1
|
Rizo‐Roca D, Henderson JD, Zierath JR. Metabolomics in cardiometabolic diseases: Key biomarkers and therapeutic implications for insulin resistance and diabetes. J Intern Med 2025; 297:584-607. [PMID: 40289598 PMCID: PMC12087830 DOI: 10.1111/joim.20090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
Cardiometabolic diseases-including Type 2 diabetes and obesity-remain leading causes of global mortality. Recent advancements in metabolomics have facilitated the identification of metabolites that are integral to the development of insulin resistance, a characteristic feature of cardiometabolic disease. Key metabolites, such as branched-chain amino acids (BCAAs), ceramides, glycine, and glutamine, have emerged as valuable biomarkers for early diagnosis, risk stratification, and potential therapeutic targets. Elevated BCAAs and ceramides are strongly associated with insulin resistance and Type 2 diabetes, whereas glycine exhibits an inverse relationship with insulin resistance, making it a promising therapeutic target. Metabolites involved in energy stress, including ketone bodies, lactate, and nicotinamide adenine dinucleotide (NAD⁺), regulate insulin sensitivity and metabolic health, with ketogenic diets and NAD⁺ precursor supplementation showing potential benefits. Additionally, the novel biomarker N-lactoyl-phenylalanine further underscores the complexity of metabolic regulation and its therapeutic potential. This review underscores the potential of metabolite-based diagnostics and precision medicine, which could enhance efforts in the prevention, diagnosis, and treatment of cardiometabolic diseases, ultimately improving patient outcomes and quality of life.
Collapse
Affiliation(s)
- David Rizo‐Roca
- Department of Physiology and Pharmacology, Integrative PhysiologyKarolinska InstitutetStockholmSweden
| | - John D. Henderson
- Novo Nordisk Foundation Center for Basic Metabolic ResearchFaculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark
| | - Juleen R. Zierath
- Department of Physiology and Pharmacology, Integrative PhysiologyKarolinska InstitutetStockholmSweden
- Novo Nordisk Foundation Center for Basic Metabolic ResearchFaculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark
- Department of Molecular Medicine and Surgery, Integrative PhysiologyKarolinska InstitutetStockholmSweden
| |
Collapse
|
2
|
Renz A, Hohner M, Jami R, Breitenbach M, Josephs-Spaulding J, Dürrwald J, Best L, Dulière V, Mialon C, Bader SM, Marinos G, Leonidou N, Cabreiro F, Pellegrini M, Doerflinger M, Rosa-Calatrava M, Pizzorno A, Dräger A, Schindler M, Kaleta C. Metabolic modeling elucidates phenformin and atpenin A5 as broad-spectrum antiviral drugs against RNA viruses. Commun Biol 2025; 8:791. [PMID: 40410544 PMCID: PMC12102274 DOI: 10.1038/s42003-025-08148-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 04/30/2025] [Indexed: 05/25/2025] Open
Abstract
The SARS-CoV-2 pandemic has reemphasized the urgent need for broad-spectrum antiviral therapies. We developed a computational workflow using scRNA-Seq data to assess cellular metabolism during viral infection. With this workflow we predicted the capacity of cells to sustain SARS-CoV-2 virion production in patients and found a tissue-wide induction of metabolic pathways that support viral replication. Expanding our analysis to influenza A and dengue viruses, we identified metabolic targets and inhibitors for potential broad-spectrum antiviral treatment. These targets were highly enriched for known interaction partners of all analyzed viruses. Indeed, phenformin, an NADH:ubiquinone oxidoreductase inhibitor, suppressed SARS-CoV-2 and dengue virus replication. Atpenin A5, blocking succinate dehydrogenase, inhibited SARS-CoV-2, dengue virus, respiratory syncytial virus, and influenza A virus with high selectivity indices. In vivo, phenformin showed antiviral activity against SARS-CoV-2 in a Syrian hamster model. Our work establishes host metabolism as druggable for broad-spectrum antiviral strategies, providing invaluable tools for pandemic preparedness.
Collapse
Affiliation(s)
- Alina Renz
- Computational Systems Biology of Infections and Antimicrobial-Resistant Pathogens, Institute for Bioinformatics and Medical Informatics (IBMI), Eberhard Karl University of Tübingen, Tübingen, Germany
| | - Mirjam Hohner
- Institute for Medical Virology and Epidemiology of Viral Diseases, University Hospital Tübingen, Tübingen, Germany
- German Center for Infection Research (DZIF), partner site, Tübingen, Germany
| | - Raphaël Jami
- Institute for Medical Virology and Epidemiology of Viral Diseases, University Hospital Tübingen, Tübingen, Germany
- German Center for Infection Research (DZIF), partner site, Tübingen, Germany
| | - Maximilian Breitenbach
- Institute for Medical Virology and Epidemiology of Viral Diseases, University Hospital Tübingen, Tübingen, Germany
- German Center for Infection Research (DZIF), partner site, Tübingen, Germany
| | - Jonathan Josephs-Spaulding
- Research Group Medical Systems Biology, Institute of Experimental Medicine, Christian-Albrechts-University Kiel & University Hospital Schleswig Holstein, Kiel, Germany
- Wolfson Wohl Cancer Research Centre, School of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - Johanna Dürrwald
- Institute for Medical Virology and Epidemiology of Viral Diseases, University Hospital Tübingen, Tübingen, Germany
| | - Lena Best
- Research Group Medical Systems Biology, Institute of Experimental Medicine, Christian-Albrechts-University Kiel & University Hospital Schleswig Holstein, Kiel, Germany
| | - Victoria Dulière
- CIRI, Centre International de Recherche en Infectiologie (Team VirPath), Université de Lyon, INSERM U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, Lyon, France
- VirNext, Faculté de Médecine RTH Laennec, Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France
- International Research Laboratory RESPIVIR France-Canada, Centre Hospitalier Universitaire de Québec - Université Laval, Québec, Canada, Centre International de Recherche en Infectiologie, Faculté de Médecine RTH Laennec, Université Claude Bernard Lyon 1, Université de Lyon, INSERM, CNRS, ENS de Lyon, Lyon, France
| | - Chloé Mialon
- CIRI, Centre International de Recherche en Infectiologie (Team VirPath), Université de Lyon, INSERM U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, Lyon, France
- VirNext, Faculté de Médecine RTH Laennec, Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France
- International Research Laboratory RESPIVIR France-Canada, Centre Hospitalier Universitaire de Québec - Université Laval, Québec, Canada, Centre International de Recherche en Infectiologie, Faculté de Médecine RTH Laennec, Université Claude Bernard Lyon 1, Université de Lyon, INSERM, CNRS, ENS de Lyon, Lyon, France
| | - Stefanie M Bader
- Division of Infectious Diseases and Immune Defense, The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
| | - Georgios Marinos
- Research Group Medical Systems Biology, Institute of Experimental Medicine, Christian-Albrechts-University Kiel & University Hospital Schleswig Holstein, Kiel, Germany
| | - Nantia Leonidou
- Computational Systems Biology of Infections and Antimicrobial-Resistant Pathogens, Institute for Bioinformatics and Medical Informatics (IBMI), Eberhard Karl University of Tübingen, Tübingen, Germany
- German Center for Infection Research (DZIF), partner site, Tübingen, Germany
- Department of Computer Science, Eberhard Karl University of Tübingen, Tübingen, Germany
- Cluster of Excellence 'Controlling Microbes to Fight Infections', Eberhard Karl University of Tübingen, Tübingen, Germany
| | - Filipe Cabreiro
- Cologne Excellence Cluster for Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Marc Pellegrini
- Division of Infectious Diseases and Immune Defense, The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
| | - Marcel Doerflinger
- Division of Infectious Diseases and Immune Defense, The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
| | - Manuel Rosa-Calatrava
- CIRI, Centre International de Recherche en Infectiologie (Team VirPath), Université de Lyon, INSERM U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, Lyon, France
- VirNext, Faculté de Médecine RTH Laennec, Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France
- International Research Laboratory RESPIVIR France-Canada, Centre Hospitalier Universitaire de Québec - Université Laval, Québec, Canada, Centre International de Recherche en Infectiologie, Faculté de Médecine RTH Laennec, Université Claude Bernard Lyon 1, Université de Lyon, INSERM, CNRS, ENS de Lyon, Lyon, France
| | - Andrés Pizzorno
- CIRI, Centre International de Recherche en Infectiologie (Team VirPath), Université de Lyon, INSERM U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, Lyon, France
- VirNext, Faculté de Médecine RTH Laennec, Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France
- International Research Laboratory RESPIVIR France-Canada, Centre Hospitalier Universitaire de Québec - Université Laval, Québec, Canada, Centre International de Recherche en Infectiologie, Faculté de Médecine RTH Laennec, Université Claude Bernard Lyon 1, Université de Lyon, INSERM, CNRS, ENS de Lyon, Lyon, France
| | - Andreas Dräger
- Computational Systems Biology of Infections and Antimicrobial-Resistant Pathogens, Institute for Bioinformatics and Medical Informatics (IBMI), Eberhard Karl University of Tübingen, Tübingen, Germany.
- German Center for Infection Research (DZIF), partner site, Tübingen, Germany.
- Data Analytics and Bioinformatics Research Group, Institute of Computer Science, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany.
| | - Michael Schindler
- Institute for Medical Virology and Epidemiology of Viral Diseases, University Hospital Tübingen, Tübingen, Germany.
- German Center for Infection Research (DZIF), partner site, Tübingen, Germany.
| | - Christoph Kaleta
- Research Group Medical Systems Biology, Institute of Experimental Medicine, Christian-Albrechts-University Kiel & University Hospital Schleswig Holstein, Kiel, Germany.
| |
Collapse
|
3
|
Welling MS, van Rossum EFC, van den Akker ELT. Antiobesity Pharmacotherapy for Patients With Genetic Obesity Due to Defects in the Leptin-Melanocortin Pathway. Endocr Rev 2025; 46:418-446. [PMID: 39929239 PMCID: PMC12063102 DOI: 10.1210/endrev/bnaf004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Indexed: 05/10/2025]
Abstract
Lifestyle interventions are the cornerstone of obesity treatment. However, insufficient long-term effects are observed in patients with genetic obesity disorders, as their hyperphagia remains untreated. Hence, patients with genetic obesity often require additional pharmacotherapy to effectively manage and treat their hyperphagia and obesity. Recent advancements in antiobesity pharmacotherapy have expanded the range of available antiobesity medications (AOM). This includes the targeted AOM setmelanotide, approved for specific genetic obesity disorders, as well as nontargeted AOMs such as naltrexone-bupropion and glucagon-like peptide-1 analogues. Targeted AOMs have demonstrated significant weight loss, reduced obesity-related comorbidities, and improved hyperphagia and quality of life in patients with specific genetic obesity disorders. Small observational studies have shown that similar benefits from nontargeted AOMs or off-label pharmacotherapies can be achieved in patients with specific genetic obesity disorders, compared to common multifactorial obesity. In the future, novel and innovative pharmacotherapeutical options, including combination therapies and possibly gene therapy, will emerge, offering promising effects on body weight, hyperphagia, and, most importantly, quality of life for patients with a variety of genetic obesity disorders.
