1
|
Schneider-Heieck K, Pérez-Schindler J, Blatter J, de Smalen LM, Duchemin W, Steurer SA, Karrer-Cardel B, Ritz D, Handschin C. Krüppel-like factor 5 remodels lipid metabolism in exercised skeletal muscle. Mol Metab 2025; 96:102154. [PMID: 40250760 PMCID: PMC12060515 DOI: 10.1016/j.molmet.2025.102154] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Revised: 04/08/2025] [Accepted: 04/11/2025] [Indexed: 04/20/2025] Open
Abstract
Regular physical activity induces a variety of health benefits, preventing and counteracting diseases caused by a sedentary lifestyle. However, the molecular underpinnings of skeletal muscle plasticity in exercise remain poorly understood. We identified a role of the Krüppel-Like Factor 5 (Klf5) in this process, in particular in the regulation of lipid homeostasis. Surprisingly, gain- and loss-of-function studies in muscle in vivo revealed seemingly opposite functions of Klf5 in the response to an acute exercise bout and chronic training, modulating lipid oxidation and synthesis, respectively. Thus, even though only transiently induced, the function of Klf5 is complex and fundamental for a normal long-term training response. These findings highlight the importance of this mediator of external stress response to adaptive remodeling of skeletal muscle tissue.
Collapse
Affiliation(s)
| | | | | | | | - Wandrille Duchemin
- sciCORE Center for Scientific Computing, University of Basel, Basel, Switzerland
| | | | | | - Danilo Ritz
- Biozentrum, University of Basel, Basel, Switzerland
| | | |
Collapse
|
2
|
Russ DW, Manickam R, Tipparaju SM. Targeting intramyocellular lipids to improve aging muscle function. Lipids Health Dis 2025; 24:197. [PMID: 40450303 DOI: 10.1186/s12944-025-02622-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2025] [Accepted: 05/22/2025] [Indexed: 06/03/2025] Open
Abstract
Decline of skeletal muscle function in old age is a significant contributor to reduced quality of life, risk of injury, comorbidity and disability and even mortality. While this loss of muscle function has traditionally been attributed to sarcopenia (loss of muscle mass), it is now generally appreciated that factors other than mass play a significant role in age-related muscle weakness. One such factor gaining increased attention is the ectopic accumulation of lipids in skeletal muscle, in particular, intramyocellular lipids (IMCLs). It has been appreciated for some time that metabolic flexibility of several tissues/organs declines with age and may be related to accumulation of IMCLs in a "vicious cycle" whereby blunted metabolic flexibility promotes accumulation of IMCLs, which leases to lipotoxicity, which can then further impair metabolic flexibility. The standard interventions for addressing lipid accumulation and muscle weakness remain diet (caloric restriction) and exercise. However, long-term compliance with both interventions in older adults is low, and in the case of caloric restriction, may be inappropriate for many older adults. Accordingly, it is important, from a public health standpoint, to pursue potential pharmacological strategies for improving muscle function. Because of the success of incretin-analog drugs in addressing obesity, these medications may potentially reduce IMCLs in aging muscles and thus improve metabolic flexibility and improve muscle health. A contrasting potential pharmacological strategy for addressing these issues might be to enhance energy provision to stimulate metabolism by increasing NAD + availability, which is known to decline with age and has been linked to reduced metabolic flexibility. In this narrative review, we present information related to IMCL accumulation and metabolic flexibility in old age and how the two major lifestyle interventions, caloric restriction and exercise, can affect these factors. Finally, we discuss the potential benefits and risks of select pharmacologic interventions in older adults.
Collapse
Affiliation(s)
- David W Russ
- School of Physical Therapy and Rehabilitation Sciences, Morsani College of Medicine, University of South Florida, 12901 Bruce B. Downs Blvd., MDC77, Tampa, FL, 33612-4799, USA.
| | - Ravikumar Manickam
- Department of Pharmaceutical Sciences, Taneja College of Pharmacy, University of South Florida, Tampa, FL, USA
| | - Srinivas M Tipparaju
- Department of Pharmaceutical Sciences, Taneja College of Pharmacy, University of South Florida, Tampa, FL, USA
| |
Collapse
|
3
|
Ningtyas MC, Ansharullah BA, Sutanto H, Prajitno JH. Beyond weight: Exploring the nexus between obesity and osteoarthritis. Semergen 2025; 51:102526. [PMID: 40449057 DOI: 10.1016/j.semerg.2025.102526] [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: 03/06/2025] [Revised: 05/02/2025] [Accepted: 05/06/2025] [Indexed: 06/02/2025]
Abstract
Obesity is a well-established risk factor for osteoarthritis (OA), traditionally attributed to increased mechanical stress on weight-bearing joints. However, recent research suggests a more complex interplay, involving metabolic, biomechanical, and inflammatory pathways. This review delves into these multifaceted connections between obesity and osteoarthritis, extending beyond the conventional understanding of weight-bearing stress. It explores the role of adipokines such as leptin, visfatin, adiponectin, and resistin in OA pathogenesis and progression, highlighting their potential as targets for novel therapeutics. The review also examines how obesity alters the biomechanics of lower extremities, contributing to changes in joint load and movement patterns. Systemic effects, including inflammation and metabolic factors, are discussed to elucidate their roles in exacerbating OA beyond joint loading. Furthermore, the impact of bariatric surgery and weight loss strategies on OA symptoms and progression is evaluated. This comprehensive review aims to provide new insights into obesity-induced OA, paving the way for more targeted and effective treatment strategies.
Collapse
Affiliation(s)
- M C Ningtyas
- Internal Medicine Study Program, Department of Internal Medicine, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia; Department of Internal Medicine, Dr. Soetomo General Academic Hospital, Surabaya, Indonesia
| | - B A Ansharullah
- Internal Medicine Study Program, Department of Internal Medicine, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia; Department of Internal Medicine, Dr. Soetomo General Academic Hospital, Surabaya, Indonesia
| | - H Sutanto
- Internal Medicine Study Program, Department of Internal Medicine, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia; Department of Internal Medicine, Dr. Soetomo General Academic Hospital, Surabaya, Indonesia.
| | - J H Prajitno
- Internal Medicine Study Program, Department of Internal Medicine, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia; Division of Endocrinology, Metabolic Diseases and Diabetes, Department of Internal Medicine, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia; Department of Internal Medicine, Airlangga University Hospital, Surabaya, Indonesia.
| |
Collapse
|
4
|
Lo CW, Lee JL, Tsai WT, Huang CS, Yang YC, Lii CK, Chen HW. Benzyl isothiocyanate ameliorates hepatic insulin resistance in mice with high-fat diet-induced nonalcoholic fatty liver disease. J Nutr Biochem 2025:109981. [PMID: 40449689 DOI: 10.1016/j.jnutbio.2025.109981] [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: 09/14/2024] [Revised: 03/01/2025] [Accepted: 05/28/2025] [Indexed: 06/03/2025]
Abstract
The global prevalence of overweight and obesity has risen sharply over the past few decades as a result of excess calorie intake and sedentary lifestyles. Obesity increases the risk for various metabolic disorders, such as hyperlipidemia, fatty liver disease, and diabetes mellitus. Isothiocyanates, which are abundant in cruciferous vegetables, have been shown to exhibit anti-cancer, anti-inflammatory, and antioxidant properties. However, the efficacy of benzyl isothiocyanate (BITC) in preventing the adverse effects of obesity, such as hepatic steatosis and insulin resistance, remains uncertain. To address this knowledge gap, we assessed whether BITC protects against hepatic insulin resistance by using primary mouse hepatocytes and AML12 cells treated with palmitic acid (PA) and mice fed a high-fat diet supplemented with cholesterol and cholic acid (HFCCD). We found that the impairments in insulin sensitivity caused by PA, such as decreases in the phosphorylation of insulin receptor substrate (IRS) 1 (Tyr608), Akt, glycogen synthase kinase (GSK) 3β, and FOXO1 and increases in the expression of glucose-6-phosphatase (G6Pase) and phosphoenolpyruvate carboxykinase 1 (PEPCK) mRNA in hepatocytes, were mitigated by pretreatment with BITC. BITC also attenuated PA-induced hepatic lipid accumulation and reactive oxygen species production. In vivo, BITC significantly reduced blood glucose levels and the HOMA-IR and inhibited hepatic lipid accumulation, IRS1 phosphorylation at Ser307, and G6Pase and PEPCK expression compared with that in mice fed the HFCCD alone. These results show that BITC ameliorates the lipotoxicity associated with insulin resistance by activating the IR/IRS/Akt/FOXO1 and GSK3β pathways, which leads to decreased gluconeogenesis and increased glycogen synthesis.
Collapse
Affiliation(s)
- Chia-Wen Lo
- Department of Nutrition, China Medical University, Taichung 406, Taiwan; Department of Nutrition, College of Medical and Health Care, Hung-Kuang University, Taichung 433, Taiwan
| | - Jyun-Lin Lee
- School of Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung 404, Taiwan
| | - Wei-Ting Tsai
- Department of Nutrition, China Medical University, Taichung 406, Taiwan
| | - Chin-Shiu Huang
- Department of Health and Nutrition Biotechnology, Asia University, Taichung 413, Taiwan
| | - Ya-Chen Yang
- Department of Health and Nutrition Biotechnology, Asia University, Taichung 413, Taiwan
| | - Chong-Kuei Lii
- Department of Nutrition, China Medical University, Taichung 406, Taiwan.
| | - Haw-Wen Chen
- Department of Nutrition, China Medical University, Taichung 406, Taiwan.
| |
Collapse
|
5
|
Yao S, Mao Z, Marron MM, Simonsick EM, Murthy VL, Shah RV, Newman AB. Metabolic Markers Demonstrate the Heterogeneity of Walking Ability in Non-Disabled Community-Dwelling Older Adults. Metabolites 2025; 15:334. [PMID: 40422910 PMCID: PMC12113439 DOI: 10.3390/metabo15050334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2025] [Revised: 05/14/2025] [Accepted: 05/15/2025] [Indexed: 05/28/2025] Open
Abstract
Background: Walking ability is important for the quality of life of older adults. A self-reported walking ability index (WAI) covering the difficulty and ease of walking captures a broader spectrum of walking ability in healthy older persons. Methods: Using metabolomics in the Health, Aging and Body Composition study, we identified Year 2 metabolites cross-sectionally and longitudinally related to WAI (0-9, higher scores indicate better walking ability) using probabilistic index models and multinomial logistic models, respectively. Results: Among 2334 participants (mean age 74.6 years, 51% women, 37% Black), 27% scored 0-5, 36% scored 6-8, and 37% scored 9 at Year 2. Over 4 years, 52% maintained a stable WAI, 6% improved, while 42% declined (22% 1-2 points and 20% >2 points decline). We identified 81 metabolites significantly associated with both poorer concurrent WAI and faster decline, including higher acylcarnitine species, shorter-chain saturated diglycerides and triglycerides, and TCA cycle intermediates (cis-aconitic, fumaric, and malic acids), and lower phospholipids levels. Eighteen additional metabolites were only associated with faster WAI decline: higher short-chain saturated triglycerides and energy metabolism markers (ATP/ADP/AMP) and lower margaric acid and glycine levels. Notably, those with improved WAI, despite poorer baseline WAI and lifestyles, showed more favorable metabolic profiles than others. Conclusions: Metabolites linked to the TCA cycle and energy metabolism, as well as inflammation and protein catabolism, were related to mobility function. Some metabolites might be particularly important for the early detection of older adults at risk of mobility decline. Metabolic profiles may also help identify older individuals (i.e., with improving WAI) with greater metabolic resilience to lifestyle risk factors and health conditions.
Collapse
Affiliation(s)
- Shanshan Yao
- Department of Epidemiology, School of Public Health, University of Pittsburgh, Pittsburgh, PA 15261, USA; (S.Y.); (Z.M.); (M.M.M.)
| | - Ziling Mao
- Department of Epidemiology, School of Public Health, University of Pittsburgh, Pittsburgh, PA 15261, USA; (S.Y.); (Z.M.); (M.M.M.)
| | - Megan M. Marron
- Department of Epidemiology, School of Public Health, University of Pittsburgh, Pittsburgh, PA 15261, USA; (S.Y.); (Z.M.); (M.M.M.)
| | - Eleanor M. Simonsick
- Intramural Research Program, National Institute on Aging, Baltimore, MD 21224, USA;
| | - Venkatesh L. Murthy
- Department of Medicine and Radiology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Ravi V. Shah
- Department of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Anne B. Newman
- Department of Epidemiology, School of Public Health, University of Pittsburgh, Pittsburgh, PA 15261, USA; (S.Y.); (Z.M.); (M.M.M.)
| |
Collapse
|
6
|
Schimmer S, Kerkmann L, Kahlert N, Jubeh SA, Werner T, Corkish C, Prendeville H, Finlay DK, Sutter K, Dittmer U, Littwitz-Salomon E. Dietary lipid overload creates a suppressive environment that impedes the antiviral functions of NK cells. iScience 2025; 28:112396. [PMID: 40352719 PMCID: PMC12063142 DOI: 10.1016/j.isci.2025.112396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 02/03/2025] [Accepted: 04/07/2025] [Indexed: 05/14/2025] Open
Abstract
Natural killer (NK) cells are innate immune cells able to recognize and eliminate virus-infected cells. NK cell activity strongly correlates with a metabolic reprogramming and breakdown of fatty acids by β-oxidation during virus infections. However, there is limited knowledge regarding the impact of obesity on antiviral NK cell functions. Here, employing the Friend retrovirus mouse model, we show that the cytotoxicity and cytokine production of NK cells was impaired in obesity, leading to higher viral loads. NK cells suppression in obesity was mediated by activated Tregs. Furthermore, obese mice that were switched back to a regular diet showed complete recovery of the NK cell activity. Interestingly, feeding mice with a high-fat diet (HFD) for just ten days caused NK cell dysfunction and increased retroviral burden. This study is the first to link the detrimental impact of an obesity-induced immunosuppressive microenvironment with NK cell dysfunction during an acute retroviral infection.
