151
|
Caloric restriction improves glycaemic control without reducing plasma branched-chain amino acids or keto-acids in obese men. Sci Rep 2022; 12:19273. [PMID: 36369511 PMCID: PMC9652417 DOI: 10.1038/s41598-022-21814-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 10/04/2022] [Indexed: 11/13/2022] Open
Abstract
Higher plasma leucine, isoleucine and valine (BCAA) concentrations are associated with diabetes, obesity and insulin resistance (IR). Here, we evaluated the effects of 6-weeks very-low calorie diet (VLCD) upon fasting BCAA in overweight (OW) non-diabetic men, to explore associations between circulating BCAA and IR, before and after a weight loss intervention. Fasting plasma BCAAs were quantified in an OW (n = 26; BMI 32.4 ± 3 kg/m2; mean age 44 ± 9 y) and a normal-weight (NW) group (n = 26; BMI 24 ± 3.1 kg/m2; mean age 32 ± 12.3 y). Ten of the OW group (BMI 32.2 ± 4 kg/m2; 46 ± 8 y) then underwent 6-weeks of VLCD (600-800 kcal/day). Fasting plasma BCAA (gas chromatography-mass spectrometry), insulin sensitivity (HOMA-IR) and body-composition (DXA) were assessed before and after VLCD. Total BCAA were higher in OW individuals (sum leucine/isoleucine/valine: 457 ± 85 µM) compared to NW control individuals (365 ± 78 µM, p < 0.001). Despite significant weight loss (baseline 103.9 ± 12.3 to 93 ± 9.6 kg and BMI 32.2 ± 4 to 28.9 ± 3.6 kg/m2), no changes were observed in BCAAs after 6-weeks of VLCD. Moreover, although VLCD resulted in a significant reduction in HOMA-IR (baseline 1.19 ± 0.62 to 0.51 ± 0.21 post-VLCD; p < 0.001), Pearson's r revealed no relationships between BCAA and HOMA-IR, either before (leucine R2: 2.49e-005, p = 0.98; isoleucine R2: 1.211-e006, p = 0.9; valine R2: 0.004, p = 0.85) or after VLCD (leucine R2: 0.003, p = 0.86; isoleucine R2: 0.006, p = 0.82; valine R2: 0.002, p = 0.65). Plasma BCAA are higher in OW compared to NW individuals. However, while 6-weeks VLCD reduced body weight and IR in OW individuals, this was not associated with reductions in BCAA. This suggests that studies demonstrating links between BCAA and insulin resistance in OW individuals, are complex and are not normalised by simply losing weight.
Collapse
|
152
|
Lennox B, Xiong W, Waters P, Coles A, Jones PB, Yeo T, May JTM, Yeeles K, Anthony D, Probert F. The serum metabolomic profile of a distinct, inflammatory subtype of acute psychosis. Mol Psychiatry 2022; 27:4722-4730. [PMID: 36131046 PMCID: PMC7613906 DOI: 10.1038/s41380-022-01784-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 09/01/2022] [Accepted: 09/05/2022] [Indexed: 12/14/2022]
Abstract
A range of studies suggest that a proportion of psychosis may have an autoimmune basis, but this has not translated through into clinical practice-there is no biochemical test able to accurately identify psychosis resulting from an underlying inflammatory cause. Such a test would be an important step towards identifying who might require different treatments and have the potential to improve outcomes for patients. To identify novel subgroups within patients with acute psychosis we measured the serum nuclear magnetic resonance (NMR) metabolite profiles of 75 patients who had identified antibodies (anti-glycine receptor [GlyR], voltage-gated potassium channel [VGKC], Contactin-associated protein-like 2 [CASPR2], leucine-rich glioma inactivated 1 [LGI1], N-methyl-D-aspartate receptor [NMDAR] antibody) and 70 antibody negative patients matched for age, gender, and ethnicity. Clinical symptoms were assessed using the positive and negative syndrome scale (PANSS). Unsupervised principal component analysis identified two distinct biochemical signatures within the cohort. Orthogonal partial least squared discriminatory analysis revealed that the serum metabolomes of NMDAR, LGI1, and CASPR2 antibody psychosis patients were indistinct from the antibody negative control group while VGKC and GlyR antibody patients had significantly decreased lipoprotein fatty acids and increased amino acid concentrations. Furthermore, these patients had more severe presentation with higher PANSS scores than either the antibody negative controls or the NMDAR, LGI1, and CASPR2 antibody groups. These results suggest that a proportion of patients with acute psychosis have a distinct clinical and biochemical phenotype that may indicate an inflammatory subtype.
Collapse
Affiliation(s)
- Belinda Lennox
- Department of Psychiatry, University of Oxford and Oxford Health NHS Foundation Trust, Oxford, UK.
| | - Wenzheng Xiong
- Department of Pharmacology, University of Oxford, Oxford, UK
- Department of Chemistry, University of Oxford, Oxford, UK
| | - Patrick Waters
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Alasdair Coles
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Peter B Jones
- Department of Psychiatry, University of Cambridge, Cambridge, UK
| | - Tianrong Yeo
- Department of Pharmacology, University of Oxford, Oxford, UK
- Department of Neurology, National Neuroscience Institute, Singapore, Singapore
- Duke-NUS Medical School, Singapore, Singapore
| | - Jeanne Tan May May
- Department of Neurology, National Neuroscience Institute, Singapore, Singapore
- Duke-NUS Medical School, Singapore, Singapore
| | - Ksenija Yeeles
- Department of Psychiatry, University of Oxford and Oxford Health NHS Foundation Trust, Oxford, UK
| | - Daniel Anthony
- Department of Pharmacology, University of Oxford, Oxford, UK
| | - Fay Probert
- Department of Chemistry, University of Oxford, Oxford, UK
| |
Collapse
|
153
|
Identifying a metabolomics profile associated with masked hypertension in two independent cohorts: Data from the African-PREDICT and SABPA studies. Hypertens Res 2022; 45:1781-1793. [PMID: 36056205 DOI: 10.1038/s41440-022-01010-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 06/27/2022] [Accepted: 07/21/2022] [Indexed: 12/15/2022]
Abstract
Individuals with masked hypertension (MHT) have a greater risk of adverse cardiovascular outcomes than normotensive (NT) individuals. Exploring metabolomic differences between NT and MHT individuals may help provide a better understanding of the etiology of MHT. We analyzed data from 910 young participants (83% NT and 17% MHT) (mean age 24 ± 3 years) from the African-PREDICT and 210 older participants (63% NT and 37% MHT) from the SABPA (mean age 42 ± 9.6 years) studies. Clinic and ambulatory blood pressures (BPs) were used to define BP phenotypes. Urinary amino acids and acylcarnitines were measured using liquid chromatography time-of-flight mass spectrometry in SABPA and liquid chromatography tandem mass spectrometry in the African-PREDICT studies. In the SABPA study, amino acids (leucine/isoleucine, valine, methionine, phenylalanine), free carnitine (C0-carnitine), and acylcarnitines C3 (propionyl)-, C4 (butyryl)-carnitine and total acylcarnitine) were higher in MHT than NT adults. In the African-PREDICT study, C0- and C5-carnitines were higher in MHT individuals. With unadjusted analyses in NT adults from the SABPA study, ambulatory SBP correlated positively with only C3-carnitine. In MHT individuals, positive correlations of ambulatory SBP with leucine/isoleucine, valine, methionine, phenylalanine, C0-carnitine and C3-carnitine were evident (all p < 0.05). In the African-PREDICT study, ambulatory SBP correlated positively with C0-carnitine (r = 0.101; p = 0.006) and C5-carnitine (r = 0.195; p < 0.001) in NT adults and C5-carnitine in MHT individuals (r = 0.169; p = 0.034). We demonstrated differences between the metabolomic profiles of NT and MHT adults, which may reflect different stages in the alteration of branched-chain amino acid metabolism early on and later in life.
Collapse
|
154
|
Lu Z, Sun GF, Pan XA, Qu XH, Yang P, Chen ZP, Han XJ, Wang T. BCATc inhibitor 2 ameliorated mitochondrial dysfunction and apoptosis in oleic acid-induced non-alcoholic fatty liver disease model. Front Pharmacol 2022; 13:1025551. [PMID: 36386234 PMCID: PMC9650408 DOI: 10.3389/fphar.2022.1025551] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 10/18/2022] [Indexed: 09/14/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a prevalent hepatic disease in the world. Disorders of branched chain amino acid (BCAA) metabolism is involved in various diseases. In this study, we aim to explore the role of BCAA metabolism in the development of NAFLD and the protective effect of BCATc Inhibitor 2, an inhibitor of cytosolic branched chain amino acid transaminase, against NAFLD as well as its underlying mechanism. It was found that oleic acid induced lipid accumulation and apoptosis in HepG2 and LO2 cells. Supplementation of BCAAs further aggravated oleic acid-induced lipid accumulation and apoptosis. In contrast, treatment of BCATc Inhibitor 2 ameliorated oleic acid-induced lipid accumulation and apoptosis. Molecularly, supplementation of BCAAs or treatment of BCATc Inhibitor 2 up-regulated or down-regulated the expression of SREBP1 and lipogenesis-related genes without affecting lipolysis-related genes. BCATc Inhibitor 2 maintained mitochondrial function by ameliorating oleic acid-induced mitochondrial ROS generation and mitochondrial membrane potential disruption. In addition, BCATc Inhibitor 2 treatment alleviated oleic acid-induced activation of JNK and AKT signaling pathway and Bcl2/Bax/Caspase axis. In conclusion, our results indicate BCAA metabolism is involved in NAFLD and BCATc Inhibitor 2 protects against oleic acid-induced lipid accumulation and apoptosis. These findings suggest that BCATc Inhibitor 2 is a promising candidate drug for the treatment of NAFLD.
Collapse
Affiliation(s)
- Zhuo Lu
- Institute of Geriatrics, Jiangxi Provincial People’s Hospital and The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Gui-Feng Sun
- Department of Pharmacology, School of Pharmaceutical Science, Nanchang University, Nanchang, China
| | - Xiao-An Pan
- Department of Pharmacology, School of Pharmaceutical Science, Nanchang University, Nanchang, China
| | - Xin-Hui Qu
- Institute of Geriatrics, Jiangxi Provincial People’s Hospital and The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
- Department of Neurology, Jiangxi Provincial People’s Hospital and The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
| | - Ping Yang
- Department of Neurology, Jiangxi Provincial People’s Hospital and The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
| | - Zhi-Ping Chen
- Department of Critical Care Medicine, Jiangxi Provincial People’s Hospital and The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
| | - Xiao-Jian Han
- Institute of Geriatrics, Jiangxi Provincial People’s Hospital and The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
- Department of Pharmacology, School of Pharmaceutical Science, Nanchang University, Nanchang, China
- Department of Neurology, Jiangxi Provincial People’s Hospital and The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
| | - Tao Wang
- Institute of Geriatrics, Jiangxi Provincial People’s Hospital and The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
| |
Collapse
|
155
|
Bartova S, Madrid-Gambin F, Fernández L, Carayol J, Meugnier E, Segrestin B, Delage P, Vionnet N, Boizot A, Laville M, Vidal H, Marco S, Hager J, Moco S. Grape polyphenols decrease circulating branched chain amino acids in overfed adults. Front Nutr 2022; 9:998044. [PMID: 36386937 PMCID: PMC9643885 DOI: 10.3389/fnut.2022.998044] [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: 07/19/2022] [Accepted: 10/10/2022] [Indexed: 12/02/2022] Open
Abstract
Introduction and aims Dietary polyphenols have long been associated with health benefits, including the prevention of obesity and related chronic diseases. Overfeeding was shown to rapidly induce weight gain and fat mass, associated with mild insulin resistance in humans, and thus represents a suitable model of the metabolic complications resulting from obesity. We studied the effects of a polyphenol-rich grape extract supplementation on the plasma metabolome during an overfeeding intervention in adults, in two randomized parallel controlled clinical trials. Methods Blood plasma samples from 40 normal weight to overweight male adults, submitted to a 31-day overfeeding (additional 50% of energy requirement by a high calorie-high fructose diet), given either 2 g/day grape polyphenol extract or a placebo at 0, 15, 21, and 31 days were analyzed (Lyon study). Samples from a similarly designed trial on females (20 subjects) were collected in parallel (Lausanne study). Nuclear magnetic resonance (NMR)-based metabolomics was conducted to characterize metabolome changes induced by overfeeding and associated effects from polyphenol supplementation. The clinical trials are registered under the numbers NCT02145780 and NCT02225457 at ClinicalTrials.gov. Results Changes in plasma levels of many metabolic markers, including branched chain amino acids (BCAA), ketone bodies and glucose in both placebo as well as upon polyphenol intervention were identified in the Lyon study. Polyphenol supplementation counterbalanced levels of BCAA found to be induced by overfeeding. These results were further corroborated in the Lausanne female study. Conclusion Administration of grape polyphenol-rich extract over 1 month period was associated with a protective metabolic effect against overfeeding in adults.
Collapse
Affiliation(s)
- Simona Bartova
- Nestlé Research, EPFL Innovation Park, Lausanne, Switzerland
| | - Francisco Madrid-Gambin
- Signal and Information Processing for Sensing Systems, Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Luis Fernández
- Signal and Information Processing for Sensing Systems, Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology, Barcelona, Spain
- Department of Electronics and Biomedical Engineering, Universitat de Barcelona, Barcelona, Spain
| | - Jerome Carayol
- Nestlé Research, EPFL Innovation Park, Lausanne, Switzerland
| | - Emmanuelle Meugnier
- University of Lyon, CarMeN Laboratory and Centre de Recherche en Nutrition Humaine Rhône-Alpes (CRNH-RA), INSERM, INRAE, Claude Bernard University Lyon 1, Pierre-Bénite, France
| | - Bérénice Segrestin
- University of Lyon, CarMeN Laboratory and Centre de Recherche en Nutrition Humaine Rhône-Alpes (CRNH-RA), INSERM, INRAE, Claude Bernard University Lyon 1, Pierre-Bénite, France
| | - Pauline Delage
- University of Lyon, CarMeN Laboratory and Centre de Recherche en Nutrition Humaine Rhône-Alpes (CRNH-RA), INSERM, INRAE, Claude Bernard University Lyon 1, Pierre-Bénite, France
| | - Nathalie Vionnet
- Service d’Endocrinologie, Diabétologie et Métabolisme, CHU de Lausanne (CHUV), Lausanne, Switzerland
| | - Alexia Boizot
- Service d’Endocrinologie, Diabétologie et Métabolisme, CHU de Lausanne (CHUV), Lausanne, Switzerland
| | - Martine Laville
- University of Lyon, CarMeN Laboratory and Centre de Recherche en Nutrition Humaine Rhône-Alpes (CRNH-RA), INSERM, INRAE, Claude Bernard University Lyon 1, Pierre-Bénite, France
| | - Hubert Vidal
- University of Lyon, CarMeN Laboratory and Centre de Recherche en Nutrition Humaine Rhône-Alpes (CRNH-RA), INSERM, INRAE, Claude Bernard University Lyon 1, Pierre-Bénite, France
| | - Santiago Marco
- Signal and Information Processing for Sensing Systems, Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology, Barcelona, Spain
- Department of Electronics and Biomedical Engineering, Universitat de Barcelona, Barcelona, Spain
| | - Jörg Hager
- Nestlé Research, EPFL Innovation Park, Lausanne, Switzerland
| | - Sofia Moco
- Nestlé Research, EPFL Innovation Park, Lausanne, Switzerland
| |
Collapse
|
156
|
Sigurdsson MI, Kobayashi H, Amrein K, Nakahira K, Rogers AJ, Pinilla-Vera M, Baron RM, Fredenburgh LE, Lasky-Su JA, Christopher KB. Circulating N-formylmethionine and metabolic shift in critical illness: a multicohort metabolomics study. Crit Care 2022; 26:321. [PMID: 36261854 PMCID: PMC9580206 DOI: 10.1186/s13054-022-04174-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Accepted: 09/20/2022] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Cell stress promotes degradation of mitochondria which release danger-associated molecular patterns that are catabolized to N-formylmethionine. We hypothesized that in critically ill adults, the response to N-formylmethionine is associated with increases in metabolomic shift-related metabolites and increases in 28-day mortality. METHODS We performed metabolomics analyses on plasma from the 428-subject Correction of Vitamin D Deficiency in Critically Ill Patients trial (VITdAL-ICU) cohort and the 90-subject Brigham and Women's Hospital Registry of Critical Illness (RoCI) cohort. In the VITdAL-ICU cohort, we analyzed 983 metabolites at Intensive Care Unit (ICU) admission, day 3, and 7. In the RoCI cohort, we analyzed 411 metabolites at ICU admission. The association between N-formylmethionine and mortality was determined by adjusted logistic regression. The relationship between individual metabolites and N-formylmethionine abundance was assessed with false discovery rate correction via linear regression, linear mixed-effects, and Gaussian graphical models. RESULTS Patients with the top quartile of N-formylmethionine abundance at ICU admission had a significantly higher adjusted odds of 28-day mortality in the VITdAL-ICU (OR, 2.4; 95%CI 1.5-4.0; P = 0.001) and RoCI cohorts (OR, 5.1; 95%CI 1.4-18.7; P = 0.015). Adjusted linear regression shows that with increases in N-formylmethionine abundance at ICU admission, 55 metabolites have significant differences common to both the VITdAL-ICU and RoCI cohorts. With increased N-formylmethionine abundance, both cohorts had elevations in individual short-chain acylcarnitine, branched chain amino acid, kynurenine pathway, and pentose phosphate pathway metabolites. CONCLUSIONS The results indicate that circulating N-formylmethionine promotes a metabolic shift with heightened mortality that involves incomplete mitochondrial fatty acid oxidation, increased branched chain amino acid metabolism, and activation of the pentose phosphate pathway.