Collapse
Affiliation(s)
- Mila S Welling
- Obesity Center CGG, Erasmus MC, University Medical Center Rotterdam, Rotterdam 3015 GD, The Netherlands
- Department of Internal Medicine, Division of Endocrinology, Erasmus MC, University Medical Center Rotterdam, Rotterdam 3015 GD, The Netherlands
- Department of Pediatrics, Division of Endocrinology, Erasmus MC-Sophia Children's Hospital, University of Medical Center Rotterdam, Rotterdam 3015 GD, The Netherlands
| | - Elisabeth F C van Rossum
- Obesity Center CGG, Erasmus MC, University Medical Center Rotterdam, Rotterdam 3015 GD, The Netherlands
- Department of Internal Medicine, Division of Endocrinology, Erasmus MC, University Medical Center Rotterdam, Rotterdam 3015 GD, The Netherlands
| | - Erica L T van den Akker
- Obesity Center CGG, Erasmus MC, University Medical Center Rotterdam, Rotterdam 3015 GD, The Netherlands
- Department of Pediatrics, Division of Endocrinology, Erasmus MC-Sophia Children's Hospital, University of Medical Center Rotterdam, Rotterdam 3015 GD, The Netherlands
| |
Collapse
|
4
|
Weber D, Ferrario PG, Bub A. Exercise intensity determines circulating levels of Lac-Phe and other exerkines: a randomized crossover trial. Metabolomics 2025; 21:63. [PMID: 40335829 PMCID: PMC12058925 DOI: 10.1007/s11306-025-02260-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 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: 12/04/2024] [Accepted: 04/18/2025] [Indexed: 05/09/2025]
Abstract
INTRODUCTION Exercise metabolomics research has revealed significant exercise-induced metabolic changes and identified several exerkines as mediators of physiological adaptations to exercise. However, the effect of exercise intensity on metabolic changes and circulating exerkine levels remains to be examined. OBJECTIVES This study compared the metabolic responses to moderate-intensity and vigorous-intensity aerobic exercise. METHODS A two-period crossover trial was conducted under controlled conditions at the Max Rubner-Institute in Karlsruhe, Germany. Seventeen young, healthy, and physically active men performed 30 min moderate-intensity (50% VO2peak) and vigorous-intensity (75% VO2peak) aerobic exercise using two bicycle ergometer protocols in a randomized sequence. Blood samples obtained immediately before exercise and at four time points after exercise were analyzed in an untargeted metabolomics approach, and separate linear mixed models were applied to over 1000 metabolites. RESULTS Vigorous-intensity exercise induced a greater metabolic response than moderate-intensity exercise. Several intensity-dependent metabolites were identified, primarily involved in amino acid metabolism and energy conversion pathways, including N-lactoyl-amino acids, TCA cycle intermediates, N-acetylated amino acids, and acylcholines. The exerkines N-lactoyl-phenylalanine, lactate, and succinate were among the most intensity-dependent metabolites. N-acetylated amino acids and acylcholines were systematically altered by exercise intensity, indicating potential physiological functions. CONCLUSION Exercise intensity significantly affects exercise-induced metabolic alterations and changes in exerkine levels. Our results expand the knowledge about exerkine dynamics and emphasize the role of exercise intensity in promoting physiological adaptations to exercise. The trial was registered on October 5, 2017, at the German Clinical Trials Register under the Registration Number DRKS00009743 (Universal Trial Number of WHO: U1111-1200-2530).
Collapse
Affiliation(s)
- Dirk Weber
- Institute of Sports and Sports Science, Karlsruhe Institute of Technology (KIT), Engler-Bunte-Ring 15, 76131, Karlsruhe, Germany.
| | - Paola G Ferrario
- Department of Physiology and Biochemistry of Nutrition, Max Rubner-Institute, Karlsruhe, Germany
| | - Achim Bub
- Institute of Sports and Sports Science, Karlsruhe Institute of Technology (KIT), Engler-Bunte-Ring 15, 76131, Karlsruhe, Germany
- Department of Physiology and Biochemistry of Nutrition, Max Rubner-Institute, Karlsruhe, Germany
| |
Collapse
|
5
|
Sellami M, Naja K, Almuraikhy S, Anwardeen N, Sultanov RI, Generozov EV, Ahmetov II, Elrayess MA. N-Lactoyl amino acids as metabolic biomarkers differentiating low and high exercise response. Biol Sport 2025; 42:331-344. [PMID: 40182705 PMCID: PMC11963115 DOI: 10.5114/biolsport.2025.145912] [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: 06/03/2024] [Revised: 09/17/2024] [Accepted: 10/27/2024] [Indexed: 04/05/2025] Open
Abstract
Aerobic physical exercise has significant benefits for cardiovascular health; however, some individuals experience no benefit or even adverse effects. One reason for poor tolerance to aerobic exercise may be a low percentage of slow-twitch (oxidative) muscle fibers. This study aims to identify the metabolic signatures associated with low and high response to exercise by comparing the metabolic profiles of participants categorized according to their improvement of the 6-minute walking distance. In this study, pre- and postexercise intervention measurements of the 6-minute walking distance were conducted in forty-three lean and overweight young women, followed by non-targeted metabolomics analysis of 1039 known metabolites. An independent validation cohort comprising 791 individuals from the GTEx project was used to assess the gene expression of selected targets. The results indicated that a low improvement in the 6-minute walking distance (Δ 6-MWD = 27 meters) was associated with higher serum levels of N-lactoyl amino acid metabolites, particularly the exercise-inducible metabolite N-lactoyl phenylalanine (Lac-Phe) (FDR = 0.016), compared to high responders. Our results were corroborated in an independent validation cohort, which showed that the gene expression of cytosolic nonspecific dipeptidase (CNDP2), the enzyme responsible for Lac-Phe synthesis, is negatively associated with the percentage of slow-twitch muscle fibers (p < 0.0001). N-lactoyl amino acids may serve as biomarkers for rapid muscle fatigue and low response to exercise, and could be used as metabolic indicators to differentiate exercise response efficacy.
Collapse
Affiliation(s)
- Maha Sellami
- Physical Education Department (PE), College of Education, Qatar University, Doha, Qatar
| | - Khaled Naja
- Biomedical Research Center, Qatar University, Doha P.O Box 2713, Qatar
| | - Shamma Almuraikhy
- Biomedical Research Center, Qatar University, Doha P.O Box 2713, Qatar
| | - Najeha Anwardeen
- Biomedical Research Center, Qatar University, Doha P.O Box 2713, Qatar
| | - Rinat I. Sultanov
- Department of Molecular Biology and Genetics, Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, Russia
| | - Eduard V. Generozov
- Department of Molecular Biology and Genetics, Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, Russia
| | - Ildus I. Ahmetov
- Department of Molecular Biology and Genetics, Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, Russia
- Laboratory of Genetics of Aging and Longevity, Kazan State Medical University, Kazan, Russia
- Research Institute for Sport and Exercise Sciences, Liverpool John Moores University, Liverpool, UK
| | - Mohamed A. Elrayess
- Biomedical Research Center, Qatar University, Doha P.O Box 2713, Qatar
- College of Medicine, QU Health, Qatar University, Doha P.O Box 2713, Qatar
| |
Collapse
|
6
|
Madsen KS, Henriksen M, Døssing A, Poulsen AS, Oscar R, Kragstrup T, Ellegaard K, Knop FK, Boesen M, Hunter DJ, Christensen R, Bliddal H. Metformin treatment for patients with hand osteoarthritis: protocol for the multicentre, randomised, placebo-controlled METRO trial. BMJ Open 2025; 15:e093831. [PMID: 40139705 PMCID: PMC11950932 DOI: 10.1136/bmjopen-2024-093831] [Citation(s) in RCA: 0] [Impact Index Per Article: 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: 09/17/2024] [Accepted: 02/21/2025] [Indexed: 03/29/2025] Open
Abstract
INTRODUCTION Hand osteoarthritis (OA) is a prevalent joint disorder with limited treatment options. Accumulating evidence suggests that the antidiabetic drug metformin has beneficial effects on knee OA and may likewise be beneficial for hand OA. The objective of this randomised, double-blinded, placebo-controlled trial is to investigate the effect of metformin 1000 mg two times a day, or maximum tolerated dose, compared with placebo on reducing finger joint pain after 16 weeks of treatment. METHODS AND ANALYSIS The participants will be enrolled from the OA clinic at the Parker Institute at Copenhagen University Hospital - Bispebjerg and Frederiksberg, Copenhagen, Denmark and from the Department of Rheumatology, Hospitalsenhed Midt, Silkeborg, Denmark. 150 participants with painful hand OA according to the American College of Rheumatology criteria will be randomly allocated in a 1:1 ratio to receive either metformin or a matching placebo for 16 weeks. The initial dose of 500 mg of metformin or placebo once daily is increased by 500 mg every week until the target dose of 1000 mg two times a day, or the maximum tolerated dose, is reached. The participants will have clinical visits every 4 weeks, except the week 12 visit, which is by telephone. The primary endpoint is the between-group difference in least squares means for the change in the Visual Analogue Scale (VAS) finger joint pain scores between the metformin and placebo groups at 16 weeks. The main analysis will be conducted on the intention-to-treat population, comprising all participants assessed and randomly assigned at baseline. Least squares means and the differences between them, along with their respective 95% CIs, will be derived from a mixed-effects model for repeated measurements (outcomes collected at baseline and at weeks 4, 8, 12 and 16). Adverse events will be registered systematically. ETHICS AND DISSEMINATION Approval has been obtained from the European Medicines Agency (EudraCT: 2023-509181-38-00), which also includes approval from the local health research ethics committee. Written informed consent will be obtained from all participants. Study findings will be published in international peer-reviewed journals and will be presented in relevant media and at international scientific conferences. TRIAL REGISTRATION NUMBER EudraCT, 2023-509181-38-00; ClinicalTrials.gov, NCT06367283.