Collapse
Affiliation(s)
- Simone Schimmer
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Leonie Kerkmann
- Institute for the Research on HIV and AIDS-associated Diseases, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Nele Kahlert
- Institute for the Research on HIV and AIDS-associated Diseases, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Shahd al Jubeh
- Institute for the Research on HIV and AIDS-associated Diseases, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Tanja Werner
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Carrie Corkish
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, 152-160 Pearse Street, Dublin 2, Ireland
| | - Hannah Prendeville
- Biomedical Engineering, School of Engineering, College of Science and Engineering, University of Galway, Galway, Ireland
| | - David K. Finlay
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, 152-160 Pearse Street, Dublin 2, Ireland
- School of Pharmacy and Pharmaceutical Sciences, Trinity Biomedical Sciences Institute, Trinity College Dublin, 152-160 Pearse Street, Dublin 2, Ireland
| | - Kathrin Sutter
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- Institute for the Research on HIV and AIDS-associated Diseases, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Ulf Dittmer
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Elisabeth Littwitz-Salomon
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- Institute for the Research on HIV and AIDS-associated Diseases, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| |
Collapse
|
7
|
Silva JG, Tavares L, Belew GD, Rodrigues JA, Araújo R, Gil AM, Jones JG. Impact of High-Fat Diet-induced Metabolic Dysfunction-associated Steatotic Liver Disease on Heart, Kidney, and Skeletal Muscle Metabolomes in Wild-Type Mice. J Proteome Res 2025; 24:2491-2504. [PMID: 40222045 DOI: 10.1021/acs.jproteome.5c00040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/15/2025]
Abstract
Background: Metabolic dysfunction-associated steatotic liver disease (MASLD) can be recapitulated in mice fed a high-fat diet. The development of MASLD and the diet per se can both perturb metabolism in key extrahepatic tissues such as the heart, kidney, and skeletal muscle. To date, these alterations have not been well described in this animal model of diet-induced MASLD. Methodology: Male C57BL/6J mice were fed either standard (SC, n = 12) or high-fat chow (HF, n = 11) for 18 weeks. Metabolites were extracted from the heart, kidney, and skeletal muscle and analyzed by 1H nuclear magnetic resonance (NMR) spectroscopy, along with multivariate and univariate statistical analyses. Results: Kidney metabolite profiles exhibited the largest differences between HF and SC diets, followed by those of skeletal muscle and then the heart. Some alterations were common across all tissues, namely decreased trimethylamine and elevated levels of linoleic acid and polyunsaturated fatty acids in HF compared to SC (p < 0.05 for all three metabolites). Overall, the metabolite variations were consistent with shifts in carbohydrate and lipid substrate selection for oxidation, increased tissue stress in the heart and kidneys, and altered choline metabolism. These findings may serve as additional important descriptors of MASLD onset and progression.
Collapse
Affiliation(s)
- João G Silva
- Institute for Interdisciplinary Research (III-UC), Centre for Innovative Biomedicine and Biotechnology (CIBB), Metabolism, Aging and Disease, University of Coimbra, Cantanhede 3060-197, Portugal
- Institute for Interdisciplinary Research (III-UC), Doctoral Programme in Experimental Biology and Biomedicine (PDBEB), University of Coimbra, Coimbra 3030-789, Portugal
- University School Vasco da Gama (EUVG), Vasco da Gama Research Center (CIVG), Coimbra 3020-210, Portugal
- Department of Chemistry and CICECO-Aveiro Institute of Materials, University of Aveiro, Aveiro 3810-193, Portugal
| | - Ludgero Tavares
- Institute for Interdisciplinary Research (III-UC), Centre for Innovative Biomedicine and Biotechnology (CIBB), Metabolism, Aging and Disease, University of Coimbra, Cantanhede 3060-197, Portugal
- University School Vasco da Gama (EUVG), Vasco da Gama Research Center (CIVG), Coimbra 3020-210, Portugal
| | - Getachew D Belew
- Institute for Interdisciplinary Research (III-UC), Centre for Innovative Biomedicine and Biotechnology (CIBB), Metabolism, Aging and Disease, University of Coimbra, Cantanhede 3060-197, Portugal
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, Ohio 45701, United States
| | - João A Rodrigues
- Department of Chemistry and CICECO-Aveiro Institute of Materials, University of Aveiro, Aveiro 3810-193, Portugal
| | - Rita Araújo
- Department of Chemistry and CICECO-Aveiro Institute of Materials, University of Aveiro, Aveiro 3810-193, Portugal
| | - Ana M Gil
- Department of Chemistry and CICECO-Aveiro Institute of Materials, University of Aveiro, Aveiro 3810-193, Portugal
| | - John G Jones
- Institute for Interdisciplinary Research (III-UC), Centre for Innovative Biomedicine and Biotechnology (CIBB), Metabolism, Aging and Disease, University of Coimbra, Cantanhede 3060-197, Portugal
| |
Collapse
|
8
|
Op den Kamp‐Bruls YMH, Op den Kamp YJM, Veeraiah P, Zapata Perez R, Phielix E, Havekes B, Schaart G, Kornips E, Berendsen BRB, Virmani A, Wildberger JE, Houtkooper RH, Hesselink MKC, Schrauwen P, Schrauwen‐Hinderling VB. Carnitine supplementation improves insulin sensitivity and skeletal muscle acetylcarnitine formation in patients with type 2 diabetes. Diabetes Obes Metab 2025; 27:2864-2877. [PMID: 40019115 PMCID: PMC11965010 DOI: 10.1111/dom.16298] [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: 10/23/2024] [Revised: 02/05/2025] [Accepted: 02/14/2025] [Indexed: 03/01/2025]
Abstract
AIM/HYPOTHESIS Recently, we reported that increasing free carnitine availability resulted in elevated skeletal muscle acetylcarnitine concentrations and restored metabolic flexibility in individuals who have impaired glucose tolerance. Metabolic flexibility is defined as the capacity to switch from predominantly fat oxidation while fasted to carbohydrate oxidation while insulin stimulated. Here we investigated if carnitine supplementation enhances the capacity of skeletal muscle to form acetylcarnitine and thereby improves insulin sensitivity and glucose homeostasis in patients with type 2 diabetes (T2DM). METHODS Thirty-two patients followed a 12-week L-carnitine treatment (2970 mg/day, orally). Insulin sensitivity was assessed by a two-step hyperinsulinemic-euglycemic clamp. In vivo skeletal muscle acetylcarnitine concentrations at rest and post-exercise (30 min, 70% Wmax) and intrahepatic lipid content (IHL) were determined by proton magnetic resonance spectroscopy (1H-MRS). All measurements were performed before and after 12 weeks of carnitine supplementation. RESULTS Compliance with the carnitine supplementation was good (as indicated by increased plasma-free carnitine levels (p < 0.01) and pill count (97.1 ± 0.7%)). Insulin-induced suppression of endogenous glucose production (31.9 ± 2.9 vs. 39.9 ± 3.2%, p = 0.020) and peripheral insulin sensitivity (Δ rate of glucose disappearance (ΔRd): 10.53 ± 1.85 vs. 13.83 ± 2.02 μmol/kg/min, p = 0.005) improved after supplementation. Resting (1.18 ± 0.13 vs. 1.54 ± 0.17 mmol/kgww, p = 0.008) and post-exercise (3.70 ± 0.22 vs. 4.53 ± 0.30 mmol/kgww, p < 0.001) skeletal muscle acetylcarnitine concentrations were both elevated after carnitine supplementation. Plasma glucose (p = 0.083) and IHL (p = 0.098) tended to be reduced after carnitine supplementation. CONCLUSION Carnitine supplementation improved insulin sensitivity and tended to lower IHL and fasting plasma glucose levels in patients with type 2 diabetes. Furthermore, carnitine supplementation increased acetylcarnitine concentration in muscle, which may underlie the beneficial effect on insulin sensitivity.
Collapse
Affiliation(s)
- Yvonne M. H. Op den Kamp‐Bruls
- Departments of Radiology and Nuclear Medicine, NUTRIM School for Nutrition and Translational Research in MetabolismMaastricht University Medical CenterMaastrichtThe Netherlands
| | - Yvo J. M. Op den Kamp
- Department of Nutrition and Movement Sciences, NUTRIM School for Nutrition and Translational Research in MetabolismMaastricht University Medical CenterMaastrichtThe Netherlands
| | - Pandichelvam Veeraiah
- Departments of Radiology and Nuclear Medicine, NUTRIM School for Nutrition and Translational Research in MetabolismMaastricht University Medical CenterMaastrichtThe Netherlands
| | - Ruben Zapata Perez
- Laboratory Genetic Metabolic Diseases, Amsterdam Gastroenterology and Metabolism, Amsterdam Cardiovascular SciencesAmsterdam University Medical Centers, University of AmsterdamAmsterdamNetherlands
| | - Esther Phielix
- Department of Nutrition and Movement Sciences, NUTRIM School for Nutrition and Translational Research in MetabolismMaastricht University Medical CenterMaastrichtThe Netherlands
| | - Bas Havekes
- Department of Internal Medicine, Division of Endocrinology, NUTRIM School for Nutrition and Translational Research in MetabolismMaastricht University Medical CenterMaastrichtThe Netherlands
| | - Gert Schaart
- Department of Nutrition and Movement Sciences, NUTRIM School for Nutrition and Translational Research in MetabolismMaastricht University Medical CenterMaastrichtThe Netherlands
| | - Esther Kornips
- Department of Nutrition and Movement Sciences, NUTRIM School for Nutrition and Translational Research in MetabolismMaastricht University Medical CenterMaastrichtThe Netherlands
| | | | | | - Joachim E. Wildberger
- Departments of Radiology and Nuclear Medicine, NUTRIM School for Nutrition and Translational Research in MetabolismMaastricht University Medical CenterMaastrichtThe Netherlands
| | - Riekelt H. Houtkooper
- Laboratory Genetic Metabolic Diseases, Amsterdam Gastroenterology and Metabolism, Amsterdam Cardiovascular SciencesAmsterdam University Medical Centers, University of AmsterdamAmsterdamNetherlands
| | - Matthijs K. C. Hesselink
- Department of Nutrition and Movement Sciences, NUTRIM School for Nutrition and Translational Research in MetabolismMaastricht University Medical CenterMaastrichtThe Netherlands
| | - Patrick Schrauwen
- Clinical EpidemiologyLeiden University Medical CenterLeidenThe Netherlands
- Institute for Clinical DiabetologyGerman Diabetes Center, Leibniz Institute for Diabetes Research at Heinrich Heine University DüsseldorfDüsseldorfGermany
| | - Vera B. Schrauwen‐Hinderling
- Departments of Radiology and Nuclear Medicine, NUTRIM School for Nutrition and Translational Research in MetabolismMaastricht University Medical CenterMaastrichtThe Netherlands
- Department of Nutrition and Movement Sciences, NUTRIM School for Nutrition and Translational Research in MetabolismMaastricht University Medical CenterMaastrichtThe Netherlands
- Institute for Clinical DiabetologyGerman Diabetes Center, Leibniz Institute for Diabetes Research at Heinrich Heine University DüsseldorfDüsseldorfGermany
- German Center for Diabetes Research (DZD)Partner DüsseldorfNeuherbergGermany
| |
Collapse
|
9
|
Heller K, Doukas P, Uhl C, Gombert A. Sex-Specific Characteristics of Perivascular Fat in Aortic Aneurysms. J Clin Med 2025; 14:3071. [PMID: 40364103 PMCID: PMC12072766 DOI: 10.3390/jcm14093071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2025] [Revised: 04/17/2025] [Accepted: 04/24/2025] [Indexed: 05/15/2025] Open
Abstract
Aortic aneurysms (AAs), the dilation or widening of the aorta, lead to dissection or rupture with high morbidity and mortality if untreated. AA displays gender disparities in its prevalence, progression and outcomes, with women having worse outcomes and faster aneurysm growth. However, current guidelines do not address gender dimorphism, emphasizing the urgent need for personalized treatment strategies and further research. Perivascular adipose tissue (PVAT), a unique type of fat surrounding blood vessels, plays a critical role in maintaining vasomotor tone and vascular homeostasis, with dysfunction associated with chronic inflammation and vessel-wall remodeling. Indeed, PVAT dysfunction promotes the development of aortic aneurysms, with hormonal and biomechanical factors exacerbating the pathological vascular microenvironment. The sexually dimorphic characteristics of PVAT include morphological, immunological, and hormonally mediated differences. Thus, targeting PVAT-mediated mechanisms may be a promising option for the (gender-specific) therapeutic management of cardiovascular pathologies. This review examines the emerging importance of PVAT in vascular health, its potential therapeutic implications for AA, and identifies gaps in the current state of research.