Collapse
Affiliation(s)
- Martin Ingi Sigurdsson
- Anesthesiology and Critical Care Medicine, Landspitali University Hospital, University of Iceland, Hringbraut 101, 101, Reykjavík, Iceland
- Faculty of Medicine, University of Iceland, Vatnsmyrarvegur 16, 101, Reykjavik, Iceland
| | - Hirotada Kobayashi
- Division of Renal Medicine, Brigham and Women's Hospital, 75 Francis Street, Boston, 02115, USA
| | - Karin Amrein
- Division of Endocrinology and Diabetology, Medical University of Graz, Auenbruggerplatz 15, 8036, Graz, Austria
| | - Kiichi Nakahira
- Nara Medical University, 840 Shijocho, Kashihara, Nara, 634-8521, Japan
- Weill Cornell Medicine, 1300 York Avenue, New York, 10065, USA
| | - Angela J Rogers
- Stanford University Medical Center, 300 Pasteur Dr. H3143, Stanford, 94305, USA
| | - Mayra Pinilla-Vera
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, 75 Francis Street, Boston, 02115, USA
| | - Rebecca M Baron
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, 75 Francis Street, Boston, 02115, USA
| | - Laura E Fredenburgh
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, 75 Francis Street, Boston, 02115, USA
| | - Jessica A Lasky-Su
- Channing Division of Network Medicine, Brigham and Women's Hospital, 181 Longwood Avenue, Boston, 02115, USA
| | - Kenneth B Christopher
- Division of Renal Medicine, Brigham and Women's Hospital, 75 Francis Street, Boston, 02115, USA.
- Channing Division of Network Medicine, Brigham and Women's Hospital, 181 Longwood Avenue, Boston, 02115, USA.
| |
Collapse
|
157
|
Yin J, Ju Y, Qian H, Wang J, Miao X, Zhu Y, Zhou L, Ye L. Nanoplastics and Microplastics May Be Damaging Our Livers. TOXICS 2022; 10:toxics10100586. [PMID: 36287866 PMCID: PMC9610555 DOI: 10.3390/toxics10100586] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 09/29/2022] [Accepted: 09/29/2022] [Indexed: 06/01/2023]
Abstract
Plastics in the environment can be degraded and even broken into pieces under the action of natural factors, and the degraded products with a particle size of less than 5 mm are called microplastics (MPs). MPs exist in a variety of environmental media that come into contact with the human body. It can enter the body through environmental media and food chains. At present, there are many studies investigating the damage of MPs to marine organisms and mammals. The liver is the largest metabolizing organ and plays an important role in the metabolism of MPs in the body. However, there is no available systematic review on the toxic effects of MPs on the liver. This paper summarizes the adverse effects and mechanisms of MPs on the liver, by searching the literature and highlighting the studies that have been published to date, and provides a scenario for the liver toxicity caused by MPs.
Collapse
Affiliation(s)
- Jianli Yin
- Department of Occupational and Environmental Health, School of Public Health, Jilin University, Changchun 130021, China
| | - Ye Ju
- School of Public Health, Jilin University, Changchun 130021, China
| | - Honghao Qian
- Department of Occupational and Environmental Health, School of Public Health, Jilin University, Changchun 130021, China
| | - Jia Wang
- Department of Occupational and Environmental Health, School of Public Health, Jilin University, Changchun 130021, China
| | - Xiaohan Miao
- Department of Occupational and Environmental Health, School of Public Health, Jilin University, Changchun 130021, China
| | - Ying Zhu
- Department of Occupational and Environmental Health, School of Public Health, Jilin University, Changchun 130021, China
| | - Liting Zhou
- Department of Occupational and Environmental Health, School of Public Health, Jilin University, Changchun 130021, China
| | - Lin Ye
- Department of Occupational and Environmental Health, School of Public Health, Jilin University, Changchun 130021, China
| |
Collapse
|
158
|
Hinkle JS, Rivera CN, Vaughan RA. Branched-Chain Amino Acids and Mitochondrial Biogenesis: An Overview and Mechanistic Summary. Mol Nutr Food Res 2022; 66:e2200109. [PMID: 36047448 PMCID: PMC9786258 DOI: 10.1002/mnfr.202200109] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 06/17/2022] [Indexed: 12/30/2022]
Abstract
Branched-chain amino acids (BCAA) are essential in the diet and promote several vital cell responses which may have benefits for health and athletic performance, as well as disease prevention. While BCAA are well-known for their ability to stimulate muscle protein synthesis, their effects on cell energetics are also becoming well-documented, but these receive less attention. In this review, much of the current evidence demonstrating BCAA ability (as individual amino acids or as part of dietary mixtures) to alter regulators of cellular energetics with an emphasis on mitochondrial biogenesis and related signaling is highlighted. Several studies have shown, both in vitro and in vivo, that BCAA (either individual or as a mixture) may promote signaling associated with increased mitochondrial biogenesis including the upregulation of master regulator of mitochondrial biogenesis peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α), as well as numerous downstream targets and related function. However, sparse data in humans and the difficulty of controlling variables associated with feeding studies leave the physiological relevance of these findings unclear. Future well-controlled diet studies will be needed to assess if BCAA consumption is associated with increased mitochondrial biogenesis and improved metabolic outcomes in healthy and/or diseased human populations.
Collapse
Affiliation(s)
- Jason S. Hinkle
- Department of Exercise ScienceHigh Point UniversityHigh PointNC27262‐3598USA
| | - Caroline N. Rivera
- Department of Exercise ScienceHigh Point UniversityHigh PointNC27262‐3598USA
| | - Roger A. Vaughan
- Department of Exercise ScienceHigh Point UniversityHigh PointNC27262‐3598USA
| |
Collapse
|
159
|
Kapralou AN, Chrousos GP. Metabolic effects of truncal vagotomy when combined with bariatric-metabolic surgery. Metabolism 2022; 135:155263. [PMID: 35835160 DOI: 10.1016/j.metabol.2022.155263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 06/30/2022] [Accepted: 07/04/2022] [Indexed: 11/25/2022]
Abstract
Bariatric-metabolic surgery (BMS) in patients with obesity frequently leads to remission of concurrent type 2 diabetes mellitus (T2DM), even before body weight loss takes place. This is probably based on the correction of a dysmetabolic cycle in the gastrointestinal physiology of T2DM that includes increased vagus-dependent exocrine pancreatic secretion (EPS) and, hence, amplified digestion and nutrient absorption. The resultant chronic exposure of tissues to high plasma levels of glucose, fatty acids and amino acids causes tissue resistance to the actions of insulin and, at a later stage, β-cell dysfunction and reduction of insulin release. We hypothesize that the addition of a surgical truncal vagotomy (TV) may improve and solidify the beneficial results of BMS on T2DM by stably decreasing EPS, - hence reducing the digestion and absorption of nutrients -, and increasing incretin secretion as a result of increased delivery of unabsorbed nutrients to the distal intestine. This hypothesis is supported by surgical data from gastrointestinal malignancies and peptic ulcer operations that include TV, as well as by vagal blockade studies. We suggest that TV may result in a stable reduction of EPS, and that its combination with the appropriate type of BΜS, may enhance and sustain the salutary effects of the latter on T2DM.
Collapse
Affiliation(s)
| | - George P Chrousos
- University Research Institute of Maternal and Child Health and Precision Medicine, UNESCO Chair on Adolescent Health Care, National and Kapodistrian University of Athens Medical School, Athens, Greece
| |
Collapse
|
160
|
Ali MK, Kadir MM, Gujral UP, Fatima SS, Iqbal R, Sun YV, Narayan KMV, Ahmad S. Obesity-associated metabolites in relation to type 2 diabetes risk: A prospective nested case-control study of the CARRS cohort. Diabetes Obes Metab 2022; 24:2008-2016. [PMID: 35676808 PMCID: PMC9543742 DOI: 10.1111/dom.14788] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 05/23/2022] [Accepted: 06/06/2022] [Indexed: 11/29/2022]
Abstract
AIMS To determine whether obesity-associated metabolites are associated with type 2 diabetes (T2DM) risk among South Asians. MATERIALS AND METHODS Serum-based nuclear magnetic resonance imaging metabolomics data were generated from two South Asian population-based prospective cohorts from Karachi, Pakistan: CARRS1 (N = 4017) and CARRS2 (N = 4802). Participants in both cohorts were followed up for 5 years and incident T2DM was ascertained. A nested case-control study approach was developed to select participants from CARRS1 (Ncases = 197 and Ncontrols = 195) and CARRS2 (Ncases = 194 and Ncontrols = 200), respectively. First, we investigated the association of 224 metabolites with general obesity based on body mass index and with central obesity based on waist-hip ratio, and then the top obesity-associated metabolites were studied in relation to incident T2DM. RESULTS In a combined sample of the CARRS1 and CARRS2 cohorts, out of 224 metabolites, 12 were associated with general obesity and, of these, one was associated with incident T2DM. Fifteen out of 224 metabolites were associated with central obesity and, of these, 10 were associated with incident T2DM. The higher level of total cholesterol in high-density lipoprotein (HDL) was associated with reduced T2DM risk (odds ratio [OR] 0.68, 95% confidence interval [CI] 0.53, 0.86; P = 1.2 × 10-3 ), while higher cholesterol esters in large very-low-density lipoprotein (VLDL) particles were associated with increased T2DM risk (OR 1.90, 95% CI 1.40, 2.58; P = 3.5 × 10-5 ). CONCLUSION Total cholesterol in HDL and cholesterol esters in large VLDL particles may be an important biomarker in the identification of early development of obesity-associated T2DM risk among South Asian adults.
Collapse
Affiliation(s)
- Mohammed K. Ali
- Hubert Department of Global HealthRollins School of Public Health, Emory UniversityAtlantaGeorgiaUSA
- Department of Family and Preventive MedicineSchool of Medicine, Emory UniversityAtlantaGeorgiaUSA
| | - M. Masood Kadir
- Department of Community Health SciencesAga Khan UniversityKarachiPakistan
| | - Unjali P. Gujral
- Hubert Department of Global HealthRollins School of Public Health, Emory UniversityAtlantaGeorgiaUSA
| | - Syeda Sadia Fatima
- Department of Biological and Biomedical SciencesAga Khan UniversityKarachiPakistan
| | - Romaina Iqbal
- Department of Community Health SciencesAga Khan UniversityKarachiPakistan
| | - Yan V. Sun
- Department of EpidemiologyRollins School of Public Health, Emory UniversityAtlantaGeorgiaUSA
| | - K. M. Venkat Narayan
- Hubert Department of Global HealthRollins School of Public Health, Emory UniversityAtlantaGeorgiaUSA
- Department of EpidemiologyRollins School of Public Health, Emory UniversityAtlantaGeorgiaUSA
| | - Shafqat Ahmad
- Department of Medical SciencesMolecular Epidemiology and Science for Life Laboratory, Uppsala UniversityUppsalaSweden
- Preventive Medicine DivisionHarvard Medical School, Brigham and Women's HospitalBostonMassachusettsUSA
| |
Collapse
|
161
|
Matye D, Gunewardena S, Chen J, Wang H, Wang Y, Hasan MN, Gu L, Clayton YD, Du Y, Chen C, Friedman JE, Lu SC, Ding WX, Li T. TFEB regulates sulfur amino acid and coenzyme A metabolism to support hepatic metabolic adaptation and redox homeostasis. Nat Commun 2022; 13:5696. [PMID: 36171419 PMCID: PMC9519740 DOI: 10.1038/s41467-022-33465-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 09/15/2022] [Indexed: 11/15/2022] Open
Abstract
Fatty liver is a highly heterogenous condition driven by various pathogenic factors in addition to the severity of steatosis. Protein insufficiency has been causally linked to fatty liver with incompletely defined mechanisms. Here we report that fatty liver is a sulfur amino acid insufficient state that promotes metabolic inflexibility via limiting coenzyme A availability. We demonstrate that the nutrient-sensing transcriptional factor EB synergistically stimulates lysosome proteolysis and methionine adenosyltransferase to increase cysteine pool that drives the production of coenzyme A and glutathione, which support metabolic adaptation and antioxidant defense during increased lipid influx. Intriguingly, mice consuming an isocaloric protein-deficient Western diet exhibit selective hepatic cysteine, coenzyme A and glutathione deficiency and acylcarnitine accumulation, which are reversed by cystine supplementation without normalizing dietary protein intake. These findings support a pathogenic link of dysregulated sulfur amino acid metabolism to metabolic inflexibility that underlies both overnutrition and protein malnutrition-associated fatty liver development.
Collapse
Affiliation(s)
- David Matye
- Harold Hamm Diabetes Center, Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Sumedha Gunewardena
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Jianglei Chen
- Harold Hamm Diabetes Center, Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Huaiwen Wang
- Laboratory for Molecular Biology and Cytometry Research, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Yifeng Wang
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Mohammad Nazmul Hasan
- Harold Hamm Diabetes Center, Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Lijie Gu
- Harold Hamm Diabetes Center, Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Yung Dai Clayton
- Harold Hamm Diabetes Center, Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Yanhong Du
- Harold Hamm Diabetes Center, Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Cheng Chen
- Harold Hamm Diabetes Center, Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Jacob E Friedman
- Harold Hamm Diabetes Center, Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Shelly C Lu
- Karsh Division of Gastroenterology and Hepatology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Wen-Xing Ding
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Tiangang Li
- Harold Hamm Diabetes Center, Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA.
| |
Collapse
|
162
|
Kivelä J, Meinilä J, Uusitupa M, Tuomilehto J, Lindström J. Longitudinal Branched-Chain Amino Acids, Lifestyle Intervention, and Type 2 Diabetes in the Finnish Diabetes Prevention Study. J Clin Endocrinol Metab 2022; 107:2844-2853. [PMID: 35917829 PMCID: PMC9516128 DOI: 10.1210/clinem/dgac463] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Indexed: 12/05/2022]
Abstract
CONTEXT Circulating branched-chain amino acids (BCAAs) are associated with the risk of type 2 diabetes (T2D). OBJECTIVE We examined to what extent lifestyle intervention aiming to prevent T2D interacts with this association and how BCAA concentrations change during the intervention. METHODS We computed trajectory clusters by k-means clustering of serum fasting BCAAs analyzed annually by mass spectrometry during a 4-year intervention. We investigated whether baseline BCAAs, BCAA trajectories, and BCAA change trajectories predicted T2D and whether BCAAs predicted T2D differently in the intervention (n = 198) and control group (n = 196). RESULTS Elevated baseline BCAAs predicted the incidence of T2D in the control group (hazard ratio [HR] 1.05 per 10 μmol/L, P = 0.01), but not in the intervention group. BCAA concentration decreased during the first year in the whole cohort (mean -14.9 μmol/L, P < 0.001), with no significant difference between the groups. We identified 5 BCAA trajectory clusters and 5 trajectory clusters for the change in BCAAs. Trajectories with high mean BCAA levels were associated with an increased HR for T2D compared with the trajectory with low BCAA levels (trajectory with highest vs lowest BCAA, HR 4.0; P = 0.01). A trajectory with increasing BCAA levels had a higher HR for T2D compared with decreasing trajectory in the intervention group only (HR 25.4, P < 0.001). CONCLUSION Lifestyle intervention modified the association of the baseline BCAA concentration and BCAA trajectories with the incidence of T2D. Our study adds to the accumulating evidence on the mechanisms behind the effect of lifestyle changes on the risk of T2D.
Collapse
Affiliation(s)
- Jemina Kivelä
- Correspondence: Jemina Kivelä, MSc, Finnish Institute for Health and Welfare (THL), Mannerheimintie 166, PO Box 30, FI-00271 Helsinki, Finland.
| | - Jelena Meinilä
- Department of Food and Nutrition, University of Helsinki, 00014 Helsinki, Finland
| | - Matti Uusitupa
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, 70211 Kuopio, Finland
| | - Jaakko Tuomilehto
- Population Health Unit, Finnish Institute of Health and Welfare, 00271 Helsinki, Finland
- Department of Public Health, University of Helsinki, 00014 Helsinki, Finland
- Saudi Diabetes Research Group, King Abdulaziz University, 80200 Jeddah, Saudi Arabia
- Department of International Health, National School of Public Health, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Jaana Lindström
- Population Health Unit, Finnish Institute of Health and Welfare, 00271 Helsinki, Finland
| |
Collapse
|
163
|
Harris CP, Ramlochansingh C, Uhl O, Demmelmair H, Heinrich J, Koletzko B, Standl M, Thiering E. Association of Maternal Diet during Pregnancy and Metabolite Profile in Cord Blood. Biomolecules 2022; 12:biom12101333. [PMID: 36291541 PMCID: PMC9599655 DOI: 10.3390/biom12101333] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 09/15/2022] [Accepted: 09/16/2022] [Indexed: 11/30/2022] Open
Abstract
Cord blood metabolites can be predictive of long-term disease risk, but how levels of different metabolites might vary with respect to maternal diet is not well understood. The aim of this study was to evaluate the associations of different dietary patterns during pregnancy with cord blood metabolites (including glycerophospholipid fatty acids, polar lipids, non-esterified fatty acids, amino acids, and the sum of hexoses). Participants from the German LISA birth cohort study, with available data on targeted cord blood metabolomics and maternal diet, were included (n = 739). Maternal diet during the last 4 weeks of pregnancy was assessed by a non-quantitative food-frequency questionnaire. Using factor analysis, ten dietary patterns were identified, which were used in linear regression models exploring associations with cord blood metabolites. After correction for multiple hypothesis testing and adjustment for basic covariates, “fish and shellfish” was associated with higher glycerophospholipid fatty acid C20:5 n3 and lower C22:5 n6, whereas the “meat and potato” pattern was directly associated with propionylcarnitine (C3:0). The observed associations highlight potential metabolic pathways involved in the early programming of health and disease through maternal diet, as well as the potential for establishing quantitative biomarkers for dietary patterns of pregnant women.