Collapse
Affiliation(s)
- Kasper Staberg Madsen
- The Parker Institute, Bispebjerg and Frederiksberg Hospital, University of Copenhagen, Frederiksberg, Copenhagen, Denmark
| | - Marius Henriksen
- The Parker Institute, Bispebjerg and Frederiksberg Hospital, University of Copenhagen, Frederiksberg, Copenhagen, Denmark
| | - Anna Døssing
- The Parker Institute, Bispebjerg and Frederiksberg Hospital, University of Copenhagen, Frederiksberg, Copenhagen, Denmark
| | - Asbjørn S Poulsen
- The Parker Institute, Bispebjerg and Frederiksberg Hospital, University of Copenhagen, Frederiksberg, Copenhagen, Denmark
| | - Rasmus Oscar
- Medical Diagnostic Center, Regional Hospital Silkeborg, Silkeborg, Denmark
| | - Tue Kragstrup
- Medical Diagnostic Center, Regional Hospital Silkeborg, Silkeborg, Denmark
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Karen Ellegaard
- The Parker Institute, Bispebjerg and Frederiksberg Hospital, University of Copenhagen, Frederiksberg, Copenhagen, Denmark
| | - Filip K Knop
- Center for Clinical Metabolic Research, Gentofte Hospital, University of Copenhagen, Hellerup, Denmark
- Novo Nordisk A/S, Søborg, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Mikael Boesen
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Radiology, Copenhagen University Hospital, Bispebjerg and Frederiksberg, Bispebjerg, Copenhagen, Denmark
| | - David J Hunter
- Rheumatology Department, Royal North Shore Hospital, St Leonards, Sydney, New South Wales, Australia
- Sydney Musculoskeletal Health, Kolling Institute of Medical Research, The University of Sydney, Sydney, New South Wales, Australia
| | - Robin Christensen
- The Parker Institute, Bispebjerg and Frederiksberg Hospital, University of Copenhagen, Frederiksberg, Copenhagen, Denmark
- Research Unit of Rheumatology, Department of Clinical Research, University of Southern Denmark, Odense University Hospital, Odense, Denmark
| | - Henning Bliddal
- The Parker Institute, Bispebjerg and Frederiksberg Hospital, University of Copenhagen, Frederiksberg, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
7
|
Xie C, Iroga P, Bound MJ, Grivell J, Huang W, Jones KL, Horowitz M, Rayner CK, Wu T. Impact of the timing of metformin administration on the plasma lactate response to intraduodenal glucose infusion in type 2 diabetes. Diabetes Obes Metab 2025; 27:1614-1617. [PMID: 39727162 DOI: 10.1111/dom.16169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 12/08/2024] [Accepted: 12/17/2024] [Indexed: 12/28/2024]
Affiliation(s)
- Cong Xie
- Adelaide Medical School and Centre of Research Excellence (CRE) in Translating Nutritional Science to Good Health, The University of Adelaide, Adelaide, South Australia, Australia
- Endocrine and Metabolic Unit, Royal Adelaide Hospital, Adelaide, South Australia, Australia
| | - Peter Iroga
- Adelaide Medical School and Centre of Research Excellence (CRE) in Translating Nutritional Science to Good Health, The University of Adelaide, Adelaide, South Australia, Australia
| | - Michelle J Bound
- Adelaide Medical School and Centre of Research Excellence (CRE) in Translating Nutritional Science to Good Health, The University of Adelaide, Adelaide, South Australia, Australia
| | - Jacqueline Grivell
- Adelaide Medical School and Centre of Research Excellence (CRE) in Translating Nutritional Science to Good Health, The University of Adelaide, Adelaide, South Australia, Australia
| | - Weikun Huang
- Adelaide Medical School and Centre of Research Excellence (CRE) in Translating Nutritional Science to Good Health, The University of Adelaide, Adelaide, South Australia, Australia
| | - Karen L Jones
- Adelaide Medical School and Centre of Research Excellence (CRE) in Translating Nutritional Science to Good Health, The University of Adelaide, Adelaide, South Australia, Australia
- Endocrine and Metabolic Unit, Royal Adelaide Hospital, Adelaide, South Australia, Australia
| | - Michael Horowitz
- Adelaide Medical School and Centre of Research Excellence (CRE) in Translating Nutritional Science to Good Health, The University of Adelaide, Adelaide, South Australia, Australia
- Endocrine and Metabolic Unit, Royal Adelaide Hospital, Adelaide, South Australia, Australia
| | - Christopher K Rayner
- Adelaide Medical School and Centre of Research Excellence (CRE) in Translating Nutritional Science to Good Health, The University of Adelaide, Adelaide, South Australia, Australia
- Department of Gastroenterology and Hepatology, Royal Adelaide Hospital, Adelaide, South Australia, Australia
| | - Tongzhi Wu
- Adelaide Medical School and Centre of Research Excellence (CRE) in Translating Nutritional Science to Good Health, The University of Adelaide, Adelaide, South Australia, Australia
- Endocrine and Metabolic Unit, Royal Adelaide Hospital, Adelaide, South Australia, Australia
| |
Collapse
|
8
|
Ocariza MGC, Paton LN, Templeton EM, Pemberton CJ, Pilbrow AP, Appleby S. CNDP2: An Enzyme Linking Metabolism and Cardiovascular Diseases? J Cardiovasc Transl Res 2025; 18:48-57. [PMID: 39349903 PMCID: PMC11885389 DOI: 10.1007/s12265-024-10560-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 09/12/2024] [Indexed: 03/09/2025]
Abstract
The heart requires a substantial amount of energy to function, utilising various substrates including lipids, glucose and lactate as energy sources. In times of increased stress, lactate becomes the primary energy source of the heart, but persistently elevated lactate levels are linked to poor patient outcomes and increased mortality. Recently, carnosine dipeptidase II (CNDP2) was discovered to catalyse the formation of Lac-Phe, an exercise-induced metabolite derived from lactate, which has been shown to suppress appetite in mice and reduce adipose tissue in humans. This review discusses CNDP2, including its role in lactate clearance, carnosine hydrolysis, oxidative stress regulation, and involvement in metabolite regulation. The association between CNDP2 and cardiometabolic and renal diseases is also explored, and knowledge gaps are highlighted. CNDP2 appears to be a complex participant in human physiological processes and disease, necessitating additional research to unveil its functions and potential therapeutic applications.
Collapse
Affiliation(s)
- Moizle Grace Castro Ocariza
- Department of Medicine, Christchurch Heart Institute, University of Otago (Christchurch), Christchurch, New Zealand.
| | - Louise Nancy Paton
- Department of Medicine, Christchurch Heart Institute, University of Otago (Christchurch), Christchurch, New Zealand
| | - Evelyn Mary Templeton
- Department of Medicine, Christchurch Heart Institute, University of Otago (Christchurch), Christchurch, New Zealand
| | - Christopher Joseph Pemberton
- Department of Medicine, Christchurch Heart Institute, University of Otago (Christchurch), Christchurch, New Zealand
| | - Anna Pauline Pilbrow
- Department of Medicine, Christchurch Heart Institute, University of Otago (Christchurch), Christchurch, New Zealand
| | - Sarah Appleby
- Department of Medicine, Christchurch Heart Institute, University of Otago (Christchurch), Christchurch, New Zealand
| |
Collapse
|
9
|
Moya-Garzon MD, Wang M, Li VL, Lyu X, Wei W, Tung ASH, Raun SH, Zhao M, Coassolo L, Islam H, Oliveira B, Dai Y, Spaas J, Delgado-Gonzalez A, Donoso K, Alvarez-Buylla A, Franco-Montalban F, Letian A, Ward CP, Liu L, Svensson KJ, Goldberg EL, Gardner CD, Little JP, Banik SM, Xu Y, Long JZ. A β-hydroxybutyrate shunt pathway generates anti-obesity ketone metabolites. Cell 2025; 188:175-186.e20. [PMID: 39536746 PMCID: PMC11724754 DOI: 10.1016/j.cell.2024.10.032] [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: 07/23/2023] [Revised: 06/12/2024] [Accepted: 10/16/2024] [Indexed: 11/16/2024]
Abstract
β-Hydroxybutyrate (BHB) is an abundant ketone body. To date, all known pathways of BHB metabolism involve the interconversion of BHB and primary energy intermediates. Here, we identify a previously undescribed BHB secondary metabolic pathway via CNDP2-dependent enzymatic conjugation of BHB and free amino acids. This BHB shunt pathway generates a family of anti-obesity ketone metabolites, the BHB-amino acids. Genetic ablation of CNDP2 in mice eliminates tissue amino acid BHB-ylation activity and reduces BHB-amino acid levels. The most abundant BHB-amino acid, BHB-Phe, is a ketosis-inducible congener of Lac-Phe that activates hypothalamic and brainstem neurons and suppresses feeding. Conversely, CNDP2-KO mice exhibit increased food intake and body weight following exogenous ketone ester supplementation or a ketogenic diet. CNDP2-dependent amino acid BHB-ylation and BHB-amino acid metabolites are also conserved in humans. Therefore, enzymatic amino acid BHB-ylation defines a ketone shunt pathway and bioactive ketone metabolites linked to energy balance.
Collapse
Affiliation(s)
- Maria Dolores Moya-Garzon
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA; Sarafan ChEM-H, Stanford University, Stanford, CA, USA; Wu Tsai Human Performance Alliance, Stanford University, Stanford, CA, USA
| | - Mengjie Wang
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Veronica L Li
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA; Department of Chemistry, Stanford University, Stanford, CA, USA; Sarafan ChEM-H, Stanford University, Stanford, CA, USA; Wu Tsai Human Performance Alliance, Stanford University, Stanford, CA, USA
| | - Xuchao Lyu
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA; Sarafan ChEM-H, Stanford University, Stanford, CA, USA; Wu Tsai Human Performance Alliance, Stanford University, Stanford, CA, USA
| | - Wei Wei
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA; Department of Biology, Stanford University, Stanford, CA, USA; Sarafan ChEM-H, Stanford University, Stanford, CA, USA
| | - Alan Sheng-Hwa Tung
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA; Department of Biology, Stanford University, Stanford, CA, USA; Sarafan ChEM-H, Stanford University, Stanford, CA, USA
| | - Steffen H Raun
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Meng Zhao
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA; Stanford Diabetes Research Center, Stanford University, Stanford, CA, USA; Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Laetitia Coassolo
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA; Stanford Diabetes Research Center, Stanford University, Stanford, CA, USA; Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Hashim Islam
- School of Health and Exercise Sciences, University of British Columbia, Kelowna, BC, Canada
| | - Barbara Oliveira
- School of Health and Exercise Sciences, University of British Columbia, Kelowna, BC, Canada
| | - Yuqin Dai
- Sarafan ChEM-H, Stanford University, Stanford, CA, USA
| | - Jan Spaas
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA; Sarafan ChEM-H, Stanford University, Stanford, CA, USA
| | | | - Kenyi Donoso
- Department of Urology, Stanford University School of Medicine, Stanford, CA, USA
| | | | - Francisco Franco-Montalban
- Departamento de Química Farmacéutica y Orgánica, Universidad de Granada, Campus de Cartuja sn, 18071 Granada, Spain
| | - Anudari Letian
- Department of Physiology, University of California, San Francisco, San Francisco, CA, USA
| | - Catherine P Ward
- Stanford Prevention Research Center, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Lichao Liu
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Katrin J Svensson
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA; Stanford Diabetes Research Center, Stanford University, Stanford, CA, USA; Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Emily L Goldberg
- Department of Physiology, University of California, San Francisco, San Francisco, CA, USA
| | - Christopher D Gardner
- Stanford Prevention Research Center, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Jonathan P Little
- School of Health and Exercise Sciences, University of British Columbia, Kelowna, BC, Canada
| | - Steven M Banik
- Department of Chemistry, Stanford University, Stanford, CA, USA; Sarafan ChEM-H, Stanford University, Stanford, CA, USA
| | - Yong Xu
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA.
