific activator for PKM2, restored the tetramer formation and the activity of PKM2, glucose-induced ATP production, and biphasic insulin secretion in L-cysteine-treated cells. Collectively, our results demonstrate that impaired insulin secretion due to exposure to L-cysteine resulted from its direct binding and inactivation of PKM2 and suggest that PKM2 is a potential therapeutic target for T2D.
Collapse
Affiliation(s)
- Daiki Nakatsu
- Department of Life Sciences, Graduate School of Arts and Sciences, University of Tokyo, 3-8-1 Komaba, Meguro-ku, Tokyo 153-8902, Japan
| | - Yuta Horiuchi
- Department of Life Sciences, Graduate School of Arts and Sciences, University of Tokyo, 3-8-1 Komaba, Meguro-ku, Tokyo 153-8902, Japan
| | - Fumi Kano
- Department of Life Sciences, Graduate School of Arts and Sciences, University of Tokyo, 3-8-1 Komaba, Meguro-ku, Tokyo 153-8902, Japan; Precursory Research for Embryonic Science and Technology (PRESTO), Japan Science and Technology Agency, 4-1-8 Honcho, Kawaguchi, Saitama 332-0012, Japan
| | - Yoshiyuki Noguchi
- Department of Life Sciences, Graduate School of Arts and Sciences, University of Tokyo, 3-8-1 Komaba, Meguro-ku, Tokyo 153-8902, Japan
| | - Taichi Sugawara
- Department of Life Sciences, Graduate School of Arts and Sciences, University of Tokyo, 3-8-1 Komaba, Meguro-ku, Tokyo 153-8902, Japan
| | - Iseki Takamoto
- Department of Diabetes and Metabolic Diseases, Graduate School of Medicine, University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Naoto Kubota
- Department of Diabetes and Metabolic Diseases, Graduate School of Medicine, University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan; Department of Clinical Nutrition Therapy, University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan; and Laboratory for Metabolic Homeostasis, RIKEN Center for Integrative Medical Sciences, 1-7-22 Suehiro-cho, Turumi-ku, Yokohama, Kanagawa 230-0045, Japan
| | - Takashi Kadowaki
- Department of Diabetes and Metabolic Diseases, Graduate School of Medicine, University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Masayuki Murata
- Department of Life Sciences, Graduate School of Arts and Sciences, University of Tokyo, 3-8-1 Komaba, Meguro-ku, Tokyo 153-8902, Japan;
| |
Collapse
|
43
|
Elevated serum levels of cysteine and tyrosine: early biomarkers in asymptomatic adults at increased risk of developing metabolic syndrome. BIOMED RESEARCH INTERNATIONAL 2015; 2015:418681. [PMID: 25821801 PMCID: PMC4364114 DOI: 10.1155/2015/418681] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Accepted: 01/15/2015] [Indexed: 12/13/2022]
Abstract
As there is effective intervention for delaying or preventing metabolic diseases, which are often present for years before becoming clinically apparent, novel biomarkers that would mark metabolic complications before the onset of metabolic disease should be identified. We investigated the role of fasting serum amino acids and their associations with inflammatory markers, adipokines, and metabolic syndrome (MetS) components in subjects prior to the onset of insulin resistance (IR). Anthropometric measurements, food records, adipokines, biochemical markers, and serum levels of amino acids were determined in 96 asymptomatic subjects aged 25–49 years divided into three groups according to the number of MetS components present. Cysteine and tyrosine were significantly higher already in group with one component of MetS present compared to subjects without MetS components. Serum amino acid levels correlated with markers of inflammation and adipokines. Alanine and glycine explained 10% of insulin resistance variability. The role of tyrosine and cysteine, that were higher already with 1 component of MetS present, should be further investigated as they might point to future insulin disturbances.
Collapse
|
44
|
McGavigan AK, O'Hara HC, Amin A, Kinsey-Jones J, Spreckley E, Alamshah A, Agahi A, Banks K, France R, Hyberg G, Wong C, Bewick GA, Gardiner JV, Lehmann A, Martin NM, Ghatei MA, Bloom SR, Murphy KG. L-cysteine suppresses ghrelin and reduces appetite in rodents and humans. Int J Obes (Lond) 2014; 39:447-55. [PMID: 25219528 PMCID: PMC4276721 DOI: 10.1038/ijo.2014.172] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2014] [Revised: 08/28/2014] [Accepted: 09/08/2014] [Indexed: 12/13/2022]
Abstract
BACKGROUND High-protein diets promote weight loss and subsequent weight maintenance, but are difficult to adhere to. The mechanisms by which protein exerts these effects remain unclear. However, the amino acids produced by protein digestion may have a role in driving protein-induced satiety. METHODS We tested the effects of a range of amino acids on food intake in rodents and identified l-cysteine as the most anorexigenic. Using rodents we further studied the effect of l-cysteine on food intake, behaviour and energy expenditure. We proceeded to investigate its effect on neuronal activation in the hypothalamus and brainstem before investigating its effect on gastric emptying and gut hormone release. The effect of l-cysteine on appetite scores and gut hormone release was then investigated in humans. RESULTS l-Cysteine dose-dependently decreased food intake in both rats and mice following oral gavage and intraperitoneal administration. This effect did not appear to be secondary to behavioural or aversive side effects. l-Cysteine increased neuronal activation in the area postrema and delayed gastric emptying. It suppressed plasma acyl ghrelin levels and did not reduce food intake in transgenic ghrelin-overexpressing mice. Repeated l-cysteine administration decreased food intake in rats and obese mice. l-Cysteine reduced hunger and plasma acyl ghrelin levels in humans. CONCLUSIONS Further work is required to determine the chronic effect of l-cysteine in rodents and humans on appetite and body weight, and whether l-cysteine contributes towards protein-induced satiety.
