1
|
Leite JSM, Vilas-Boas EA, Takahashi HK, Munhoz AC, Araújo LCC, Carvalho CR, Jr JD, Curi R, Carpinelli AR, Cruzat V. Liver lipid metabolism, oxidative stress, and inflammation in glutamine-supplemented ob/ob mice. J Nutr Biochem 2025; 138:109842. [PMID: 39824260 DOI: 10.1016/j.jnutbio.2025.109842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 12/21/2024] [Accepted: 01/13/2025] [Indexed: 01/20/2025]
Abstract
Glutamine availability may be reduced in chronic diseases, such as type 2 diabetes mellitus (T2DM)-induced by obesity. Herein, the antioxidant, anti-inflammatory and lipid metabolism effects of chronic oral glutamine supplementation in its free and dipeptide form were assessed in ob/ob mice. Adult male C57BL/6J ob/ob mice were supplemented with L-alanyl-L-glutamine (DIP) or free L-glutamine (GLN) in the drinking water for 40 days, whilst C57BL/6J Wild-type lean (WT) and control ob/ob mice (CTRL) received fresh water only. Plasma and tissue (skeletal muscle and liver) glutamine levels, and insulin resistance parameters (e.g., GTT, ITT, insulin) were determined. Oxidative stress (e.g., GSH system, Nrf2 translocation), inflammatory (e.g., NFkB translocation, TNF-α gene expression) and lipid metabolism parameters (e.g., plasma and liver triglyceride levels, SRBP-1, FAS, ACC, and ChRBP gene expression) were also analyzed. CTRL ob/ob mice showed lower glutamine levels in plasma and tissue, as well as increased insulin resistance and fat in the liver. Conversely, chronic DIP supplementation restored glutamine levels in plasma and tissues, improved glucose homeostasis and reduced plasma and liver lipid levels. Also, Nrf2 restoration, reduced NFkB translocation, and lower TNF-α gene expression was observed in the DIP group. Interestingly, chronic free GLN only increased muscle glutamine stores but reduced overall insulin resistance, and attenuated plasma and liver lipid metabolic biomarkers. The results presented herein indicate that restoration of body glutamine levels reduces oxidative stress and inflammation in obese and T2DM ob/ob mice. This effect attenuated hepatic lipid metabolic changes observed in obesity.
Collapse
Affiliation(s)
- Jaqueline Santos Moreira Leite
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, São Paulo, Brazil
| | - Eloisa Aparecida Vilas-Boas
- Department of Clinical and Toxicological Analysis, Faculty of Pharmaceutical Sciences, University of Sao Paulo, São Paulo, São Paulo, Brazil
| | - Hilton K Takahashi
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, São Paulo, Brazil
| | - Ana Cláudia Munhoz
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, São Paulo, Brazil
| | - Layanne C C Araújo
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, São Paulo, Brazil
| | - Carla Roberta Carvalho
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, São Paulo, Brazil
| | - Jose Donato Jr
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, São Paulo, Brazil
| | - Rui Curi
- Interdisciplinary Post-graduate Program in Health Sciences, ICAFE, Universidade Cruzeiro do Sul, São Paulo, São Paulo, Brazil; Instituto Butantan, São Paulo, São Paulo, Brazil
| | - Angelo Rafael Carpinelli
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, São Paulo, Brazil
| | - Vinicius Cruzat
- Faculty of Health, Southern Cross University, Gold Coast, Queensland, Australia.
| |
Collapse
|
2
|
Martínez-González MA, Planes FJ, Ruiz-Canela M, Toledo E, Estruch R, Salas-Salvadó J, Valdés-Más R, Mena P, Castañer O, Fitó M, Clish C, Landberg R, Wittenbecher C, Liang L, Guasch-Ferré M, Lamuela-Raventós RM, Wang DD, Forouhi N, Razquin C, Hu FB. Recent advances in precision nutrition and cardiometabolic diseases. REVISTA ESPANOLA DE CARDIOLOGIA (ENGLISH ED.) 2025; 78:263-271. [PMID: 39357800 PMCID: PMC11875914 DOI: 10.1016/j.rec.2024.09.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Accepted: 09/17/2024] [Indexed: 10/04/2024]
Abstract
A growing body of research on nutrition omics has led to recent advances in cardiovascular disease epidemiology and prevention. Within the PREDIMED trial, significant associations between diet-related metabolites and cardiovascular disease were identified, which were subsequently replicated in independent cohorts. Some notable metabolites identified include plasma levels of ceramides, acyl-carnitines, branched-chain amino acids, tryptophan, urea cycle pathways, and the lipidome. These metabolites and their related pathways have been associated with incidence of both cardiovascular disease and type 2 diabetes. Future directions in precision nutrition research include: a) developing more robust multimetabolomic scores to predict long-term risk of cardiovascular disease and mortality; b) incorporating more diverse populations and a broader range of dietary patterns; and c) conducting more translational research to bridge the gap between precision nutrition studies and clinical applications.
Collapse
Affiliation(s)
- Miguel A Martínez-González
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Spain; Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Navarra, Spain; Universidad de Navarra, Departamento de Medicina Preventiva y Salud Pública, Pamplona, Navarra, Spain; Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States.