| | - Jonathan Z Long
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA; Sarafan ChEM-H, Stanford University, Stanford, CA, USA; Stanford Diabetes Research Center, Stanford University, Stanford, CA, USA; Wu Tsai Human Performance Alliance, Stanford University, Stanford, CA, USA; The Phil & Penny Knight Initiative for Brain Resilience at the Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA.
| |
Collapse
|
10
|
Zhang Y, Spitzer BW, Zhang Y, Wallace DA, Yu B, Qi Q, Argos M, Avilés-Santa ML, Boerwinkle E, Daviglus ML, Kaplan R, Cai J, Redline S, Sofer T. Untargeted metabolome atlas for sleep-related phenotypes in the Hispanic community health study/study of Latinos. EBioMedicine 2025; 111:105507. [PMID: 39693737 PMCID: PMC11722176 DOI: 10.1016/j.ebiom.2024.105507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 11/25/2024] [Accepted: 12/04/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND Sleep is essential to maintaining health and wellbeing of individuals, influencing a variety of outcomes from mental health to cardiometabolic disease. This study aims to assess the relationships between various sleep-related phenotypes and blood metabolites. METHODS Utilising data from the Hispanic Community Health Study/Study of Latinos, we performed association analyses between 40 sleep-related phenotypes, grouped in several domains (sleep disordered breathing (SDB), sleep duration, sleep timing, self-reported insomnia symptoms, excessive daytime sleepiness (EDS), and heart rate during sleep), and 768 metabolites measured via untargeted metabolomics profiling. Network analysis was employed to visualise and interpret the associations between sleep phenotypes and metabolites. FINDINGS The patterns of statistically significant associations between sleep phenotypes and metabolites differed by superpathways, and highlighted subpathways of interest for future studies. For example, primary bile acid metabolism showed the highest cumulative percentage of statistically significant associations across all sleep phenotype domains except for SDB and EDS phenotypes. Several metabolites were associated with multiple sleep phenotypes, from a few domains. Glycochenodeoxycholate, vanillyl mandelate (VMA) and 1-stearoyl-2-oleoyl-GPE (18:0/18:1) were associated with the highest number of sleep phenotypes, while pregnenolone sulfate was associated with all sleep phenotype domains except for sleep duration. N-lactoyl amino acids such as N-lactoyl phenylalanine (lac-Phe), were associated with sleep duration, SDB, sleep timing and heart rate during sleep. INTERPRETATION This atlas of sleep-metabolite associations will facilitate hypothesis generation and further study of the metabolic underpinnings of sleep health. FUNDING R01HL161012, R35HL135818, R01AG80598.
Collapse
Affiliation(s)
- Ying Zhang
- Division of Sleep Medicine and Circadian Disorders, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Brian W Spitzer
- CardioVascular Institute, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Yu Zhang
- CardioVascular Institute, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Danielle A Wallace
- Division of Sleep Medicine and Circadian Disorders, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA; CardioVascular Institute, Beth Israel Deaconess Medical Center, Boston, MA, USA; Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Bing Yu
- Department of Epidemiology, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Qibin Qi
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Maria Argos
- Department of Epidemiology and Biostatistics, School of Public Health, University of Illinois Chicago, Chicago, IL, USA; Department of Environmental Health, School of Public Health, Boston University, Boston, MA, USA
| | - M Larissa Avilés-Santa
- Division of Clinical and Health Services Research, National Institute on Minority Health and Health Disparities, National Institutes of Health, Bethesda, MD, USA
| | - Eric Boerwinkle
- Department of Epidemiology, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Martha L Daviglus
- Institute for Minority Health Research, University of Illinois at Chicago, Chicago, IL, USA
| | - Robert Kaplan
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, USA; Division of Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Jianwen Cai
- Collaborative Studies Coordinating Center, Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Susan Redline
- Division of Sleep Medicine and Circadian Disorders, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA; Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Tamar Sofer
- Division of Sleep Medicine and Circadian Disorders, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA; CardioVascular Institute, Beth Israel Deaconess Medical Center, Boston, MA, USA; Department of Medicine, Harvard Medical School, Boston, MA, USA; Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA.
| |
Collapse
|
11
|
Reczek CR, Chakrabarty RP, D'Alessandro KB, Sebo ZL, Grant RA, Gao P, Budinger GR, Chandel NS. Metformin targets mitochondrial complex I to lower blood glucose levels. SCIENCE ADVANCES 2024; 10:eads5466. [PMID: 39693440 DOI: 10.1126/sciadv.ads5466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 11/19/2024] [Indexed: 12/20/2024]
Abstract
Metformin is among the most prescribed antidiabetic drugs, but the primary molecular mechanism by which metformin lowers blood glucose levels is unknown. Previous studies have proposed numerous mechanisms by which acute metformin lowers blood glucose, including the inhibition of mitochondrial complex I of the electron transport chain (ETC). Here, we used transgenic mice that globally express the Saccharomyces cerevisiae internal alternative NADH dehydrogenase (NDI1) protein to determine whether the glucose-lowering effect of acute oral administration of metformin requires inhibition of mitochondrial complex I of the ETC in vivo. NDI1 is a yeast NADH dehydrogenase enzyme that complements the loss of mammalian mitochondrial complex I electron transport function and is insensitive to pharmacologic mitochondrial complex I inhibitors including metformin. We demonstrate that NDI1 expression attenuates metformin's ability to lower blood glucose levels under standard chow and high-fat diet conditions. Our results indicate that acute oral administration of metformin targets mitochondrial complex I to lower blood glucose.
Collapse
Affiliation(s)
- Colleen R Reczek
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Ram P Chakrabarty
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Karis B D'Alessandro
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Zachary L Sebo
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Rogan A Grant
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Peng Gao
- Robert H. Lurie Cancer Center Metabolomics Core, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - G R Budinger
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Navdeep S Chandel
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Chan Zuckerberg Biohub, Chicago, IL, USA
| |
Collapse
|
12
|
Green DR. Meeting metformin again for the first time. SCIENCE ADVANCES 2024; 10:eadu7436. [PMID: 39693446 PMCID: PMC11654691 DOI: 10.1126/sciadv.adu7436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Accepted: 12/03/2024] [Indexed: 12/20/2024]
Abstract
New evidence convincingly shows that metformin, a drug that reduces circulating glucose, acts by inhibiting mitochondrial complex I.
Collapse
Affiliation(s)
- Douglas R. Green
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, TN 38139, USA
| |
Collapse
|
13
|
Ahmad S, Wu T, Arnold M, Hankemeier T, Ghanbari M, Roshchupkin G, Uitterlinden AG, Neitzel J, Kraaij R, Van Duijn CM, Arfan Ikram M, Kaddurah-Daouk R, Kastenmüller G, Alzheimer’s Disease Metabolomics Consortium. The blood metabolome of cognitive function and brain health in middle-aged adults - influences of genes, gut microbiome, and exposome. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.12.16.24317793. [PMID: 39763567 PMCID: PMC11702749 DOI: 10.1101/2024.12.16.24317793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/11/2025]
Abstract
Increasing evidence suggests the involvement of metabolic alterations in neurological disorders, including Alzheimer's disease (AD), and highlights the significance of the peripheral metabolome, influenced by genetic factors and modifiable environmental exposures, for brain health. In this study, we examined 1,387 metabolites in plasma samples from 1,082 dementia-free middle-aged participants of the population-based Rotterdam Study. We assessed the relation of metabolites with general cognition (G-factor) and magnetic resonance imaging (MRI) markers using linear regression and estimated the variance of these metabolites explained by genes, gut microbiome, lifestyle factors, common clinical comorbidities, and medication using gradient boosting decision tree analysis. Twenty-one metabolites and one metabolite were significantly associated with total brain volume and total white matter lesions, respectively. Fourteen metabolites showed significant associations with G-factor, with ergothioneine exhibiting the largest effect (adjusted mean difference = 0.122, P = 4.65×10-7). Associations for nine of the 14 metabolites were replicated in an independent, older cohort. The metabolite signature of incident AD in the replication cohort resembled that of cognition in the discovery cohort, emphasizing the potential relevance of the identified metabolites to disease pathogenesis. Lifestyle, clinical variables, and medication were most important in determining these metabolites' blood levels, with lifestyle, explaining up to 28.6% of the variance. Smoking was associated with ten metabolites linked to G-factor, while diabetes and antidiabetic medication were associated with 13 metabolites linked to MRI markers, including N-lactoyltyrosine. Antacid medication strongly affected ergothioneine levels. Mediation analysis revealed that lower ergothioneine levels may partially mediate negative effects of antacids on cognition (31.5%). Gut microbial factors were more important for the blood levels of metabolites that were more strongly associated with cognition and incident AD in the older replication cohort (beta-cryptoxanthin, imidazole propionate), suggesting they may be involved later in the disease process. The detailed results on how multiple modifiable factors affect blood levels of cognition- and brain imaging-related metabolites in dementia-free participants may help identify new AD prevention strategies.
Collapse
Affiliation(s)
- Shahzad Ahmad
- Department of Epidemiology, Erasmus MC, University Medical Centre, Rotterdam, The Netherlands
- Division of Systems Biomedicine and Pharmacology, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Tong Wu
- Institute of Computational Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Matthias Arnold
- Institute of Computational Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Department of Psychiatry and Behavioral Sciences, Duke University, Durham, NC, USA
| | - Thomas Hankemeier
- Division of Systems Biomedicine and Pharmacology, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Mohsen Ghanbari
- Department of Epidemiology, Erasmus MC, University Medical Centre, Rotterdam, The Netherlands
| | - Gennady Roshchupkin
- Department of Epidemiology, Erasmus MC, University Medical Centre, Rotterdam, The Netherlands
| | - André G. Uitterlinden
- Department of Internal Medicine, Erasmus MC, University Medical Centre, Rotterdam, The Netherlands
| | - Julia Neitzel
- Department of Epidemiology, Erasmus MC, University Medical Centre, Rotterdam, The Netherlands
- Department of Radiology and Nuclear Medicine, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
- Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Robert Kraaij
- Department of Internal Medicine, Erasmus MC, University Medical Centre, Rotterdam, The Netherlands
| | - Cornelia M. Van Duijn
- Department of Epidemiology, Erasmus MC, University Medical Centre, Rotterdam, The Netherlands
- Nuffield Department of Population Health, Oxford University, Oxford, UK
| | - M. Arfan Ikram
- Department of Epidemiology, Erasmus MC, University Medical Centre, Rotterdam, The Netherlands
| | - Rima Kaddurah-Daouk
- Department of Psychiatry and Behavioral Sciences, Duke University, Durham, NC, USA
- Duke Institute of Brain Sciences, Duke University, Durham, NC, USA
- Department of Medicine, Duke University, Durham, NC, USA
| | - Gabi Kastenmüller
- Institute of Computational Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | | |
Collapse
|
14
|
Kim HS, Jung S, Kim MJ, Jeong JY, Hwang IM, Lee JH. Comparative Analysis of N-Lactoyl-phenylalanine and 3-Phenyllactic Acid Production in Lactic Acid Bacteria from Kimchi: Metabolic Insights and Influencing Factors. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:27177-27186. [PMID: 39606886 DOI: 10.1021/acs.jafc.4c07158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
N-Lactoyl-phenylalanine (Lac-Phe) is a metabolite known for its appetite-suppressing and antiobesity properties, while phenyllactic acid (PLA) is recognized for its antibacterial activity. Both metabolites are derived from phenylalanine and lactic acid metabolism through peptidase and dehydrogenase activities. The aim of this study was to investigate the production of Lac-Phe and PLA in kimchi, focusing on the role of lactic acid bacteria (LAB). Ultrahigh performance liquid chromatography coupled with time-of-flight mass spectrometry was used to quantify these metabolites in homemade and commercial kimchi. Lac-Phe and PLA were detected in both kimchi sample types. Various genera, including Lactobacillus, Leuconostoc, and Weissella, were evaluated for Lac-Phe and PLA production. LAB strains exhibiting high Lac-Phe production generally showed lower PLA production, indicating an inverse relationship between these two metabolites. Analysis of dipeptidase sequences revealed that the presence of carnosine dipeptidase 2 (CNDP2)-like M20 metallopeptidase is crucial for Lac-Phe production in LAB. Additionally, phenylalanine was identified as a major factor for both Lac-Phe and PLA production, whereas lactic acid supplementation did not significantly affect their production levels.