Collapse
Affiliation(s)
- Katja Heller
- Department of Vascular Surgery, RWTH Aachen University Hospital, 52074 Aachen, Germany; (P.D.); (C.U.); (A.G.)
| | | | | | | |
Collapse
|
10
|
Kassim Y, Sheng H, Xu G, Jin H, Iqbal T, Elashry M, Zhang K. Integrated Multi-Omics Analysis Reveals Key Regulators of Bovine Oocyte Maturation. Int J Mol Sci 2025; 26:3973. [PMID: 40362214 PMCID: PMC12071811 DOI: 10.3390/ijms26093973] [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: 03/22/2025] [Revised: 04/11/2025] [Accepted: 04/15/2025] [Indexed: 05/15/2025] Open
Abstract
A well-regulated metabolism is crucial for optimal oocyte development and embryonic health. However, the metabolic framework governing oocyte maturation remains poorly understood. Using bovine oocytes as a model, we examined metabolomic and transcriptomic alterations during the transition from the germinal vesicle (GV) to the metaphase II (MII) stage. Our findings reveal distinct metabolic shifts, including suppressed β-oxidation combined with the accumulation of long-chain fatty acids (LCFAs). Notably, progesterone emerged as a key regulator of meiotic resumption through its influence on cAMP levels. We also observed enhanced glycolysis, moderate activation of the citric acid cycle (TCA cycle), and suppression of oxidative phosphorylation (OXPHOS), alongside reduced urea cycle flux and shifts in amino acid metabolism favoring glutamate synthesis. Intriguingly, discrepancies between metabolic and transcriptional activities in pathways such as the TCA cycle and nucleotide metabolism suggest asynchronous regulation. These findings provide a comprehensive multi-omics resource, advancing our understanding of the dynamic metabolic and transcriptional landscape during bovine oocyte maturation.
Collapse
Affiliation(s)
- Yassin Kassim
- Key Laboratory of Dairy Cow Genetic Improvement and Milk Quality Research of Zhejiang Province, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
- Department of Animal and Poultry Production, Faculty of Agriculture, Minia University, El-Minya 61519, Egypt
| | - Hao Sheng
- Key Laboratory of Dairy Cow Genetic Improvement and Milk Quality Research of Zhejiang Province, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Guangjun Xu
- Key Laboratory of Dairy Cow Genetic Improvement and Milk Quality Research of Zhejiang Province, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Hao Jin
- Key Laboratory of Dairy Cow Genetic Improvement and Milk Quality Research of Zhejiang Province, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Tariq Iqbal
- Key Laboratory of Dairy Cow Genetic Improvement and Milk Quality Research of Zhejiang Province, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Mostafa Elashry
- Key Laboratory of Dairy Cow Genetic Improvement and Milk Quality Research of Zhejiang Province, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Kun Zhang
- Key Laboratory of Dairy Cow Genetic Improvement and Milk Quality Research of Zhejiang Province, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
11
|
Xiao M, Zhou N, Tian Z, Sun C. Endogenous Metabolites in Metabolic Diseases: Pathophysiologic Roles and Therapeutic Implications. J Nutr 2025:S0022-3166(25)00227-5. [PMID: 40250565 DOI: 10.1016/j.tjnut.2025.04.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2025] [Accepted: 04/14/2025] [Indexed: 04/20/2025] Open
Abstract
Breakthroughs in metabolomics technology have revealed the direct regulatory role of metabolites in physiology and disease. Recent data have highlighted the bioactive metabolites involved in the etiology and prevention and treatment of metabolic diseases such as obesity, nonalcoholic fatty liver disease, type 2 diabetes mellitus, and atherosclerosis. Numerous studies reveal that endogenous metabolites biosynthesized by host organisms or gut microflora regulate metabolic responses and disorders. Lipids, amino acids, and bile acids, as endogenous metabolic modulators, regulate energy metabolism, insulin sensitivity, and immune response through multiple pathways, such as insulin signaling cascade, chemical modifications, and metabolite-macromolecule interactions. Furthermore, the gut microbial metabolites short-chain fatty acids, as signaling regulators have a variety of beneficial impacts in regulating energy metabolic homeostasis. In this review, we will summarize information about the roles of bioactive metabolites in the pathogenesis of many metabolic diseases. Furthermore, we discuss the potential value of metabolites in the promising preventive and therapeutic perspectives of human metabolic diseases.
Collapse
Affiliation(s)
- Mengjie Xiao
- National Key Discipline, Department of Nutrition and Food Hygiene, School of Public Health, Harbin Medical University, Harbin, P. R. China; Department of Nutrition and Food Hygiene, School of Public Health, Key Laboratory of Precision Nutrition and Health, Ministry of Education, Harbin Medical University, Heilongjiang, Harbin, P. R. China
| | - Ning Zhou
- National Key Discipline, Department of Nutrition and Food Hygiene, School of Public Health, Harbin Medical University, Harbin, P. R. China; Department of Nutrition and Food Hygiene, School of Public Health, Key Laboratory of Precision Nutrition and Health, Ministry of Education, Harbin Medical University, Heilongjiang, Harbin, P. R. China
| | - Zhen Tian
- National Key Discipline, Department of Nutrition and Food Hygiene, School of Public Health, Harbin Medical University, Harbin, P. R. China; Department of Nutrition and Food Hygiene, School of Public Health, Key Laboratory of Precision Nutrition and Health, Ministry of Education, Harbin Medical University, Heilongjiang, Harbin, P. R. China.
| | - Changhao Sun
- National Key Discipline, Department of Nutrition and Food Hygiene, School of Public Health, Harbin Medical University, Harbin, P. R. China; Department of Nutrition and Food Hygiene, School of Public Health, Key Laboratory of Precision Nutrition and Health, Ministry of Education, Harbin Medical University, Heilongjiang, Harbin, P. R. China.
| |
Collapse
|
12
|
Rahim M, Bednarski TK, Hasenour CM, Banerjee DR, Trenary I, Young JD. Simultaneous in vivo multi-organ fluxomics reveals divergent metabolic adaptations in liver, heart, and skeletal muscle during obesity. Cell Rep 2025; 44:115591. [PMID: 40244853 DOI: 10.1016/j.celrep.2025.115591] [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: 02/03/2024] [Revised: 02/23/2025] [Accepted: 03/28/2025] [Indexed: 04/19/2025] Open
Abstract
We present an isotope-based metabolic flux analysis (MFA) approach to simultaneously quantify metabolic fluxes in the liver, heart, and skeletal muscle of individual mice. The platform was scaled to examine metabolic flux adaptations in age-matched cohorts of mice exhibiting varying levels of chronic obesity. We found that severe obesity increases hepatic gluconeogenesis and citric acid cycle flux, accompanied by elevated glucose oxidation in the heart that compensates for impaired fatty acid oxidation. In contrast, skeletal muscle fluxes exhibit an overall reduction in substrate oxidation. These findings demonstrate the dichotomy in fuel utilization between cardiac and skeletal muscle during worsening metabolic disease and demonstrate the divergent effects of obesity on metabolic fluxes in different organs. This multi-tissue MFA technology can be extended to address important questions about in vivo regulation of metabolism and its dysregulation in disease, which cannot be fully answered through studies of single organs or isolated cells/tissues.
Collapse
Affiliation(s)
- Mohsin Rahim
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Tomasz K Bednarski
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Clinton M Hasenour
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Deveena R Banerjee
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Irina Trenary
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Jamey D Young
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA; Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA.
| |
Collapse
|
13
|
Qin W, Zheng S, Zhou L, Liu X, Chen T, Wang X, Li Q, Zhao Y, Wang D, Xu G. High-Coverage Metabolomics Reveals Gut Microbiota-Related Metabolic Traits of Type-2 Diabetes in Serum. J Proteome Res 2025; 24:1649-1661. [PMID: 40130449 DOI: 10.1021/acs.jproteome.4c00507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/26/2025]
Abstract
Metabolic perturbations of the gut microbiome have been implicated in the pathogenesis of multiple human diseases, including type-2 diabetes (T2D). However, our understanding of the global metabolic alterations of the gut microbiota in T2D and their functional roles remains limited. To address this, we conducted a high-coverage metabolomics profiling analysis of serum samples from 1282 Chinese individuals with and without T2D. Among the 220 detected microbiota-associated compounds detected, 111 were significantly altered, forming a highly interactive regulatory network associated with T2D development. Pathway enrichment and correlation analyses revealed aberrant metabolic pathways, primarily including the activation of pyrimidine metabolism, unsaturated fatty acid biosynthesis, and diverse amino acid metabolisms such as Tryptophan metabolism, Lysine metabolism, and Branched-chain amino acid biosynthesis. A microbiota-dependent biomarker panel, comprising pipecolinic acid, methoxysalicylic acid, N-acetylhistamine, and 3-hydroxybutyrylcarnitine, was defined and validated with satisfactory sensitivity (>78%) for large-scale, population-based T2D screening. The functional role of a gut microbial product, N-acetylhistamine, was further elucidated in T2D progression through its inhibition of adenosine monophosphate-activated protein kinase phosphorylation. Overall, this study expands our understanding of gut microbiota-driven metabolic dysregulation in T2D and suggests that monitoring these metabolic changes could facilitate the diagnosis and treatment of T2D.
Collapse
Affiliation(s)
- Wangshu Qin
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
- Liaoning Province Key Laboratory of Metabolomics, Dalian 116023, China
| | - Sijia Zheng
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
- Liaoning Province Key Laboratory of Metabolomics, Dalian 116023, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Lina Zhou
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
- Liaoning Province Key Laboratory of Metabolomics, Dalian 116023, China
| | - Xinyu Liu
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
- Liaoning Province Key Laboratory of Metabolomics, Dalian 116023, China
| | - Tiantian Chen
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
- Liaoning Province Key Laboratory of Metabolomics, Dalian 116023, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiaolin Wang
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
- Liaoning Province Key Laboratory of Metabolomics, Dalian 116023, China
| | - Qi Li
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
- Liaoning Province Key Laboratory of Metabolomics, Dalian 116023, China
| | - Ying Zhao
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
- Liaoning Province Key Laboratory of Metabolomics, Dalian 116023, China
| | - Difei Wang
- Department of Gerontology and Geriatrics, Shengjing Hospital of China Medical University, Shenyang 110022, China
| | - Guowang Xu
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
- Liaoning Province Key Laboratory of Metabolomics, Dalian 116023, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
14
|
Alcover S, Ramos-Regalado L, Girón G, Muñoz-García N, Vilahur G. HDL-Cholesterol and Triglycerides Dynamics: Essential Players in Metabolic Syndrome. Antioxidants (Basel) 2025; 14:434. [PMID: 40298782 PMCID: PMC12024175 DOI: 10.3390/antiox14040434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Revised: 03/27/2025] [Accepted: 03/31/2025] [Indexed: 04/30/2025] Open
Abstract
Metabolic syndrome (MetS) is a complex cluster of interrelated metabolic disorders that significantly elevate the risk of cardiovascular disease, making it a pressing public health concern worldwide. Among the key features of MetS, dyslipidemia-characterized by altered levels of high-density lipoprotein cholesterol (HDL-C) and triglycerides (TG)-plays a crucial role in the disorder's progression. This review aims to elucidate the intricate interplay between HDL-C and TG within the context of lipid metabolism and cardiovascular health, while also addressing the detrimental impact of various cardiovascular risk factors and associated comorbidities. The dynamics of HDL-C and TG are explored, highlighting their reciprocal relationship and respective contributions to the pathophysiology of MetS. Elevated levels of TGs are consistently associated with reduced concentrations of HDL-C, resulting in a lipid profile that promotes the development of vascular disease. Specifically, as TG levels rise, the protective cardiovascular effects of HDL-C are diminished, leading to the increased accumulation of pro-atherogenic TG-rich lipoproteins and low-density lipoprotein particles within the vascular wall, contributing to the progression of atheromas, which can ultimately result in significant ischemic cardiovascular events. Ultimately, this paper underscores the significance of HDL and TG as essential targets for therapeutic intervention, emphasizing their potential in effectively managing MetS and reducing cardiovascular risk.