| | - Francisco J Planes
- Tecnun Escuela de Ingeniería, Departamento de Ingeniería Biomédica y Ciencias, Universidad de Navarra, San Sebastián, Guipúzcoa, Spain
| | - Miguel Ruiz-Canela
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Spain; Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Navarra, Spain; Universidad de Navarra, Departamento de Medicina Preventiva y Salud Pública, Pamplona, Navarra, Spain
| | - Estefanía Toledo
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Spain; Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Navarra, Spain; Universidad de Navarra, Departamento de Medicina Preventiva y Salud Pública, Pamplona, Navarra, Spain
| | - Ramón Estruch
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Spain; Departamento de Medicina Interna, Instituto de Investigaciones Biomédicas August Pi Sunyer (IDIBAPS), Hospital Clínico, Universidad de Barcelona, Barcelona, Spain
| | - Jordi Salas-Salvadó
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Spain; Instituto de Investigación Sanitaria Pere i Virgili, Departamento de Bioquímica y Biotecnología, Unidad de Nutrición Humana Universidad Rovira i Virgili, Reus, Tarragona, Spain
| | - Rafael Valdés-Más
- Immunology Department, Weizmann Institute of Science, Rehovot, Israel
| | - Pedro Mena
- Dipartimento di Scienze degli Alimenti e del Farmaco, Universitá di Parma, Parma, Italy
| | - Olga Castañer
- Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Spain
| | - Montse Fitó
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Spain; Unidad de Riesgo Cardiovascular y Nutrición, Instituto Hospital del Mar de Investigaciones Médicas (IMIM), Barcelona, Spain
| | - Clary Clish
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States
| | - Rikard Landberg
- Division of Food and Nutrition Science, Department of Life Sciences, Chalmers University of Technology, Gothenburg, Sweden
| | - Clemens Wittenbecher
- Department of Life Sciences, SciLifeLab, Chalmers University of Technology, Gothenburg, Sweden
| | - Liming Liang
- Departments of Epidemiology and Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States
| | - Marta Guasch-Ferré
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States; Department of Public Health and Novo Nordisk Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Rosa M Lamuela-Raventós
- Grup de recerca antioxidants naturals: polifenols, Departament de Nutrició, Ciències de l'Alimentació i Gastronomia, Facultat de Farmàcia, Universitat de Barcelona, Barcelona, Spain; Institut de Nutrició i Seguretat Alimentària (INSA), Universitat de Barcelona (UB), Barcelona, Spain
| | - Dong D Wang
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States; Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States; Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, United States
| | - Nita Forouhi
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom
| | - Cristina Razquin
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Spain; Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Navarra, Spain; Universidad de Navarra, Departamento de Medicina Preventiva y Salud Pública, Pamplona, Navarra, Spain
| | - Frank B Hu
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States; Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, United States
| |
Collapse
|
3
|
Martínez-González MA, Planes FJ, Ruiz-Canela M, Toledo E, Estruch R, Salas-Salvadó J, Valdés-Más R, Mena P, Castañer O, Fitó M, Clish C, Landberg R, Wittenbecher C, Liang L, Guasch-Ferré M, Lamuela-Raventós RM, Wang DD, Forouhi N, Razquin C, Hu FB. Recent advances in precision nutrition and cardiometabolic diseases. Rev Esp Cardiol 2025; 78:263-271. [PMID: 39357800 DOI: 10.1016/j.recesp.2024.09.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Accepted: 09/17/2024] [Indexed: 01/11/2025]
Abstract
A growing body of research on nutrition omics has led to recent advances in cardiovascular disease epidemiology and prevention. Within the PREDIMED trial, significant associations between diet-related metabolites and cardiovascular disease were identified, which were subsequently replicated in independent cohorts. Some notable metabolites identified include plasma levels of ceramides, acyl-carnitines, branched-chain amino acids, tryptophan, urea cycle pathways, and the lipidome. These metabolites and their related pathways have been associated with incidence of both cardiovascular disease and type 2 diabetes. Future directions in precision nutrition research include: a) developing more robust multimetabolomic scores to predict long-term risk of cardiovascular disease and mortality; b) incorporating more diverse populations and a broader range of dietary patterns; and c) conducting more translational research to bridge the gap between precision nutrition studies and clinical applications.
Collapse
Affiliation(s)
- Miguel A Martínez-González
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Spain; Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Navarra, Spain; Universidad de Navarra, Departamento de Medicina Preventiva y Salud Pública, Pamplona, Navarra, Spain; Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States.
| | - Francisco J Planes
- Tecnun Escuela de Ingeniería, Departamento de Ingeniería Biomédica y Ciencias, Universidad de Navarra, San Sebastián, Guipúzcoa, Spain
| | - Miguel Ruiz-Canela
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Spain; Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Navarra, Spain; Universidad de Navarra, Departamento de Medicina Preventiva y Salud Pública, Pamplona, Navarra, Spain
| | - Estefanía Toledo
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Spain; Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Navarra, Spain; Universidad de Navarra, Departamento de Medicina Preventiva y Salud Pública, Pamplona, Navarra, Spain
| | - Ramón Estruch
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Spain; Departamento de Medicina Interna, Instituto de Investigaciones Biomédicas August Pi Sunyer (IDIBAPS), Hospital Clínico, Universidad de Barcelona, Barcelona, Spain
| | - Jordi Salas-Salvadó
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Spain; Instituto de Investigación Sanitaria Pere i Virgili, Departamento de Bioquímica y Biotecnología, Unidad de Nutrición Humana Universidad Rovira i Virgili, Reus, Tarragona, Spain
| | - Rafael Valdés-Más
- Immunology Department, Weizmann Institute of Science, Rehovot, Israel
| | - Pedro Mena
- Dipartimento di Scienze degli Alimenti e del Farmaco, Universitá di Parma, Parma, Italy
| | - Olga Castañer
- Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Spain
| | - Montse Fitó
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Spain; Unidad de Riesgo Cardiovascular y Nutrición, Instituto Hospital del Mar de Investigaciones Médicas (IMIM), Barcelona, Spain
| | - Clary Clish
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States
| | - Rikard Landberg
- Division of Food and Nutrition Science, Department of Life Sciences, Chalmers University of Technology, Gothenburg, Sweden
| | - Clemens Wittenbecher
- Department of Life Sciences, SciLifeLab, Chalmers University of Technology, Gothenburg, Sweden
| | - Liming Liang
- Departments of Epidemiology and Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States
| | - Marta Guasch-Ferré
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States; Department of Public Health and Novo Nordisk Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Rosa M Lamuela-Raventós
- Grup de recerca antioxidants naturals: polifenols, Departament de Nutrició, Ciències de l'Alimentació i Gastronomia, Facultat de Farmàcia, Universitat de Barcelona, Barcelona, Spain; Institut de Nutrició i Seguretat Alimentària (INSA), Universitat de Barcelona (UB), Barcelona, Spain
| | - Dong D Wang
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States; Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States; Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, United States
| | - Nita Forouhi
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom
| | - Cristina Razquin
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Spain; Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Navarra, Spain; Universidad de Navarra, Departamento de Medicina Preventiva y Salud Pública, Pamplona, Navarra, Spain
| | - Frank B Hu
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States; Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, United States
| |
Collapse
|
4
|
Hunter B, Li M, Parker BL, Koay YC, Harney DJ, Pearson E, Cao J, Chen GT, Guneratne O, Smyth GK, Larance M, O'Sullivan JF, Lal S. Proteomic and metabolomic analyses of the human adult myocardium reveal ventricle-specific regulation in end-stage cardiomyopathies. Commun Biol 2024; 7:1666. [PMID: 39702518 DOI: 10.1038/s42003-024-07306-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 11/22/2024] [Indexed: 12/21/2024] Open
Abstract
The left and right ventricles of the human heart are functionally and developmentally distinct such that genetic or acquired insults can cause dysfunction in one or both ventricles resulting in heart failure. To better understand ventricle-specific molecular changes influencing heart failure development, we first performed unbiased quantitative mass spectrometry on pre-mortem non-diseased human myocardium to compare the metabolome and proteome between the normal left and right ventricles. Constituents of gluconeogenesis, glycolysis, lipogenesis, lipolysis, fatty acid catabolism, the citrate cycle and oxidative phosphorylation were down-regulated in the left ventricle, while glycogenesis, pyruvate and ketone metabolism were up-regulated. Inter-ventricular significance of these metabolic pathways was then found to be diminished within end-stage dilated cardiomyopathy and ischaemic cardiomyopathy, while heart failure-associated pathways were increased in the left ventricle relative to the right within ischaemic cardiomyopathy, such as fluid sheer-stress, increased glutamine-glutamate ratio, and down-regulation of contractile proteins, indicating a left ventricular pathological bias.