Collapse
Affiliation(s)
- Carla P. Harris
- Institute of Epidemiology, Helmholtz Zentrum München GmbH, German Research Center for Environmental Health, 85764 Neuherberg, Germany
- Dr. von Hauner Children’s Hospital, University Hospital, LMU Munich, 80337 Munich, Germany
- Correspondence:
| | - Carlana Ramlochansingh
- Institute of Epidemiology, Helmholtz Zentrum München GmbH, German Research Center for Environmental Health, 85764 Neuherberg, Germany
- Institute of Medical Informatics, Biometry and Epidemiology (IBE), LMU Munich, 81377 Munich, Germany
| | - Olaf Uhl
- Dr. von Hauner Children’s Hospital, University Hospital, LMU Munich, 80337 Munich, Germany
| | - Hans Demmelmair
- Dr. von Hauner Children’s Hospital, University Hospital, LMU Munich, 80337 Munich, Germany
| | - Joachim Heinrich
- Institute of Epidemiology, Helmholtz Zentrum München GmbH, German Research Center for Environmental Health, 85764 Neuherberg, Germany
- Institute and Outpatient Clinic for Occupational, Social and Environmental Medicine, LMU University Hospitals, 80336 Munich, Germany
- Allergy and Lung Health Unit, Melbourne School of Population and Global Health, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Berthold Koletzko
- Dr. von Hauner Children’s Hospital, University Hospital, LMU Munich, 80337 Munich, Germany
| | - Marie Standl
- Institute of Epidemiology, Helmholtz Zentrum München GmbH, German Research Center for Environmental Health, 85764 Neuherberg, Germany
| | - Elisabeth Thiering
- Institute of Epidemiology, Helmholtz Zentrum München GmbH, German Research Center for Environmental Health, 85764 Neuherberg, Germany
- Dr. von Hauner Children’s Hospital, University Hospital, LMU Munich, 80337 Munich, Germany
| |
Collapse
|
164
|
Mednova IA, Chernonosov AA, Kornetova EG, Semke AV, Bokhan NA, Koval VV, Ivanova SA. Levels of Acylcarnitines and Branched-Chain Amino Acids in Antipsychotic-Treated Patients with Paranoid Schizophrenia with Metabolic Syndrome. Metabolites 2022; 12:metabo12090850. [PMID: 36144254 PMCID: PMC9504797 DOI: 10.3390/metabo12090850] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Revised: 09/04/2022] [Accepted: 09/06/2022] [Indexed: 11/25/2022] Open
Abstract
Several studies have shown that patients with schizophrenia are at high risk for metabolic syndrome (MetS) and bioenergetic dysfunction. Because acylcarnitines are involved in bioenergetic pathways and reflect the functioning of mitochondria, we hypothesized that these compounds are biomarkers of MetS in schizophrenia. The aim of this work was to quantify acylcarnitines and branched-chain amino acids in patients with schizophrenia comorbid with MetS. The study included 112 patients with paranoid schizophrenia treated with antipsychotics. Among them, 39 subjects met criteria of MetS. Concentrations of 30 acylcarnitines and three amino acids in dry serum spots were measured by liquid chromatography coupled with tandem mass spectrometry. MetS patients were found to have higher levels of valeryl carnitine (C5), leucine/isoleucine, and alanine as compared with patients without MetS, indicating possible participation of these compounds in the pathogenesis of metabolic disorders in schizophrenia. In patients with paranoid schizophrenia with or without MetS, lower levels of carnitines C10, C10:1, C12, and C18 were recorded as compared with the healthy individuals (n = 70), implying deterioration of energy metabolism. We believe that this finding can be explained by effects of antipsychotic medication on an enzyme called carnitine-palmitoyl transferase I.
Collapse
Affiliation(s)
- Irina A. Mednova
- Mental Health Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Aleutskaya Str. 4, 634014 Tomsk, Russia
- Correspondence:
| | - Alexander A. Chernonosov
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of Russian Academy of Sciences, Lavrentyev Avenue 8, 630090 Novosibirsk, Russia
| | - Elena G. Kornetova
- Mental Health Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Aleutskaya Str. 4, 634014 Tomsk, Russia
- Siberian State Medical University Hospital, Moskovsky Trakt 2, 634050 Tomsk, Russia
| | - Arkadiy V. Semke
- Mental Health Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Aleutskaya Str. 4, 634014 Tomsk, Russia
| | - Nikolay A. Bokhan
- Mental Health Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Aleutskaya Str. 4, 634014 Tomsk, Russia
- Department of Psychiatry, Addictology and Psychotherapy, Siberian State Medical University, Moskovsky Trakt 2, 634050 Tomsk, Russia
| | - Vladimir V. Koval
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of Russian Academy of Sciences, Lavrentyev Avenue 8, 630090 Novosibirsk, Russia
| | - Svetlana A. Ivanova
- Mental Health Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Aleutskaya Str. 4, 634014 Tomsk, Russia
- Department of Psychiatry, Addictology and Psychotherapy, Siberian State Medical University, Moskovsky Trakt 2, 634050 Tomsk, Russia
| |
Collapse
|
165
|
Palacios-González B, León-Reyes G, Rivera-Paredez B, Ibarra-González I, Vela-Amieva M, Flores YN, Canizales-Quinteros S, Salmerón J, Velázquez-Cruz R. Targeted Metabolomics Revealed a Sex-Dependent Signature for Metabolic Syndrome in the Mexican Population. Nutrients 2022; 14:nu14183678. [PMID: 36145054 PMCID: PMC9504093 DOI: 10.3390/nu14183678] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 09/01/2022] [Accepted: 09/02/2022] [Indexed: 11/26/2022] Open
Abstract
Metabolic syndrome (MetS) is a group of several metabolic conditions predisposing to chronic diseases. Individuals diagnosed with MetS are physiologically heterogeneous, with significant sex-specific differences. Therefore, we aimed to investigate the potential sex-specific serum modifications of amino acids and acylcarnitines (ACs) and their relationship with MetS in the Mexican population. This study included 602 participants from the Health Workers Cohort Study. Forty serum metabolites were analyzed using a targeted metabolomics approach. Multivariate regression models were used to test associations of clinical and biochemical parameters with metabolomic profiles. Our findings showed a serum amino acid signature (citrulline and glycine) and medium-chain ACs (AC14:1, AC10, and AC18:10H) associated with MetS. Glycine and AC10 were specific metabolites representative of discrimination according to sex-dependent MetS. In addition, we found that glycine and short-chain ACs (AC2, AC3, and AC8:1) are associated with age-dependent MetS. We also reported a significant correlation between body fat and metabolites associated with sex-age-dependent MetS. In conclusion, the metabolic profile varies by MetS status, and these differences are sex-age-dependent in the Mexican population.
Collapse
Affiliation(s)
| | - Guadalupe León-Reyes
- Genomics of Bone Metabolism Laboratory, National Institute of Genomic Medicine (INMEGEN), Mexico City 14610, Mexico
| | - Berenice Rivera-Paredez
- Research Center in Policies, Population and Health, School of Medicine, National Autonomous University of Mexico (UNAM), Mexico City 04510, Mexico
| | | | - Marcela Vela-Amieva
- Laboratory of Inborn Errors of Metabolism, National Pediatrics Institute (INP), Mexico City 04530, Mexico
| | - Yvonne N. Flores
- Epidemiological and Health Services Research Unit, Morelos Mexican Institute of Social Security, Cuernavaca 62000, Mexico
- Department of Health Policy and Management and UCLA-Kaiser Permanente Center for Health Equity, Fielding School of Public Health, University of California, Los Angeles, CA 90095, USA
- UCLA Center for Cancer Prevention and Control Research, Fielding School of Public Health and Jonsson Comprehensive Cancer Center, Los Angeles, CA 90095, USA
| | - Samuel Canizales-Quinteros
- Unit of Genomics of Population Applied to Health, Faculty of Chemistry, National Autonomous University of Mexico (UNAM), Mexico City 04510, Mexico
- National Institute of Genomic Medicine (INMEGEN), Mexico City 14610, Mexico
| | - Jorge Salmerón
- Research Center in Policies, Population and Health, School of Medicine, National Autonomous University of Mexico (UNAM), Mexico City 04510, Mexico
| | - Rafael Velázquez-Cruz
- Genomics of Bone Metabolism Laboratory, National Institute of Genomic Medicine (INMEGEN), Mexico City 14610, Mexico
- Correspondence: ; Tel./Fax: +52-(55)-5350-1900
| |
Collapse
|
166
|
Arjmand B, Ebrahimi Fana S, Ghasemi E, Kazemi A, Ghodssi-Ghassemabadi R, Dehghanbanadaki H, Najjar N, Kakaii A, Forouzanfar K, Nasli-Esfahani E, Farzadfar F, Larijani B, Razi F. Metabolic signatures of insulin resistance in non-diabetic individuals. BMC Endocr Disord 2022; 22:212. [PMID: 36002887 PMCID: PMC9404631 DOI: 10.1186/s12902-022-01130-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 08/18/2022] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Insulin resistance (IR) evolved from excessive energy intake and poor energy expenditure, affecting the patient's quality of life. Amino acid and acylcarnitine metabolomic profiles have identified consistent patterns associated with metabolic disease and insulin sensitivity. Here, we have measured a wide array of metabolites (30 acylcarnitines and 20 amino acids) with the MS/MS and investigated the association of metabolic profile with insulin resistance. METHODS The study population (n = 403) was randomly chosen from non-diabetic participants of the Surveillance of Risk Factors of NCDs in Iran Study (STEPS 2016). STEPS 2016 is a population-based cross-sectional study conducted periodically on adults aged 18-75 years in 30 provinces of Iran. Participants were divided into two groups according to the optimal cut-off point determined by the Youden index of HOMA-IR for the diagnosis of metabolic syndrome. Associations were investigated using regression models adjusted for age, sex, and body mass index (BMI). RESULTS People with high IR were significantly younger, and had higher education level, BMI, waist circumference, FPG, HbA1c, ALT, triglyceride, cholesterol, non-HDL cholesterol, uric acid, and a lower HDL-C level. We observed a strong positive association of serum BCAA (valine and leucine), AAA (tyrosine, tryptophan, and phenylalanine), alanine, and C0 (free carnitine) with IR (HOMA-IR); while C18:1 (oleoyl L-carnitine) was inversely correlated with IR. CONCLUSIONS In the present study, we identified specific metabolites linked to HOMA-IR that improved IR prediction. In summary, our study adds more evidence that a particular metabolomic profile perturbation is associated with metabolic disease and reemphasizes the significance of understanding the biochemistry and physiology which lead to these associations.
Collapse
Affiliation(s)
- Babak Arjmand
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran, Iran
- Diabetes Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Saeed Ebrahimi Fana
- Department of Clinical Biochemistry, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Erfan Ghasemi
- Non-Communicable Diseases Research Center, Endocrinology and Metabolism Population Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Ameneh Kazemi
- Non-Communicable Diseases Research Center, Endocrinology and Metabolism Population Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Hojat Dehghanbanadaki
- Diabetes Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Niloufar Najjar
- Metabolomics and Genomics Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Ardeshir Kakaii
- Metabolic Disorders Research Center, Endocrinology and Metabolism Molecular -Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Katayoon Forouzanfar
- Metabolomics and Genomics Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Ensieh Nasli-Esfahani
- Diabetes Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Farshad Farzadfar
- Non-Communicable Diseases Research Center, Endocrinology and Metabolism Population Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Bagher Larijani
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Farideh Razi
- Metabolomics and Genomics Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
167
|
Meneses MJ, Sousa-Lima I, Jarak I, Raposo JF, Alves MG, Macedo MP. Distinct impacts of fat and fructose on the liver, muscle, and adipose tissue metabolome: An integrated view. Front Endocrinol (Lausanne) 2022; 13:898471. [PMID: 36060961 PMCID: PMC9428722 DOI: 10.3389/fendo.2022.898471] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 07/12/2022] [Indexed: 11/17/2022] Open
Abstract
Objective In the last years, changes in dietary habits have contributed to the increasing prevalence of metabolic disorders, such as non-alcoholic fatty liver disease (NAFLD) and type 2 diabetes mellitus (T2DM). The differential burden of lipids and fructose on distinct organs needs to be unveiled. Herein, we hypothesized that high-fat and high-fructose diets differentially affect the metabolome of insulin-sensitive organs such as the liver, muscle, and different adipose tissue depots. Methods We have studied the impact of 12 weeks of a control (11.50% calories from fat, 26.93% from protein, and 61.57% from carbohydrates), high-fat/sucrose (HFat), or high-fructose (HFruct) feeding on C57Bl/6J male mice. Besides glucose homeostasis, we analyzed the hepatic levels of glucose and lipid-metabolism-related genes and the metabolome of the liver, the muscle, and white (WAT) and brown adipose tissue (BAT) depots. Results HFat diet led to a more profound impact on hepatic glucose and lipid metabolism than HFruct, with mice presenting glucose intolerance, increased saturated fatty acids, and no glycogen pool, yet both HFat and HFruct presented hepatic insulin resistance. HFat diet promoted a decrease in glucose and lactate pools in the muscle and an increase in glutamate levels. While HFat had alterations in BAT metabolites that indicate increased thermogenesis, HFruct led to an increase in betaine, a protective metabolite against fructose-induced inflammation. Conclusions Our data illustrate that HFat and HFruct have a negative but distinct impact on the metabolome of the liver, muscle, WAT, and BAT.
Collapse
Affiliation(s)
- Maria João Meneses
- iNOVA4Health, NOVA Medical School/Faculdade de Ciências Médicas (NMS/FCM), Universidade Nova de Lisboa, Lisbon, Portugal
- Portuguese Diabetes Association - Education and Research Center (APDP-ERC), Lisbon, Portugal
| | - Inês Sousa-Lima
- iNOVA4Health, NOVA Medical School/Faculdade de Ciências Médicas (NMS/FCM), Universidade Nova de Lisboa, Lisbon, Portugal
| | - Ivana Jarak
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal
- Department of Anatomy and Unit for Multidisciplinary Research in Biomedicine (UMIB), Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Porto, Portugal
| | - João F. Raposo
- iNOVA4Health, NOVA Medical School/Faculdade de Ciências Médicas (NMS/FCM), Universidade Nova de Lisboa, Lisbon, Portugal
- Portuguese Diabetes Association - Education and Research Center (APDP-ERC), Lisbon, Portugal
| | - Marco G. Alves
- Department of Anatomy and Unit for Multidisciplinary Research in Biomedicine (UMIB), Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Porto, Portugal
| | - Maria Paula Macedo
- iNOVA4Health, NOVA Medical School/Faculdade de Ciências Médicas (NMS/FCM), Universidade Nova de Lisboa, Lisbon, Portugal
- Portuguese Diabetes Association - Education and Research Center (APDP-ERC), Lisbon, Portugal
- Medical Sciences Department, University of Aveiro, Aveiro, Portugal
| |
Collapse
|
168
|
Vanweert F, Schrauwen P, Phielix E. Role of branched-chain amino acid metabolism in the pathogenesis of obesity and type 2 diabetes-related metabolic disturbances BCAA metabolism in type 2 diabetes. Nutr Diabetes 2022; 12:35. [PMID: 35931683 PMCID: PMC9356071 DOI: 10.1038/s41387-022-00213-3] [Citation(s) in RCA: 115] [Impact Index Per Article: 38.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 06/15/2022] [Accepted: 07/05/2022] [Indexed: 12/23/2022] Open
Abstract
Branched-chain amino acid (BCAA) catabolism has been considered to have an emerging role in the pathogenesis of metabolic disturbances in obesity and type 2 diabetes (T2D). Several studies showed elevated plasma BCAA levels in humans with insulin resistance and patients with T2D, although the underlying reason is unknown. Dysfunctional BCAA catabolism could theoretically be an underlying factor. In vitro and animal work collectively show that modulation of the BCAA catabolic pathway alters key metabolic processes affecting glucose homeostasis, although an integrated understanding of tissue-specific BCAA catabolism remains largely unknown, especially in humans. Proof-of-concept studies in rodents -and to a lesser extent in humans – strongly suggest that enhancing BCAA catabolism improves glucose homeostasis in metabolic disorders, such as obesity and T2D. In this review, we discuss several hypothesized mechanistic links between BCAA catabolism and insulin resistance and overview current available tools to modulate BCAA catabolism in vivo. Furthermore, this review considers whether enhancing BCAA catabolism forms a potential future treatment strategy to promote metabolic health in insulin resistance and T2D.
Collapse
Affiliation(s)
- Froukje Vanweert
- Department of Nutrition and Movement Sciences, NUTRIM, School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Patrick Schrauwen
- Department of Nutrition and Movement Sciences, NUTRIM, School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Esther Phielix
- Department of Nutrition and Movement Sciences, NUTRIM, School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, The Netherlands.
| |
Collapse
|
169
|
Bonomini-Gnutzmann R, Plaza-Díaz J, Jorquera-Aguilera C, Rodríguez-Rodríguez A, Rodríguez-Rodríguez F. Effect of Intensity and Duration of Exercise on Gut Microbiota in Humans: A Systematic Review. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:9518. [PMID: 35954878 PMCID: PMC9368618 DOI: 10.3390/ijerph19159518] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 07/17/2022] [Accepted: 07/25/2022] [Indexed: 02/05/2023]
Abstract
(1) Background: The gut microbiota might play a part in affecting athletic performance and is of considerable importance to athletes. The aim of this study was to search the recent knowledge of the protagonist played by high-intensity and high-duration aerobic exercise on gut microbiota composition in athletes and how these effects could provide disadvantages in sports performance. (2) Methods: This systematic review follows the PRISMA guidelines. An exhaustive bibliographic search in Web of Science, PubMed, and Scopus was conducted considering the articles published in the last 5 years. The selected articles were categorized according to the type of study. The risk of bias was assessed using the Joanna Briggs Institute's Critical Appraisal Tool for Systematic Reviews. (3) Results: Thirteen studies had negative effects of aerobic exercise on intestinal microbiota such as an upsurge in I-FABP, intestinal distress, and changes in the gut microbiota, such as an increase in Prevotella, intestinal permeability and zonulin. In contrast, seven studies observed positive effects of endurance exercise, including an increase in the level of bacteria such as increased microbial diversity and increased intestinal metabolites. (4) Conclusions: A large part of the studies found reported adverse effects on the intestinal microbiota when performing endurance exercises. In studies carried out on athletes, more negative effects on the microbiota were found than in those carried out on non-athletic subjects.