Collapse
Affiliation(s)
- Hyun-Sung Kim
- Fermentation Regulation Research Group, Technology Innovation Research Division, World Institute of Kimchi, Gwangju 61755, Republic of Korea
| | - Sera Jung
- Fermentation Regulation Research Group, Technology Innovation Research Division, World Institute of Kimchi, Gwangju 61755, Republic of Korea
| | - Min Ji Kim
- Fermentation Regulation Research Group, Technology Innovation Research Division, World Institute of Kimchi, Gwangju 61755, Republic of Korea
| | - Ji Young Jeong
- Fermentation Regulation Research Group, Technology Innovation Research Division, World Institute of Kimchi, Gwangju 61755, Republic of Korea
| | - In Min Hwang
- Fermentation Regulation Research Group, Technology Innovation Research Division, World Institute of Kimchi, Gwangju 61755, Republic of Korea
| | - Jong-Hee Lee
- Fermentation Regulation Research Group, Technology Innovation Research Division, World Institute of Kimchi, Gwangju 61755, Republic of Korea
| |
Collapse
|
15
|
Hinton A, Neikirk K, Le H, Oliver A, Harris C, Martin P, Gaye A. N-lactoyl phenylalanine suppresses appetite and obesity with important implications for aging and age-related diseases. AGING ADVANCES 2024; 1:172-173. [PMID: 39845129 PMCID: PMC11752169 DOI: 10.4103/agingadv.agingadv-d-24-00011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 11/21/2024] [Indexed: 01/24/2025]
Affiliation(s)
- Antentor Hinton
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Kit Neikirk
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Han Le
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Ashton Oliver
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, TN, USA; Department of Biomedical Sciences, Meharry Medical College, Nashville, TN, USA
| | - Chanel Harris
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, TN, USA; Department of Biomedical Sciences, Meharry Medical College, Nashville, TN, USA
| | - Pamela Martin
- Department of Biomedical Sciences, Meharry Medical College, Nashville, TN, USA
| | - Amadou Gaye
- Department of Integrative Genomics and Epidemiology, Meharry Medical College, Nashville, TN, USA
| |
Collapse
|
16
|
Sharma S, Dong Q, Haid M, Adam J, Bizzotto R, Fernandez-Tajes JJ, Jones AG, Tura A, Artati A, Prehn C, Kastenmüller G, Koivula RW, Franks PW, Walker M, Forgie IM, Giordano G, Pavo I, Ruetten H, Dermitzakis M, McCarthy MI, Pedersen O, Schwenk JM, Tsirigos KD, De Masi F, Brunak S, Viñuela A, Mari A, McDonald TJ, Kokkola T, Adamski J, Pearson ER, Grallert H. Role of human plasma metabolites in prediabetes and type 2 diabetes from the IMI-DIRECT study. Diabetologia 2024; 67:2804-2818. [PMID: 39349772 PMCID: PMC11604760 DOI: 10.1007/s00125-024-06282-6] [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: 02/28/2024] [Accepted: 07/29/2024] [Indexed: 11/29/2024]
Abstract
AIMS/HYPOTHESIS Type 2 diabetes is a chronic condition that is caused by hyperglycaemia. Our aim was to characterise the metabolomics to find their association with the glycaemic spectrum and find a causal relationship between metabolites and type 2 diabetes. METHODS As part of the Innovative Medicines Initiative - Diabetes Research on Patient Stratification (IMI-DIRECT) consortium, 3000 plasma samples were measured with the Biocrates AbsoluteIDQ p150 Kit and Metabolon analytics. A total of 911 metabolites (132 targeted metabolomics, 779 untargeted metabolomics) passed the quality control. Multivariable linear and logistic regression analysis estimates were calculated from the concentration/peak areas of each metabolite as an explanatory variable and the glycaemic status as a dependent variable. This analysis was adjusted for age, sex, BMI, study centre in the basic model, and additionally for alcohol, smoking, BP, fasting HDL-cholesterol and fasting triacylglycerol in the full model. Statistical significance was Bonferroni corrected throughout. Beyond associations, we investigated the mediation effect and causal effects for which causal mediation test and two-sample Mendelian randomisation (2SMR) methods were used, respectively. RESULTS In the targeted metabolomics, we observed four (15), 34 (99) and 50 (108) metabolites (number of metabolites observed in untargeted metabolomics appear in parentheses) that were significantly different when comparing normal glucose regulation vs impaired glucose regulation/prediabetes, normal glucose regulation vs type 2 diabetes, and impaired glucose regulation vs type 2 diabetes, respectively. Significant metabolites were mainly branched-chain amino acids (BCAAs), with some derivatised BCAAs, lipids, xenobiotics and a few unknowns. Metabolites such as lysophosphatidylcholine a C17:0, sum of hexoses, amino acids from BCAA metabolism (including leucine, isoleucine, valine, N-lactoylvaline, N-lactoylleucine and formiminoglutamate) and lactate, as well as an unknown metabolite (X-24295), were associated with HbA1c progression rate and were significant mediators of type 2 diabetes from baseline to 18 and 48 months of follow-up. 2SMR was used to estimate the causal effect of an exposure on an outcome using summary statistics from UK Biobank genome-wide association studies. We found that type 2 diabetes had a causal effect on the levels of three metabolites (hexose, glutamate and caproate [fatty acid (FA) 6:0]), whereas lipids such as specific phosphatidylcholines (PCs) (namely PC aa C36:2, PC aa C36:5, PC ae C36:3 and PC ae C34:3) as well as the two n-3 fatty acids stearidonate (18:4n3) and docosapentaenoate (22:5n3) potentially had a causal role in the development of type 2 diabetes. CONCLUSIONS/INTERPRETATION Our findings identify known BCAAs and lipids, along with novel N-lactoyl-amino acid metabolites, significantly associated with prediabetes and diabetes, that mediate the effect of diabetes from baseline to follow-up (18 and 48 months). Causal inference using genetic variants shows the role of lipid metabolism and n-3 fatty acids as being causal for metabolite-to-type 2 diabetes whereas the sum of hexoses is causal for type 2 diabetes-to-metabolite. Identified metabolite markers are useful for stratifying individuals based on their risk progression and should enable targeted interventions.
Collapse
Affiliation(s)
- Sapna Sharma
- Research Unit of Molecular Epidemiology, Institute of Epidemiology, German Research Center for Environmental Health, Helmholtz Zentrum München, Neuherberg, Germany.
| | - Qiuling Dong
- Research Unit of Molecular Epidemiology, Institute of Epidemiology, German Research Center for Environmental Health, Helmholtz Zentrum München, Neuherberg, Germany
- Faculty of Medicine, Ludwig-Maximilians-University München, Munich, Germany
| | - Mark Haid
- Metabolomics and Proteomics Core, German Research Center for Environmental Health, Helmholtz Zentrum München, Neuherberg, Germany
| | - Jonathan Adam
- Research Unit of Molecular Epidemiology, Institute of Epidemiology, German Research Center for Environmental Health, Helmholtz Zentrum München, Neuherberg, Germany
- German Center for Diabetes Research (DZD), München Neuherberg, Germany
| | - Roberto Bizzotto
- Institute of Neuroscience, National Research Council, Padova, Italy
| | | | - Angus G Jones
- Department of Clinical and Biomedical Sciences, University of Exeter College of Medicine & Health, Exeter, UK
| | - Andrea Tura
- Institute of Neuroscience, National Research Council, Padova, Italy
| | - Anna Artati
- Metabolomics and Proteomics Core, German Research Center for Environmental Health, Helmholtz Zentrum München, Neuherberg, Germany
| | - Cornelia Prehn
- Metabolomics and Proteomics Core, German Research Center for Environmental Health, Helmholtz Zentrum München, Neuherberg, Germany
| | - Gabi Kastenmüller
- Institute of Computational Biology, Helmholtz Zentrum München, Munich, Germany
| | - Robert W Koivula
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK
| | - Paul W Franks
- Department of Clinical Science, Genetic and Molecular Epidemiology, Lund University Diabetes Centre, Malmö, Sweden
| | - Mark Walker
- Translational and Clinical Research Institute, Faculty of Medical Sciences, University of Newcastle, Newcastle upon Tyne, UK
| | - Ian M Forgie
- Population Health and Genomics, Ninewells Hospital and Medical School, University of Dundee, Dundee, UK
| | - Giuseppe Giordano
- Department of Clinical Science, Genetic and Molecular Epidemiology, Lund University Diabetes Centre, Malmö, Sweden
| | - Imre Pavo
- Eli Lilly Regional Operations GmbH, Vienna, Austria
| | - Hartmut Ruetten
- Sanofi Partnering, Sanofi-Aventis Deutschland GmbH, Frankfurt am Main, Germany
| | - Manolis Dermitzakis
- Department of Genetic Medicine and Development, University of Geneva Medical School, Geneva, Switzerland
- Institute for Genetics and Genomics in Geneva (iGE3), University of Geneva, Geneva, Switzerland
- Swiss Institute of Bioinformatics, Geneva, Switzerland
| | - Mark I McCarthy
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Oluf Pedersen
- Center for Clinical Metabolic Research, Herlev and Gentofte University Hospital, Copenhagen, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jochen M Schwenk
- Science for Life Laboratory, School of Biotechnology, KTH - Royal Institute of Technology, Solna, Sweden
| | | | - Federico De Masi
- Department of Health Technology, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Soren Brunak
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Health Technology, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Ana Viñuela
- Biosciences Institute, Faculty of Medical Sciences, University of Newcastle, Newcastle upon Tyne, UK
| | - Andrea Mari
- Institute of Neuroscience, National Research Council, Padova, Italy
| | | | - Tarja Kokkola
- Internal Medicine, Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland
| | - Jerzy Adamski
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Institute of Experimental Genetics, German Research Center for Environmental Health, Helmholtz Zentrum München, Neuherberg, Germany
- Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Ewan R Pearson
- Population Health and Genomics, Ninewells Hospital and Medical School, University of Dundee, Dundee, UK
| | - Harald Grallert
- Research Unit of Molecular Epidemiology, Institute of Epidemiology, German Research Center for Environmental Health, Helmholtz Zentrum München, Neuherberg, Germany.