Collapse
Affiliation(s)
- Sebastià Alcover
- Research Institute Sant Pau (IR SANT PAU), 08041 Barcelona, Spain; (S.A.); (L.R.-R.); (G.G.); (N.M.-G.)
- Facultat de Biologia, Universitat de Barcelona, 08028 Barcelona, Spain
| | - Lisaidy Ramos-Regalado
- Research Institute Sant Pau (IR SANT PAU), 08041 Barcelona, Spain; (S.A.); (L.R.-R.); (G.G.); (N.M.-G.)
- Facultat de Biologia, Universitat de Barcelona, 08028 Barcelona, Spain
| | - Gabriela Girón
- Research Institute Sant Pau (IR SANT PAU), 08041 Barcelona, Spain; (S.A.); (L.R.-R.); (G.G.); (N.M.-G.)
- Facultat de Biociències, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain
| | - Natàlia Muñoz-García
- Research Institute Sant Pau (IR SANT PAU), 08041 Barcelona, Spain; (S.A.); (L.R.-R.); (G.G.); (N.M.-G.)
- Facultat de Biologia, Universitat de Barcelona, 08028 Barcelona, Spain
| | - Gemma Vilahur
- Research Institute Sant Pau (IR SANT PAU), 08041 Barcelona, Spain; (S.A.); (L.R.-R.); (G.G.); (N.M.-G.)
- Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares, Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
15
|
Shastry A, Wilkinson MS, Miller DM, Kuriakose M, Veeneman JLMH, Smith MR, Hindmarch CCT, Dunham-Snary KJ. Multi-tissue metabolomics reveal mtDNA- and diet-specific metabolite profiles in a mouse model of cardiometabolic disease. Redox Biol 2025; 81:103541. [PMID: 39983345 PMCID: PMC11893332 DOI: 10.1016/j.redox.2025.103541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Accepted: 02/08/2025] [Indexed: 02/23/2025] Open
Abstract
RATIONALE Excess consumption of sugar- and fat-rich foods has heightened the prevalence of cardiometabolic disease, which remains a driver of cardiovascular disease- and type II diabetes-related mortality globally. Skeletal muscle insulin resistance is an early feature of cardiometabolic disease and is a precursor to diabetes. Insulin resistance risk varies with self-reported race, whereby African-Americans have a greater risk of diabetes development relative to their White counterparts. Self-reported race is strongly associated with mitochondrial DNA (mtDNA) haplogroups, and previous reports have noted marked differences in bioenergetic and metabolic parameters in cells belonging to distinct mtDNA haplogroups, but the mechanism of these associations remains unknown. Additionally, distinguishing nuclear DNA (nDNA) and mtDNA contributions to cardiometabolic disease remains challenging in humans. The Mitochondrial-Nuclear eXchange (MNX) mouse model enables in vivo preclinical investigation of the role of mtDNA in cardiometabolic disease development, and has been implemented in studies of insulin resistance, fatty liver disease, and obesity in previous reports. METHODS Six-week-old male C57nDNA:C57mtDNA and C3HnDNA:C3HmtDNA wild-type mice, and C57nDNA:C3HmtDNA and C3HnDNA:C57mtDNA MNX mice, were fed sucrose-matched high-fat (45% kcal fat) or control diet (10% kcal fat) until 12 weeks of age (n = 5/group). Mice were weighed weekly and total body fat was collected at euthanasia. Gastrocnemius skeletal muscle and plasma metabolomes were characterized using untargeted dual-chromatography mass spectrometry; both hydrophilic interaction liquid chromatography (HILIC) and C18 columns were used, in positive- and negative-ion modes, respectively. RESULTS Comparative analyses between nDNA-matched wild-type and MNX strains demonstrated significantly increased body fat percentage in mice possessing C57mtDNA regardless of nDNA background. High-fat diet in mice possessing C57mtDNA was associated with differential abundance of phosphatidylcholines, lysophosphatidylcholines, phosphatidylethanolamines, and glucose. Conversely, high-fat diet in mice possessing C3HmtDNA was associated with differential abundance of phosphatidylcholines, cardiolipins, and alanine. Glycerophospholipid metabolism and beta-alanine signaling pathways were enriched in skeletal muscle and plasma, indicating mtDNA-directed priming of mitochondria towards oxidative stress and increased fatty acid oxidation in C57nDNA:C57mtDNA wild-type and C3HnDNA:C57mtDNA MNX mice, relative to their nDNA-matched counterparts. In mtDNA-matched mice, C57mtDNA was associated with metabolite co-expression related to the pentose phosphate pathway and sugar-related metabolism; C3HmtDNA was associated with branched chain amino acid metabolite co-expression. CONCLUSIONS These results reveal novel nDNA-mtDNA interactions that drive significant changes in metabolite levels. Alterations to key metabolites involved in mitochondrial bioenergetic dysfunction and electron transport chain activity are implicated in elevated beta-oxidation during high-fat diet feeding; abnormally elevated rates of beta-oxidation may be a key driver of insulin resistance. The results reported here support the hypothesis that mtDNA influences cardiometabolic disease-susceptibility by modulating mitochondrial function and metabolic pathways.
Collapse
Affiliation(s)
- Abhishek Shastry
- Department of Medicine, Queen's University, Kingston, ON, K7L 3N6, Canada
| | - Mia S Wilkinson
- Department of Medicine, Queen's University, Kingston, ON, K7L 3N6, Canada
| | - Dalia M Miller
- Department of Medicine, Queen's University, Kingston, ON, K7L 3N6, Canada
| | - Michelle Kuriakose
- Department of Biomedical & Molecular Sciences, Queen's University, Kingston, ON, K7L 3N6, Canada
| | | | - Matthew Ryan Smith
- Atlanta Veterans Affairs Health Care System, Decatur, GA, 30033, USA; Department of Medicine, Emory University, Atlanta, GA, 30322, USA
| | - Charles C T Hindmarch
- Department of Medicine, Queen's University, Kingston, ON, K7L 3N6, Canada; Department of Biomedical & Molecular Sciences, Queen's University, Kingston, ON, K7L 3N6, Canada; Queen's CardioPulmonary Unit, Queen's University, Kingston, ON, K7L 3N6, Canada
| | - Kimberly J Dunham-Snary
- Department of Medicine, Queen's University, Kingston, ON, K7L 3N6, Canada; Department of Biomedical & Molecular Sciences, Queen's University, Kingston, ON, K7L 3N6, Canada.
| |
Collapse
|
16
|
Xu M, Lv D, Wei H, Li Z, Jin S, Liu Q, Zhang Y, Liu Y. Effects of antidiabetic agents on lipid metabolism of skeletal muscle: A narrative review. Diabetes Obes Metab 2025; 27:1693-1707. [PMID: 39807619 DOI: 10.1111/dom.16189] [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: 09/27/2024] [Revised: 12/17/2024] [Accepted: 12/26/2024] [Indexed: 01/16/2025]
Abstract
Metabolic syndrome-related diseases frequently involve disturbances in skeletal muscle lipid metabolism. The accumulation of lipid metabolites, lipid-induced mitochondrial stress in skeletal muscle cells, as well as the inflammation of adjacent adipose tissue, are associated with the development of insulin resistance and metabolic dysfunction. Consequently, when antidiabetic medications are used to treat various chronic conditions related to hyperglycaemia, the impact on skeletal muscle lipid metabolism should not be overlooked. However, current research has predominantly focused on muscle mass rather than skeletal muscle lipid metabolism and its interplay with glucose metabolism. In this review, we summarised the latest research on the effects of antidiabetic drugs and certain natural compounds with antidiabetic activity on skeletal muscle lipid metabolism, focusing on data from preclinical to clinical studies. Given the widespread use of antidiabetic drugs, a better understanding of their effects on skeletal muscle lipid metabolism merits further attention in future research.
Collapse
Affiliation(s)
- Ming Xu
- Department of Endocrinology, First Hospital of Shanxi Medical University, Taiyuan, China
- The First Clinical Medical College, Shanxi Medical University, Taiyuan, China
| | - Dongqing Lv
- Department of Endocrinology, First Hospital of Shanxi Medical University, Taiyuan, China
- The First Clinical Medical College, Shanxi Medical University, Taiyuan, China
| | - Hongxia Wei
- Department of Endocrinology, First Hospital of Shanxi Medical University, Taiyuan, China
- The First Clinical Medical College, Shanxi Medical University, Taiyuan, China
| | - Zhe Li
- Department of Endocrinology, First Hospital of Shanxi Medical University, Taiyuan, China
- The First Clinical Medical College, Shanxi Medical University, Taiyuan, China
| | - Shuqing Jin
- Department of Endocrinology, First Hospital of Shanxi Medical University, Taiyuan, China
- The First Clinical Medical College, Shanxi Medical University, Taiyuan, China
| | - Qinhao Liu
- Department of Endocrinology, First Hospital of Shanxi Medical University, Taiyuan, China
- The First Clinical Medical College, Shanxi Medical University, Taiyuan, China
| | - Yi Zhang
- Department of Pharmacology, Shanxi Medical University, Taiyuan, China
- Medicinal Basic Research Innovation Center of Chronic Kidney Disease, Ministry of Education, Shanxi Medical University, Taiyuan, China
| | - Yunfeng Liu
- Department of Endocrinology, First Hospital of Shanxi Medical University, Taiyuan, China
- The First Clinical Medical College, Shanxi Medical University, Taiyuan, China
| |
Collapse
|
17
|
Xiang F, Zhang Z, Xie J, Xiong S, Yang C, Liao D, Xia B, Lin L. Comprehensive review of the expanding roles of the carnitine pool in metabolic physiology: beyond fatty acid oxidation. J Transl Med 2025; 23:324. [PMID: 40087749 PMCID: PMC11907856 DOI: 10.1186/s12967-025-06341-5] [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: 11/09/2024] [Accepted: 03/01/2025] [Indexed: 03/17/2025] Open
Abstract
Traditionally, the carnitine pool is closely related to fatty acid metabolism. However, with increasing research, the pleiotropic effects of the carnitine pool have gradually emerged. The purpose of this review is to comprehensively investigate of the emerging understanding of the pleiotropic role of the carnitine pool, carnitine/acylcarnitines are not only auxiliaries or metabolites of fatty acid oxidation, but also play more complex and diverse roles, including energy metabolism, mitochondrial homeostasis, epigenetic regulation, regulation of inflammation and the immune system, tumor biology, signal transduction, and neuroprotection. This review provides an overview of the complex network of carnitine synthesis, transport, shuttle, and regulation, carnitine/acylcarnitines have the potential to be used as communication molecules, biomarkers and therapeutic targets for multiple diseases, with profound effects on intercellular communication, metabolic interactions between organs and overall metabolic health. The purpose of this review is to comprehensively summarize the multidimensional biological effects of the carnitine pool beyond its traditional role in fatty acid oxidation and to summarize the systemic effects mediated by carnitine/acylcarnitine to provide new perspectives for pharmacological research and treatment innovation and new strategies for the prevention and treatment of a variety of diseases.
Collapse
Affiliation(s)
- Feng Xiang
- Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Zhimin Zhang
- Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Jingchen Xie
- Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Suhui Xiong
- Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Chen Yang
- Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Duanfang Liao
- Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Bohou Xia
- Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, China.
| | - Limei Lin
- Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, China.
| |
Collapse
|
18
|
Lee WD, Weilandt DR, Liang L, MacArthur MR, Jaiswal N, Ong O, Mann CG, Chu Q, Hunter CJ, Ryseck RP, Lu W, Oschmann AM, Cowan AJ, TeSlaa TA, Bartman CR, Jang C, Baur JA, Titchenell PM, Rabinowitz JD. Lactate homeostasis is maintained through regulation of glycolysis and lipolysis. Cell Metab 2025; 37:758-771.e8. [PMID: 39889702 PMCID: PMC11926601 DOI: 10.1016/j.cmet.2024.12.009] [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: 04/30/2024] [Revised: 09/19/2024] [Accepted: 12/17/2024] [Indexed: 02/03/2025]
Abstract
Lactate is among the highest flux circulating metabolites. It is made by glycolysis and cleared by both tricarboxylic acid (TCA) cycle oxidation and gluconeogenesis. Severe lactate elevations are life-threatening, and modest elevations predict future diabetes. How lactate homeostasis is maintained, however, remains poorly understood. Here, we identify, in mice, homeostatic circuits regulating lactate production and consumption. Insulin induces lactate production by upregulating glycolysis. We find that hyperlactatemia inhibits insulin-induced glycolysis, thereby suppressing excess lactate production. Unexpectedly, insulin also promotes lactate TCA cycle oxidation. The mechanism involves lowering circulating fatty acids, which compete with lactate for mitochondrial oxidation. Similarly, lactate can promote its own consumption by lowering circulating fatty acids via the adipocyte-expressed G-protein-coupled receptor hydroxycarboxylic acid receptor 1 (HCAR1). Quantitative modeling suggests that these mechanisms suffice to produce lactate homeostasis, with robustness to noise and perturbation of individual regulatory mechanisms. Thus, through regulation of glycolysis and lipolysis, lactate homeostasis is maintained.