Collapse
Affiliation(s)
| | - Julio Plaza-Díaz
- Children’s Hospital of Eastern Ontario Research Institute, Ottawa, ON K1H 8L1, Canada
- Department of Biochemistry and Molecular Biology II, School of Pharmacy, University of Granada, 18071 Granada, Spain
- Instituto de Investigación Biosanitaria IBS.GRANADA, Complejo Hospitalario Universitario de Granada, 18014 Granada, Spain
| | - Carlos Jorquera-Aguilera
- Escuela de Nutrición y Dietética, Facultad de Ciencias, Universidad Mayor, Santiago 8580745, Chile;
| | - Andrés Rodríguez-Rodríguez
- Gastric Cancer Research Group—Laboratory of Oncology, UC Center for Investigational Oncology (CITO), Pontificia Universidad Católica de Chile, Santiago 8331150, Chile;
| | - Fernando Rodríguez-Rodríguez
- IRyS Group, Physical Education School, Pontificia Universidad Católica de Valparaíso, Valparaíso 2374631, Chile;
| |
Collapse
|
170
|
Associations of serum amino acids with insulin resistance among people with and without overweight or obesity: A prospective study in Japan. Clin Nutr 2022; 41:1827-1833. [PMID: 35839544 DOI: 10.1016/j.clnu.2022.06.039] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 06/15/2022] [Accepted: 06/21/2022] [Indexed: 01/27/2023]
Abstract
BACKGROUND & AIMS Limited evidence exists regarding the prospective associations between amino acids and insulin resistance. In addition, amino acids have been suggested to promote insulin resistance with the requirement of obesity in animal studies, but the interaction between amino acids and obesity on the development of insulin resistance has not been examined in epidemiological studies. We aimed to investigate the differences in the prospective associations of serum amino acids with insulin resistance among adults with and without overweight or obesity. METHODS Fasting serum concentrations of 25 amino acids were quantified in 1131 non-diabetic Japanese workers aged 22-71 years at baseline. The homeostasis model assessment of insulin resistance (HOMA-IR) was estimated at baseline and the 3-year follow-up. Generalized linear models were used to assess the associations between amino acids at baseline and HOMA-IR at follow-up with adjustment for potential confounding factors. A Bonferroni-corrected threshold of p = 0.001 was considered significant for multiple tests. RESULTS The associations for the following amino acids with HOMA-IR at the 3-year follow-up significantly varied by obesity status: isoleucine, valine, tyrosine, alanine, and methionine (all p for interaction <0.05). Higher concentrations of serum isoleucine, valine, tyrosine, and alanine (per 1SD) were significantly associated with higher HOMA-IR levels in overweight/obese participants (multivariable-adjusted β coefficients ranging from 0.09 to 0.12; all p < 0.001), but no association was observed in the underweight/normal-weight participants. The associations for serum methionine were direct among overweight/obese participants, but inverse among underweight/normal-weight participants (all p < 0.001). CONCLUSIONS This study demonstrated the prospective associations of different individual serum amino acids with insulin resistance, with most pronounced associations being for overweight/obese adults. Our findings support the possibility of heterogeneous effects of individual amino acids, as well as their interplay with obesity in the progression of insulin resistance.
Collapse
|
171
|
An Investigation into the Correlation of Intestinal Flora with Obesity and Gestational Diabetes Mellitus. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2022; 2022:5677073. [PMID: 35880087 PMCID: PMC9308517 DOI: 10.1155/2022/5677073] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 06/13/2022] [Accepted: 06/15/2022] [Indexed: 11/22/2022]
Abstract
Method Thirty-two pregnant women aged 25-35 who were hospitalized in Shanxi Maternal and Child Health Hospital from January 2019 to December 2019 were included for evaluation, including 15 normal pregnant women (NG_NO group), 6 pregnant women with GDM alone (G_NO group), and 7 pregnant women with overweight alone (NG_O group). Stools were collected from pregnant women at 24 and 37 weeks of gestation and newborns' first meconium. The v3-v4 variable region of the gut flora 16s rRNA was double-ended sequenced and bioinformatically analyzed using the Illumina MiSeq PE300 sequencing platform. Results In the third trimester of pregnancy, there were significant differences in the composition of intestinal flora between the simple overweight group, simple GDM group, and normal pregnant group. From the second trimester to the third trimester, there was no significant change in the relative distribution of intestinal flora at the phyla classification level in normal pregnant women. The relative distribution of intestinal flora at the phylum level of newborns was significantly different from that of their mothers. The characteristic intestinal microbes of newborns in simple GDM group were g_Diaphorobacter, while the simple recombinant neonates were Nocardiaceae (f_Nocardioidaceae). In addition, the results showed significant differences in intestinal flora among the normal pregnant women group, simple GDM group, simple overweight group, and GDM overweight group. The results of β diversity analysis showed a significant difference in intestinal microflora species composition structure between the simple overweight group and the normal pregnant group in the second trimester of pregnancy. The species composition structure of intestinal flora was similar between the simple GDM group and the normal pregnant group. In the third trimester of pregnancy, there was no significant difference in the β diversity index among the groups, and the composition and structure of intestinal flora were similar. There were significant differences in the composition structure (β diversity) of intestinal flora between pregnant women and their newborns in each group (P < 0.05). Correlation analysis showed that the blood glucose values of oral glucose tolerance test (OGTT)_1 h and OGTT_2 h were positively correlated with Bacteroides (Bacteroides) and negatively correlated with Proteus (Prevotella), prepregnancy BMI was negatively correlated with Bacteroides, and weight gain during pregnancy was negatively correlated with Vibrio (Desulfovibrio) in Proteus. The birth weight of newborns was positively correlated with Actinomycetes (Actinomyces), Bacteroides (Faecalibacterium), and microbacilli (Dialister) and negatively correlated with Rolston (Ralstonia). Conclusion Gut microbiota is strongly linked to obesity and gestational diabetes.
Collapse
|
172
|
Xiong Y, Jiang L, Li T. Aberrant branched-chain amino acid catabolism in cardiovascular diseases. Front Cardiovasc Med 2022; 9:965899. [PMID: 35911554 PMCID: PMC9334649 DOI: 10.3389/fcvm.2022.965899] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 06/29/2022] [Indexed: 01/04/2023] Open
Abstract
Globally, cardiovascular diseases are the leading cause of death. Research has focused on the metabolism of carbohydrates, fatty acids, and amino acids to improve the prognosis of cardiovascular diseases. There are three types of branched-chain amino acids (BCAAs; valine, leucine, and isoleucine) required for protein homeostasis, energy balance, and signaling pathways. Increasing evidence has implicated BCAAs in the pathogenesis of multiple cardiovascular diseases. This review summarizes the biological origin, signal transduction pathways and function of BCAAs as well as their significance in cardiovascular diseases, including myocardial hypertrophy, heart failure, coronary artery disease, diabetic cardiomyopathy, dilated cardiomyopathy, arrhythmia and hypertension.
Collapse
Affiliation(s)
- Yixiao Xiong
- Department of Anesthesiology, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital of Sichuan University, Chengdu, China
- Laboratory of Mitochondria and Metabolism, West China Hospital of Sichuan University, Chengdu, China
| | - Ling Jiang
- Department of Anesthesiology, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital of Sichuan University, Chengdu, China
- Laboratory of Mitochondria and Metabolism, West China Hospital of Sichuan University, Chengdu, China
| | - Tao Li
- Department of Anesthesiology, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital of Sichuan University, Chengdu, China
- Laboratory of Mitochondria and Metabolism, West China Hospital of Sichuan University, Chengdu, China
- *Correspondence: Tao Li,
| |
Collapse
|
173
|
Effect of the BCAT2 polymorphism (rs11548193) on plasma branched-chain amino acid concentrations after dietary intervention in subjects with obesity and insulin resistance. Br J Nutr 2022; 128:43-54. [PMID: 34340727 DOI: 10.1017/s0007114521002920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Branched-chain amino acids (BCAA) are considered markers of insulin resistance (IR) in subjects with obesity. In this study, we evaluated whether the presence of the SNP of the branched-chain aminotransferase 2 (BCAT2) gene can modify the effect of a dietary intervention (DI) on the plasma concentration of BCAA in subjects with obesity and IR. A prospective cohort study of adult subjects with obesity, BMI ≥ 30 kg/m2, homeostatic model assessment-insulin resistance (HOMA-IR ≥ 2·5) no diagnosed chronic disease, underwent a DI with an energy restriction of 3140 kJ/d and nutritional education for 1 month. Anthropometric measurements, body composition, blood pressure, resting energy expenditure, oral glucose tolerance test results, serum biochemical parameters and the plasma amino acid profile were evaluated before and after the DI. SNP were assessed by the TaqMan SNP genotyping assay. A total of eighty-two subjects were included, and fifteen subjects with a BCAT2 SNP had a greater reduction in leucine, isoleucine, valine and the sum of BCAA. Those subjects also had a greater reduction in skeletal muscle mass, fat-free mass, total body water, blood pressure, muscle strength and biochemical parameters after 1 month of the DI and adjusting for age and sex. This study demonstrated that the presence of the BCAT2 SNP promotes a greater reduction in plasma BCAA concentration after adjusting for age and sex, in subjects with obesity and IR after a 1-month energy-restricted DI.
Collapse
|
174
|
Remchak MME, Heiston EM, Ballantyne A, Dotson BL, Stewart NR, Spaeth AM, Malin SK. Insulin Sensitivity and Metabolic Flexibility Parallel Plasma TCA Levels in Early Chronotype With Metabolic Syndrome. J Clin Endocrinol Metab 2022; 107:e3487-e3496. [PMID: 35429387 PMCID: PMC9282268 DOI: 10.1210/clinem/dgac233] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Indexed: 12/15/2022]
Abstract
CONTEXT People characterized as late chronotype have elevated type 2 diabetes and cardiovascular disease risk compared to early chronotype. It is unclear how chronotype is associated with insulin sensitivity, metabolic flexibility, or plasma TCA cycle intermediates concentration, amino acids (AA), and/or beta-oxidation. OBJECTIVE This study examined these metabolic associations with chronotype. METHODS The Morningness-Eveningness Questionnaire (MEQ) was used to classify adults with metabolic syndrome (ATP III criteria) as either early (n = 15 [13F], MEQ = 64.7 ± 1.4) or late (n = 19 [16F], MEQ = 45.5 ± 1.3) chronotype. Fasting bloods determined hepatic (HOMA-IR) and adipose insulin resistance (Adipose-IR) while a 120-minute euglycemic clamp (40 mU/m2/min, 5 mmoL/L) was performed to test peripheral insulin sensitivity (glucose infusion rate). Carbohydrate (CHOOX) and fat oxidation (FOX), as well as nonoxidative glucose disposal (NOGD), were also estimated (indirect calorimetry). Plasma tricarboxylic acid cycle (TCA) intermediates, AA, and acyl-carnitines were measured along with VO2max and body composition (DXA). RESULTS There were no statistical differences in age, BMI, fat-free mass, VO2max, or ATP III criteria between groups. Early chronotype, however, had higher peripheral insulin sensitivity (P = 0.009) and lower HOMA-IR (P = 0.02) and Adipose-IR (P = 0.05) compared with late chronotype. Further, early chronotype had higher NOGD (P = 0.008) and greater insulin-stimulated CHOOX (P = 0.02). While fasting lactate (P = 0.01), TCA intermediates (isocitrate, α-ketoglutarate, succinate, fumarate, malate; all P ≤ 0.04) and some AA (proline, isoleucine; P = 0.003-0.05) were lower in early chronotype, other AA (threonine, histidine, arginine; all P ≤ 0.05) and most acyl-carnitines were higher (P ≤ 0.05) compared with late chronotype. CONCLUSION Greater insulin sensitivity and metabolic flexibility relates to plasma TCA concentration in early chronotype.
Collapse
Affiliation(s)
| | - Emily M Heiston
- University of Virginia, Charlottesville, VA, USA
- Virginia Commonwealth University, Richmond, VA, USA
| | | | | | - Nathan R Stewart
- Rutgers University, New Brunswick, NJ, USA
- University of Virginia, Charlottesville, VA, USA
| | | | - Steven K Malin
- Correspondence: Steven K. Malin, PhD, Department of Kinesiology & Health, 70 Lipman Dr Loree Gymnasium, New Brunswick, NJ 08091, USA.
| |
Collapse
|
175
|
Dar MA, Arafah A, Bhat KA, Khan A, Khan MS, Ali A, Ahmad SM, Rashid SM, Rehman MU. Multiomics technologies: role in disease biomarker discoveries and therapeutics. Brief Funct Genomics 2022; 22:76-96. [PMID: 35809340 DOI: 10.1093/bfgp/elac017] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 05/21/2022] [Accepted: 06/14/2022] [Indexed: 11/13/2022] Open
Abstract
Medical research has been revolutionized after the publication of the full human genome. This was the major landmark that paved the way for understanding the biological functions of different macro and micro molecules. With the advent of different high-throughput technologies, biomedical research was further revolutionized. These technologies constitute genomics, transcriptomics, proteomics, metabolomics, etc. Collectively, these high-throughputs are referred to as multi-omics technologies. In the biomedical field, these omics technologies act as efficient and effective tools for disease diagnosis, management, monitoring, treatment and discovery of certain novel disease biomarkers. Genotyping arrays and other transcriptomic studies have helped us to elucidate the gene expression patterns in different biological states, i.e. healthy and diseased states. Further omics technologies such as proteomics and metabolomics have an important role in predicting the role of different biological molecules in an organism. It is because of these high throughput omics technologies that we have been able to fully understand the role of different genes, proteins, metabolites and biological pathways in a diseased condition. To understand a complex biological process, it is important to apply an integrative approach that analyses the multi-omics data in order to highlight the possible interrelationships of the involved biomolecules and their functions. Furthermore, these omics technologies offer an important opportunity to understand the information that underlies disease. In the current review, we will discuss the importance of omics technologies as promising tools to understand the role of different biomolecules in diseases such as cancer, cardiovascular diseases, neurodegenerative diseases and diabetes. SUMMARY POINTS
Collapse
|
176
|
Vineis P, Barouki R. The exposome as the science of social-to-biological transitions. ENVIRONMENT INTERNATIONAL 2022; 165:107312. [PMID: 35635963 DOI: 10.1016/j.envint.2022.107312] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 04/19/2022] [Accepted: 05/17/2022] [Indexed: 06/15/2023]
Abstract
The understanding of disease etiology and pathogenesis has radically changed as a consequence of the new challenges posed by climate change, environmental degradation and emerging infectious diseases. The awareness of the influence of distal causes (e.g. planetary changes at the roots of new pandemics), of the social environment and of early life exposures calls for innovative models of disease onset. Here we propose a scheme for the practice of epidemiology and toxicology that incorporates new recent advancements in both disciplines, under the general umbrella of the "exposome". The exposome approach to disease encompasses a lifecourse perspective from conception onwards, and the investigation of the role played by all exposures individuals undergo in their lives. These include social inequalities and psychosocial influences, in addition to chemical, biological and physical exposures. We stress the role played by social differences and inequalities in the course of life as an overarching factor that influences downstream layers (including behaviours). We show that the idea of "lifecourse exposome" is compatible with the current interpretation of Adverse Outcome Pathways in toxicology, and in fact we propose an extension of the concept towards "lifecourse Adverse Outcome Pathways". We propose to merge different research perspectives and promote an encounter between the sociological perspective of "biography" (using Pierre Bourdieu's conceptual framework) and biology, according to the idea of accumulated biological capital of individuals. We also propose to treat social capital (including inequalities) no longer as a confounding factor but as an overarching determinant, perhaps the most important of all because it is the one that influences all other exposures downstream. The importance of early exposures in a lifecourse perspective leads to policy implications, i.e. investing more in the various forms of capital (social, economic, cultural) in early life.
Collapse
Affiliation(s)
- Paolo Vineis
- MRC Centre for Environment and Health, School of Public Health, Imperial College London, United Kingdom.
| | | |
Collapse
|
177
|
Bowman ER, Wilson M, Riedl KM, MaWhinney S, Jankowski CM, Funderburg NT, Erlandson KM. Lipidome Alterations with Exercise Among People With and Without HIV: An Exploratory Study. AIDS Res Hum Retroviruses 2022; 38:544-551. [PMID: 35302400 PMCID: PMC9297322 DOI: 10.1089/aid.2021.0154] [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] [Indexed: 11/13/2022] Open
Abstract
Age-related comorbidities and physical function impairments in aging people with HIV (PWH) can be improved through exercise interventions. The mechanisms underlying these improvements, including lipidomic changes, are unknown. Sedentary adults (50-75 years old) with or without HIV participated in supervised endurance/resistance exercise for 24 weeks. Plasma lipid concentrations (∼1,200 lipid species from 13 lipid classes) at baseline and week 24 were measured by mass spectrometry. Given multiple comparisons, unadjusted and Benjamini-Hochberg corrected p values are reported. Analyses are considered exploratory. Twenty-five PWH and 24 controls had paired samples at baseline and week 24. The change in total triacylglycerol (TAG) concentrations after exercise intervention differed between groups (unadj-p = 0.006, adj-p = 0.078) with concentrations increasing among controls, but not among PWH. Changes in concentrations of TAG species composed of long-chain fatty acids differed between groups (unadj-p < 0.04) with increases among controls, but not among PWH. Changes in total diacylglycerol (DAG) concentration from baseline to week 24 differed between groups (unadj-p = 0.03, adj-p = 0.2) with an increase in PWH and a nonsignificant decrease in controls. Baseline to week 24 changes in DAGs composed of palmitic acid (16:0), palmitoleic acid (16:1), and stearic acid (18:0) differed by serostatus (unadj-p = 0.009-0.03; adj-p 0.10-0.12), with nonsignificant increases and decreases in concentrations in PWH and controls, respectively. Concentrations of individual lysophosphatidylcholine (LPC) and ceramide (CER) species also differed by HIV serostatus (unadj-p < = 0.05). Although exploratory, the effects of exercise on the lipidome may differ among people with and without HIV, potentially due to underlying alterations in lipid processing and fatty acid oxidation in PWH. Clinical Trials NCT02404792.