Collapse
Affiliation(s)
- Benjamin Hunter
- Precision Cardiovascular Laboratory, The University of Sydney, Sydney, NSW, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Mengbo Li
- Bioinformatics Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, VIC, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, Australia
| | - Benjamin L Parker
- Department of Anatomy and Physiology, The University of Melbourne, Parkville, VIC, Australia
| | - Yen Chin Koay
- Precision Cardiovascular Laboratory, The University of Sydney, Sydney, NSW, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
- Heart Research Institute, Newtown, NSW, Australia
| | - Dylan J Harney
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
- School of Life and Environmental Sciences, Faculty of Science, The University of Sydney, Sydney, NSW, Australia
| | - Evangeline Pearson
- Paediatric Oncology and Haematology, Oxford Children's Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, England
| | - Jacob Cao
- Central Clinical School, Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Gavin T Chen
- Department of Cardiology, Royal Prince Alfred Hospital, Camperdown, NSW, Australia
| | - Oneka Guneratne
- Kolling Institute, Royal North Shore Hospital, and Charles Perkins Centre, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Gordon K Smyth
- Bioinformatics Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, VIC, Australia
- School of Mathematics and Statistics, The University of Melbourne, Parkville, VIC, Australia
| | - Mark Larance
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - John F O'Sullivan
- Precision Cardiovascular Laboratory, The University of Sydney, Sydney, NSW, Australia.
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia.
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia.
- Heart Research Institute, Newtown, NSW, Australia.
- Central Clinical School, Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia.
- Department of Cardiology, Royal Prince Alfred Hospital, Camperdown, NSW, Australia.
- Faculty of Medicine, TU Dresden, Dresden, Germany.
| | - Sean Lal
- Precision Cardiovascular Laboratory, The University of Sydney, Sydney, NSW, Australia.
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia.
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia.
- Central Clinical School, Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia.
- Department of Cardiology, Royal Prince Alfred Hospital, Camperdown, NSW, Australia.
- The Baird Institute for Applied Heart and Lung Surgical Research, Sydney, NSW, Australia.
| |
Collapse
|
5
|
Tan Y, Li M, Li H, Guo Y, Zhang B, Wu G, Li J, Zhang Q, Sun Y, Gao F, Yi W, Zhang X. Cardiac Urea Cycle Activation by Time-Restricted Feeding Protects Against Pressure Overload-Induced Heart Failure. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2407677. [PMID: 39467073 DOI: 10.1002/advs.202407677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 09/14/2024] [Indexed: 10/30/2024]
Abstract
Heart failure is a leading cause of mortality worldwide, necessitating the development of novel therapeutic and lifestyle interventions. Recent studies highlight a potential role of time-restricted feeding (TRF) in the prevention and treatment of cardiac diseases. Here, it is found that TRF protected against heart failure at different stages in mice. Metabolomic profiling revealed that TRF upregulated most circulating amino acids, and amino acid supplementation protected against heart failure. In contrast, TRF showed a mild effect on cardiac amino acid profile, but increased cardiac amino acid utilization and activated the cardiac urea cycle through upregulating argininosuccinate lyase (ASL) expression. Cardiac-specific ASL knockout abolished the cardioprotective effects afforded by TRF. Circulating amino acids also protected against heart failure through activation of the urea cycle. Additionally, TRF upregulated cardiac ASL expression through transcription factor Yin Yang 1, and urea cycle-derived NO contributes to TRF-afforded cardioprotection. Furthermore, arteriovenous gradients of circulating metabolites across the human hearts were measured, and found that amino acid utilization and urea cycle activity were impaired in patients with decreased cardiac function. These results suggest that TRF is a promising intervention for heart failure, and highlight the importance of urea cycle in regulation of cardiac function.
Collapse
Affiliation(s)
- Yanzhen Tan
- Key Laboratory of Ministry of Education, School of Aerospace Medicine, Fourth Military Medical University, Xi'an, 710032, China
- Department of Cardiovascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Min Li
- Key Laboratory of Ministry of Education, School of Aerospace Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Han Li
- Key Laboratory of Ministry of Education, School of Aerospace Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Yongzheng Guo
- Division of Cardiology, The First Affiliated Hospital, Cardiovascular Disease Laboratory, Chongqing Medical University, Chongqing, 400016, China
| | - Bing Zhang
- Department of Cardiovascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Guiling Wu
- Key Laboratory of Ministry of Education, School of Aerospace Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Jia Li
- Key Laboratory of Ministry of Education, School of Aerospace Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Qian Zhang
- Key Laboratory of Ministry of Education, School of Aerospace Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Yang Sun
- Department of Geriatrics, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Feng Gao
- Key Laboratory of Ministry of Education, School of Aerospace Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Wei Yi
- Department of Cardiovascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Xing Zhang
- Key Laboratory of Ministry of Education, School of Aerospace Medicine, Fourth Military Medical University, Xi'an, 710032, China
- Department of Rehabilitation, Air Force Medical Center, Beijing, 100142, China
| |
Collapse
|
6
|
Zhang Y, Qiu J, Sun S, Fang X. Altered amino acid levels in young hypopituitarism: impact of NAFLD and insulin resistance. Amino Acids 2024; 56:65. [PMID: 39580591 PMCID: PMC11585508 DOI: 10.1007/s00726-024-03429-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Accepted: 11/17/2024] [Indexed: 11/25/2024]
Abstract
Elevated concentrations of amino acids (AAs) are commonly observed in patients with nonalcoholic fatty liver disease (NAFLD). Individuals with hypopituitarism (HP) are at a heightened risk of developing NAFLD due to factors such as visceral obesity, increased insulin resistance (IR), and disturbances in lipid metabolism. However, the changes in AAs concentrations associated with HP remain poorly understood. Therefore, our study aimed to investigate whether individuals with HP, who were not receiving growth hormone replacement therapy (GHRT), exhibited altered AAs compared to controls (CTs), and whether these AAs were associated with IR, the presence of NAFLD, and the Metabolic Syndrome (MetS) score. The AAs profiles of 133 young males with HP (age: 24.5 ± 5.9; 57 with NAFLD and 76 without NAFLD) and 90 age and BMI-matched CTs were analyzed using untargeted metabolomics. The results revealed that most AAs were found to be elevated in subjects with HPs compared to CTs. Glutamate, glutamine, norleucine, and branched-chain amino acids (BCAAs) (leucine and valine) were correlated with the homeostasis model assessment of insulin resistance (HOMA-IR), with glutamate and norleucine showing independent linkage. Glutamate and proline levels were specifically associated with MetS score, while alanine and proline linked to NAFLD. Given that elevated glutamate and BCAAs levels have higher prevalence of NAFLD, we hypothesized that the changes in AAs observed in HPs may be attributed to the impact of NAFLD and IR.