Collapse
Affiliation(s)
- Won Dong Lee
- Department of Chemistry, Princeton University, Princeton, NJ, USA; Lewis-Sigler Institute of Integrative Genomics, Princeton University, Princeton, NJ, USA; Ludwig Institute for Cancer Research, Princeton University, Princeton, NJ, USA
| | - Daniel R Weilandt
- Department of Chemistry, Princeton University, Princeton, NJ, USA; Lewis-Sigler Institute of Integrative Genomics, Princeton University, Princeton, NJ, USA; Ludwig Institute for Cancer Research, Princeton University, Princeton, NJ, USA
| | - Lingfan Liang
- Department of Chemistry, Princeton University, Princeton, NJ, USA; Lewis-Sigler Institute of Integrative Genomics, Princeton University, Princeton, NJ, USA; Ludwig Institute for Cancer Research, Princeton University, Princeton, NJ, USA
| | - Michael R MacArthur
- Department of Chemistry, Princeton University, Princeton, NJ, USA; Lewis-Sigler Institute of Integrative Genomics, Princeton University, Princeton, NJ, USA; Ludwig Institute for Cancer Research, Princeton University, Princeton, NJ, USA
| | - Natasha Jaiswal
- Department of Health and Kinesiology, Purdue University, West Lafayette, IN, USA
| | - Olivia Ong
- Department of Health and Kinesiology, Purdue University, West Lafayette, IN, USA
| | - Charlotte G Mann
- Department of Health Sciences and Technology, ETH Zürich, Schwerzenbach, Switzerland
| | - Qingwei Chu
- Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Craig J Hunter
- Department of Chemistry, Princeton University, Princeton, NJ, USA; Lewis-Sigler Institute of Integrative Genomics, Princeton University, Princeton, NJ, USA; Ludwig Institute for Cancer Research, Princeton University, Princeton, NJ, USA
| | - Rolf-Peter Ryseck
- Department of Chemistry, Princeton University, Princeton, NJ, USA; Lewis-Sigler Institute of Integrative Genomics, Princeton University, Princeton, NJ, USA; Ludwig Institute for Cancer Research, Princeton University, Princeton, NJ, USA
| | - Wenyun Lu
- Department of Chemistry, Princeton University, Princeton, NJ, USA; Lewis-Sigler Institute of Integrative Genomics, Princeton University, Princeton, NJ, USA; Ludwig Institute for Cancer Research, Princeton University, Princeton, NJ, USA
| | - Anna M Oschmann
- Department of Chemistry, Princeton University, Princeton, NJ, USA; Lewis-Sigler Institute of Integrative Genomics, Princeton University, Princeton, NJ, USA; Ludwig Institute for Cancer Research, Princeton University, Princeton, NJ, USA
| | - Alexis J Cowan
- Lewis-Sigler Institute of Integrative Genomics, Princeton University, Princeton, NJ, USA
| | - Tara A TeSlaa
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Caroline R Bartman
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Cholsoon Jang
- Department of Biological Chemistry, University of California Irvine, Irvine, CA, USA
| | - Joseph A Baur
- Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Paul M Titchenell
- Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Joshua D Rabinowitz
- Department of Chemistry, Princeton University, Princeton, NJ, USA; Lewis-Sigler Institute of Integrative Genomics, Princeton University, Princeton, NJ, USA; Ludwig Institute for Cancer Research, Princeton University, Princeton, NJ, USA.
| |
Collapse
|
19
|
Bork J, Markus MRP, Ewert R, Nauck M, Templin C, Völzke H, Kastenmüller G, Artati A, Adamski J, Dörr M, Friedrich N, Bahls M. The Metabolic Signature of Cardiorespiratory Fitness. Scand J Med Sci Sports 2025; 35:e70034. [PMID: 40072034 PMCID: PMC11899505 DOI: 10.1111/sms.70034] [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: 09/19/2024] [Revised: 02/23/2025] [Accepted: 02/28/2025] [Indexed: 03/15/2025]
Abstract
High cardiorespiratory fitness (CRF) is associated with better overall health. This study aimed to find a metabolic signature associated with CRF to identify health-promoting effects. CRF based on cardiopulmonary exercise testing, targeted and untargeted metabolomics approaches based on mass spectrometry, and clinical data from two independent cohorts of the Study of Health in Pomerania (SHIP) were used. Sex-stratified linear regression models were adjusted for age, smoking, and height to relate CRF with individual metabolites. A total of 132 (SHIP-START-2: 483 men with a median age of 58 years and 450 women with a median age of 56 years) and 118 (SHIP-TREND-0: 341 men and 371 women both with a median age of 51 years) metabolites were associated with CRF. Lipids showed bidirectional relations to CRF independent of sex. Specific subsets of sphingomyelins were positively related to CRF in men (SM (OH) C14:1, SM(OH)C22:2 SM C16:0, SM C20:2 SM(OH)C24:1) and inversely in women (SM C16:1, SM C18:0, SM C18:1). Metabolites involved in energy production (citrate and succinylcarnitine) were only associated with CRF in men. In women, xenobiotics (hippurate, stachydrine) were related to CRF. The sex-specific metabolic signature of CRF is influenced by sphingomyelins, energy substrates, and xenobiotics. The greater effect estimates seen in women may emphasize the important role of CRF in maintaining metabolic health. Future research should explore how this profile changes with different types of exercise interventions or diseases in diverse populations and how these metabolites could be implemented in primary prevention settings.
Collapse
Affiliation(s)
- Julia Bork
- Department of Internal Medicine BUniversity Medicine GreifswaldGreifswaldGermany
- German Centre for Cardiovascular Research (DZHK)Partner Site GreifswaldGreifswaldGermany
| | - Marcello R. P. Markus
- Department of Internal Medicine BUniversity Medicine GreifswaldGreifswaldGermany
- German Centre for Cardiovascular Research (DZHK)Partner Site GreifswaldGreifswaldGermany
- German Center for Diabetes Research (DZD)Partner Site GreifswaldGreifswaldGermany
| | - Ralf Ewert
- Department of Internal Medicine BUniversity Medicine GreifswaldGreifswaldGermany
| | - Matthias Nauck
- German Centre for Cardiovascular Research (DZHK)Partner Site GreifswaldGreifswaldGermany
- Institute of Clinical Chemistry and Laboratory MedicineUniversity Medicine GreifswaldGreifswaldGermany
| | - Christian Templin
- Department of Internal Medicine BUniversity Medicine GreifswaldGreifswaldGermany
- German Centre for Cardiovascular Research (DZHK)Partner Site GreifswaldGreifswaldGermany
| | - Henry Völzke
- German Centre for Cardiovascular Research (DZHK)Partner Site GreifswaldGreifswaldGermany
- Institute for Community Medicine, SHIP‐KEFUniversity Medicine GreifswaldGreifswaldGermany
| | - Gabi Kastenmüller
- Institute of Bioinformatics and Systems BiologyHelmholtz Zentrum MünchenNeuherbergGermany
| | - Anna Artati
- Metabolomics and Proteomics CoreHelmholtz Center MunichNeuherbergGermany
| | - Jerzy Adamski
- Institute of Experimental Genetics, Helmholtz Zentrum MünchenGerman Research Center for Environmental HealthNeuherbergGermany
- Department of Biochemistry, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- Institute of Biochemistry, Faculty of MedicineUniversity of LjubljanaLjubljanaSlovenia
| | - Marcus Dörr
- Department of Internal Medicine BUniversity Medicine GreifswaldGreifswaldGermany
- German Centre for Cardiovascular Research (DZHK)Partner Site GreifswaldGreifswaldGermany
| | - Nele Friedrich
- German Centre for Cardiovascular Research (DZHK)Partner Site GreifswaldGreifswaldGermany
- Institute of Clinical Chemistry and Laboratory MedicineUniversity Medicine GreifswaldGreifswaldGermany
| | - Martin Bahls
- Department of Internal Medicine BUniversity Medicine GreifswaldGreifswaldGermany
- German Centre for Cardiovascular Research (DZHK)Partner Site GreifswaldGreifswaldGermany
| |
Collapse
|
20
|
Abushamat LA, Jia X, Xu L, Cheng C, Ndumele CE, Sun C, Windham BG, Matsushita K, Yu B, Nambi V, Bozkurt B, Reusch JEB, Rebholz CM, Selvin E, Ballantyne CM, Hoogeveen RC. Does Adiponectin Inform Cardiovascular Risk in Older Adults?: The ARIC Study. JACC. ADVANCES 2025; 4:101625. [PMID: 39983616 PMCID: PMC11891717 DOI: 10.1016/j.jacadv.2025.101625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 01/07/2025] [Accepted: 01/17/2025] [Indexed: 02/23/2025]
Abstract
BACKGROUND Adiponectin, an atheroprotective adipokine, is associated with adverse outcomes in older age. It is unclear whether this is due to overlapping pathophysiological pathways with N-terminal pro-B-type natriuretic peptide (NT-proBNP). OBJECTIVES The authors investigated adiponectin's associations with cardiovascular disease (CVD) risk in older adults. METHODS Among Atherosclerosis Risk in Communities prospective cohort study participants without baseline CVD at visit 5 (n = 4,729, mean age 75), adiponectin and adiponectin/NT-proBNP category associations with incident CVD events (heart failure [HF], atherosclerotic cardiovascular disease, and death during median follow-up of 5.5 years) and echocardiographic parameters were assessed. Metabolomic signatures of adiponectin/NT-proBNP categories were explored. RESULTS Higher adiponectin was associated with older age, female sex, and less obesity, diabetes, and hypertension but increased risk for incident HF (HR: 1.91 [95% CI: 1.49-2.44], per natural-log unit increase) and CVD death (HR: 1.67 [95% CI: 1.19-2.32]). Interaction of NT-proBNP with adiponectin was significant for HF (P-interaction = 0.03). There was no significant association between adiponectin and heart failure with preserved ejection fraction after adjusting for NT-proBNP. Elevations of both biomarkers (A+ [upper tertile]/N+ [≥125 pg/mL]) had higher risk (vs A+/N-; HF: HR 5.41 [95% CI: 2.72-10.78]; CVD death: HR 3.50 [95% CI: 1.48-8.24]). Compared with A+/N-, A-/N+ had increased risk for HF (HR 2.84 [95% CI: 1.41-5.72]) while A-/N- had no increased event risk. A+/N+'s metabolomic signature (88% similar to NT-proBNP's) showed acylcarnitine species consistent with incomplete beta-oxidation; top-associated metabolites were significantly associated with HF and CVD death. CONCLUSIONS Elevated adiponectin and NT-proBNP in older adults are associated with increased risk for HF and CVD death beyond traditional risk factors.
Collapse
Affiliation(s)
- Layla A Abushamat
- Section of Cardiovascular Research, Population Science, Department of Medicine, Baylor College of Medicine, Houston, Texas, USA.
| | - Xiaoming Jia
- Section of Cardiovascular Research, Population Science, Department of Medicine, Baylor College of Medicine, Houston, Texas, USA; Section of Cardiology, and Population Science, Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - Lu Xu
- Section of Cardiovascular Research, Population Science, Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - Chao Cheng
- Section of Epidemiology and Population Science, Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - Chiadi E Ndumele
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA; Johns Hopkins Ciccarone Center for the Prevention of Cardiovascular Disease, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Caroline Sun
- Section of Cardiovascular Research, Population Science, Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - B Gwen Windham
- Department of Medicine, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Kunihiro Matsushita
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Bing Yu
- Department of Epidemiology, Human Genetics and Environmental Sciences, School of Public Health, University of Texas Health Science Center, Houston, Texas, USA
| | - Vijay Nambi
- Section of Cardiovascular Research, Population Science, Department of Medicine, Baylor College of Medicine, Houston, Texas, USA; Section of Cardiology, and Population Science, Department of Medicine, Baylor College of Medicine, Houston, Texas, USA; Section of Cardiology, Michael E. DeBakey Veterans Affairs Medical Center, Houston, Texas, USA
| | - Biykem Bozkurt
- Section of Cardiology, and Population Science, Department of Medicine, Baylor College of Medicine, Houston, Texas, USA; Section of Cardiology, Michael E. DeBakey Veterans Affairs Medical Center, Houston, Texas, USA; Winters Center for Heart Failure Research, Baylor College of Medicine, Houston, Texas, USA
| | - Jane E B Reusch
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Casey M Rebholz
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Elizabeth Selvin
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Christie M Ballantyne
- Section of Cardiovascular Research, Population Science, Department of Medicine, Baylor College of Medicine, Houston, Texas, USA; Section of Cardiology, and Population Science, Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - Ron C Hoogeveen
- Section of Cardiovascular Research, Population Science, Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
21
|
Xie C, Qi C, Zhang J, Wang W, Meng X, Aikepaer A, Lin Y, Su C, Liu Y, Feng X, Gao H. When short-chain fatty acids meet type 2 diabetes mellitus: Revealing mechanisms, envisioning therapies. Biochem Pharmacol 2025; 233:116791. [PMID: 39894305 DOI: 10.1016/j.bcp.2025.116791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 01/19/2025] [Accepted: 01/30/2025] [Indexed: 02/04/2025]
Abstract
Evidence is accumulating that short-chain fatty acids (SCFAs) produced by the gut microbiota play pivotal roles in host metabolism. They contribute to the metabolic regulation and energy homeostasis of the host not only by preserving intestinal health and serving as energy substrates but also by entering the systemic circulation as signaling molecules, affecting the gut-brain axis and neuroendocrine-immune network. This review critically summarizes the current knowledge regarding the effects of SCFAs in the fine-tuning of the pathogenesis of type 2 diabetes mellitus (T2DM) and insulin resistance, with an emphasis on the complex relationships among diet, microbiota-derived metabolites, T2DM inflammation, glucose metabolism, and the underlying mechanisms involved. We hold an optimistic view that elucidating how diet can influence gut bacterial composition and activity, SCFA production, and metabolic functions in the host will advance our understanding of the mutual interactions of the intestinal microbiota with other metabolically active organs, and may pave the way for harnessing these pathways to develop novel personalized therapeutics for glucometabolic disorders.