Collapse
Affiliation(s)
- Emily R. Bowman
- College of Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Melissa Wilson
- Department of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, Colorado, USA
| | - Kenneth M. Riedl
- College of Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Samantha MaWhinney
- Department of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, Colorado, USA
| | - Catherine M. Jankowski
- Department of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, Colorado, USA
| | | | - Kristine M. Erlandson
- Department of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, Colorado, USA
| |
Collapse
|
178
|
Long SE, Jacobson MH, Wang Y, Liu M, Afanasyeva Y, Sumner SJ, McRitchie S, Kirchner DR, Brubaker SG, Mehta-Lee SS, Kahn LG, Trasande L. Longitudinal associations of pre-pregnancy BMI and gestational weight gain with maternal urinary metabolites: an NYU CHES study. Int J Obes (Lond) 2022; 46:1332-1340. [PMID: 35411100 PMCID: PMC9581342 DOI: 10.1038/s41366-022-01116-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 03/22/2022] [Accepted: 03/23/2022] [Indexed: 12/02/2022]
Abstract
BACKGROUND/OBJECTIVES Excessive gestational weight gain (GWG) and pre-pregnancy obesity affect a significant portion of the US pregnant population and are linked with negative maternal and child health outcomes. The objective of this study was to explore associations of pre-pregnancy body mass index (pBMI) and GWG with longitudinally measured maternal urinary metabolites throughout pregnancy. SUBJECTS/METHODS Among 652 participants in the New York University Children's Health and Environment Study, a longitudinal pregnancy cohort, targeted metabolomics were measured in serially collected urine samples throughout pregnancy. Metabolites were measured at median 10 (T1), 21 (T2), and 29 (T3) weeks gestation using the Biocrates AbsoluteIDQ® p180 Urine Extension kit. Acylcarnitine, amino acid, biogenic amine, phosphatidylcholine, lysophosphatidylcholine, sphingolipid, and sugar levels were quantified. Pregnant people 18 years or older, without type 1 or 2 diabetes and with singleton live births and valid pBMI and metabolomics data were included. GWG and pBMI were calculated using weight and height data obtained from electronic health records. Linear mixed effects models with interactions with time were fit to determine the gestational age-specific associations of categorical pBMI and continuous interval-specific GWG with urinary metabolites. All analyses were corrected for false discovery rate. RESULTS Participants with obesity had lower long-chain acylcarnitine levels throughout pregnancy and lower phosphatidylcholine and glucogenic amino acids and higher phenylethylamine concentrations in T2 and T3 compared with participants with normal/underweight pBMI. GWG was associated with taurine in T2 and T3 and C5 acylcarnitine species, C5:1, C5-DC, and C5-M-DC, in T2. CONCLUSIONS pBMI and GWG were associated with the metabolic environment of pregnant individuals, particularly in relation to mid-pregnancy. These results highlight the importance of both preconception and prenatal maternal health.
Collapse
Affiliation(s)
- Sara E Long
- Division of Environmental Pediatrics, Department of Pediatrics, NYU Langone Medical Center, New York, NY, USA.
| | - Melanie H Jacobson
- Division of Environmental Pediatrics, Department of Pediatrics, NYU Langone Medical Center, New York, NY, USA
| | - Yuyan Wang
- Department of Population Health, NYU Langone Medical Center, New York, NY, USA
| | - Mengling Liu
- Department of Population Health, NYU Langone Medical Center, New York, NY, USA
- Department of Environmental Medicine, NYU Langone Medical Center, New York, NY, USA
| | - Yelena Afanasyeva
- Division of Environmental Pediatrics, Department of Pediatrics, NYU Langone Medical Center, New York, NY, USA
- Department of Population Health, NYU Langone Medical Center, New York, NY, USA
| | - Susan J Sumner
- Department of Nutrition, UNC Chapel Hill, Chapel Hill, NC, USA
- UNC Chapel Hill Nutrition Research Institute, Kannapolis, NC, USA
| | - Susan McRitchie
- UNC Chapel Hill Nutrition Research Institute, Kannapolis, NC, USA
| | - David R Kirchner
- UNC Chapel Hill Nutrition Research Institute, Kannapolis, NC, USA
| | - Sara G Brubaker
- Department of Obstetrics and Gynecology, NYU Langone Medical Center, New York, NY, 10016, USA
| | - Shilpi S Mehta-Lee
- Department of Obstetrics and Gynecology, NYU Langone Medical Center, New York, NY, 10016, USA
| | - Linda G Kahn
- Division of Environmental Pediatrics, Department of Pediatrics, NYU Langone Medical Center, New York, NY, USA
- Department of Population Health, NYU Langone Medical Center, New York, NY, USA
| | - Leonardo Trasande
- Division of Environmental Pediatrics, Department of Pediatrics, NYU Langone Medical Center, New York, NY, USA
- Department of Population Health, NYU Langone Medical Center, New York, NY, USA
- Department of Environmental Medicine, NYU Langone Medical Center, New York, NY, USA
- NYU Wagner School of Public Service, New York, NY, USA
- NYU College of Global Public Health, New York, NY, USA
| |
Collapse
|
179
|
Dietary Protein Restriction Improves Metabolic Dysfunction in Patients with Metabolic Syndrome in a Randomized, Controlled Trial. Nutrients 2022; 14:nu14132670. [PMID: 35807851 PMCID: PMC9268415 DOI: 10.3390/nu14132670] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 06/15/2022] [Accepted: 06/16/2022] [Indexed: 11/28/2022] Open
Abstract
Dietary restriction (DR) reduces adiposity and improves metabolism in patients with one or more symptoms of metabolic syndrome. Nonetheless, it remains elusive whether the benefits of DR in humans are mediated by calorie or nutrient restriction. This study was conducted to determine whether isocaloric dietary protein restriction is sufficient to confer the beneficial effects of dietary restriction in patients with metabolic syndrome. We performed a prospective, randomized controlled dietary intervention under constant nutritional and medical supervision. Twenty-one individuals diagnosed with metabolic syndrome were randomly assigned for caloric restriction (CR; n = 11, diet of 5941 ± 686 KJ per day) or isocaloric dietary protein restriction (PR; n = 10, diet of 8409 ± 2360 KJ per day) and followed for 27 days. Like CR, PR promoted weight loss due to a reduction in adiposity, which was associated with reductions in blood glucose, lipid levels, and blood pressure. More strikingly, both CR and PR improved insulin sensitivity by 62.3% and 93.2%, respectively, after treatment. Fecal microbiome diversity was not affected by the interventions. Adipose tissue bulk RNA-Seq data revealed minor changes elicited by the interventions. After PR, terms related to leukocyte proliferation were enriched among the upregulated genes. Protein restriction is sufficient to confer almost the same clinical outcomes as calorie restriction without the need for a reduction in calorie intake. The isocaloric characteristic of the PR intervention makes this approach a more attractive and less drastic dietary strategy in clinical settings and has more significant potential to be used as adjuvant therapy for people with metabolic syndrome.
Collapse
|
180
|
Liu Y, Zhang C, Zhang Y, Jiang X, Liang Y, Wang H, Li Y, Sun G. Association between Excessive Dietary Branched-Chain Amino Acids Intake and Hypertension Risk in Chinese Population. Nutrients 2022; 14:nu14132582. [PMID: 35807761 PMCID: PMC9268479 DOI: 10.3390/nu14132582] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Revised: 06/16/2022] [Accepted: 06/20/2022] [Indexed: 01/27/2023] Open
Abstract
The dietary intake of branched-chain amino acids (BCAAs) has been reported to be associated with both elevated blood pressure (BP) and hypertension risk, while published findings were inconsistent, and the causality has never been well disclosed. We performed this prospective study aiming to find out the relationship between dietary BCAAs intake and hypertension risk in the Chinese population. A total of 8491 participants (40,285 person-years) were selected. The levels of dietary BCAAs intake were estimated using the 24-h Food Frequency Questionnaire. Associations of both BP values and hypertension risk with per standard deviation increase of BCAAs were estimated using linear and COX regression analysis, respectively. The hazard ratios and 95% confidence interval were given. Restricted cubic spline analysis (RCS) was used to estimate the nonlinearity. Both systolic and diastolic BP values at the end points of follow-up were positively associated with dietary BCAAs intake. Positive associations between BCAAs intake and hypertension risk were shown in both men and women. By performing a RCS analysis, the nonlinear relationship between BCAAs intake and hypertension was shown. As the intake levels of Ile, Leu, and Val, respectively, exceeded 2.49 g/day, 4.91 g/day, and 2.88 g/day in men (2.16 g/day, 3.84 g/day, and 2.56 g/day in women), the hypertension risk increased. Our findings could provide some concrete evidence in the primary prevention of hypertension based on dietary interventions.
Collapse
Affiliation(s)
- Yuyan Liu
- Department of Clinical Epidemiology, The Fourth Affiliated Hospital of China Medical University, Shenyang 110031, China;
| | - Chengwen Zhang
- Research Center of Environmental and Non-Communicable Disease, School of Public Health, China Medical University, Shenyang 110000, China; (C.Z.); (Y.Z.); (X.J.); (Y.L.); (H.W.); (G.S.)
| | - Yuan Zhang
- Research Center of Environmental and Non-Communicable Disease, School of Public Health, China Medical University, Shenyang 110000, China; (C.Z.); (Y.Z.); (X.J.); (Y.L.); (H.W.); (G.S.)
| | - Xuheng Jiang
- Research Center of Environmental and Non-Communicable Disease, School of Public Health, China Medical University, Shenyang 110000, China; (C.Z.); (Y.Z.); (X.J.); (Y.L.); (H.W.); (G.S.)
| | - Yuanhong Liang
- Research Center of Environmental and Non-Communicable Disease, School of Public Health, China Medical University, Shenyang 110000, China; (C.Z.); (Y.Z.); (X.J.); (Y.L.); (H.W.); (G.S.)
| | - Huan Wang
- Research Center of Environmental and Non-Communicable Disease, School of Public Health, China Medical University, Shenyang 110000, China; (C.Z.); (Y.Z.); (X.J.); (Y.L.); (H.W.); (G.S.)
| | - Yongfang Li
- Research Center of Environmental and Non-Communicable Disease, School of Public Health, China Medical University, Shenyang 110000, China; (C.Z.); (Y.Z.); (X.J.); (Y.L.); (H.W.); (G.S.)
- Correspondence:
| | - Guifan Sun
- Research Center of Environmental and Non-Communicable Disease, School of Public Health, China Medical University, Shenyang 110000, China; (C.Z.); (Y.Z.); (X.J.); (Y.L.); (H.W.); (G.S.)
| |
Collapse
|
181
|
Yu L, Song P, Zhu Q, Li Y, Jia S, Zhang S, Wang Z, Zhang J. The Dietary Branched-Chain Amino Acids Transition and Risk of Type 2 Diabetes Among Chinese Adults From 1997 to 2015: Based on Seven Cross-Sectional Studies and a Prospective Cohort Study. Front Nutr 2022; 9:881847. [PMID: 35677550 PMCID: PMC9168595 DOI: 10.3389/fnut.2022.881847] [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: 02/23/2022] [Accepted: 04/25/2022] [Indexed: 11/13/2022] Open
Abstract
Background The situation is grim for the prevention and control of type 2 diabetes (T2D) and prediabetes in China. Serum and dietary branched-chain amino acids (BCAAs) were risk factors for T2D. However, there is a lack of information on trends in consumption of BCAAs and the risk of T2D associated with BCAAs intake, based on nationally representative data in China. Thus, we aimed to comprehensively describe the dietary BCAAs transition and risk of T2D, at a national level among Chinese adults from 1997 to 2015. Methods The data sources were the China Health and Nutrition Survey (CHNS) and China Nutrition and Health Survey (CNHS). Cross-sectional data on intake were obtained from CHNS (1997, n = 9,404), CHNS (2000, n = 10,291), CHNS (2004, n = 9,682), CHNS (2006, n = 9,553), CHNS (2009, n = 9,811), CHNS (2011, n = 12,686) and CNHS (2015, n = 71,695). Prospective cohort data were obtained CHNS (1997-2015, n = 15,508). Results From 1997 to 2015, there was a significant decreasing trend in the BCAAs intake of Chinese adults in all subgroups (P < 0.0001) except for Leu in 80 or older, and a decreasing trend in the consumption of BCAAs after 40 years old (P < 0.05). The mean intake of BCAAs in the population of cohort study was 11.83 ± 3.77g/day. The 95% CI was above the HR of 1.0, when the consumptions were higher than 14.01, 3.75, 6.07, 4.21 g/day in BCAAs, Ile, Leu and Val, based on RCS curves. According to the Cox proportional hazards models, Compared with individuals with BCAAs consumption of 10.65-12.37 g/day, the multivariable-adjusted HR for diabetes was 2.26 (95% CI 1.45 to 3.51) for individuals with consumption of BCAAs more than 18.52 g/day. A statistically significant positive association between BCAAs intake and risk of T2D was observed in males or participants aged 45 years and older, but not in females or participants younger than 45 years. Conclusion Our results reveal a trend toward decreased BCAAs intake in Chinese from 1997 to 2015. After 40 years of age, consumption of BCAAs declined with increasing age. Higher BCAAs intake was associated with higher risk of T2D. This relationship is more stable among men and middle-aged and elderly people.
Collapse
Affiliation(s)
- Lianlong Yu
- National Institute for Nutrition and Health, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Pengkun Song
- National Institute for Nutrition and Health, Chinese Center for Disease Control and Prevention, Beijing, China.,NHC Key Laboratory of Trace Element Nutrition, National Institute for Nutrition and Health, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Qianrang Zhu
- National Institute for Nutrition and Health, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Yuqian Li
- National Institute for Nutrition and Health, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Shanshan Jia
- National Institute for Nutrition and Health, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Shixiu Zhang
- Department of Nutrition and Food Hygiene, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Zhihong Wang
- National Institute for Nutrition and Health, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Jian Zhang
- National Institute for Nutrition and Health, Chinese Center for Disease Control and Prevention, Beijing, China
| |
Collapse
|
182
|
Horgusluoglu E, Neff R, Song W, Wang M, Wang Q, Arnold M, Krumsiek J, Galindo‐Prieto B, Ming C, Nho K, Kastenmüller G, Han X, Baillie R, Zeng Q, Andrews S, Cheng H, Hao K, Goate A, Bennett DA, Saykin AJ, Kaddurah‐Daouk R, Zhang B, for the Alzheimer's Disease Neuroimaging Initiative (ADNI), the Alzheimer Disease Metabolomics Consortium. Integrative metabolomics-genomics approach reveals key metabolic pathways and regulators of Alzheimer's disease. Alzheimers Dement 2022; 18:1260-1278. [PMID: 34757660 PMCID: PMC9085975 DOI: 10.1002/alz.12468] [Citation(s) in RCA: 93] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 04/14/2021] [Accepted: 04/17/2021] [Indexed: 12/29/2022]
Abstract
Metabolites, the biochemical products of the cellular process, can be used to measure alterations in biochemical pathways related to the pathogenesis of Alzheimer's disease (AD). However, the relationships between systemic abnormalities in metabolism and the pathogenesis of AD are poorly understood. In this study, we aim to identify AD-specific metabolomic changes and their potential upstream genetic and transcriptional regulators through an integrative systems biology framework for analyzing genetic, transcriptomic, metabolomic, and proteomic data in AD. Metabolite co-expression network analysis of the blood metabolomic data in the Alzheimer's Disease Neuroimaging Initiative (ADNI) shows short-chain acylcarnitines/amino acids and medium/long-chain acylcarnitines are most associated with AD clinical outcomes, including episodic memory scores and disease severity. Integration of the gene expression data in both the blood from the ADNI and the brain from the Accelerating Medicines Partnership Alzheimer's Disease (AMP-AD) program reveals ABCA1 and CPT1A are involved in the regulation of acylcarnitines and amino acids in AD. Gene co-expression network analysis of the AMP-AD brain RNA-seq data suggests the CPT1A- and ABCA1-centered subnetworks are associated with neuronal system and immune response, respectively. Increased ABCA1 gene expression and adiponectin protein, a regulator of ABCA1, correspond to decreased short-chain acylcarnitines and amines in AD in the ADNI. In summary, our integrated analysis of large-scale multiomics data in AD systematically identifies novel metabolites and their potential regulators in AD and the findings pave a way for not only developing sensitive and specific diagnostic biomarkers for AD but also identifying novel molecular mechanisms of AD pathogenesis.