Collapse
Affiliation(s)
- Yuwen Zhang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Jiting Qiu
- Department of Neurosurgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Shouyue Sun
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Xuqian Fang
- Department of Pathology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| |
Collapse
|
7
|
Longo S, Cicalini I, Pieragostino D, De Laurenzi V, Legramante JM, Menghini R, Rizza S, Federici M. A Metabolomic Approach to Unexplained Syncope. Biomedicines 2024; 12:2641. [PMID: 39595205 PMCID: PMC11591916 DOI: 10.3390/biomedicines12112641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 11/11/2024] [Accepted: 11/12/2024] [Indexed: 11/28/2024] Open
Abstract
Background: This study aims to identify a metabolomic signature that facilitates the classification of syncope and the categorization of the unexplained syncope (US) to aid in its management. Methods: We compared a control group (CTRL, n = 10) with a transient loss of consciousness (TLC) group divided into the OH group (n = 23) for orthostatic syncope, the NMS group (n = 26) for neuromediated syncope, the CS group (n = 9) for cardiological syncope, and the US group (n = 27) for US defined as syncope without a precise categorization after first- and second-level diagnostic approaches. Results: The CTRL and the TLC groups significantly differed in metabolic profile. A new logistic regression model has been developed to predict how the US will be clustered. Using differences in lysophosphatidylcholine with 22 carbon atom (C22:0-LPC) levels, 96% of the US belongs to the NMS and 4% to the CS subgroup. Differences in glutamine and lysine (GLN/LYS) levels clustered 95% of the US in the NMS and 5% in the CS subgroup. Conclusions: We hypothesize a possible role of C22:0 LPC and GLN/LYS in re-classifying US and differentiating it from cardiological syncope.
Collapse
Affiliation(s)
- Susanna Longo
- Department of Systems Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; (S.L.); (J.M.L.); (R.M.); (S.R.)
| | - Ilaria Cicalini
- Department of Innovative Technologies in Medicine and Dentistry, “G. d‘Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy; (I.C.); (D.P.); (V.D.L.)
- Center for Advanced Studies and Technology (CAST), “G. d‘Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| | - Damiana Pieragostino
- Department of Innovative Technologies in Medicine and Dentistry, “G. d‘Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy; (I.C.); (D.P.); (V.D.L.)
- Center for Advanced Studies and Technology (CAST), “G. d‘Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| | - Vincenzo De Laurenzi
- Department of Innovative Technologies in Medicine and Dentistry, “G. d‘Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy; (I.C.); (D.P.); (V.D.L.)
- Center for Advanced Studies and Technology (CAST), “G. d‘Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| | - Jacopo M. Legramante
- Department of Systems Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; (S.L.); (J.M.L.); (R.M.); (S.R.)
| | - Rossella Menghini
- Department of Systems Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; (S.L.); (J.M.L.); (R.M.); (S.R.)
| | - Stefano Rizza
- Department of Systems Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; (S.L.); (J.M.L.); (R.M.); (S.R.)
| | - Massimo Federici
- Department of Systems Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; (S.L.); (J.M.L.); (R.M.); (S.R.)
| |
Collapse
|
8
|
Chi K, Liu J, Li X, Wang H, Li Y, Liu Q, Zhou Y, Ge Y. Biomarkers of heart failure: advances in omics studies. Mol Omics 2024; 20:169-183. [PMID: 38224222 DOI: 10.1039/d3mo00173c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2024]
Abstract
Heart failure is a complex syndrome characterized by progressive circulatory dysfunction, manifesting clinically as pulmonary and systemic venous congestion, alongside inadequate tissue perfusion. The early identification of HF, particularly at the mild and moderate stages (stages B and C), presents a clinical challenge due to the overlap of signs, symptoms, and natriuretic peptide levels with other cardiorespiratory pathologies. Nonetheless, early detection coupled with timely pharmacological intervention is imperative for enhancing patient outcomes. Advances in high-throughput omics technologies have enabled researchers to analyze patient-derived biofluids and tissues, discovering biomarkers that are sensitive and specific for HF diagnosis. Due to the diversity of HF etiology, it is insufficient to study the diagnostic data of early HF using a single omics technology. This study reviewed the latest progress in genomics, transcriptomics, proteomics, and metabolomics for the identification of HF biomarkers, offering novel insights into the early clinical diagnosis of HF. However, the validity of biomarkers depends on the disease status, intervention time, genetic diversity and comorbidities of the subjects. Moreover, biomarkers lack generalizability in different clinical settings. Hence, it is imperative to conduct multi-center, large-scale and standardized clinical trials to enhance the diagnostic accuracy and utility of HF biomarkers.
Collapse
Affiliation(s)
- Kuo Chi
- Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China.
| | - Jing Liu
- Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China.
| | - Xinghua Li
- Changzhi People's Hospital Affiliated to Changzhi Medical College, Changzhi, Shanxi, China.
| | - He Wang
- Department of Cardiovascular Disease II, First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China.
| | - Yanliang Li
- Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China.
| | - Qingnan Liu
- Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China.
| | - Yabin Zhou
- Department of Cardiovascular Disease II, First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China.
| | - Yuan Ge
- Department of Cardiovascular Disease II, First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China.