Collapse
Affiliation(s)
- Cong Xie
- Department of Endocrinology, Yuquan Hospital, School of Clinical Medicine, Tsinghua University, Beijing 100040 China
| | - Cong Qi
- Department of Endocrinology, Yuquan Hospital, School of Clinical Medicine, Tsinghua University, Beijing 100040 China
| | - Jianwen Zhang
- Department of Endocrinology, Yuquan Hospital, School of Clinical Medicine, Tsinghua University, Beijing 100040 China; School of Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617 China
| | - Wei Wang
- Department of Endocrinology, Yuquan Hospital, School of Clinical Medicine, Tsinghua University, Beijing 100040 China
| | - Xing Meng
- Department of Endocrinology, Yuquan Hospital, School of Clinical Medicine, Tsinghua University, Beijing 100040 China; School of Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617 China
| | - Aifeila Aikepaer
- Department of Endocrinology, Yuquan Hospital, School of Clinical Medicine, Tsinghua University, Beijing 100040 China; Dongzhimen Hospital, the First Clinical Medical School of Beijing University of Chinese Medicine, Beijing 100700 China
| | - Yuhan Lin
- Department of Endocrinology, Yuquan Hospital, School of Clinical Medicine, Tsinghua University, Beijing 100040 China; Dongzhimen Hospital, the First Clinical Medical School of Beijing University of Chinese Medicine, Beijing 100700 China
| | - Chang Su
- Life Science and Engineering College, Northwest Minzu University, Lanzhou 730124 China
| | - Yunlu Liu
- Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700 China
| | - Xingzhong Feng
- Department of Endocrinology, Yuquan Hospital, School of Clinical Medicine, Tsinghua University, Beijing 100040 China.
| | - Huijuan Gao
- Department of Endocrinology, Yuquan Hospital, School of Clinical Medicine, Tsinghua University, Beijing 100040 China.
| |
Collapse
|
22
|
Abushamat LA, Yu B, Hoogeveen RC, Sun C, Cheng C, Hartig SM, Herman MA, Balasubramanyam A, Reusch JE, Selvin E, Ndumele CE, Nambi V, Ballantyne CM. Erythritol, Erythronate, and Cardiovascular Outcomes in Older Adults in the ARIC Study. JACC. ADVANCES 2025; 4:101605. [PMID: 39983608 PMCID: PMC11889355 DOI: 10.1016/j.jacadv.2025.101605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Revised: 01/08/2025] [Accepted: 01/10/2025] [Indexed: 02/23/2025]
Abstract
BACKGROUND Circulating erythritol, an endogenously produced metabolite and an artificial sweetener, is associated with cardiovascular outcomes. OBJECTIVES The authors assessed associations of erythritol and its downstream metabolite, erythronate, with cardiovascular risk factors and events in older adults in the ARIC (Atherosclerosis Risk In Communities) study (visit 5, 2011-2013). METHODS We included 4,006 participants without prevalent cardiovascular disease and with metabolomic profiling. Erythritol and erythronate were measured by mass spectrometry. We analyzed associations of log-transformed erythritol and erythronate with cardiovascular risk factors and events using Cox proportional hazard models. RESULTS Participants in the highest tertiles of erythritol or erythronate were older, more likely to have diabetes, hypertension, hyperlipidemia, or microalbuminuria, and had higher body mass index and cardiac biomarkers and lower estimated glomerular filtration rate (P < 0.001). Over median follow-up of 8.41 (7.62, 8.93) years, higher erythritol and erythronate concentrations were significantly associated with heart failure (HF) hospitalization, HF with preserved ejection fraction, cardiovascular death, and total mortality after adjustment for demographics and traditional cardiovascular risk factors. Erythronate was additionally significantly associated with coronary heart disease (HR: 1.30 [95% CI: 1.04-1.61], P = 0.02), stroke (1.40 [95% CI: 1.08-1.83], P = 0.012), and HF with reduced ejection fraction (1.38 [95% CI: 1.09-1.74], P = 0.007). Diabetes status did not modify any of these associations (P for interaction >0.20). CONCLUSIONS Circulating erythritol and erythronate levels are markers of cardiometabolic health and cardiovascular outcomes in an older adult population. In particular, erythronate is associated with all cardiovascular outcomes assessed. Future studies should assess the role of erythronate and its related pathways in cardiovascular disease.
Collapse
Affiliation(s)
- Layla A Abushamat
- Section of Cardiovascular Research, Department of Medicine, Baylor College of Medicine, Houston, Texas, USA.
| | - Bing Yu
- Department of Epidemiology, School of Public Health, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Ron C Hoogeveen
- Section of Cardiovascular Research, Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - Caroline Sun
- Section of Cardiovascular Research, Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - Chao Cheng
- Section of Epidemiology and Population Science, Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - Sean M Hartig
- Section of Endocrinology, Diabetes and Metabolism, Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - Mark A Herman
- Section of Endocrinology, Diabetes and Metabolism, Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - Ashok Balasubramanyam
- Section of Endocrinology, Diabetes and Metabolism, Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - Jane Eb Reusch
- Division of Endocrinology, Metabolism, and Diabetes, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA; Rocky Mountain Regional VAMC, Aurora, Colorado, USA
| | - Elizabeth Selvin
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Chiadi E Ndumele
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA; Johns Hopkins Ciccarone Center for the Prevention of Cardiovascular Disease, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Vijay Nambi
- Section of Cardiovascular Research, Department of Medicine, Baylor College of Medicine, Houston, Texas, USA; Section of Cardiology, Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - Christie M Ballantyne
- Section of Cardiovascular Research, Department of Medicine, Baylor College of Medicine, Houston, Texas, USA; Section of Cardiology, Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
23
|
An J, Astapova I, Zhang G, Cangelosi AL, Ilkayeva O, Marchuk H, Muehlbauer MJ, George T, Brozinick J, Herman MA, Newgard CB. Integration of metabolomic and transcriptomic analyses reveals regulatory functions of the ChREBP transcription factor in energy metabolism. Cell Rep 2025; 44:115278. [PMID: 39921857 DOI: 10.1016/j.celrep.2025.115278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 12/03/2024] [Accepted: 01/16/2025] [Indexed: 02/10/2025] Open
Abstract
The transcription factor carbohydrate response element binding protein (ChREBP) activates genes of glucose, fructose, and lipid metabolism in response to carbohydrate feeding. Integrated transcriptomic and metabolomic analyses in rats with GalNac-siRNA-mediated suppression of ChREBP expression in liver reveal other ChREBP functions. GalNac-siChREBP treatment reduces expression of genes involved in coenzyme A (CoA) biosynthesis, with lowering of CoA and short-chain acyl-CoA levels. Despite suppression of pyruvate kinase, pyruvate levels are maintained, possibly via increased expression of pyruvate and amino acid transporters. In addition, expression of multiple anaplerotic enzymes is decreased by GalNac-siChREBP treatment, affecting TCA cycle intermediates. Finally, GalNAc-siChREBP treatment suppresses late steps in purine and NAD synthesis, with increases in precursors and lowering of end products in both pathways. In sum, our study reveals functions of ChREBP beyond its canonical roles in carbohydrate and lipid metabolism to include regulation of substrate transport, mitochondrial function, and energy balance.
Collapse
Affiliation(s)
- Jie An
- Sarah W. Stedman Nutrition and Metabolism Center & Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC, USA
| | - Inna Astapova
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Guofang Zhang
- Sarah W. Stedman Nutrition and Metabolism Center & Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC, USA; Division of Endocrinology, Metabolism and Nutrition, Department of Medicine, Duke University Medical Center, Durham, NC, USA
| | - Andrew L Cangelosi
- Sarah W. Stedman Nutrition and Metabolism Center & Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC, USA
| | - Olga Ilkayeva
- Sarah W. Stedman Nutrition and Metabolism Center & Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC, USA; Division of Endocrinology, Metabolism and Nutrition, Department of Medicine, Duke University Medical Center, Durham, NC, USA
| | - Hannah Marchuk
- Sarah W. Stedman Nutrition and Metabolism Center & Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC, USA
| | - Michael J Muehlbauer
- Sarah W. Stedman Nutrition and Metabolism Center & Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC, USA
| | - Tabitha George
- Sarah W. Stedman Nutrition and Metabolism Center & Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC, USA
| | | | - Mark A Herman
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Christopher B Newgard
- Sarah W. Stedman Nutrition and Metabolism Center & Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC, USA; Division of Endocrinology, Metabolism and Nutrition, Department of Medicine, Duke University Medical Center, Durham, NC, USA; Department of Pharmacology & Cancer Biology, Duke University Medical Center, Durham, NC, USA.
| |
Collapse
|
24
|
Ahmed N, Walker S, Roma A, Minden MD, Spagnuolo PA. Dietary Modulation of Fatty Acid Oxidation Imparts Stem Cell Protection in Bone Marrow. Nutr Cancer 2025; 77:530-536. [PMID: 39887185 DOI: 10.1080/01635581.2025.2459445] [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: 09/26/2024] [Revised: 01/21/2025] [Accepted: 01/22/2025] [Indexed: 02/01/2025]
Abstract
Hematopoietic stem cells (HSCs) maintain production of all functional blood cells and are located within the bone marrow. In pathological conditions, such as obesity or leukemia, changes in these cells contribute to disease pathophysiology. In this study, we examined the impact of metabolic modulation of stem and progenitor cells within the bone marrow during diet-induced obesity (DIO) and leukemia relapse. Avocatin B (Avo), an inhibitor of fatty acid oxidation (FAO), was provided in the diet and its impact on stem cells using two disease models was tested. In DIO, high fat diet(HFD)-induced alterations in HSC number and function were attenuated with Avo (HFD: 46.9% decrease compared to control; p < 0.001; whereas DIO + Avo: 58.8% recovery; p < 0.05). In leukemia relapse, dietary Avo delayed disease reoccurrence. Taken together, addition of Avo into the diet imparts protection in the bone marrow.
Collapse
Affiliation(s)
- Nawaz Ahmed
- Department of Food Science, University of Guelph, Guelph, Ontario, Canada
| | - Sarah Walker
- Department of Food Science, University of Guelph, Guelph, Ontario, Canada
| | - Alessia Roma
- Department of Food Science, University of Guelph, Guelph, Ontario, Canada
| | - Mark D Minden
- Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - Paul A Spagnuolo
- Department of Food Science, University of Guelph, Guelph, Ontario, Canada
| |
Collapse
|
25
|
Larrea D, Tamucci KA, Kabra K, Velasco KR, Yun TD, Pera M, Montesinos J, Agrawal RR, Paradas C, Smerdon JW, Lowry ER, Stepanova A, Yoval-Sanchez B, Galkin A, Wichterle H, Area-Gomez E. Altered mitochondria-associated ER membrane (MAM) function shifts mitochondrial metabolism in amyotrophic lateral sclerosis (ALS). Nat Commun 2025; 16:379. [PMID: 39753538 PMCID: PMC11699139 DOI: 10.1038/s41467-024-51578-1] [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: 10/27/2022] [Accepted: 08/12/2024] [Indexed: 01/06/2025] Open
Abstract
Mitochondrial function is modulated by its interaction with the endoplasmic reticulum (ER). Recent research indicates that these contacts are disrupted in familial models of amyotrophic lateral sclerosis (ALS). We report here that this impairment in the crosstalk between mitochondria and the ER impedes the use of glucose-derived pyruvate as mitochondrial fuel, causing a shift to fatty acids to sustain energy production. Over time, this deficiency alters mitochondrial electron flow and the active/dormant status of complex I in spinal cord tissues, but not in the brain. These findings suggest mitochondria-associated ER membranes (MAM domains) play a crucial role in regulating cellular glucose metabolism and that MAM dysfunction may underlie the bioenergetic deficits observed in ALS.