Collapse
Affiliation(s)
- Emrin Horgusluoglu
- Department of Genetics and Genomic SciencesMount Sinai Center for Transformative Disease ModelingIcahn School of Medicine at Mount SinaiIcahn Institute of Genomics and Multiscale BiologyNew YorkNew YorkUSA
| | - Ryan Neff
- Department of Genetics and Genomic SciencesMount Sinai Center for Transformative Disease ModelingIcahn School of Medicine at Mount SinaiIcahn Institute of Genomics and Multiscale BiologyNew YorkNew YorkUSA
| | - Won‐Min Song
- Department of Genetics and Genomic SciencesMount Sinai Center for Transformative Disease ModelingIcahn School of Medicine at Mount SinaiIcahn Institute of Genomics and Multiscale BiologyNew YorkNew YorkUSA
| | - Minghui Wang
- Department of Genetics and Genomic SciencesMount Sinai Center for Transformative Disease ModelingIcahn School of Medicine at Mount SinaiIcahn Institute of Genomics and Multiscale BiologyNew YorkNew YorkUSA
| | - Qian Wang
- Department of Genetics and Genomic SciencesMount Sinai Center for Transformative Disease ModelingIcahn School of Medicine at Mount SinaiIcahn Institute of Genomics and Multiscale BiologyNew YorkNew YorkUSA
| | - Matthias Arnold
- Institute of Computational BiologyHelmholtz Zentrum MünchenGerman Research Center for Environmental HealthNeuherbergGermany
- Department of Psychiatry and Behavioral SciencesDuke UniversityDurhamNorth CarolinaUSA
| | - Jan Krumsiek
- Department of Physiology and BiophysicsWeill Cornell MedicineInstitute for Computational BiomedicineEnglander Institute for Precision MedicineNew YorkNew YorkUSA
| | - Beatriz Galindo‐Prieto
- Department of Physiology and BiophysicsWeill Cornell MedicineInstitute for Computational BiomedicineEnglander Institute for Precision MedicineNew YorkNew YorkUSA
- Helen and Robert Appel Alzheimer's Disease Research InstituteBrain and Mind Research InstituteWeill Cornell MedicineNew YorkNew YorkUSA
| | - Chen Ming
- Department of Genetics and Genomic SciencesMount Sinai Center for Transformative Disease ModelingIcahn School of Medicine at Mount SinaiIcahn Institute of Genomics and Multiscale BiologyNew YorkNew YorkUSA
| | - Kwangsik Nho
- Department of Radiology and Imaging Sciences; Indiana Alzheimer Disease CenterIndiana University School of MedicineIndianapolisIndianaUSA
| | - Gabi Kastenmüller
- Institute of Computational BiologyHelmholtz Zentrum MünchenGerman Research Center for Environmental HealthNeuherbergGermany
| | - Xianlin Han
- Barshop Institute for Longevity and Aging StudiesUniversity of Texas Health Science Center at San AntonioSan AntonioTexasUSA
| | | | - Qi Zeng
- Department of Genetics and Genomic SciencesMount Sinai Center for Transformative Disease ModelingIcahn School of Medicine at Mount SinaiIcahn Institute of Genomics and Multiscale BiologyNew YorkNew YorkUSA
| | - Shea Andrews
- Department of NeuroscienceRonald M. Loeb Center for Alzheimer's DiseaseIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Haoxiang Cheng
- Department of Genetics and Genomic SciencesMount Sinai Center for Transformative Disease ModelingIcahn School of Medicine at Mount SinaiIcahn Institute of Genomics and Multiscale BiologyNew YorkNew YorkUSA
| | - Ke Hao
- Department of Genetics and Genomic SciencesMount Sinai Center for Transformative Disease ModelingIcahn School of Medicine at Mount SinaiIcahn Institute of Genomics and Multiscale BiologyNew YorkNew YorkUSA
| | - Alison Goate
- Department of NeuroscienceRonald M. Loeb Center for Alzheimer's DiseaseIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - David A. Bennett
- Rush Alzheimer's Disease CenterRush University Medical CenterChicagoIllinoisUSA
| | - Andrew J. Saykin
- Department of Radiology and Imaging Sciences; Indiana Alzheimer Disease CenterIndiana University School of MedicineIndianapolisIndianaUSA
| | - Rima Kaddurah‐Daouk
- Department of Psychiatry and Behavioral SciencesDuke UniversityDurhamNorth CarolinaUSA
- Duke Institute of Brain SciencesDuke UniversityDurhamNorth CarolinaUSA
- Department of MedicineDuke UniversityDurhamNorth CarolinaUSA
| | - Bin Zhang
- Department of Genetics and Genomic SciencesMount Sinai Center for Transformative Disease ModelingIcahn School of Medicine at Mount SinaiIcahn Institute of Genomics and Multiscale BiologyNew YorkNew YorkUSA
| | | | | |
Collapse
|
183
|
Missawi O, Venditti M, Cappello T, Zitouni N, Marco GDE, Boughattas I, Bousserrhine N, Belbekhouche S, Minucci S, Maisano M, Banni M. Autophagic event and metabolomic disorders unveil cellular toxicity of environmental microplastics on marine polychaete Hediste diversicolor. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2022; 302:119106. [PMID: 35248622 DOI: 10.1016/j.envpol.2022.119106] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 02/26/2022] [Accepted: 03/03/2022] [Indexed: 06/14/2023]
Abstract
Although the hazards of microplastics (MPs) have been quite well explored, the aberrant metabolism and the involvement of the autophagy pathway as an adverse response to environmental MPs in benthic organisms are still unclear. The present work aims to assess the impact of different environmental MPs collected from the south coast of the Mediterranean Sea, composed by polyethylene (PE), polyethylene vinyl acetate (PEVA), low-density polyethylene (LDPE), high-density polyethylene (HDPE), polypropylene (PP) and polyamide (PA) on the metabolome and proteome of the marine polychaete Hediste diversicolor. As a result, all the microplastic types were detected with Raman microspectroscopy in polychaetes tissues, causing cytoskeleton damage and induced autophagy pathway manifested by immunohistochemical labeling of specific targeted proteins, through Tubulin (Tub), Microtubule-associated protein light chain 3 (LC3), and p62 (also named Sequestosome 1). Metabolomics was conducted to further investigate the metabolic alterations induced by the environmental MPs-mixture in polychaetes. A total of 28 metabolites were differentially expressed between control and MPs-treated polychaetes, which showed elevated levels of amino acids, glucose, ATP/ADP, osmolytes, glutathione, choline and phosphocholine, and reduced concentration of aspartate. These novel findings extend our understanding given the toxicity of environmental microplastics and unravel their underlying mechanisms.
Collapse
Affiliation(s)
- Omayma Missawi
- University of Sousse, Laboratory of Agrobiodiversity and Ecotoxicology, Higher Institute of Agronomy, Sousse, Tunisia.
| | - Massimo Venditti
- Department of Experimental Medicine, Section Human Physiology and Integrated Biological Functions "F. Bottazzi", University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Tiziana Cappello
- University of Messina, Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, 98166 Messina, Italy
| | - Nesrine Zitouni
- University of Sousse, Laboratory of Agrobiodiversity and Ecotoxicology, Higher Institute of Agronomy, Sousse, Tunisia
| | - Giuseppe DE Marco
- University of Messina, Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, 98166 Messina, Italy
| | - Iteb Boughattas
- University of Sousse, Laboratory of Agrobiodiversity and Ecotoxicology, Higher Institute of Agronomy, Sousse, Tunisia; Regional Field Crops Research Center of Beja, Tunisia
| | - Noureddine Bousserrhine
- University Paris-Est Creteil, Laboratory of Water, Environment and Urban Systems, Faculty of Science and Technology, Creteil Cedex, France
| | - Sabrina Belbekhouche
- CNRS, University of Paris-Est Creteil, Institute of Chemistry and Materials Paris-Est ICMPE, UMR7182, 94320 Thiais, France
| | - Sergio Minucci
- Department of Experimental Medicine, Section Human Physiology and Integrated Biological Functions "F. Bottazzi", University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Maria Maisano
- University of Messina, Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, 98166 Messina, Italy
| | - Mohamed Banni
- University of Sousse, Laboratory of Agrobiodiversity and Ecotoxicology, Higher Institute of Agronomy, Sousse, Tunisia; Higher Institute of Biotechnology Monastir, University of Monastir, Monastir, Tunisia
| |
Collapse
|
184
|
Fang S, Wade KH, Hughes DA, Fitzgibbon S, Yip V, Timpson NJ, Corbin LJ. A multivariant recall-by-genotype study of the metabolomic signature of BMI. Obesity (Silver Spring) 2022; 30:1298-1310. [PMID: 35598895 PMCID: PMC9324973 DOI: 10.1002/oby.23441] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 03/14/2022] [Accepted: 03/15/2022] [Indexed: 12/12/2022]
Abstract
OBJECTIVE This study estimated the effect of BMI on circulating metabolites in young adults using a recall-by-genotype study design. METHODS A recall-by-genotype study was implemented in the Avon Longitudinal Study of Parents and Children. Samples from 756 participants were selected for untargeted metabolomics analysis based on low versus high genetic liability for higher BMI defined by a genetic risk score (GRS). Regression analyses were performed to investigate associations between BMI GRS group and relative abundance of 973 metabolites. RESULTS After correction for multiple testing, 29 metabolites were associated with BMI GRS group. Bilirubin was among the most strongly associated metabolites, with reduced levels measured in individuals in the high-BMI GRS group (β = -0.32, 95% CI: -0.46 to -0.18, Benjamini-Hochberg adjusted p = 0.005). This study observed associations between BMI GRS group and the levels of several potentially diet-related metabolites, including hippurate, which had lower mean abundance in individuals in the high-BMI GRS group (β = -0.29, 95% CI: -0.44 to -0.15, Benjamini-Hochberg adjusted p = 0.008). CONCLUSIONS Together with existing literature, these results suggest that a genetic predisposition to higher BMI captures differences in metabolism leading to adiposity gain. In the absence of prospective data, separating these effects from the downstream consequences of weight gain is challenging.
Collapse
Affiliation(s)
- Si Fang
- MRC Integrative Epidemiology Unit at the University of BristolBristolUK
- Population Health ScienceBristol Medical SchoolUniversity of BristolBristolUK
| | - Kaitlin H. Wade
- MRC Integrative Epidemiology Unit at the University of BristolBristolUK
- Population Health ScienceBristol Medical SchoolUniversity of BristolBristolUK
| | - David A. Hughes
- MRC Integrative Epidemiology Unit at the University of BristolBristolUK
- Population Health ScienceBristol Medical SchoolUniversity of BristolBristolUK
| | - Sophie Fitzgibbon
- Bristol Bioresource LaboratoriesPopulation Health ScienceBristol Medical SchoolUniversity of BristolBristolUK
| | - Vikki Yip
- Bristol Bioresource LaboratoriesPopulation Health ScienceBristol Medical SchoolUniversity of BristolBristolUK
| | - Nicholas J. Timpson
- MRC Integrative Epidemiology Unit at the University of BristolBristolUK
- Population Health ScienceBristol Medical SchoolUniversity of BristolBristolUK
| | - Laura J. Corbin
- MRC Integrative Epidemiology Unit at the University of BristolBristolUK
- Population Health ScienceBristol Medical SchoolUniversity of BristolBristolUK
| |
Collapse
|
185
|
Zhang H, Xiang L, Huo M, Wu Y, Yu M, Lau CW, Tian D, Gou L, Huang Y, Luo JY, Wang L, Song W, Huang J, Cai Z, Chen S, Tian XY, Huang Y. Branched-chain amino acid supplementation impairs insulin sensitivity and promotes lipogenesis during exercise in diet-induced obese mice. Obesity (Silver Spring) 2022; 30:1205-1218. [PMID: 35357085 DOI: 10.1002/oby.23394] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 01/07/2022] [Accepted: 01/10/2022] [Indexed: 12/14/2022]
Abstract
OBJECTIVE Branched-chain amino acids (BCAAs) are popular dietary supplements for exercise. However, increased BCAA levels positively correlate with obesity and diabetes. The metabolic impact of BCAA supplementation on insulin sensitivity during exercise is less understood. METHODS Male C57BL/6 mice were fed for 12 weeks with a high-fat diet, normal chow diet, or BCAA-restricted high-fat diet. They were subjected to running exercise with or without BCAA treatment for another 12 weeks. RESULTS Exercise reduced body weight, improved insulin sensitivity, lowered BCAAs in plasma, and inhibited the upregulation of BCAAs and metabolites caused by BCAA supplementation in the subcutaneous white adipose tissue (sWAT) of obese mice. BCAA supplementation reversed insulin sensitivity ameliorated by exercise. The phosphorylation of protein kinase B (Ser473 and Ser474) was decreased by BCAAs in the sWAT of obese mice. However, BCAA supplementation had no such effects in lean mice. BCAAs also increased the expression of fatty acid synthase and other lipogenesis genes in the sWAT of exercised obese mice. BCAA restriction had no effect on body weight and insulin sensitivity in obese mice. CONCLUSIONS BCAA supplementation impaired the beneficial effect of exercise on glycolipid metabolism in obese but not lean mice. Caution should be taken regarding the use of BCAAs for individuals with obesity who exercise.
Collapse
Affiliation(s)
- Hongsong Zhang
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
- School of Biomedical Sciences, Chinese University of Hong Kong, Hong Kong, China
| | - Li Xiang
- State Key Laboratory of Environmental and Biological Analysis, Department of Chemistry, Hong Kong Baptist University, Hong Kong, China
| | - Mingyu Huo
- School of Biomedical Sciences, Chinese University of Hong Kong, Hong Kong, China
| | - Yalan Wu
- School of Biomedical Sciences, Chinese University of Hong Kong, Hong Kong, China
| | - Mingyang Yu
- School of Biomedical Sciences, Chinese University of Hong Kong, Hong Kong, China
| | - Chi Wai Lau
- School of Biomedical Sciences, Chinese University of Hong Kong, Hong Kong, China
| | - Danyang Tian
- School of Biomedical Sciences, Chinese University of Hong Kong, Hong Kong, China
| | - Lingshan Gou
- School of Biomedical Sciences, Chinese University of Hong Kong, Hong Kong, China
| | - Yuhong Huang
- School of Biomedical Sciences, Chinese University of Hong Kong, Hong Kong, China
| | - Jiang-Yun Luo
- School of Biomedical Sciences, Chinese University of Hong Kong, Hong Kong, China
| | - Li Wang
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
| | - Wencong Song
- School of Biomedical Sciences, Chinese University of Hong Kong, Hong Kong, China
| | - Juan Huang
- School of Biomedical Sciences, Chinese University of Hong Kong, Hong Kong, China
| | - Zongwei Cai
- State Key Laboratory of Environmental and Biological Analysis, Department of Chemistry, Hong Kong Baptist University, Hong Kong, China
| | - Shaoliang Chen
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xiao Yu Tian
- School of Biomedical Sciences, Chinese University of Hong Kong, Hong Kong, China
| | - Yu Huang
- School of Biomedical Sciences, Chinese University of Hong Kong, Hong Kong, China
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
| |
Collapse
|
186
|
Involvement of Gut Microbial Metabolites Derived from Diet on Host Energy Homeostasis. Int J Mol Sci 2022; 23:ijms23105562. [PMID: 35628369 PMCID: PMC9146040 DOI: 10.3390/ijms23105562] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 05/14/2022] [Accepted: 05/15/2022] [Indexed: 12/12/2022] Open
Abstract
Due to the excess energy intake, which is a result of a high fat and high carbohydrate diet, dysfunction of energy balance leads to metabolic disorders such as obesity and type II diabetes mellitus (T2DM). Since obesity can be a risk factor for various diseases, including T2DM, hypertension, hyperlipidemia, and metabolic syndrome, novel prevention and treatment are expected. Moreover, host diseases linked to metabolic disorders are associated with changes in gut microbiota profile. Gut microbiota is affected by diet, and nutrients are used as substrates by gut microbiota for produced metabolites, such as short-chain and long-chain fatty acids, that may modulate host energy homeostasis. These free fatty acids are not only essential energy sources but also signaling molecules via G-protein coupled receptors (GPCRs). Some GPCRs are critical for metabolic functions, such as hormone secretion and immune function in various types of cells and tissues and contribute to energy homeostasis. The current studies have shown that GPCRs for gut microbial metabolites improved host energy homeostasis and systemic metabolic disorders. Here, we will review the association between diet, gut microbiota, and host energy homeostasis.
Collapse
|
187
|
Lian N, Luo K, Xie H, Kang Y, Tang K, Lu P, Li T. Obesity by High-Fat Diet Increases Pain Sensitivity by Reprogramming Branched-Chain Amino Acid Catabolism in Dorsal Root Ganglia. Front Nutr 2022; 9:902635. [PMID: 35634382 PMCID: PMC9133809 DOI: 10.3389/fnut.2022.902635] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 04/26/2022] [Indexed: 11/13/2022] Open
Abstract
Obesity is a significant health concern as a result of poor-quality diet, for example, high-fat diet (HFD). Although multiple biological and molecular changes have been identified to contribute to HFD-induced pain susceptibility, the mechanisms are not fully understood. Here, we show that mice under 8 weeks of HFD were sensitive to mechanical and thermal stimuli, which was coupled with an accumulation of branched-chain amino acids (BCAAs) in lumbar dorsal root ganglia (DRG) due to local BCAA catabolism deficiency. This HFD-induced hyperalgesic phenotype could be exacerbated by supply of excessive BCAAs or mitigated by promotion of BCAA catabolism via BT2 treatment. In addition, our results suggested that HFD-related pain hypersensitivity was associated with a pro-inflammatory status in DRG, which could be regulated by BCAA abundance. Therefore, our study demonstrates that defective BCAA catabolism in DRG facilitates HFD-induced pain hypersensitivity by triggering inflammation. These findings not only reveal metabolic underpinnings for the pathogenesis of HFD-related hyperalgesia but also offer potential targets for developing diet-based therapy of chronic pain.