| |
Collapse
|
9
|
Chen Z, Hu Y, Hu FB, Manson JE, Rimm EB, Doria A, Sun Q. Dietary Glutamine and Glutamate in Relation to Cardiovascular Disease Incidence and Mortality in the United States Men and Women with Diabetes Mellitus. J Nutr 2023; 153:3247-3258. [PMID: 37660951 PMCID: PMC10687617 DOI: 10.1016/j.tjnut.2023.08.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 08/03/2023] [Accepted: 08/24/2023] [Indexed: 09/05/2023] Open
Abstract
BACKGROUND Evidence regarding the potential health effects of dietary amino acids glutamine and glutamate among individuals with type 2 diabetes (T2D) is limited. OBJECTIVES The aim was to examine dietary glutamine and glutamate in relation to subsequent risk of cardiovascular disease (CVD) and mortality among individuals with T2D. METHODS We prospectively followed 15,040 men and women with T2D at baseline or diagnosed during follow-up (Nurses' Health Study: 1980-2014 and Health Professionals Follow-Up Study: 1986-2018). Diet was repeatedly assessed using validated food frequency questionnaires every 2-4 y. Associations of energy-adjusted glutamine and glutamate intake, as well as their ratio, with CVD risk and mortality, were assessed using Cox proportional-hazards models with adjustments for demographics, dietary and lifestyle factors, and medical history. RESULTS During 196,955 and 225,371 person-years of follow-up in participants with T2D, there were 2927 incident CVD cases and 4898 deaths, respectively. Higher intake of glutamine was associated with lower risk of CVD incidence, CVD mortality, and total mortality: comparing extreme quintiles, the hazard ratios (HRs) (95% confidence intervals [CIs]) were 0.88 (0.77, 0.99), 0.78 (0.65, 0.92), and 0.84 (0.76, 0.92), respectively (all P-trend < 0.05). In contrast, higher intake of glutamate was associated with a higher risk of CVD incidence, CVD mortality, and total mortality; the HRs were 1.30 (1.15, 1.46), 1.46 (1.24, 1.72), and 1.20 (1.09, 1.32), respectively (all P-trend < 0.05). Furthermore, comparing extreme quintiles, a higher dietary glutamine-to-glutamate ratio was associated with a lower risk of CVD incidence (0.84 [0.75, 0.95]), CVD mortality (0.66 [0.57, 0.77]), and total mortality (0.82 [0.75, 0.90]). In addition, compared with participants with stable or decreased consumption of glutamine-to-glutamate ratio from prediabetes to postdiabetes diagnosis, those who increased the ratio had a 17% (5%, 27%) lower CVD mortality. CONCLUSIONS In adults with T2D, dietary glutamine was associated with a lower risk of CVD incidence and mortality, whereas the opposite was observed for glutamate intake.
Collapse
Affiliation(s)
- Zhangling Chen
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, United States; Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China; Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Yang Hu
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, United States
| | - Frank B Hu
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, United States; Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, United States; Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States
| | - JoAnn E Manson
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, United States; Division of Preventive Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States
| | - Eric B Rimm
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, United States; Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, United States; Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States
| | - Alessandro Doria
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, United States; Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States; Joslin Diabetes Center, Boston, MA, United States
| | - Qi Sun
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, United States; Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, United States; Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States; Joslin Diabetes Center, Boston, MA, United States.
| |
Collapse
|
10
|
Zhang P, Wang TY, Luo ZY, Ding JC, Yang Q, Hu PF. Identification of Key Immune-Related Genes in the Treatment of Heart Failure After Myocardial Infarction with Empagliflozin Based on RNA-Seq. J Inflamm Res 2023; 16:4679-4696. [PMID: 37872957 PMCID: PMC10590601 DOI: 10.2147/jir.s428747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 10/10/2023] [Indexed: 10/25/2023] Open
Abstract
Purpose Heart failure is a serious complication after acute myocardial infarction (AMI). It is crucial to investigate the mechanism of action of empagliflozin in the treatment of heart failure. Methods A total of 20 wild type (WT) male C57BL6/J mice were used to establish a model of heart failure after myocardial infarction and randomly divided into 2 groups: treatment group and control group. The treatment group was treated with empagliflozin, and the control group was treated with placebo. After 8 weeks of treatment, mouse heart tissues were collected for next generation sequencing. Bioinformatics methods were used to screen the key genes. Finally, the correlation between clinical data and gene expression was analyzed. Quantitative real-time polymerase chain reaction (qRT-PCR) was used to verify the expression of key genes. Results A mouse model of heart failure was successfully constructed. By DEG analysis, a total of 740 DEGs in the treatment group vs the control group were obtained. Dendritic cells, granulocytes, follicular B, plasma cell, cDC1, cDC2, pDC and neutrophils were 8 different immune cells identified by immunoinfiltration analysis. Through WGCNA, the turquoise module with the highest correlation with the above differential immune cells was selected. One hundred and forty-two immune-related DEGs were obtained by taking intersection of the DEGs and the genes of the turquoise module. Col17a1 and Gria4 were finally screened out as key immune-related genes via PPI analysis and machine learning. Col17a1 was significantly up-regulated, while Gria4 was significantly down-regulated in the treatment group. At the same time, the expression level of Col17a1 was significantly correlated with left ventricular ejection fraction (LVEF), left ventricular fraction shortening (LVFS) and left ventricular internal dimension systole (LVIDs). Conclusion Col17a1 and Gria4 are key immune-related genes of empagliflozin in the treatment of heart failure after myocardial infarction. This study provides a scientific basis for elucidating the mechanism of action of empagliflozin in treating heart failure after myocardial infarction.
Collapse
Affiliation(s)
- Pei Zhang
- Department of Cardiology, Sir Run Run Shaw Hospital, College of Medicine Zhejiang University, Hangzhou, Zhejiang Province, 310018, People’s Republic of China
| | - Tian-Yu Wang
- Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, 310053, People’s Republic of China
| | - Zi-Yue Luo
- Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, 310053, People’s Republic of China
| | - Jun-Can Ding
- Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, 310053, People’s Republic of China
| | - Qiang Yang
- Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, 310053, People’s Republic of China
| | - Peng-Fei Hu
- Department of Cardiology, the Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, 310005, People’s Republic of China
| |
Collapse
|
11
|
Sun J, Ma X, Yang L, Jin X, Zhao M, Xi B, Song S. The number of metabolic syndrome risk factors predicts alterations in gut microbiota in Chinese children from the Huantai study. BMC Pediatr 2023; 23:191. [PMID: 37085796 PMCID: PMC10120097 DOI: 10.1186/s12887-023-04017-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 04/14/2023] [Indexed: 04/23/2023] Open
Abstract
BACKGROUND Evidence on the effect of gut microbiota on the number of metabolic syndrome (MetS) risk factors among children is scarce. We aimed to examine the alterations of gut microbiota with different numbers of MetS risk factors among children. METHODS Data were collected from a nested case-control study at the baseline of the Huantai Childhood Cardiovascular Health Cohort Study in Zibo, China. We compared the differences in gut microbiota based on 16S rRNA gene sequencing among 72 children with different numbers of MetS risk factors matched by age and sex (i.e., none, one, and two-or-more MetS risk factors; 24 children for each group). RESULTS The community richness (i.e., the total number of species in the community) and diversity (i.e., the richness and evenness of species in the community) of gut microbiota decreased with an increased number of MetS risk factors in children (P for trend < 0.05). Among genera with a relative abundance greater than 0.01%, the relative abundance of Lachnoclostridium (PFDR = 0.009) increased in the MetS risk groups, whereas Alistipes (PFDR < 0.001) and Lachnospiraceae_NK4A136_group (PFDR = 0.043) decreased in the MetS risk groups compared to the non-risk group. The genus Christensenellaceae_R-7_group excelled at distinguishing one and two-or-more risk groups from the non-risk group (area under the ROC curve [AUC]: 0.84 - 0.92), while the genera Family_XIII_AD3011_group (AUC: 0.73 - 0.91) and Lachnoclostridium (AUC: 0.77 - 0.80) performed moderate abilities in identifying none, one, and two-or-more MetS risk factors in children. CONCLUSIONS Based on the nested case-control study and the 16S rRNA gene sequencing technology, we found that dysbiosis of gut microbiota, particularly for the genera Christensenellaceae_R-7_group, Family_XIII_AD3011_group, and Lachnoclostridium may contribute to the early detection and the accumulation of MetS risk factors in childhood.