Collapse
Affiliation(s)
- Delfina Larrea
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA.
| | - Kirstin A Tamucci
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
- Institute of Human Nutrition, Columbia University Irving Medical Center, New York, NY, USA
| | - Khushbu Kabra
- Institute of Human Nutrition, Columbia University Irving Medical Center, New York, NY, USA
| | - Kevin R Velasco
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
| | - Taekyung D Yun
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
| | - Marta Pera
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
| | - Jorge Montesinos
- Department of Biomedicine, Centro de Investigaciones Biológicas Margarita Salas (CSIC), Madrid, Spain
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Rishi R Agrawal
- Institute of Human Nutrition, Columbia University Irving Medical Center, New York, NY, USA
| | - Carmen Paradas
- Department of Neurology, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío, Seville, Spain
| | - John W Smerdon
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Emily R Lowry
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Anna Stepanova
- Brain and Mind Research Institute, Weill Cornell Medical College, New York, NY, USA
| | - Belem Yoval-Sanchez
- Brain and Mind Research Institute, Weill Cornell Medical College, New York, NY, USA
| | - Alexander Galkin
- Brain and Mind Research Institute, Weill Cornell Medical College, New York, NY, USA
| | - Hynek Wichterle
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Estela Area-Gomez
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA.
- Department of Biomedicine, Centro de Investigaciones Biológicas Margarita Salas (CSIC), Madrid, Spain.
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.
| |
Collapse
|
26
|
Schleh MW, Ryan BJ, Ahn C, Ludzki AC, Van Pelt DW, Pitchford LM, Chugh OK, Luker AT, Luker KE, Samovski D, Abumrad NA, Burant CF, Horowitz JF. Impaired suppression of fatty acid release by insulin is a strong predictor of reduced whole-body insulin-mediated glucose uptake and skeletal muscle insulin receptor activation. Acta Physiol (Oxf) 2025; 241:e14249. [PMID: 39487600 DOI: 10.1111/apha.14249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 09/06/2024] [Accepted: 09/24/2024] [Indexed: 11/04/2024]
Abstract
AIM To examine factors underlying why most, but not all, adults with obesity exhibit impaired insulin-mediated glucose uptake, we compared: (1) adipose tissue fatty acid (FA) release, (2) skeletal muscle lipid droplet (LD) characteristics, and (3) insulin signalling events, in skeletal muscle of adults with obesity with relatively high versus low insulin-mediated glucose uptake. METHODS Seventeen adults with obesity (BMI: 36 ± 3 kg/m2) completed a 2 h hyperinsulinemic-euglycemic clamp with stable isotope tracer infusions to measure glucose rate of disappearance (glucose Rd) and FA rate of appearance (FA Ra). Skeletal muscle biopsies were collected at baseline and 30 min into the insulin infusion. Participants were stratified into HIGH (n = 7) and LOW (n = 10) insulin sensitivity cohorts by their glucose Rd during the hyperinsulinemic clamp (LOW< 400; HIGH >550 nmol/kgFFM/min/[μU/mL]). RESULTS Insulin-mediated suppression of FA Ra was lower in LOW compared with HIGH (p < 0.01). In skeletal muscle, total intramyocellular lipid content did not differ between cohorts. However, the size of LDs in the subsarcolemmal region (SS) of type II muscle fibres was larger in LOW compared with HIGH (p = 0.01). Additionally, insulin receptor-β (IRβ) interactions with regulatory proteins CD36 and Fyn were lower in LOW versus HIGH (p < 0.01), which aligned with attenuated insulin-mediated Tyr phosphorylation of IRβ and downstream insulin-signalling proteins in LOW. CONCLUSION Collectively, reduced ability for insulin to suppress FA mobilization, with accompanying modifications in intramyocellular LD size and distribution, and diminished IRβ interaction with key regulatory proteins may be key contributors to impaired insulin-mediated glucose uptake commonly found in adults with obesity.
Collapse
Affiliation(s)
- Michael W Schleh
- Substrate Metabolism Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Benjamin J Ryan
- Substrate Metabolism Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Cheehoon Ahn
- Substrate Metabolism Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Alison C Ludzki
- Substrate Metabolism Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Douglas W Van Pelt
- Substrate Metabolism Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Lisa M Pitchford
- Substrate Metabolism Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Olivia K Chugh
- Substrate Metabolism Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Austin T Luker
- Substrate Metabolism Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Kathryn E Luker
- Center for Molecular Imaging, Department of Radiology, University of Michigan, Ann Arbor, Michigan, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, Michigan, USA
| | - Dmitri Samovski
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Nada A Abumrad
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Charles F Burant
- Division of Metabolism, Endocrinology, and Diabetes, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Jeffrey F Horowitz
- Substrate Metabolism Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
27
|
Sadri H, Ghaffari MH, Sauerwein H, Schuchardt S, Martín-Tereso J, Doelman J, Daniel JB. Longitudinal characterization of the muscle metabolome in dairy cows during the transition from lactation cessation to lactation resumption. J Dairy Sci 2025; 108:1062-1077. [PMID: 39343201 DOI: 10.3168/jds.2024-25324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 09/05/2024] [Indexed: 10/01/2024]
Abstract
Skeletal muscle is vital in maintaining metabolic homeostasis and adapting to the physiological needs of pregnancy and lactation. Despite advancements in understanding metabolic changes in dairy cows around calving and early lactation, there are still gaps in our knowledge, especially concerning muscle metabolism and the changes associated with drying off. This study aimed to characterize the skeletal muscle metabolome in the context of the dietary and metabolic changes occurring during the transition from the cessation of lactation to the resumption of lactation in dairy cows. Twelve Holstein dairy cows housed in tiestalls were dried off 6 wk before the expected calving date. Cows were individually fed ad libitum TMR composed of grass silage, corn silage, and concentrate during lactation and of corn silage, barley straw, and concentrate during the dry period. The metabolome was characterized in skeletal muscle samples (M. longissimus dorsi) collected on wk -7 (9 d before dry-off), -5 (6 d after dry-off), and wk -1, and wk 1 relative to calving. The targeted metabolomics approach was conducted using the MxP Quant 500 kit (Biocrates Life Sciences AG) with liquid chromatography, flow injection, and electrospray ionization triple quadrupole mass spectrometry. Statistical analysis on the muscle metabolite data was performed using MetaboAnalyst 5.0, which allowed us to conduct various multivariate analyses such as principal component analysis (PCA), partial least squares discriminant analysis (PLS-DA), informative heat map generation, and hierarchical clustering. The statistical analysis revealed a clear separation between pregnancy (wk -7, -5, and -1) and postcalving (wk 1). Starting 5 wk before calving and continuing through the first week thereafter, the concentration of 3-methylhistidine (3-MH) in the muscle increased. This coincided with an increase in the concentrations of 11 AA (Phe, His, Tyr, Trp, Arg, Asn, Leu, Ile, Gly, Ser, and Thr) in the first week after calving, whereas Gln decreased. l-Arginine pathway metabolites (homoarginine, ornithine, citrulline, and asymmetric dimethylarginine), betaine, and sarcosine followed a similar pattern, increasing from wk -7 to -5, but decreasing from wk -1 to 1. The transition from pregnancy to lactation was associated with an increase in concentrations of the long-chain acylcarnitine species C16, C16:1, C18, and C18:1 in the muscle, whereas the concentrations of phosphatidylcholine and sphingomyelin in the muscle remained stable. The significant changes observed in the metabolome mainly concerned the AA and AA-related metabolites, indicating muscle protein breakdown in the first week after calving. The metabolites produced by the l-Arg pathway might contribute to regulating skeletal muscle mass and function in periparturient dairy cows. The elevated concentrations of long-chain acylcarnitine species in the muscle in the first week after calving suggest incomplete fatty acid oxidation, likely due to insufficient metabolic adaptation in response to the fatty acid load around the time of calving.
Collapse
Affiliation(s)
- H Sadri
- Department of Clinical Science, Faculty of Veterinary Medicine, University of Tabriz, 5166616471 Tabriz, Iran; Institute of Animal Science, Physiology Unit, University of Bonn, 53115 Bonn, Germany.
| | - M H Ghaffari
- Institute of Animal Science, Physiology Unit, University of Bonn, 53115 Bonn, Germany
| | - H Sauerwein
- Institute of Animal Science, Physiology Unit, University of Bonn, 53115 Bonn, Germany
| | - S Schuchardt
- Fraunhofer Institute for Toxicology and Experimental Medicine, 30625 Hannover, Germany
| | | | - John Doelman
- Trouw Nutrition R&D, 3800 AG, Amersfoort, the Netherlands
| | - J B Daniel
- Trouw Nutrition R&D, 3800 AG, Amersfoort, the Netherlands.
| |
Collapse
|
28
|
Wu Y, Zhang X, Sun L, Wu Q, Liu X, Deng Y, Lu Z, Li Z, Deng C, He R, Zhang L, Zeng R, Zhang X, Chen L, Lin X. Two-dimensional Health State Map to define metabolic health using separated static and dynamic homeostasis features: a proof-of-concept study. Natl Sci Rev 2025; 12:nwae425. [PMID: 39816947 PMCID: PMC11734281 DOI: 10.1093/nsr/nwae425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Revised: 10/25/2024] [Accepted: 11/05/2024] [Indexed: 01/18/2025] Open
Abstract
Defining metabolic health is critical for the earlier reversing of metabolic dysfunction and disease, and fasting-based diagnosis may not adequately assess an individual's metabolic adaptivity under stress. We constructed a novel Health State Map (HSM) comprising a Health Phenotype Score (HPS) with fasting features alone and a Homeostatic Resilience Score (HRS) with five time-point features only (t = 30, 60, 90, 180, 240 min) following a standardized mixed macronutrient tolerance test (MMTT). Among 111 Chinese adults, when the same set of fasting and post-MMTT data as for the HSM was used, the mixed-score was highly correlated with the HPS. The HRS was significantly associated with metabolic syndrome prevalence, independently of the HPS (OR [95% CI]: 0.41 [0.18, 0.92]) and the mixed-score (0.34 [0.15, 0.69]). Moreover, the HRS could discriminate metabolic characteristics unseparated by the HPS and the mixed-score. Participants with higher HRSs had better metabolic traits than those with lower HRSs. Large interpersonal variations were also evidenced by evaluating postprandial homeostatic resiliencies for glucose, lipids and amino acids when participants had similar overall HRSs. Additionally, the HRS was positively associated with physical activity level and specific gut microbiome structure. Collectively, our HSM model might offer a novel approach to precisely define an individual's metabolic health and nutritional capacity.
Collapse
Affiliation(s)
- Yanpu Wu
- Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai 200031, China
- BYHEALTH Institute of Nutrition & Health, Guangzhou 510799, China
| | - Xinyan Zhang
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
- Key Laboratory of Systems Biology, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Liang Sun
- Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai 200031, China
- Department of Nutrition and Food Hygiene, School of Public Health, Institute of Nutrition, Fudan University, Shanghai 200032, China
| | - Qingqing Wu
- Key Laboratory of Systems Biology, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Xiaoping Liu
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
| | - Yueyi Deng
- Department of Nephrology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200030, China
| | - Zhenzhen Lu
- Department of Nephrology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200030, China
| | - Zhongxia Li
- BYHEALTH Institute of Nutrition & Health, Guangzhou 510799, China
| | - Chaoming Deng
- BYHEALTH Institute of Nutrition & Health, Guangzhou 510799, China
| | - Ruikun He
- BYHEALTH Institute of Nutrition & Health, Guangzhou 510799, China
| | - Luyun Zhang
- Department of Nephrology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200030, China
| | - Rong Zeng
- Key Laboratory of Systems Biology, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Xuguang Zhang
- Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai 200031, China
- BYHEALTH Institute of Nutrition & Health, Guangzhou 510799, China
| | - Luonan Chen
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
- Key Laboratory of Systems Biology, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai 200031, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Xu Lin
- Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai 200031, China
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
| |
Collapse
|
29
|
Imierska M, Zabielski P, Roszczyc-Owsiejczuk K, Pogodzińska K, Błachnio-Zabielska A. Impact of reduced hepatic ceramide levels in high-fat diet mice on glucose metabolism. J Nutr Biochem 2025; 135:109785. [PMID: 39427846 DOI: 10.1016/j.jnutbio.2024.109785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 09/27/2024] [Accepted: 10/15/2024] [Indexed: 10/22/2024]
Abstract
Dysregulation of insulin action in hepatocytes, common in obesity, significantly contributes to insulin resistance, type 2 diabetes, and metabolic syndrome. Previous research highlights ceramides' role in these conditions. This study explores the impact of ceramides by silencing the serine palmitoyltransferase (Sptlc2) gene, crucial for the initial ceramide biosynthesis, using hydrodynamic gene delivery. Male C57BL/6 mice were randomly divided into three groups: one on a low-fat diet (LFD) receiving scrambled shRNA plasmids, another on a high-fat diet (HFD) with scrambled shRNA plasmids, and a third on HFD with a plasmid targeting Sptlc2. Analyses included RT-PCR for gene expression, western blot for protein levels, and UHPLC/MS/MS for lipid profiling. Glucose metabolism was evaluated via oral glucose tolerance tests, homeostatic model assessment of insulin resistance, and glucose-6-phosphate analysis. Results showed that HFD induces insulin resistance by inhibiting insulin signaling and increasing active lipid levels in hepatocytes. Sptlc2 silencing reduced ceramide accumulation, improving insulin signaling and glucose metabolism. Notably, ceramide synthesis inhibition did not significantly affect other lipid levels, highlighting ceramide's critical role in hepatic insulin resistance.