Collapse
Affiliation(s)
- A K McGavigan
- Department of Investigative Medicine, Imperial College London, London, UK
| | - H C O'Hara
- Department of Investigative Medicine, Imperial College London, London, UK
| | - A Amin
- Department of Investigative Medicine, Imperial College London, London, UK
| | - J Kinsey-Jones
- Department of Investigative Medicine, Imperial College London, London, UK
| | - E Spreckley
- Department of Investigative Medicine, Imperial College London, London, UK
| | - A Alamshah
- Department of Investigative Medicine, Imperial College London, London, UK
| | - A Agahi
- Department of Investigative Medicine, Imperial College London, London, UK
| | - K Banks
- Department of Investigative Medicine, Imperial College London, London, UK
| | - R France
- Department of Investigative Medicine, Imperial College London, London, UK
| | - G Hyberg
- AstraZeneca R&D, Mölndal, Sweden
| | - C Wong
- Department of Investigative Medicine, Imperial College London, London, UK
| | - G A Bewick
- 1] Department of Investigative Medicine, Imperial College London, London, UK [2] Division of Diabetes & Nutritional Sciences, King's College London, London, UK
| | - J V Gardiner
- Department of Investigative Medicine, Imperial College London, London, UK
| | - A Lehmann
- 1] AstraZeneca R&D, Mölndal, Sweden [2] NextRx, Gothenburg, Sweden
| | - N M Martin
- Department of Investigative Medicine, Imperial College London, London, UK
| | - M A Ghatei
- Department of Investigative Medicine, Imperial College London, London, UK
| | - S R Bloom
- Department of Investigative Medicine, Imperial College London, London, UK
| | - K G Murphy
- Department of Investigative Medicine, Imperial College London, London, UK
| |
Collapse
|
45
|
DietaryL-Cysteine Improves the Antioxidative Potential and Lipid Metabolism in Rats Fed a Normal Diet. Biosci Biotechnol Biochem 2014; 77:1430-4. [DOI: 10.1271/bbb.130083] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
|
46
|
Vidal K, Breuillé D, Serrant P, Denis P, Glomot F, Béchereau F, Papet I. Long-term cysteine fortification impacts cysteine/glutathione homeostasis and food intake in ageing rats. Eur J Nutr 2013; 53:963-71. [DOI: 10.1007/s00394-013-0600-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2013] [Accepted: 10/08/2013] [Indexed: 12/20/2022]
|
47
|
Elshorbagy AK, Valdivia-Garcia M, Mattocks DAL, Plummer JD, Orentreich DS, Orentreich N, Refsum H, Perrone CE. Effect of taurine and N-acetylcysteine on methionine restriction-mediated adiposity resistance. Metabolism 2013; 62:509-17. [PMID: 23154184 DOI: 10.1016/j.metabol.2012.10.005] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2012] [Revised: 09/07/2012] [Accepted: 10/09/2012] [Indexed: 12/16/2022]
Abstract
OBJECTIVES Methionine-restricted (MR) rats, which are lean and insulin sensitive, have low serum total cysteine (tCys) and taurine and decreased hepatic expression and activity indices of stearoyl-coenzyme A desaturase-1 (SCD1). These effects are partly or completely reversed by cysteine supplementation. We investigated whether reversal of MR phenotypes can be achieved by other sulfur compounds, namely taurine or N-acetylcysteine (NAC). METHODS MR and control-fed (CF) rats were supplemented with taurine (0.5%) or NAC (0.5%) for 12weeks. Adiposity, serum sulfur amino acids (SAA), Scd1 gene expression in liver and white adipose tissue, and SCD1 activity indices (calculated from serum fatty acid profile) were monitored. RESULTS Taurine supplementation of MR rats did not restore weight gain or hepatic Scd1 expression or indices to CF levels, but further decreased adiposity. Taurine supplementation of CF rats did not affect adiposity, but lowered triglyceridemia. NAC supplementation in MR rats raised tCys and partly or completely reversed MR effects on weight, fat %, Scd1 expression in liver and white adipose tissue, and estimated SCD1 activity. In CF rats, NAC decreased body fat % and lowered SCD1-18 activity index (P<0.001). Serum triglycerides and leptin were over 40% lower in CF+NAC relative to CF rats (P≤0.003 for both). In all groups, change in tCys correlated with change in SCD1-16 index (partial r=0.60, P<0.001) independent of other SAA. CONCLUSION The results rule out taurine as a mediator of increased adiposity produced by cysteine in MR, and show that NAC, similar to L-cysteine, blocks anti-obesity effects of MR. Our data show that dietary SAA can influence adiposity in part through mechanisms that converge on SCD1 function. This may have implications for understanding and preventing human obesity.
Collapse
|
48
|
|