Collapse
Affiliation(s)
- Jiahong Sun
- Department of Epidemiology, School of Public Health, Cheeloo College of Medicine, Shandong University, 44 Wen Hua Xi Road, Jinan, 250012, China
| | - Xiaoyun Ma
- Department of Epidemiology, School of Public Health, Cheeloo College of Medicine, Shandong University, 44 Wen Hua Xi Road, Jinan, 250012, China
| | - Liu Yang
- Department of Epidemiology, School of Public Health, Cheeloo College of Medicine, Shandong University, 44 Wen Hua Xi Road, Jinan, 250012, China
| | - Xuli Jin
- Department of Epidemiology, School of Public Health, Cheeloo College of Medicine, Shandong University, 44 Wen Hua Xi Road, Jinan, 250012, China
| | - Min Zhao
- Department of Nutrition and Food Hygiene, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Bo Xi
- Department of Epidemiology, School of Public Health, Cheeloo College of Medicine, Shandong University, 44 Wen Hua Xi Road, Jinan, 250012, China.
| | - Suhang Song
- Taub Institute for Research in Alzheimer 's disease and the Aging Brain, Columbia University, New York, NY, USA
| |
Collapse
|
12
|
Jeong S, Jang HB, Kim HJ, Lee HJ. Identification of Biomarkers Related to Metabolically Unhealthy Obesity in Korean Obese Adolescents: A Cross-Sectional Study. CHILDREN (BASEL, SWITZERLAND) 2023; 10:children10020322. [PMID: 36832451 PMCID: PMC9955165 DOI: 10.3390/children10020322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 01/25/2023] [Accepted: 02/02/2023] [Indexed: 02/11/2023]
Abstract
BACKGROUND The current study aimed to screen for relationships and different potential metabolic biomarkers involved between metabolically healthy obesity (MHO) and metabolically unhealthy obesity (MUO) in adolescents. METHODS The study included 148 obese adolescents aged between 14 and 16. The study participants were divided into MUO and MHO groups based on the age-specific adolescent metabolic syndrome (MetS) criteria of the International Diabetes Federation. The current study was conducted to investigate the clinical and metabolic differences between the MHO and MUO groups. Multivariate analyses were conducted to investigate the metabolites as independent predictors for the odds ratio and the presence of the MetS. RESULTS There were significant differences in the three acylcarnitines, five amino acids, glutamine/glutamate ratio, three biogenic amines, two glycerophospholipids, and the triglyceride-glucose index between the MUO group and those in the MHO group. Moreover, several metabolites were associated with the prevalence of MUO. Additionally, several metabolites were inversely correlated with MHO in the MUO group. CONCLUSIONS In this study, the biomarkers found in this study have the potential to reflect the clinical outcomes of the MUO group. These biomarkers will lead to a better understanding of MetS in obese adolescents.
Collapse
Affiliation(s)
| | | | | | - Hye-Ja Lee
- Correspondence: ; Tel.: +82-43-719-8452; Fax: +82-43-719-8709
| |
Collapse
|
13
|
Fanni G, Eriksson JW, Pereira MJ. Several Metabolite Families Display Inflexibility during Glucose Challenge in Patients with Type 2 Diabetes: An Untargeted Metabolomics Study. Metabolites 2023; 13:metabo13010131. [PMID: 36677056 PMCID: PMC9863788 DOI: 10.3390/metabo13010131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 01/06/2023] [Accepted: 01/12/2023] [Indexed: 01/18/2023] Open
Abstract
Metabolic inflexibility is a hallmark of insulin resistance and can be extensively explored with high-throughput metabolomics techniques. However, the dynamic regulation of the metabolome during an oral glucose tolerance test (OGTT) in subjects with type 2 diabetes (T2D) is largely unknown. We aimed to identify alterations in metabolite responses to OGTT in subjects with T2D using untargeted metabolomics of both plasma and subcutaneous adipose tissue (SAT) samples. Twenty subjects with T2D and twenty healthy controls matched for sex, age, and body mass index (BMI) were profiled with untargeted metabolomics both in plasma (755 metabolites) and in the SAT (588) during an OGTT. We assessed metabolite concentration changes 90 min after the glucose load, and those responses were compared between patients with T2D and controls. Post-hoc analyses were performed to explore the associations between glucose-induced metabolite responses and markers of obesity and glucose metabolism, sex, and age. During the OGTT, T2D subjects had an impaired reduction in plasma levels of several metabolite families, including acylcarnitines, amino acids, acyl ethanolamines, and fatty acid derivates (p < 0.05), compared to controls. Additionally, patients with T2D had a greater increase in plasma glucose and fructose levels during the OGTT compared to controls (p < 0.05). The plasma concentration change of most metabolites after the glucose load was mainly associated with indices of hyperglycemia rather than insulin resistance, insulin secretion, or BMI. In multiple linear regression analyses, hyperglycemia indices (glucose area under the curve (AUC) during OGTT and glycosylated hemoglobin (HbA1c)) were the strongest predictors of plasma metabolite changes during the OGTT. No differences were found in the adipose tissue metabolome in response to the glucose challenge between T2D and controls. Using a metabolomics approach, we show that T2D patients display attenuated responses in several circulating metabolite families during an OGTT. Besides the well-known increase in monosaccharides, the glucose-induced lowering of amino acids, acylcarnitines, and fatty acid derivatives was attenuated in T2D subjects compared to controls. These data support the hypothesis of inflexibility in several metabolic pathways, which may contribute to dysregulated substrate partitioning and turnover in T2D. These findings are not directly associated with changes in adipose tissue metabolism; therefore, other tissues, such as muscle and liver, are probably of greater importance.