- German Center for Diabetes Research (DZD), München Neuherberg, Germany.
| |
Collapse
|
17
|
Rasouli MA, Dumesic DA, Singhal V. Male infertility and obesity. Curr Opin Endocrinol Diabetes Obes 2024; 31:203-209. [PMID: 39253759 DOI: 10.1097/med.0000000000000883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/11/2024]
Abstract
PURPOSE OF REVIEW The increasing rate of obesity is having an adverse impact on male reproduction. RECENT FINDINGS The negative effect of reactive oxygen species on male reproductive tissues and the age of onset of obesity are new areas of research on male infertility. SUMMARY This review highlights how obesity impairs male reproduction through complex mechanisms, including metabolic syndrome, lipotoxicity, sexual dysfunction, hormonal and adipokine alterations as well as epigenetic changes, and how new management strategies may improve the reproductive health of men throughout life.
Collapse
Affiliation(s)
| | | | - Vibha Singhal
- Division of Endocrinology, Department of Pediatrics, University of California, Los Angeles, Los Angeles, California, USA
| |
Collapse
|
18
|
Ye J, Yin J. Type 2 diabetes: a sacrifice program handling energy surplus. LIFE METABOLISM 2024; 3:loae033. [PMID: 39873003 PMCID: PMC11748514 DOI: 10.1093/lifemeta/loae033] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 07/30/2024] [Accepted: 09/12/2024] [Indexed: 01/30/2025]
Abstract
Type 2 diabetes mellitus (T2DM) is closely associated with obesity, while interactions between the two diseases remain to be fully elucidated. To this point, we offer this perspective to introduce a set of new insights into the interpretation of T2DM spanning the etiology, pathogenesis, and treatment approaches. These include a definition of T2DM as an energy surplus-induced diabetes characterized by the gradual decline of β cell insulin secretion function, which ultimately aims to prevent the onset of severe obesity through mechanisms of weight loss. The body employs three adaptive strategies in response to energy surplus: the first one is adipose tissue expansion to store the energy for weight gain under normal weight conditions; the second one is insulin resistance to slow down adipose tissue expansion and weight gain under overweight conditions; and the third one is the onset of T2DM following β cell failure to reverse the weight gain in obese conditions. The primary signaling molecules driving the compensatory responses are adenosine derivatives, such as adenosine triphosphate (ATP), acetyl coenzyme A (acetyl-CoA), and reduced nicotinamide adenine dinucleotide (NADH). These molecules exert their effects through allosteric, post-translational, and transcriptional regulation of metabolic pathways. The insights suggest that insulin resistance and T2DM are protective mechanisms in the defense against excessive adiposity to avert severe obesity. The perspective provides a unified framework explaining the interactions between the two diseases and opens new avenues in the study of T2DM.
Collapse
Affiliation(s)
- Jianping Ye
- Metabolic Disease Research Center, Zhengzhou Key Laboratory of Obesity Research, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, Henan 450007, China
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Jun Yin
- Department of Endocrinology and Metabolism, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai Clinical Center for Diabetes, Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai 200233, China
| |
Collapse
|
19
|
Yang Y, Zhang X, Zhang Y, Zhao J, Jia J, Liu H, Song S. Metformin treatment improves depressive symptoms associated with type 2 diabetes: A 24-week longitudinal study. J Affect Disord 2024; 365:80-86. [PMID: 39147157 DOI: 10.1016/j.jad.2024.08.071] [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: 04/03/2024] [Revised: 06/23/2024] [Accepted: 08/12/2024] [Indexed: 08/17/2024]
Abstract
OBJECTIVE Metformin is a medication that is widely used for lowering blood sugar in patients with type 2 diabetes. Metformin was shown to have significant antidepressant effects; however, it is not clear whether metformin treatment improves outcomes in patients with type 2 diabetes who have concomitant depressive symptoms. METHODS A total of 475 patients with type 2 diabetes mellitus with depressive symptoms were included in this study and divided into metformin and nonmetformin groups according to whether they were taking metformin. The DASS-21 was used to assess patients' depression and anxiety scores before and after a 24-week intervention. In addition, general information about whether the patients had developed complications from diabetes and whether they had been diagnosed with other diseases was assessed. RESULTS (1) After 24 weeks, anxiety and depression scores were significantly lower in the metformin group than in the nonmetformin group. (2) The prevalence of depressive symptoms was significantly greater in female type 2 diabetic patients than in male patients (OR = 2.039, 95 % CI = 1.160-3.568). (3) People with type 2 diabetes who develop complications from diabetes (OR = 1.794, 95 % CI = 1.015-3.171) and those diagnosed with other conditions are more likely to experience depressive symptoms. CONCLUSION Metformin has an ameliorative effect on type 2 diabetes. However, women, those with diabetes complications, and those with type 2 diabetes who are also diagnosed with other conditions are more likely to experience depressive symptoms.
Collapse
Affiliation(s)
- Yating Yang
- The Second People's Hospital of Huizhou, Huizhou 512200, Guangdong, China
| | - Xi Zhang
- Department of Psychiatry, Chaohu Hospital of Anhui Medical University, Hefei 238000, China; School of Mental Health and Psychological Sciences, Anhui Medical University, Hefei, 238000, China; Anhui Provincial Key Laboratory for Brain Bank Construction and Resource Utilization, Hefei 23800, China
| | - Yun Zhang
- Department of Psychiatry, Chaohu Hospital of Anhui Medical University, Hefei 238000, China; School of Mental Health and Psychological Sciences, Anhui Medical University, Hefei, 238000, China; Anhui Provincial Key Laboratory for Brain Bank Construction and Resource Utilization, Hefei 23800, China
| | - Jianyong Zhao
- Department of Endocrinology, Chaohu Hospital of Anhui Medical University, Hefei 238000, China
| | - Jingfang Jia
- Department of Endocrinology, Chaohu Hospital of Anhui Medical University, Hefei 238000, China
| | - Huanzhong Liu
- Department of Psychiatry, Chaohu Hospital of Anhui Medical University, Hefei 238000, China; Anhui Provincial Key Laboratory for Brain Bank Construction and Resource Utilization, Hefei 23800, China.
| | - Suqi Song
- Department of Psychiatry, Chaohu Hospital of Anhui Medical University, Hefei 238000, China; Anhui Provincial Key Laboratory for Brain Bank Construction and Resource Utilization, Hefei 23800, China.
| |
Collapse
|
20
|
Wissbroecker KB, Zmuda AJ, Karumanchi H, Niehaus TD. Biochemical and genomic evidence for converging metabolic routes of metformin and biguanide breakdown in environmental Pseudomonads. J Biol Chem 2024; 300:107935. [PMID: 39476966 DOI: 10.1016/j.jbc.2024.107935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 10/18/2024] [Accepted: 10/21/2024] [Indexed: 12/01/2024] Open
Abstract
Metformin is commonly used to lower blood glucose levels and is one of the most widely used pharmaceuticals worldwide. Typical doses are high (0.5-2.0 g day-1) and the majority travels through the digestive system unabsorbed and enters the wastewater system. Metformin is not removed by standard wastewater treatments and eventually enters freshwater systems, where it can form N-chloro-derivatives that are toxic to fish and human cells. Thus, metformin is one of the most prevalent anthropogenic pollutants worldwide and there has been considerable interest in finding ways to remove it. We recently isolated Pseudomonads capable of growing on metformin as the sole nitrogen source. We identified candidate genes involved in metformin breakdown through genomic analyses informed by feeding studies. One candidate, a pair of genes that are located on ∼80kb extra-genomic plasmids, was shown to encode a heteromeric Ni-dependent hydrolase that converts metformin to guanylurea and dimethylamine. Metforminase activity of these gene products is now well established as our results confirm three recently published independent studies. Our isolated Pseudomonads also grow on biguanide, suggesting the existence of an additional breakdown enzyme. Another candidate gene located on the ∼80kb plasmids was shown to encode an aminohydrolase that converts biguanide to guanylurea. Biguanide may arise through successive N-demethylations of metformin or come from other sources. Our results suggest that the recent evolution of metforminase and biguanide hydrolase enzymes allow Pseudomonads to convert either metformin or biguanide to guanylurea, which can be assimilated by existing pathways.
Collapse
Affiliation(s)
- Katie B Wissbroecker
- The Department of Plant and Microbial Biology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Anthony J Zmuda
- The Department of Plant and Microbial Biology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Harsheeth Karumanchi
- The Department of Plant and Microbial Biology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Thomas D Niehaus
- The Department of Plant and Microbial Biology, University of Minnesota, Minneapolis, Minnesota, USA.
| |
Collapse
|
21
|
Ottosen RN, Seefeldt JM, Hansen J, Nielsen R, Møller N, Johannsen M, Poulsen TB. Preparation and Preclinical Characterization of a Simple Ester for Dual Exogenous Supply of Lactate and Beta-hydroxybutyrate. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:19883-19890. [PMID: 39214666 PMCID: PMC11403612 DOI: 10.1021/acs.jafc.4c04849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Elevation of the plasma levels of (S)-lactate (Lac) and/or (R)-beta-hydroxybutyrate (BHB) occurs naturally in response to strenuous exercise and prolonged fasting, respectively, resulting in millimolar concentrations of these two metabolites. It is increasingly appreciated that Lac and BHB have wide-ranging beneficial physiological effects, suggesting that novel nutritional solutions, compatible with high-level and/or sustained consumption, which allow direct control of plasma levels of Lac and BHB, are of strong interest. In this study, we present a molecular hybrid between (S)-lactate and the BHB-precursor (R)-1,3-butanediol in the form of a simple ester referred to as LaKe. We show that LaKe can be readily prepared on the kilogram scale and undergoes rapid hydrolytic conversion under a variety of physiological conditions to release its two constituents. Oral ingestion of LaKe, in rats, resulted in dose-dependent elevation of plasma levels of Lac and BHB triggering expected physiological responses such as reduced lipolysis and elevation of the appetite-suppressing compound N-L-lactoyl-phenylalanine (Lac-Phe).