Collapse
Affiliation(s)
- Nan Lian
- Department of Anesthesiology, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital of Sichuan University, Chengdu, China
- Laboratory of Mitochondria and Metabolism, West China Hospital of Sichuan University, Chengdu, China
- Department of Radiology, Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital of Sichuan University, Chengdu, China
| | - Kaiteng Luo
- Department of Anesthesiology, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital of Sichuan University, Chengdu, China
- Laboratory of Mitochondria and Metabolism, West China Hospital of Sichuan University, Chengdu, China
| | - Huijing Xie
- Department of Anesthesiology, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital of Sichuan University, Chengdu, China
- Laboratory of Mitochondria and Metabolism, West China Hospital of Sichuan University, Chengdu, China
| | - Yi Kang
- Department of Anesthesiology, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital of Sichuan University, Chengdu, China
- Laboratory of Mitochondria and Metabolism, West China Hospital of Sichuan University, Chengdu, China
| | - Kuo Tang
- Department of Anesthesiology, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital of Sichuan University, Chengdu, China
- Laboratory of Mitochondria and Metabolism, West China Hospital of Sichuan University, Chengdu, China
| | - Peilin Lu
- Department of Anesthesiology, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital of Sichuan University, Chengdu, China
- Laboratory of Mitochondria and Metabolism, West China Hospital of Sichuan University, Chengdu, China
- Peilin Lu,
| | - Tao Li
- Department of Anesthesiology, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital of Sichuan University, Chengdu, China
- Laboratory of Mitochondria and Metabolism, West China Hospital of Sichuan University, Chengdu, China
- *Correspondence: Tao Li,
| |
Collapse
|
188
|
Peng ML, Zhang Z, Zhou M, He C, Xiao L, Yin H, Zhao K. Identification of differential metabolites using untargeted metabolomics between gestational diabetes and normal pregnant women. Int J Gynaecol Obstet 2022; 159:903-911. [PMID: 35514238 DOI: 10.1002/ijgo.14253] [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: 10/23/2021] [Revised: 02/26/2022] [Accepted: 05/03/2022] [Indexed: 11/08/2022]
Abstract
OBJECTIVE To study the metabonomics differences between pregnant women with gestational diabetes mellitus (GDM) in the third trimester and those in a group without GDM by screening a group of highly efficient and sensitive markers for GDM and validating previously published early metabolic markers of GDM. METHODS A cross-sectional cohort study based on ultra performance liquid chromatography tandem mass spectrometry untargeted metabolomics analysis of serum samples collected from 59 pregnant women with GDM and 59 pregnant women without GDM. RESULTS A total of 121 metabolites were detected, and 27 were identified as differential metabolites between GDM and control. The combination of 27 metabolic peaks had area under curve (AUC) values of 0.90, 0.92, and 0.93 in the prediction models using support vector machine, partial least squares, and random forest, respectively. Finally, five metabolite biomarkers were selected to construct logistic regression models: L-valine, hypoxanthine, eicosapentaenoic acid, 2-amino-1,3,4-octadecanotriol, and choline. The AUC value of these metabolites was 0.769 between the GDM group and the control group. CONCLUSIONS The discovery of a group of differential metabolites in pregnant women with GDM in the third trimester and in pregnant women without GDM may facilitate the study of the pathologic mechanism of GDM; it may be possible to find an efficient and sensitive alternative GDM detection method.
Collapse
Affiliation(s)
- Mei Lin Peng
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zheng Zhang
- Central China Normal University, School of Life Sciences, Wuhan, China.,Wuhan Prevention and Treatment Center for Occupational Diseases, Wuhan, China
| | - Minqi Zhou
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chao He
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lin Xiao
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Heng Yin
- Department of Obstetrics, Maternal and Child Health Hospital of Hubei Province, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kai Zhao
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
189
|
Perng W, Hivert MF, Michelotti G, Oken E, Dabelea D. Metabolomic Predictors of Dysglycemia in Two U.S. Youth Cohorts. Metabolites 2022; 12:404. [PMID: 35629908 PMCID: PMC9147862 DOI: 10.3390/metabo12050404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 04/26/2022] [Accepted: 04/28/2022] [Indexed: 01/27/2023] Open
Abstract
Here, we seek to identify metabolite predictors of dysglycemia in youth. In the discovery analysis among 391 youth in the Exploring Perinatal Outcomes among CHildren (EPOCH) cohort, we used reduced rank regression (RRR) to identify sex-specific metabolite predictors of impaired fasting glucose (IFG) and elevated fasting glucose (EFG: Q4 vs. Q1 fasting glucose) 6 years later and compared the predictive capacity of four models: Model 1: ethnicity, parental diabetes, in utero exposure to diabetes, and body mass index (BMI); Model 2: Model 1 covariates + baseline waist circumference, insulin, lipids, and Tanner stage; Model 3: Model 2 + baseline fasting glucose; Model 4: Model 3 + baseline metabolite concentrations. RRR identified 19 metabolite predictors of fasting glucose in boys and 14 metabolite predictors in girls. Most compounds were on lipid, amino acid, and carbohydrate metabolism pathways. In boys, no improvement in aurea under the receiver operating characteristics curve AUC occurred until the inclusion of metabolites in Model 4, which increased the AUC for prediction of IFG (7.1%) from 0.81 to 0.97 (p = 0.002). In girls, %IFG was too low for regression analysis (3.1%), but we found similar results for EFG. We replicated the results among 265 youth in the Project Viva cohort, focusing on EFG due to low %IFG, suggesting that the metabolite profiles identified herein have the potential to improve the prediction of glycemia in youth.
Collapse
Affiliation(s)
- Wei Perng
- Lifcourse Epidemiology of Adiposity and Diabetes (LEAD) Center, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Department of Epidemiology, Colorado School of Public Health, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA;
| | - Marie-France Hivert
- Division of Chronic Disease Research Across the Lifecourse (CoRAL), Department of Population Medicine, Harvard Pilgrim Health Care Institute, Harvard Medical School, Boston, MA 02215, USA; (M.-F.H.); (E.O.)
- Diabetes Unit, Massachusetts General Hospital, Boston, MA 02114, USA
| | | | - Emily Oken
- Division of Chronic Disease Research Across the Lifecourse (CoRAL), Department of Population Medicine, Harvard Pilgrim Health Care Institute, Harvard Medical School, Boston, MA 02215, USA; (M.-F.H.); (E.O.)
- Department of Nutrition, T. H. Chan Harvard School of Public Health, Boston, MA 02115, USA
| | - Dana Dabelea
- Department of Epidemiology, Colorado School of Public Health, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA;
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO 80045, USA
| |
Collapse
|
190
|
Lin S, Miao Y, Zheng X, Dong Y, Yang Q, Yang Q, Du S, Xu J, Zhou S, Yuan T. ANGPTL4 negatively regulates the progression of osteosarcoma by remodeling branched-chain amino acid metabolism. Cell Death Dis 2022; 8:225. [PMID: 35461343 PMCID: PMC9035178 DOI: 10.1038/s41420-022-01029-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 04/08/2022] [Accepted: 04/11/2022] [Indexed: 01/18/2023]
Abstract
Angiopoietin-like-4 (ANGPTL4), a secreted glycoprotein that is mainly known as a regulator in lipid metabolism, now, is also indicated to be involved in the regulation of cancer progression and metastasis. However, little is known about not only biological functions, but also underlying mechanism of ANGPTL4 in the progression of osteosarcoma (OS). Here, we discovered that ANGPTL4 is downregulated in OS, and is associated with branched-chain amino acid (BCAA) metabolism. The BCAAs (valine, leucine, and isoleucine) are essential amino acids that play an important role in metabolic regulation. Aberrant BCAA metabolism is also found in various cancers and is associated with tumor progression, including proliferation, invasion, and metastasis. In this study, we indicated that the negative relation between the expression of ANGPTL4 and BCAA catabolism in OS samples and cell lines. The knockdown of ANGPTL4 in OS cells resulted in the accumulation of BCAAs, which in turn activated the mTOR signaling pathway, enhancing OS cell proliferation. Thus, reduced expression of ANGPTL4 is associated with the progression of OS. Taken together, our results demonstrated that the ANGPTL4/BCAA/mTOR axis is an important pathway in OS progression and may be a potential therapeutic target to slow OS progression.
Collapse
|
191
|
Type 2 Diabetes Induced by Changes in Proteomic Profiling of Zebrafish Chronically Exposed to a Mixture of Organochlorine Pesticides at Low Concentrations. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:ijerph19094991. [PMID: 35564385 PMCID: PMC9100612 DOI: 10.3390/ijerph19094991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 04/07/2022] [Accepted: 04/15/2022] [Indexed: 02/04/2023]
Abstract
Effect of organochlorine pesticides (OCPs) mixtures on development of type 2 diabetes mellitus (T2DM) and the underlying mechanism, especially at protein levels, are largely unknown. We exposed a mixture of five OCPs to zebrafish at concentrations of 0, 0.05, 0.25, 2.5, and 25 μg/L for 12 weeks. Differentially expressed proteins (DEPs) were quantitatively identified in female zebrafish livers, and its functional study was conducted. The significantly high glucose and low insulin levels were observed only at 0.05 μg/L, linking to the different pattern of DEPs than other concentrations. A total of 1082 proteins was quantified, of which 321 proteins formed 6 clusters in protein dynamics analysis. The enriched pathways in cluster 3 showing distinct pattern of DEPs could explain the nonlinear response at 0.05 μg/L, indicating that OCP mixtures adversely affected proteins associated with mitochondrial function and energy metabolism. We proposed a feasible mechanism that decrease in expression of aldehyde dehydrogenase led to abnormal accumulation of aldehydes, reducing expression of glyceraldehyde 3-phosphate dehydrogenase, and resulting in disruption of glucose homeostasis. Our findings help to better understand the causality of T2DM by exposure to OCP mixtures and to identify biomarkers in the protein expression level.
Collapse
|
192
|
Metabolomic Analysis of Serum and Tear Samples from Patients with Obesity and Type 2 Diabetes Mellitus. Int J Mol Sci 2022; 23:ijms23094534. [PMID: 35562924 PMCID: PMC9105607 DOI: 10.3390/ijms23094534] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 04/13/2022] [Accepted: 04/14/2022] [Indexed: 12/14/2022] Open
Abstract
Metabolomics strategies are widely used to examine obesity and type 2 diabetes (T2D). Patients with obesity (n = 31) or T2D (n = 26) and sex- and age-matched controls (n = 28) were recruited, and serum and tear samples were collected. The concentration of 23 amino acids and 10 biogenic amines in serum and tear samples was analyzed. Statistical analysis and Pearson correlation analysis along with network analysis were carried out. Compared to controls, changes in the level of 6 analytes in the obese group and of 10 analytes in the T2D group were statistically significant. For obesity, the energy generation, while for T2D, the involvement of NO synthesis and its relation to insulin signaling and inflammation, were characteristic. We found that BCAA and glutamine metabolism, urea cycle, and beta-oxidation make up crucial parts of the metabolic changes in T2D. According to our data, the retromer-mediated retrograde transport, the ethanolamine metabolism, and, consequently, the endocannabinoid signaling and phospholipid metabolism were characteristic of both conditions and can be relevant pathways to understanding and treating insulin resistance. By providing potential therapeutic targets and new starting points for mechanistic studies, our results emphasize the importance of complex data analysis procedures to better understand the pathomechanism of obesity and diabetes.
Collapse
|
193
|
Cuomo P, Capparelli R, Iannelli A, Iannelli D. Role of Branched-Chain Amino Acid Metabolism in Type 2 Diabetes, Obesity, Cardiovascular Disease and Non-Alcoholic Fatty Liver Disease. Int J Mol Sci 2022; 23:ijms23084325. [PMID: 35457142 PMCID: PMC9030262 DOI: 10.3390/ijms23084325] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 04/06/2022] [Accepted: 04/11/2022] [Indexed: 12/17/2022] Open
Abstract
Branched-chain amino acids (BCAAs) include leucine, isoleucine, and valine. Mammalians cannot synthesize these amino acids de novo and must acquire them through their diet. High levels of BCAAs are associated with insulin resistance; type 2 diabetes; obesity; and non-metabolic diseases, including several forms of cancer. BCAAs—in particular leucine—activate the rapamycin complex1 mTORC1, which regulates cell growth and metabolism, glucose metabolism and several more essential physiological processes. Diets rich in BCAAs are associated with metabolic diseases (listed above), while diets low in BCAAs are generally reported to promote metabolic health. As for the dysregulation of the metabolism caused by high levels of BCAAs, recent studies propose that the accumulation of acyl-carnitine and diacyl-CoA in muscles alters lipid metabolism. However, this suggestion is not broadly accepted. On clinical grounds, pre- and post-operative metabolic profiles of candidate patients for bariatric surgery are being used to select the optimal procedure for each individual patient.
Collapse
Affiliation(s)
- Paola Cuomo
- Department of Agricultural Sciences, University of Naples “Federico II”, Via Università, 100-Portici, 80055 Naples, Italy;
| | - Rosanna Capparelli
- Department of Agricultural Sciences, University of Naples “Federico II”, Via Università, 100-Portici, 80055 Naples, Italy;
- Correspondence: (R.C.); (D.I.)
| | - Antonio Iannelli
- Department of Digestive Surgery, Université Côte d’ Azur, F-06108 Nice, France;
- Centre Hospitalier Universitaire de Nice-Digestive Surgery and Liver Transplantation Unit, Archet 2 Hospital, 151 Route de Saint Antoine de Ginestiere, F-062024 Nice, France
- Inserm, U1065, Team 8 “Hepatic Complications of Obesity and Alcohol”, F-062024 Nice, France
| | - Domenico Iannelli
- Department of Agricultural Sciences, University of Naples “Federico II”, Via Università, 100-Portici, 80055 Naples, Italy;
- Correspondence: (R.C.); (D.I.)
| |
Collapse
|
194
|
Liu M, Huang Y, Zhang H, Aitken D, Nevitt MC, Rockel JS, Pelletier JP, Lewis CE, Torner J, Rampersaud YR, Perruccio AV, Mahomed NN, Furey A, Randell EW, Rahman P, Sun G, Martel-Pelletier J, Kapoor M, Jones G, Felson D, Qi D, Zhai G. Restricting Branched-Chain Amino Acids within a High-Fat Diet Prevents Obesity. Metabolites 2022; 12:334. [PMID: 35448521 PMCID: PMC9030079 DOI: 10.3390/metabo12040334] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 04/01/2022] [Accepted: 04/04/2022] [Indexed: 02/01/2023] Open
Abstract
Obesity is a global pandemic, but there is yet no effective measure to control it. Recent metabolomics studies have identified a signature of altered amino acid profiles to be associated with obesity, but it is unclear whether these findings have actionable clinical potential. The aims of this study were to reveal the metabolic alterations of obesity and to explore potential strategies to mitigate obesity. We performed targeted metabolomic profiling of the plasma/serum samples collected from six independent cohorts and conducted an individual data meta-analysis of metabolomics for body mass index (BMI) and obesity. Based on the findings, we hypothesized that restriction of branched-chain amino acids (BCAAs), phenylalanine, or tryptophan may prevent obesity and tested our hypothesis in a dietary restriction trial with eight groups of 4-week-old male C57BL/6J mice (n = 5/group) on eight different types of diets, respectively, for 16 weeks. A total of 3397 individuals were included in the meta-analysis. The mean BMI was 30.7 ± 6.1 kg/m2, and 49% of participants were obese. Fifty-eight metabolites were associated with BMI and obesity (all p ≤ 2.58 × 10-4), linked to alterations of the BCAA, phenylalanine, tryptophan, and phospholipid metabolic pathways. The restriction of BCAAs within a high-fat diet (HFD) maintained the mice's weight, fat and lean volume, subcutaneous and visceral adipose tissue weight, and serum glucose and insulin at levels similar to those in the standard chow group, and prevented obesity, adipocyte hypertrophy, adipose inflammation, and insulin resistance induced by HFD. Our data suggest that four metabolic pathways, BCAA, phenylalanine, tryptophan, and phospholipid metabolic pathways, are altered in obesity and restriction of BCAAs within a HFD can prevent the development of obesity and insulin resistance in mice, providing a promising strategy to potentially mitigate diet-induced obesity.
Collapse
Affiliation(s)
- Ming Liu
- Division of Biomedical Sciences (Genetics), Faculty of Medicine, Memorial University of Newfoundland, St. John’s, NL A1B 3V6, Canada;
| | - Yiheng Huang
- College of Pharmacy, University of Manitoba, Winnipeg, MB R3E 0T5, Canada; (Y.H.); (D.Q.)
| | - Hongwei Zhang
- Discipline of Medicine, Faculty of Medicine, Memorial University of Newfoundland, St. John’s, NL A1B 3V6, Canada; (H.Z.); (P.R.); (G.S.)
| | - Dawn Aitken
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS 7000, Australia; (D.A.); (G.J.)
| | - Michael C. Nevitt
- Department of Epidemiology and Biostatistics, University of California, San Francisco, CA 94158, USA;
| | - Jason S. Rockel
- Osteoarthritis Research Program, Division of Orthopaedics, Schroeder Arthritis Institute, University Health Network, Toronto, ON M5T 2S8, Canada; (J.S.R.); (Y.R.R.); (A.V.P.); (N.N.M.); (M.K.)
| | - Jean-Pierre Pelletier
- Osteoarthritis Research Unit, University of Montreal Hospital Research Centre (CRCHUM), Montreal, QC H2X 0A9, Canada; (J.-P.P.); (J.M.-P.)
| | - Cora E. Lewis
- Department of Epidemiology, University of Alabama, Birmingham, AL 35233, USA;
| | - James Torner
- Department of Epidemiology, University of Iowa, Iowa City, IA 52242, USA;
| | - Yoga Raja Rampersaud
- Osteoarthritis Research Program, Division of Orthopaedics, Schroeder Arthritis Institute, University Health Network, Toronto, ON M5T 2S8, Canada; (J.S.R.); (Y.R.R.); (A.V.P.); (N.N.M.); (M.K.)
| | - Anthony V. Perruccio
- Osteoarthritis Research Program, Division of Orthopaedics, Schroeder Arthritis Institute, University Health Network, Toronto, ON M5T 2S8, Canada; (J.S.R.); (Y.R.R.); (A.V.P.); (N.N.M.); (M.K.)
- Institute of Health Policy, Management and Evaluation, Dalla Lana School of Public Health, University of Toronto, Toronto, ON M5T 2S8, Canada
| | - Nizar N. Mahomed
- Osteoarthritis Research Program, Division of Orthopaedics, Schroeder Arthritis Institute, University Health Network, Toronto, ON M5T 2S8, Canada; (J.S.R.); (Y.R.R.); (A.V.P.); (N.N.M.); (M.K.)