Collapse
|
14
|
Li W, Li S, Cao Z, Sun Y, Qiu W, Jia M, Su M. Exploration of the amino acid metabolic signature in anthracycline-induced cardiotoxicity using an optimized targeted metabolomics approach based on UPLC-MS/MS. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2022; 395:1209-1224. [PMID: 35879430 DOI: 10.1007/s00210-022-02271-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 07/08/2022] [Indexed: 10/16/2022]
Abstract
Although anthracyclines improve the long-term survival rate of patients with cancer, severe and irreversible myocardial damage limits their clinical application. Amino acid (AA) metabolism in cardiomyocytes can be altered under pathological conditions. Therefore, exploring the AA metabolic signature in anthracycline-induced cardiotoxicity (AIC) is important for identifying novel mechanisms. We established mouse and cellular models of Adriamycin (ADR)-induced cardiac injury. We observed a decreased expression of troponins I (cTnI) after ADR treatment and ADR accelerated the degradation of cTnI, implying that AA metabolism could be altered in AIC. Using a targeted AA metabolomics approach based on ultra-performance liquid chromatography-tandem mass spectrometry (UPLC-MS/MS), the AA metabolic signatures in the sera of AIC mice and supernatant samples of ADR-treated H9c2 cardiomyocytes were analyzed. The levels of 14 AA metabolites were altered in ADR-treated mice (p < 0.05). Via bioinformatics analysis, we identified nine differential AA metabolites in mice and five differential AA metabolites in ADR-treated H9c2 cardiomyocytes. Three AAs with increased levels (L-glutamate, L-serine, and L-tyrosine) overlapped in the two models, suggesting a possible mechanism of AA metabolic impairment during AIC. The metabolic pathways perturbed by AIC involved aminoacyl-tRNA biosynthesis and alanine, aspartate, and glutamate metabolism. Our data suggests that ADR perturbed AA metabolism in AIC models. Moreover, the targeted AA metabolomics approach based on UPLC-MS/MS can be a unique platform to provide new clues for the prevention and treatment of AIC.
Collapse
Affiliation(s)
- Wendi Li
- Department of Clinical Laboratory, Peking University People's Hospital, No. 11 Xizhimen South Street, Beijing, 100044, People's Republic of China
| | - Shanshan Li
- Department of Clinical Laboratory, Peking University People's Hospital, No. 11 Xizhimen South Street, Beijing, 100044, People's Republic of China
| | - Zhenju Cao
- Department of Clinical Laboratory, Peking University People's Hospital, No. 11 Xizhimen South Street, Beijing, 100044, People's Republic of China
| | - Yi Sun
- Department of Clinical Laboratory, Peking University People's Hospital, No. 11 Xizhimen South Street, Beijing, 100044, People's Republic of China
| | - Wei Qiu
- Department of Urology, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, People's Republic of China.
| | - Mei Jia
- Department of Clinical Laboratory, Peking University People's Hospital, No. 11 Xizhimen South Street, Beijing, 100044, People's Republic of China.
| | - Ming Su
- Department of Clinical Laboratory, Peking University People's Hospital, No. 11 Xizhimen South Street, Beijing, 100044, People's Republic of China.
| |
Collapse
|
15
|
Identification of Single and Combined Serum Metabolites Associated with Food Intake. Metabolites 2022; 12:metabo12100908. [PMID: 36295810 PMCID: PMC9607433 DOI: 10.3390/metabo12100908] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Revised: 09/16/2022] [Accepted: 09/23/2022] [Indexed: 11/17/2022] Open
Abstract
Assessment of dietary intake is challenging. Traditional methods suffer from both random and systematic errors; thus objective measures are important complements in monitoring dietary exposure. The study presented here aims to identify serum metabolites associated with reported food intake and to explore whether combinations of metabolites may improve predictive models. Fasting blood samples and a 4-day weighed food diary were collected from healthy Swedish subjects (n = 119) self-defined as having habitual vegan, vegetarian, vegetarian + fish, or omnivore diets. Serum was analyzed for metabolites by 1H-nuclear magnetic resonance spectroscopy. Associations between single and combined metabolites and 39 foods and food groups were explored. Area under the curve (AUC) was calculated for prediction models. In total, 24 foods or food groups associated with serum metabolites using the criteria of rho > 0.2, p < 0.01 and AUC ≥ 0.7 were identified. For the consumption of soybeans, citrus fruits and marmalade, nuts and almonds, green tea, red meat, poultry, total fish and shellfish, dairy, fermented dairy, cheese, eggs, and beer the final models included two or more metabolites. Our results indicate that a combination of metabolites improve the possibilities to use metabolites to identify several foods included in the current diet. Combined metabolite models should be confirmed in dose−response intervention studies.
Collapse
|
16
|
Singh RB, Fedacko J, Pella D, Fatima G, Elkilany G, Moshiri M, Hristova K, Jakabcin P, Vaňova N. High Exogenous Antioxidant, Restorative Treatment (Heart) for Prevention of the Six Stages of Heart Failure: The Heart Diet. Antioxidants (Basel) 2022; 11:1464. [PMID: 36009183 PMCID: PMC9404840 DOI: 10.3390/antiox11081464] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 07/08/2022] [Accepted: 07/19/2022] [Indexed: 01/06/2023] Open
Abstract
The exact pathophysiology of heart failure (HF) is not yet known. Western diet, characterized by highly sweetened foods, as well as being rich in fat, fried foods, red meat and processed meat, eggs, and sweet beverages, may cause inflammation, leading to oxidative dysfunction in the cardiac ultra-structure. Oxidative function of the myocardium and how oxidative dysfunction causes physio-pathological remodeling, leading to HF, is not well known. Antioxidants, such as polyphenolics and flavonoids, omega-3 fatty acids, and other micronutrients that are rich in Indo-Mediterranean-type diets, could be protective in sustaining the oxidative functions of the heart. The cardiomyocytes use glucose and fatty acids for the physiological functions depending upon the metabolic requirements of the heart. Apart from toxicity due to glucose, lipotoxicity also adversely affects the cardiomyocytes, which worsen in the presence of deficiency of endogenous antioxidants and deficiency of exogenous antioxidant nutrients in the diet. The high-sugar-and-high-fat-induced production of ceramide, advanced glycation end products (AGE) and triamino-methyl-N-oxide (TMAO) can predispose individuals to oxidative dysfunction and Ca-overloading. The alteration in the biology may start with normal cardiac cell remodeling to biological remodeling due to inflammation. An increase in the fat content of a diet in combination with inducible nitric oxide synthase (NOSi) via N-arginine methyl ester has been found to preserve the ejection fraction in HF. It is proposed that a greater intake of high exogenous antioxidant restorative treatment (HEART) diet, polyphenolics and flavonoids, as well as cessation of red meat intake and egg, can cause improvement in the oxidative function of the heart, by inhibiting oxidative damage to lipids, proteins and DNA in the cell, resulting in beneficial effects in the early stage of the Six Stages of HF. There is an unmet need to conduct cohort studies and randomized, controlled studies to demonstrate the role of the HEART diet in the treatment of HF.