Collapse
Affiliation(s)
- Rasmus N Ottosen
- Department of Chemistry, Aarhus University, Langelandsgade 140, Aarhus C DK-8000, Denmark
| | - Jacob M Seefeldt
- Department of Cardiology, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, Aarhus N DK-8200, Denmark
| | - Jakob Hansen
- Department of Forensic Medicine, Aarhus University, Palle Juul-Jensens Boulevard. 99, Aarhus N DK-8200, Denmark
| | - Roni Nielsen
- Department of Cardiology, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, Aarhus N DK-8200, Denmark
| | - Niels Møller
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Palle Juul-Jensens Boulevard 11, Aarhus N DK-8200, Denmark
- Department of Clinical Medicine, Aarhus University, Palle Juul-Jensens Boulevard 82, Aarhus N DK-8200, Denmark
| | - Mogens Johannsen
- Department of Forensic Medicine, Aarhus University, Palle Juul-Jensens Boulevard. 99, Aarhus N DK-8200, Denmark
| | - Thomas B Poulsen
- Department of Chemistry, Aarhus University, Langelandsgade 140, Aarhus C DK-8000, Denmark
| |
Collapse
|
22
|
Moya-Garzon MD, Wang M, Li VL, Lyu X, Wei W, Tung ASH, Raun SH, Zhao M, Coassolo L, Islam H, Oliveira B, Dai Y, Spaas J, Delgado-Gonzalez A, Donoso K, Alvarez-Buylla A, Franco-Montalban F, Letian A, Ward C, Liu L, Svensson KJ, Goldberg EL, Gardner CD, Little JP, Banik SM, Xu Y, Long JZ. A secondary β-hydroxybutyrate metabolic pathway linked to energy balance. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.09.612087. [PMID: 39314488 PMCID: PMC11418978 DOI: 10.1101/2024.09.09.612087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
β-hydroxybutyrate (BHB) is an abundant ketone body. To date, all known pathways of BHB metabolism involve interconversion of BHB and primary energy intermediates. Here we show that CNDP2 controls a previously undescribed secondary BHB metabolic pathway via enzymatic conjugation of BHB and free amino acids. This BHB-ylation reaction produces a family of endogenous ketone metabolites, the BHB-amino acids. Genetic ablation of CNDP2 in mice eliminates tissue amino acid BHB-ylation activity and reduces BHB-amino acid levels. Administration of BHB-Phe, the most abundant BHB-amino acid, to obese mice activates neural populations in the hypothalamus and brainstem and suppresses feeding and body weight. Conversely, CNDP2-KO mice exhibit increased food intake and body weight upon ketosis stimuli. CNDP2-dependent amino acid BHB-ylation and BHB-amino acid metabolites are also conserved in humans. Therefore, the metabolic pathways of BHB extend beyond primary metabolism and include secondary ketone metabolites linked to energy balance.
Collapse
Affiliation(s)
- Maria Dolores Moya-Garzon
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
- Sarafan ChEM-H, Stanford University, Stanford, CA, USA
- Wu Tsai Human Performance Alliance, Stanford University, Stanford, CA, USA
| | - Mengjie Wang
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Veronica L Li
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Chemistry, Stanford University, Stanford, CA, USA
- Sarafan ChEM-H, Stanford University, Stanford, CA, USA
- Wu Tsai Human Performance Alliance, Stanford University, Stanford, CA, USA
| | - Xuchao Lyu
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
- Sarafan ChEM-H, Stanford University, Stanford, CA, USA
- Wu Tsai Human Performance Alliance, Stanford University, Stanford, CA, USA
| | - Wei Wei
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Biology, Stanford University, Stanford, CA, USA
- Sarafan ChEM-H, Stanford University, Stanford, CA, USA
| | - Alan Sheng-Hwa Tung
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Biology, Stanford University, Stanford, CA, USA
- Sarafan ChEM-H, Stanford University, Stanford, CA, USA
| | - Steffen H Raun
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Meng Zhao
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Diabetes Research Center, Stanford University, Stanford, CA, USA
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Laetitia Coassolo
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Diabetes Research Center, Stanford University, Stanford, CA, USA
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Hashim Islam
- School of Health and Exercise Sciences, University of British Columbia, Kelowna, British Columbia, Canada
| | - Barbara Oliveira
- School of Health and Exercise Sciences, University of British Columbia, Kelowna, British Columbia, Canada
| | - Yuqin Dai
- Sarafan ChEM-H, Stanford University, Stanford, CA, USA
| | - Jan Spaas
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
- Sarafan ChEM-H, Stanford University, Stanford, CA, USA
| | | | - Kenyi Donoso
- Department of Urology, Stanford University School of Medicine, Stanford, CA, USA
| | | | - Francisco Franco-Montalban
- Departamento de Química Farmacéutica y Orgánica, Universidad de Granada, Campus de Cartuja sn, 18011, Granada, Spain
| | - Anudari Letian
- Department of Physiology, University of California, San Francisco, San Francisco, CA, USA
| | - Catherine Ward
- Stanford Prevention Research Center, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Lichao Liu
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA, USA
| | - Katrin J Svensson
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Diabetes Research Center, Stanford University, Stanford, CA, USA
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Emily L Goldberg
- Department of Physiology, University of California, San Francisco, San Francisco, CA, USA
| | - Christopher D Gardner
- Stanford Prevention Research Center, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Jonathan P Little
- School of Health and Exercise Sciences, University of British Columbia, Kelowna, British Columbia, Canada
| | - Steven M Banik
- Department of Chemistry, Stanford University, Stanford, CA, USA
- Sarafan ChEM-H, Stanford University, Stanford, CA, USA
| | - Yong Xu
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Jonathan Z Long
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
- Sarafan ChEM-H, Stanford University, Stanford, CA, USA
- Stanford Diabetes Research Center, Stanford University, Stanford, CA, USA
- Wu Tsai Human Performance Alliance, Stanford University, Stanford, CA, USA
- The Phil & Penny Knight Initiative for Brain Resilience at the Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
| |
Collapse
|
23
|
Sirtori CR, Castiglione S, Pavanello C. METFORMIN: FROM DIABETES TO CANCER TO PROLONGATION OF LIFE. Pharmacol Res 2024; 208:107367. [PMID: 39191336 DOI: 10.1016/j.phrs.2024.107367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 08/12/2024] [Accepted: 08/21/2024] [Indexed: 08/29/2024]
Abstract
The metformin molecule dates back to over a century, but its clinical use started in the '50s. Since then, its use in diabetics has grown constantly, with over 150 million users today. The therapeutic profile also expanded, with improved understanding of novel mechanisms. Metformin has a major activity on insulin resistance, by acting on the insulin receptors and mitochondria, most likely by activation of the adenosine monophosphate-activated kinase. These and associated mechanisms lead to significant lipid lowering and body weight loss. An anti-cancer action has come up in recent years, with mechanisms partly dependent on the mitochondrial activity and also on phosphatidylinositol 3-kinase resistance occurring in some malignant tumors. The potential of metformin to raise life-length is the object of large ongoing studies and of several basic and clinical investigations. The present review article will attempt to investigate the basic mechanisms behind these diverse activities and the potential clinical benefits. Metformin may act on transcriptional activity by histone modification, DNA methylation and miRNAs. An activity on age-associated inflammation (inflammaging) may occur via activation of the nuclear factor erythroid 2 related factor and changes in gut microbiota. A senolytic activity, leading to reduction of cells with the senescent associated secretory phenotype, may be crucial in lifespan prolongation as well as in ancillary properties in age-associated diseases, such as Parkinson's disease. Telomere prolongation may be related to the activity on mitochondrial respiratory factor 1 and on peroxisome gamma proliferator coactivator 1-alpha. Very recent observations on the potential to act on the most severe neurological disorders, such as amyotrophic lateral sclerosis and frontotemporal dementia, have raised considerable hope.
Collapse
Affiliation(s)
- Cesare R Sirtori
- Center of Dyslipidemias, ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy; Centro E. Grossi Paoletti, Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy.
| | - Sofia Castiglione
- Center of Dyslipidemias, ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy; Centro E. Grossi Paoletti, Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Chiara Pavanello
- Center of Dyslipidemias, ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy; Centro E. Grossi Paoletti, Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| |
Collapse
|
24
|
Li VL, Xiao S, Schlosser P, Scherer N, Wiggenhorn AL, Spaas J, Tung ASH, Karoly ED, Köttgen A, Long JZ. SLC17A1/3 transporters mediate renal excretion of Lac-Phe in mice and humans. Nat Commun 2024; 15:6895. [PMID: 39134528 PMCID: PMC11319466 DOI: 10.1038/s41467-024-51174-3] [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: 11/13/2023] [Accepted: 08/01/2024] [Indexed: 08/15/2024] Open
Abstract
N-lactoyl-phenylalanine (Lac-Phe) is a lactate-derived metabolite that suppresses food intake and body weight. Little is known about the mechanisms that mediate Lac-Phe transport across cell membranes. Here we identify SLC17A1 and SLC17A3, two kidney-restricted plasma membrane-localized solute carriers, as physiologic urine Lac-Phe transporters. In cell culture, SLC17A1/3 exhibit high Lac-Phe efflux activity. In humans, levels of Lac-Phe in urine exhibit a strong genetic association with the SLC17A1-4 locus. Urine Lac-Phe levels are increased following a Wingate sprint test. In mice, genetic ablation of either SLC17A1 or SLC17A3 reduces urine Lac-Phe levels. Despite these differences, both knockout strains have normal blood Lac-Phe and body weights, demonstrating SLC17A1/3-dependent de-coupling of urine and plasma Lac-Phe pools. Together, these data establish SLC17A1/3 family members as the physiologic urine Lac-Phe transporters and uncover a biochemical pathway for the renal excretion of this signaling metabolite.
Collapse
Affiliation(s)
- Veronica L Li
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Chemistry, Stanford University, Stanford, CA, USA
- Sarafan ChEM-H, Stanford University, Stanford, CA, USA
- Stanford Diabetes Research Center, Stanford University, Stanford, CA, USA
| | - Shuke Xiao
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
- Sarafan ChEM-H, Stanford University, Stanford, CA, USA
- Stanford Diabetes Research Center, Stanford University, Stanford, CA, USA
| | - Pascal Schlosser
- Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany
- Department of Epidemiology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, USA
- Centre for Integrative Biological Signaling Studies (CIBSS), University of Freiburg, Freiburg, Germany
| | - Nora Scherer
- Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany
- Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, Freiburg, Germany
| | - Amanda L Wiggenhorn
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Chemistry, Stanford University, Stanford, CA, USA
- Sarafan ChEM-H, Stanford University, Stanford, CA, USA
- Stanford Diabetes Research Center, Stanford University, Stanford, CA, USA
| | - Jan Spaas
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
- Sarafan ChEM-H, Stanford University, Stanford, CA, USA
- Stanford Diabetes Research Center, Stanford University, Stanford, CA, USA
| | - Alan Sheng-Hwa Tung
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
- Sarafan ChEM-H, Stanford University, Stanford, CA, USA
- Stanford Diabetes Research Center, Stanford University, Stanford, CA, USA
| | | | - Anna Köttgen
- Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany
- Department of Epidemiology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, USA
- Centre for Integrative Biological Signaling Studies (CIBSS), University of Freiburg, Freiburg, Germany
| | - Jonathan Z Long
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA.