- Department of Surgery, University of Toronto, Toronto, ON M5T 2S8, Canada
| | - Andrew Furey
- Discipline of Surgery, Faculty of Medicine, Memorial University of Newfoundland, St. John’s, NL A1B 3V6, Canada;
- Office of the Premier, Government of Newfoundland and Labrador, St. John’s, NL A1B 4J6, Canada
| | - Edward W. Randell
- Discipline of Laboratory Medicine, Faculty of Medicine, Memorial University of Newfoundland, St. John’s, NL A1B 3V6, Canada;
| | - Proton Rahman
- Discipline of Medicine, Faculty of Medicine, Memorial University of Newfoundland, St. John’s, NL A1B 3V6, Canada; (H.Z.); (P.R.); (G.S.)
| | - Guang Sun
- Discipline of Medicine, Faculty of Medicine, Memorial University of Newfoundland, St. John’s, NL A1B 3V6, Canada; (H.Z.); (P.R.); (G.S.)
| | - Johanne Martel-Pelletier
- Osteoarthritis Research Unit, University of Montreal Hospital Research Centre (CRCHUM), Montreal, QC H2X 0A9, Canada; (J.-P.P.); (J.M.-P.)
| | - Mohit Kapoor
- Osteoarthritis Research Program, Division of Orthopaedics, Schroeder Arthritis Institute, University Health Network, Toronto, ON M5T 2S8, Canada; (J.S.R.); (Y.R.R.); (A.V.P.); (N.N.M.); (M.K.)
| | - Graeme Jones
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS 7000, Australia; (D.A.); (G.J.)
| | - David Felson
- Department of Rheumatology, Boston University School of Medicine, Boston, MA 02118, USA;
- NIHR Manchester Biomedical Research Centre, Manchester Academic Health Science Centre, Manchester University NHS Foundation Trust, Manchester M13 9WL, UK
| | - Dake Qi
- College of Pharmacy, University of Manitoba, Winnipeg, MB R3E 0T5, Canada; (Y.H.); (D.Q.)
| | - Guangju Zhai
- Division of Biomedical Sciences (Genetics), Faculty of Medicine, Memorial University of Newfoundland, St. John’s, NL A1B 3V6, Canada;
| |
Collapse
|
195
|
Dimou A, Tsimihodimos V, Bairaktari E. The Critical Role of the Branched Chain Amino Acids (BCAAs) Catabolism-Regulating Enzymes, Branched-Chain Aminotransferase (BCAT) and Branched-Chain α-Keto Acid Dehydrogenase (BCKD), in Human Pathophysiology. Int J Mol Sci 2022; 23:ijms23074022. [PMID: 35409380 PMCID: PMC8999875 DOI: 10.3390/ijms23074022] [Citation(s) in RCA: 88] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Revised: 03/31/2022] [Accepted: 04/03/2022] [Indexed: 12/26/2022] Open
Abstract
Branched chain amino acids (BCAAs), leucine, isoleucine and valine, are essential amino acids widely studied for their crucial role in the regulation of protein synthesis mainly through the activation of the mTOR signaling pathway and their emerging recognition as players in the regulation of various physiological and metabolic processes, such as glucose homeostasis. BCAA supplementation is primarily used as a beneficial nutritional intervention in chronic liver and kidney disease as well as in muscle wasting disorders. However, downregulated/upregulated plasma BCAAs and their defective catabolism in various tissues, mainly due to altered enzymatic activity of the first two enzymes in their catabolic pathway, BCAA aminotransferase (BCAT) and branched-chain α-keto acid dehydrogenase (BCKD), have been investigated in many nutritional and disease states. The current review focused on the underlying mechanisms of altered BCAA catabolism and its contribution to the pathogenesis of a numerous pathological conditions such as diabetes, heart failure and cancer. In addition, we summarize findings that indicate that the recovery of the dysregulated BCAA catabolism may be associated with an improved outcome and the prevention of serious disease complications.
Collapse
Affiliation(s)
- Aikaterini Dimou
- Laboratory of Clinical Chemistry, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece;
| | - Vasilis Tsimihodimos
- Department of Internal Medicine, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece;
| | - Eleni Bairaktari
- Laboratory of Clinical Chemistry, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece;
- Correspondence: ; Tel.: +30-26510-07620
| |
Collapse
|
196
|
Rivera ME, Rivera CN, Vaughan RA. Excess branched-chain amino acids alter myotube metabolism and substrate preference which is worsened by concurrent insulin resistance. Endocrine 2022; 76:18-28. [PMID: 34811646 DOI: 10.1007/s12020-021-02939-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 11/02/2021] [Indexed: 11/26/2022]
Abstract
PURPOSE Branched-chain amino acids (BCAA) have been shown to enhance several cellular signaling pathways including protein synthesis and mitochondrial biogenesis, yet population data demonstrate a correlation between circulating BCAA and severity of insulin resistance which has been hypothesized to be, in part, a byproduct of BCAA inhibition of mitochondrial function. The purpose of this study is to examine the effect of a BCAA mixture on muscle metabolism and related gene expression in vitro. METHODS C2C12 myotubes were treated with a BCAA mixture containing leucine:isoleucine:valine at a ratio of 2:1:1 at 0.2, 2, or 20 mM (based on leucine content) for 6 days. qRT-PCR was used to measure metabolic gene expression. Oxygen consumption and extracellular acidification were used to assess mitochondrial and glycolytic metabolism, respectively. Mitochondrial content was determined via mitochondrial-specific staining. RESULTS Despite significantly elevated mitochondrial staining, 6-day BCAA treatment reduced basal mitochondrial metabolism at a supraphysiological concentration (20 mM) in both insulin sensitive and resistant cells. Peak mitochondrial capacity was also reduced in insulin-resistant (but not insulin sensitive) cells. Conversely, basal glycolytic metabolism was elevated following 20 mM BCAA treatment, regardless of insulin resistance. In addition, insulin-resistant cells treated with 20 mM BCAA exhibited reduced gene expression of Ppargc1a, Cytc, Atp5b, Glut4, and several glycolytic enzymes versus insulin sensitive cells treated with 20 mM BCAA. CONCLUSIONS Collectively, these findings suggest BCAA at supraphysiologically high levels may negatively alter mitochondrial metabolism, and concurrent insulin resistance may also diminish peak mitochondrial capacity, as well as impede molecular adaptations that support a transition to a glycolytic preference/compensation.
Collapse
Affiliation(s)
- Madison E Rivera
- Department of Exercise Science, High Point University, High Point, NC, USA
| | - Caroline N Rivera
- Department of Exercise Science, High Point University, High Point, NC, USA
| | - Roger A Vaughan
- Department of Exercise Science, High Point University, High Point, NC, USA.
| |
Collapse
|
197
|
Palmer BF, Clegg DJ. Metabolic Flexibility and Its Impact on Health Outcomes. Mayo Clin Proc 2022; 97:761-776. [PMID: 35287953 DOI: 10.1016/j.mayocp.2022.01.012] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 01/04/2022] [Accepted: 01/12/2022] [Indexed: 02/06/2023]
Abstract
A metabolically flexible state exists when there is a rapid switch between glucose and fatty acids during the transition between the fed and fasting state. This flexibility in fuel choice serves to prevent hyperglycemia following a meal and simultaneously ensures an adequate amount of blood glucose is available for delivery to the brain and exclusively glycolytic tissues during fasting. The modern era is characterized by chronic overnutrition in which a mixture of fuels is delivered to the mitochondria in an unabated manner thereby uncoupling the feast and famine situation. The continuous influx of fuel leads to accumulation of reducing equivalents in the mitochondria and an increase in the mitochondrial membrane potential. These changes create a microenvironment fostering the generation of reactive oxygen species and other metabolites leading to deleterious protein modification, cell injury, and ultimately clinical disease. Insulin resistance may also play a primary role in this deleterious effect. The imbalance between mitochondrial energy delivery and use is made worse with a sedentary lifestyle. Maneuvers that restore energy balance across the mitochondria activate pathways that remove or repair damaged molecules and restore the plasticity characteristic of normal energy metabolism. Readily available strategies to maintain energy balance across the mitochondria include exercise, various forms of caloric restriction, administration of sodium-glucose cotransporter-2 inhibitors, cold exposure, and hypobaric hypoxia.
Collapse
Affiliation(s)
- Biff F Palmer
- Department of Medicine, Division of Nephrology, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| | | |
Collapse
|
198
|
Cataldi S, Bonavolontà V, Poli L, Clemente FM, De Candia M, Carvutto R, Silva AF, Badicu G, Greco G, Fischetti F. The Relationship between Physical Activity, Physical Exercise, and Human Gut Microbiota in Healthy and Unhealthy Subjects: A Systematic Review. BIOLOGY 2022; 11:479. [PMID: 35336852 PMCID: PMC8945171 DOI: 10.3390/biology11030479] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 03/18/2022] [Accepted: 03/20/2022] [Indexed: 02/06/2023]
Abstract
Several studies have been conducted to find at least an association between physical activity (PA)/ physical exercise (PE) and the possibility to modulate the gut microbiome (GM). However, the specific effects produced on the human GM by different types of PA/PE, different training modalities, and their age-related effects are not yet fully understood. Therefore, this systematic review aims to evaluate and summarize the current scientific evidence investigating the bi-directional relationship between PA/PE and the human GM, with a specific focus on the different types/variables of PA/PE and age-related effects, in healthy and unhealthy people. A systematic search was conducted across four databases (Web of Science, Medline (PubMed), Google Scholar, and Cochrane Library). Information was extracted using the populations, exposure, intervention, comparison, outcomes (PICOS) format. The Oxford Quality Scoring System Scale, the Risk of Bias in Non-Randomized Studies of Interventions (ROBINS-I) tool, and the JBI Critical Appraisal Checklist for Analytical Cross-Sectional Studies were used as a qualitative measure of the review. The protocol was registered in PROSPERO (code: CRD42022302725). The following data items were extracted: author, year of publication, study design, number and age of participants, type of PA/PE carried out, protocol/workload and diet assessment, duration of intervention, measurement tools used, and main outcomes. Two team authors reviewed 694 abstracts for inclusion and at the end of the screening process, only 76 full texts were analyzed. Lastly, only 25 research articles met the eligibility criteria. The synthesis of these findings suggests that GM diversity is associated with aerobic exercise contrary to resistance training; abundance of Prevotella genus seems to be correlated with training duration; no significant change in GM richness and diversity are detected when exercising according to the minimum dose recommended by the World Health Organizations; intense and prolonged PE can induce a higher abundance of pro-inflammatory bacteria; PA does not lead to significant GM α/β-diversity in elderly people (60+ years). The heterogeneity of the training parameters used in the studies, diet control, and different sequencing methods are the main confounders. Thus, this systematic review can provide an in-depth overview of the relationship between PA/PE and the human intestinal microbiota and, at the same time, provide indications from the athletic and health perspective.
Collapse
Affiliation(s)
- Stefania Cataldi
- Department of Basic Medical Sciences, Neuroscience and Sense Organs, University of Study of Bari, 70124 Bari, Italy; (S.C.); (V.B.); (L.P.); (M.D.C.); (R.C.); (F.F.)
| | - Valerio Bonavolontà
- Department of Basic Medical Sciences, Neuroscience and Sense Organs, University of Study of Bari, 70124 Bari, Italy; (S.C.); (V.B.); (L.P.); (M.D.C.); (R.C.); (F.F.)
| | - Luca Poli
- Department of Basic Medical Sciences, Neuroscience and Sense Organs, University of Study of Bari, 70124 Bari, Italy; (S.C.); (V.B.); (L.P.); (M.D.C.); (R.C.); (F.F.)
| | - Filipe Manuel Clemente
- Escola Superior Desporto e Lazer, Instituto Politécnico de Viana do Castelo, Rua Escola Industrial e Comercial de Nun’Álvares, 4900-347 Viana do Castelo, Portugal; (F.M.C.); (A.F.S.)
- Research Center in Sports Performance, Recreation, Innovation and Technology (SPRINT), 4960-320 Melgaço, Portugal
- Instituto de Telecomunicações, Delegação da Covilhã, 1049-001 Lisboa, Portugal
| | - Michele De Candia
- Department of Basic Medical Sciences, Neuroscience and Sense Organs, University of Study of Bari, 70124 Bari, Italy; (S.C.); (V.B.); (L.P.); (M.D.C.); (R.C.); (F.F.)
| | - Roberto Carvutto
- Department of Basic Medical Sciences, Neuroscience and Sense Organs, University of Study of Bari, 70124 Bari, Italy; (S.C.); (V.B.); (L.P.); (M.D.C.); (R.C.); (F.F.)
| | - Ana Filipa Silva
- Escola Superior Desporto e Lazer, Instituto Politécnico de Viana do Castelo, Rua Escola Industrial e Comercial de Nun’Álvares, 4900-347 Viana do Castelo, Portugal; (F.M.C.); (A.F.S.)
- Research Center in Sports Performance, Recreation, Innovation and Technology (SPRINT), 4960-320 Melgaço, Portugal
- The Research Centre in Sports Sciences, Health Sciences and Human Development (CIDESD), 5001-801 Vila Real, Portugal
| | - Georgian Badicu
- Department of Physical Education and Special Motricity, Transilvania University of Brasov, 500068 Brasov, Romania;
| | - Gianpiero Greco
- Department of Basic Medical Sciences, Neuroscience and Sense Organs, University of Study of Bari, 70124 Bari, Italy; (S.C.); (V.B.); (L.P.); (M.D.C.); (R.C.); (F.F.)
| | - Francesco Fischetti
- Department of Basic Medical Sciences, Neuroscience and Sense Organs, University of Study of Bari, 70124 Bari, Italy; (S.C.); (V.B.); (L.P.); (M.D.C.); (R.C.); (F.F.)
| |
Collapse
|
199
|
Mashiko T, Tsukada K, Takada H, Wu SH, Kanayama K, Asahi R, Mori M, Kurisaki A, Oka S, Yoshimura K. Genetic and cytometric analyses of subcutaneous adipose tissue in patients with hemophilia and HIV-associated lipodystrophy. AIDS Res Ther 2022; 19:14. [PMID: 35246167 PMCID: PMC8895510 DOI: 10.1186/s12981-022-00432-9] [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: 08/21/2021] [Accepted: 01/31/2022] [Indexed: 11/15/2022] Open
Abstract
Background The authors recently performed plastic surgeries for a small number of patients with hemophilia, HIV infection, and morphologic evidence of lipodystrophy. Because the pathophysiological mechanism of HIV-associated lipodystrophy remains to be elucidated, we analyzed subcutaneous adipose tissues from the patients. Methods All six patients had previously been treated with older nucleoside analogue reverse-transcriptase inhibitors (NRTIs; stavudine, didanosine or zidovudine). Abdominal and inguinal subcutaneous fat samples were obtained from the HIV+ patients with hemophilia and HIV− healthy volunteers (n = 6 per group), and analyzed via DNA microarray, real-time PCR, flow cytometry and immunohistochemistry. Results The time from initial NRTI treatment to collecting samples were 21.7 years in average. Cytometric analysis revealed infiltration of inflammatory M1 macrophages into HIV-infected adipose tissue and depletion of adipose-derived stem cells, possibly due to exhaustion following sustained adipocyte death. Genetic analysis revealed that adipose tissue from HIV+ group had increased immune activation, mitochondrial toxicity, chronic inflammation, progressive fibrosis and adipocyte dysfunction (e.g. insulin resistance, inhibited adipocyte differentiation and accelerated apoptosis). Of note, both triglyceride synthesis and lipolysis were inhibited in adipose tissue from patients with HIV. Conclusions Our findings provide important insights into the pathogenesis of HIV-associated lipodystrophy, suggesting that fat redistribution may critically depend on adipocytes’ sensitivity to drug-induced mitochondrial toxicity, which may lead either to atrophy or metabolic complications. Supplementary Information The online version contains supplementary material available at 10.1186/s12981-022-00432-9.
Collapse
|
200
|
Nichols CG, York NW, Remedi MS. ATP-Sensitive Potassium Channels in Hyperinsulinism and Type 2 Diabetes: Inconvenient Paradox or New Paradigm? Diabetes 2022; 71:367-375. [PMID: 35196393 PMCID: PMC8893938 DOI: 10.2337/db21-0755] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 11/28/2021] [Indexed: 11/13/2022]
Abstract
Secretion of insulin from pancreatic β-cells is complex, but physiological glucose-dependent secretion is dominated by electrical activity, in turn controlled by ATP-sensitive potassium (KATP) channel activity. Accordingly, loss-of-function mutations of the KATP channel Kir6.2 (KCNJ11) or SUR1 (ABCC8) subunit increase electrical excitability and secretion, resulting in congenital hyperinsulinism (CHI), whereas gain-of-function mutations cause underexcitability and undersecretion, resulting in neonatal diabetes mellitus (NDM). Thus, diazoxide, which activates KATP channels, and sulfonylureas, which inhibit KATP channels, have dramatically improved therapies for CHI and NDM, respectively. However, key findings do not fit within this simple paradigm: mice with complete absence of β-cell KATP activity are not hyperinsulinemic; instead, they are paradoxically glucose intolerant and prone to diabetes, as are older human CHI patients. Critically, despite these advances, there has been little insight into any role of KATP channel activity changes in the development of type 2 diabetes (T2D). Intriguingly, the CHI progression from hypersecretion to undersecretion actually mirrors the classical response to insulin resistance in the progression of T2D. In seeking to explain the progression of CHI, multiple lines of evidence lead us to propose that underlying mechanisms are also similar and that development of T2D may involve loss of KATP activity.
Collapse
Affiliation(s)
- Colin G Nichols
- Center for the Investigation of Membrane Excitability Diseases, Washington University School of Medicine, St. Louis, MO
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO
| | - Nathaniel W York
- Center for the Investigation of Membrane Excitability Diseases, Washington University School of Medicine, St. Louis, MO
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO
| | - Maria S Remedi
- Center for the Investigation of Membrane Excitability Diseases, Washington University School of Medicine, St. Louis, MO
- Division of Endocrinology Metabolism and Lipid Research, Department of Medicine, Washington University School of Medicine, St. Louis, MO
| |
Collapse
|