Collapse
Affiliation(s)
- Ram B. Singh
- Halberg Hospital and Research Institute, Moradabad 244001, India;
| | - Jan Fedacko
- Department of Gerontology and Geriatric, Medipark, University Research Park, PJ Safarik University, 040-11 Kosice, Slovakia
| | - Dominik Pella
- Department of Cardiology, Faculty of Medicine and East Slovak, Institute for Cardiovascular Disease, PJ Safarik University, 040-11 Kosice, Slovakia;
| | - Ghizal Fatima
- Department of Biotechnology, Era University, Lucknow 226001, India;
| | - Galal Elkilany
- International College of Cardiology, Laplace, LA 90001, USA;
| | - Mahmood Moshiri
- International College of Cardiology, Richmond Hill, ON LL-9955, Canada;
| | - Krasimira Hristova
- Department of Cardiology, National University Hospital, 1000 Sofia, Bulgaria;
| | - Patrik Jakabcin
- Department of Social and Clinical Pharmacy, Faculty of Pharmacy in Hradec Králové, Charles University, 10000 Prague, Czech Republic;
| | - Natalia Vaňova
- Department of Internal Medicine UPJS MF and AGEL Hospital, Research Park, PJ Safaric University, 040-11 Kosice, Slovakia;
| |
Collapse
|
17
|
Basic fibroblast growth factor alleviates metabolic abnormalities in the heart of streptozotocin-induced diabetic rats. Int J Diabetes Dev Ctries 2022. [DOI: 10.1007/s13410-022-01059-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
|
18
|
Ogilvie AR, Watford M, Wu G, Sukumar D, Kwon J, Shapses SA. Decreased fasting serum glucogenic amino acids with a higher compared to normal protein diet during energy restriction in women: a randomized controlled trial. Amino Acids 2021; 53:1467-1472. [PMID: 34338883 DOI: 10.1007/s00726-021-03053-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 07/20/2021] [Indexed: 11/25/2022]
Abstract
Dietary protein alters circulating amino acid (AAs) levels and higher protein intake (HP) is one means of losing weight. We examined 34 overweight and obese women (57 ± 4 years) during 6 months of energy restriction (7.3 ± 3.8% weight loss) divided into groups consuming either normal protein (NP; 18.6 energy% protein) or HP (24.3 energy% protein). There was a reduction in fasting serum glucogenic AAs (p = 0.015) that also associated with greater weight loss (p < 0.05) in the HP group, but not in the NP group. These findings have implications for nutrient prioritization during energy restriction.
Collapse
Affiliation(s)
- A R Ogilvie
- Department of Nutritional Sciences, Rutgers University and NJ-Institute of Food Health and Nutrition, New Brunswick, NJ, USA
| | - M Watford
- Department of Nutritional Sciences, Rutgers University and NJ-Institute of Food Health and Nutrition, New Brunswick, NJ, USA
| | - G Wu
- Department of Animal Science, Texas A&M University, College Station, TX, USA
| | - D Sukumar
- Department of Nutrition Sciences, Drexel University, Philadelphia, PA, USA
| | - J Kwon
- Department of Nutritional Sciences, Rutgers University and NJ-Institute of Food Health and Nutrition, New Brunswick, NJ, USA
| | - S A Shapses
- Department of Nutritional Sciences, Rutgers University and NJ-Institute of Food Health and Nutrition, New Brunswick, NJ, USA.
- Department of Medicine, Rutgers-RWJ Medical School, New Brunswick, NJ, USA.
| |
Collapse
|
19
|
Carioca AAF, Steluti J, Carvalho AMD, Silva AM, Silva IDCGD, Fisberg RM, Marchioni DM. Plasma metabolomics are associated with metabolic syndrome: A targeted approach. Nutrition 2020; 83:111082. [PMID: 33360505 DOI: 10.1016/j.nut.2020.111082] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 10/19/2020] [Accepted: 11/05/2020] [Indexed: 12/15/2022]
Abstract
OBJECTIVES Advances in metabolomic tools have allowed us to gain a more comprehensive understanding of metabolic syndrome (MetS). The aim of this study was to evaluate the association between plasma metabolomic profiles and MetS. METHODS For this study, adults without diabetes, chronic kidney disease, stroke, heart disease, or cancer and with full metabolomics, biochemical, and dietetic data available, representing a subsample of the Health Survey of Sao Paulo study (ISA-Capital; N = 130), were included. The joint interim statement consensus criteria were used for diagnosing MetS. Absolute quantification (µmol/L) of blood metabolites was achieved by targeted quantitative profiling of annotated metabolites by electrospray ionization tandem mass spectrometry in plasma samples. Mean differences in the compounds for MetS were evaluated by linear regression adjusted for confounding factors. RESULTS Serine was inversely associated with MetS (β = -15.04; P = 0.014). In glycerophospholipids with acyl-alkyl bonds, there was an inverse association with MetS, including phosphatidylcholine (PC) ae C42:5 (β = -0.15; P = 0.040), PC ae C44:5 (β = -0.15; P = 0.046), PC ae C40:4 (β = -0.21; P = 0.014) and PC ae C44:4 (β = -0.04; P = 0.032). CONCLUSION Plasma metabolomic profiles were associated with MetS, especially the amino acid serine and some acyl-alkyl PCs.
Collapse
Affiliation(s)
- Antonio Augusto Ferreira Carioca
- Department of Nutrition, School of Public Health, University of Sao Paulo, SP, Brazil; University of Fortaleza (UNIFOR), Nutrition Course, Fortaleza, Brazil.
| | - Josiane Steluti
- Department of Nutrition, School of Public Health, University of Sao Paulo, SP, Brazil
| | | | | | | | - Regina Mara Fisberg
- Department of Nutrition, School of Public Health, University of Sao Paulo, SP, Brazil
| | - Dirce Maria Marchioni
- Department of Nutrition, School of Public Health, University of Sao Paulo, SP, Brazil
| |
Collapse
|