- Sarafan ChEM-H, Stanford University, Stanford, CA, USA.
- Stanford Diabetes Research Center, Stanford University, Stanford, CA, USA.
- The Phil & Penny Knight Initiative for Brain Resilience at the Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA.
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA.
| |
Collapse
|
25
|
Xiao S, Li VL, Long JZ. Lac-Phe (N-lactoyl-phenylalanine). Trends Endocrinol Metab 2024; 35:758-759. [PMID: 39137723 PMCID: PMC11446501 DOI: 10.1016/j.tem.2024.05.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 05/14/2024] [Accepted: 05/15/2024] [Indexed: 08/15/2024]
Affiliation(s)
- Shuke Xiao
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA; Sarafan ChEM-H, Stanford University, Stanford, CA, USA; Stanford Diabetes Research Center, Stanford University, Stanford, CA, USA
| | - Veronica L Li
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA; Sarafan ChEM-H, Stanford University, Stanford, CA, USA; Stanford Diabetes Research Center, Stanford University, Stanford, CA, USA; Department of Chemistry, Stanford University, Stanford, CA, USA; Wu Tsai Human Performance Alliance, Stanford University, Stanford, CA, USA
| | - Jonathan Z Long
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA; Sarafan ChEM-H, Stanford University, Stanford, CA, USA; Stanford Diabetes Research Center, Stanford University, Stanford, CA, USA; Wu Tsai Human Performance Alliance, Stanford University, Stanford, CA, USA; Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA; The Phil and Penny Knight Initiative for Brain Resilience at the Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA.
| |
Collapse
|
26
|
Starling S. Metformin acts through appetite-suppressing metabolite: Lac-Phe. Nat Rev Endocrinol 2024; 20:319. [PMID: 38565678 DOI: 10.1038/s41574-024-00986-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
|
27
|
Nakamura M. Lipotoxicity as a therapeutic target in obesity and diabetic cardiomyopathy. JOURNAL OF PHARMACY & PHARMACEUTICAL SCIENCES : A PUBLICATION OF THE CANADIAN SOCIETY FOR PHARMACEUTICAL SCIENCES, SOCIETE CANADIENNE DES SCIENCES PHARMACEUTIQUES 2024; 27:12568. [PMID: 38706718 PMCID: PMC11066298 DOI: 10.3389/jpps.2024.12568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 04/09/2024] [Indexed: 05/07/2024]
Abstract
Unhealthy sources of fats, ultra-processed foods with added sugars, and a sedentary lifestyle make humans more susceptible to developing overweight and obesity. While lipids constitute an integral component of the organism, excessive and abnormal lipid accumulation that exceeds the storage capacity of lipid droplets disrupts the intracellular composition of fatty acids and results in the release of deleterious lipid species, thereby giving rise to a pathological state termed lipotoxicity. This condition induces endoplasmic reticulum stress, mitochondrial dysfunction, inflammatory responses, and cell death. Recent advances in omics technologies and analytical methodologies and clinical research have provided novel insights into the mechanisms of lipotoxicity, including gut dysbiosis, epigenetic and epitranscriptomic modifications, dysfunction of lipid droplets, post-translational modifications, and altered membrane lipid composition. In this review, we discuss the recent knowledge on the mechanisms underlying the development of lipotoxicity and lipotoxic cardiometabolic disease in obesity, with a particular focus on lipotoxic and diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Michinari Nakamura
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Rutgers Biomedical and Health Sciences, Newark, United States
| |
Collapse
|
28
|
Li VL, Xiao S, Schlosser P, Scherer N, Wiggenhorn AL, Spaas J, Tung ASH, Karoly ED, Köttgen A, Long JZ. SLC17 transporters mediate renal excretion of Lac-Phe in mice and humans. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.18.589815. [PMID: 38659895 PMCID: PMC11042375 DOI: 10.1101/2024.04.18.589815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
N-lactoyl-phenylalanine (Lac-Phe) is a lactate-derived metabolite that suppresses food intake and body weight. Little is known about the mechanisms that mediate Lac-Phe transport across cell membranes. Here we identify SLC17A1 and SLC17A3, two kidney-restricted plasma membrane-localized solute carriers, as physiologic urine Lac-Phe transporters. In cell culture, SLC17A1/3 exhibit high Lac-Phe efflux activity. In humans, levels of Lac-Phe in urine exhibit a strong genetic association with the SLC17A1-4 locus. Urine Lac-Phe levels are also increased following a Wingate sprint test. In mice, genetic ablation of either SLC17A1 or SLC17A3 reduces urine Lac-Phe levels. Despite these differences, both knockout strains have normal blood Lac-Phe and body weights, demonstrating that urine and plasma Lac-Phe pools are functionally and biochemically de-coupled. Together, these data establish SLC17 family members as the physiologic urine transporters for Lac-Phe and uncover a biochemical pathway for the renal excretion of this signaling metabolite.
Collapse
|
29
|
TeSlaa T. Metformin induces a Lac-Phe gut-brain signalling axis. Nat Metab 2024; 6:603-605. [PMID: 38499764 DOI: 10.1038/s42255-024-01014-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/20/2024]
Affiliation(s)
- Tara TeSlaa
- Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, CA, USA.
- Molecular Biology Institute, University of California Los Angeles, Los Angeles, CA, USA.
- Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
30
|
Xiao S, Li VL, Lyu X, Chen X, Wei W, Abbasi F, Knowles JW, Tung ASH, Deng S, Tiwari G, Shi X, Zheng S, Farrell L, Chen ZZ, Taylor KD, Guo X, Goodarzi MO, Wood AC, Chen YDI, Lange LA, Rich SS, Rotter JI, Clish CB, Tahir UA, Gerszten RE, Benson MD, Long JZ. Lac-Phe mediates the effects of metformin on food intake and body weight. Nat Metab 2024; 6:659-669. [PMID: 38499766 PMCID: PMC11062621 DOI: 10.1038/s42255-024-00999-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Accepted: 01/30/2024] [Indexed: 03/20/2024]
Abstract
Metformin is a widely prescribed anti-diabetic medicine that also reduces body weight. There is ongoing debate about the mechanisms that mediate metformin's effects on energy balance. Here, we show that metformin is a powerful pharmacological inducer of the anorexigenic metabolite N-lactoyl-phenylalanine (Lac-Phe) in cells, in mice and two independent human cohorts. Metformin drives Lac-Phe biosynthesis through the inhibition of complex I, increased glycolytic flux and intracellular lactate mass action. Intestinal epithelial CNDP2+ cells, not macrophages, are the principal in vivo source of basal and metformin-inducible Lac-Phe. Genetic ablation of Lac-Phe biosynthesis in male mice renders animals resistant to the effects of metformin on food intake and body weight. Lastly, mediation analyses support a role for Lac-Phe as a downstream effector of metformin's effects on body mass index in participants of a large population-based observational cohort, the Multi-Ethnic Study of Atherosclerosis. Together, these data establish Lac-Phe as a critical mediator of the body weight-lowering effects of metformin.
Collapse
Affiliation(s)
- Shuke Xiao
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
- Sarafan ChEM-H, Stanford University, Stanford, CA, USA
- Stanford Diabetes Research Center, Stanford University, Stanford, CA, USA
| | - Veronica L Li
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
- Sarafan ChEM-H, Stanford University, Stanford, CA, USA
- Stanford Diabetes Research Center, Stanford University, Stanford, CA, USA
- Department of Chemistry, Stanford University, Stanford, CA, USA
- Wu Tsai Human Performance Alliance, Stanford University, Stanford, CA, USA
| | - Xuchao Lyu
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
- Sarafan ChEM-H, Stanford University, Stanford, CA, USA
- Stanford Diabetes Research Center, Stanford University, Stanford, CA, USA
- Wu Tsai Human Performance Alliance, Stanford University, Stanford, CA, USA
| | - Xudong Chen
- Department of Molecular and Cellular Physiology, Stanford University, Stanford, CA, USA
- Howard Hughes Medical Institute, Stanford University, Stanford, CA, USA
| | - Wei Wei
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
- Sarafan ChEM-H, Stanford University, Stanford, CA, USA
- Stanford Diabetes Research Center, Stanford University, Stanford, CA, USA
| | - Fahim Abbasi
- Stanford Diabetes Research Center, Stanford University, Stanford, CA, USA
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Joshua W Knowles
- Stanford Diabetes Research Center, Stanford University, Stanford, CA, USA
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Prevention Research Center, Stanford University School of Medicine, Stanford, CA, USA
| | - Alan Sheng-Hwa Tung
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
- Sarafan ChEM-H, Stanford University, Stanford, CA, USA
- Stanford Diabetes Research Center, Stanford University, Stanford, CA, USA
| | - Shuliang Deng
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Gaurav Tiwari
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Xu Shi
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Shuning Zheng
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Laurie Farrell
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Zsu-Zsu Chen
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Kent D Taylor
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Xiuqing Guo
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Mark O Goodarzi
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Alexis C Wood
- USDA/ARS Children's Nutrition Research Center, Baylor College of Medicine, Houston, TX, USA
| | - Yii-Der Ida Chen
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Leslie A Lange
- Division of Biomedical Informatics and Personalized Medicine, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Stephen S Rich
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, USA
| | - Jerome I Rotter
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Clary B Clish
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Usman A Tahir
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Robert E Gerszten
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Mark D Benson
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Jonathan Z Long
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA.
- Sarafan ChEM-H, Stanford University, Stanford, CA, USA.
- Stanford Diabetes Research Center, Stanford University, Stanford, CA, USA.
- Wu Tsai Human Performance Alliance, Stanford University, Stanford, CA, USA.
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
31
|
Skinnider MA. Hiding in plain sight: a research parasite's perspective on new lessons in old data. Gigascience 2024; 13:giae097. [PMID: 39657102 DOI: 10.1093/gigascience/giae097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 09/23/2024] [Accepted: 11/04/2024] [Indexed: 12/17/2024] Open
Abstract
High-throughput techniques that measure thousands of analytes at once have become ubiquitous features of biological research. The increasing expectation that the raw data generated by these techniques be deposited to public repositories creates rich opportunities for secondary analysis of these datasets. Such opportunities can take multiple forms. As the recipient of the 2023 Junior Research Parasite Award, I was asked to comment on the role of so-called research parasites within the ecosystem of secondary data analysis. Drawing on my own experiences, I discuss mechanisms by which reanalysis of published datasets can catalyze biological discoveries, produce resources that would be impossible to generate within a single laboratory, and drive the refinement of computational methods.
Collapse
Affiliation(s)
- Michael A Skinnider
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08540, USA
- Ludwig Institute for Cancer Research, Princeton University, Princeton, NJ 08540, USA
| |
Collapse
|