Collapse
Affiliation(s)
- Monika Imierska
- Department of Hygiene, Epidemiology and Metabolic Disorders, Medical University of Bialystok, Bialystok, Poland
| | - Piotr Zabielski
- Department of Medical Biology, Medical University of Bialystok, Bialystok, Poland
| | - Kamila Roszczyc-Owsiejczuk
- Department of Hygiene, Epidemiology and Metabolic Disorders, Medical University of Bialystok, Bialystok, Poland
| | - Karolina Pogodzińska
- Department of Hygiene, Epidemiology and Metabolic Disorders, Medical University of Bialystok, Bialystok, Poland
| | - Agnieszka Błachnio-Zabielska
- Department of Hygiene, Epidemiology and Metabolic Disorders, Medical University of Bialystok, Bialystok, Poland.
| |
Collapse
|
30
|
Dajnowicz-Brzezik P, Żebrowska E, Maciejczyk M, Zalewska A, Chabowski A. α -lipoic acid supplementation reduces oxidative stress and inflammation in red skeletal muscle of insulin-resistant rats. Biochem Biophys Res Commun 2025; 742:151107. [PMID: 39667068 DOI: 10.1016/j.bbrc.2024.151107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Revised: 11/16/2024] [Accepted: 11/30/2024] [Indexed: 12/14/2024]
Abstract
α -lipoic acid (ALA) is an eight-carbon saturated fatty acid with strong antioxidant activity. Despite previous reports of ALA's protective properties in treating cardiovascular and metabolic diseases (including insulin resistance and diabetes), little is known about the compound's effects on skeletal muscle metabolism. In particular, the effect of ALA on glycooxidative and nitrosative damage in red muscles during insulin resistance is unknown. This study investigated the therapeutic potential of ALA on the antioxidant barrier as well as oxidative, nitrosative and carbonyl stress in the red skeletal muscle of rats with high-fat diet-induced insulin resistance. Male Wistar cmdb/outbred rats were divided into four equal groups: control diet (CTRL), high fat diet (HFD), CTRL + ALA (30 mg/kg body weight for 4 weeks; intragastrically) and HFD + ALA. Enzymatic and nonenzymatic antioxidant systems, protein and lipid glycoxidation, nitrosative stress, and selected inflammatory/apoptosis parameters were assessed using colorimetric, fluorimetric, and immune-enzymatic methods. ALA lowered body weight and glucose metabolism parameters in insulin-resistant rats. ALA not only strengthened enzymatic antioxidant defense (by increasing superoxide dismutase, catalase and glutathione peroxidase activity) but also stimulated the synthesis of non-enzymatic GSH. ALA supplementation inhibited lipid peroxidation (decreased lipid hydroperoxides and malondialdehyde content) and prevented protein oxidation (by lowering advanced oxidation protein products concentration) in red muscle. ALA's multifactorial actions on muscle tissue also included inhibition of inflammation and apoptosis, requiring further research to elucidate its effects in metabolic diseases.
Collapse
Affiliation(s)
- Patrycja Dajnowicz-Brzezik
- Department of Physiology, Medical University of Bialystok, Mickiewicza 2C st., 15-222, Bialystok, Poland.
| | - Ewa Żebrowska
- Department of Physiology, Medical University of Bialystok, Mickiewicza 2C st., 15-222, Bialystok, Poland
| | - Mateusz Maciejczyk
- Department of Hygiene, Epidemiology and Ergonomics, Medical University of Bialystok, Mickiewicza 2C st., 15-222, Bialystok, Poland
| | - Anna Zalewska
- Independent Laboratory of Experimental Dentistry, Medical University of Bialystok, M. Skłodowskiej-Curie 24A st., 15-276, Bialystok, Poland
| | - Adrian Chabowski
- Department of Physiology, Medical University of Bialystok, Mickiewicza 2C st., 15-222, Bialystok, Poland
| |
Collapse
|
31
|
Ostrom EL, Stuppard R, Mattson-Hughes A, Marcinek DJ. Inducible and reversible SOD2 knockdown in mouse skeletal muscle drives impaired pyruvate oxidation and reduced metabolic flexibility. Free Radic Biol Med 2025; 226:237-250. [PMID: 39551449 PMCID: PMC11757001 DOI: 10.1016/j.freeradbiomed.2024.10.310] [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: 09/23/2024] [Revised: 10/27/2024] [Accepted: 10/29/2024] [Indexed: 11/19/2024]
Abstract
INTRODUCTION Skeletal muscle mitochondrial dysfunction is a key characteristic of aging muscle and contributes to age related diseases such as sarcopenia, frailty, and type 2 diabetes. Mitochondrial oxidative stress has been implicated as a driving factor in these age-related diseases, however whether it is a cause, or a consequence of mitochondrial dysfunction remains to be determined. The development of flexible genetic models is an important tool to test the mechanistic role of mitochondrial oxidative stress on skeletal muscle metabolic dysfunction. We characterize a new model of inducible and reversible mitochondrial redox stress using a tetracycline controlled skeletal muscle specific short hairpin RNA targeted to superoxide dismutase 2 (iSOD2). METHODS iSOD2 KD and control (CON) animals were administered doxycycline for 3- or 12- weeks and followed for up to 24 weeks and mitochondrial respiration and muscle contraction were measured to define the time course of SOD2 KD and muscle functional changes and recovery. RESULTS Maximum knockdown of SOD2 protein occurred by 6 weeks and recovered by 24 weeks after DOX treatment. Mitochondrial aconitase activity and maximum mitochondrial respiration declined in KD muscle by 12 weeks and recovered by 24 weeks. There were no significant differences in antioxidant or mitochondrial biogenesis genes between groups. Twelve-week KD showed a small, but significant decrease in muscle fatigue resistance. The primary phenotype was reduced metabolic flexibility characterized by impaired pyruvate driven respiration when other substrates are present. The pyruvate dehydrogenase kinase inhibitor dichloroacetate partially restored pyruvate driven respiration, while the thiol reductant DTT did not. CONCLUSION We use a model of inducible and reversible skeletal muscle SOD2 knockdown to demonstrate that elevated matrix superoxide reversibly impairs mitochondrial substrate flexibility characterized by impaired pyruvate oxidation. Despite the bioenergetic effect, the limited change in gene expression suggests that the elevated redox stress in this model is confined to the mitochondrial matrix.
Collapse
Affiliation(s)
- Ethan L Ostrom
- Department of Radiology, University of Washington School of Medicine, Seattle, WA, USA.
| | - Rudy Stuppard
- Department of Radiology, University of Washington School of Medicine, Seattle, WA, USA
| | - Aurora Mattson-Hughes
- Department of Radiology, University of Washington School of Medicine, Seattle, WA, USA
| | - David J Marcinek
- Department of Radiology, University of Washington School of Medicine, Seattle, WA, USA; Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, WA, USA
| |
Collapse
|
32
|
Shmet M, Amasha M, Khattib A, Schweitzer R, Khatib S, Hamudi J, Halabi M, Khatib S. Untargeted metabolomics reveals biomarkers for the diagnosis of coronary artery plaques as observed by coronary cardiac computed tomography. Biofactors 2025; 51:e2156. [PMID: 39878362 DOI: 10.1002/biof.2156] [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: 06/24/2024] [Accepted: 12/18/2024] [Indexed: 01/31/2025]
Abstract
Atherosclerosis is a major cause of morbidity and mortality worldwide; in Israel, ischemic heart disease is the second leading cause of death for both genders aged 45 and above. Atherosclerosis involves stiffening of the arteries due to the accumulation of lipids and oxidized lipids on the blood vessel walls, triggering the development of artery plaque. Coronary artery disease (CAD) is the most common manifestation of atherosclerosis. The prevalence of CAD in the general population remains high, despite efforts to improve the identification of risk factors and preventive treatments. The discovery of new biomarkers is vital to improving the diagnosis of CAD and its risk factors. We aimed to identify novel biomarkers that could provide an early diagnosis of coronary artery atherosclerotic plaques, their type, and the percentage of stenosis. We used an untargeted metabolomics approach to identify potential biomarkers that could enable highly sensitive and specific CAD detection. The study consisted of 109 patients who underwent cardiac computed tomography angiography at the Cardiology Department of Ziv Medical Center. Fifty-four patients were diagnosed with coronary atherosclerotic plaques (CAD group), and 55 without plaques used control. Untargeted metabolomics using LC-MS/MS revealed 2560 metabolites in the patients' serum: 106 showed statistically significant upregulation in the serum of the CAD group compared with the healthy control group (p < 0.05). These metabolites belonged to the following chemical families: acyl-carnitines, cyclodipeptides, lysophosphatidylcholine, and primary bile acids. In contrast, 98 metabolites displayed statistically significant downregulation in the serum of the CAD group compared with the control group, belonging to the following chemical families: GABA amino acids and derivatives (inhibitory neurotransmitters), lipids, and secondary bile acids. Our comprehensive untargeted serum metabolomic analysis revealed biomarkers that can be used for the diagnosis of patients with CAD. Further cohort studies with a larger number of participants are needed to validate the detected biomarkers.
Collapse
Affiliation(s)
- Manar Shmet
- Natural Products and Analytical Chemistry Laboratory, MIGAL - Galilee Research Institute, Kiryat Shemona, Israel
- Department of Biotechnology, Tel-Hai College, Qiryat Shemona, Israel
| | | | - Ali Khattib
- Natural Products and Analytical Chemistry Laboratory, MIGAL - Galilee Research Institute, Kiryat Shemona, Israel
- The Rappaport Family Institute for Research in the Medical Science and Rambam Medical Center, Haifa, Israel
| | - Ron Schweitzer
- Natural Products and Analytical Chemistry Laboratory, MIGAL - Galilee Research Institute, Kiryat Shemona, Israel
| | | | | | | | - Soliman Khatib
- Natural Products and Analytical Chemistry Laboratory, MIGAL - Galilee Research Institute, Kiryat Shemona, Israel
- Department of Biotechnology, Tel-Hai College, Qiryat Shemona, Israel
| |
Collapse
|
33
|
Brun A, Denis P, Rambeau M, Rigaudière JP, Jouve C, Mazurak V, Capel F. Polyunsaturated fatty acids prevent myosteatosis and lipotoxicity. J Nutr Biochem 2024; 134:109722. [PMID: 39142445 DOI: 10.1016/j.jnutbio.2024.109722] [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: 12/04/2023] [Revised: 07/30/2024] [Accepted: 08/01/2024] [Indexed: 08/16/2024]
Abstract
Myosteatosis occurs in response to excess circulating fatty acids and is associated with muscle dysfunction. This study aimed to characterize the sequence of events of lipid-induced toxicity within muscle cells and the role of polyunsaturated fatty acids (PUFA) as potential preventive factors. Myosteatosis was induced in C2C12 myotubes exposed to palmitic acid (PAL 500µM). Furthermore, cells were co-incubated with PUFA (α-linolenic acid = ALA, Eicosapentaenoic acid = EPA, Docosahexaenoic acid = DHA; Arachidonic acid = ARA) over a period of 48 h. Cell viability, morphology, and measures of lipid and protein metabolism were assessed at 6, 12, 24, and 48 h. We observed that myotube integrity was rapidly and progressively disrupted by PAL treatment after 12 h, ultimately leading to cell death (41.7% cell survival at 48 h, p < .05). Cell death did not occur in cells exposed to PAL+ARA and PAL+DHA. After 6 h of PAL treatment, an accumulation of large lipid droplets was observed within the cell (6 folds, p < .05). This was associated with an increase in ceramides (CER x3 fold change) and diacylglycerol (DAG x150 fold change) contents (p < .05). At the same time, insulin was no longer able to stimulate protein synthesis (p < .05) nor leverage autophagic flux (p < .05). DHA and ARA were able to completely reverse the defect in protein synthesis and partially modulate the accumulation of CER and DAG. These findings present new and intriguing research avenues in the field of muscle metabolism and nutrition, particularly in the context of aging, chronic muscle disorders, and insulin resistance.
Collapse
Affiliation(s)
- Aurélien Brun
- UMR1019 Unité de Nutrition Humaine, Université Clermont Auvergne, INRAE, CRNH Auvergne, Clermont-Ferrand, France
| | - Philippe Denis
- UMR1019 Unité de Nutrition Humaine, Université Clermont Auvergne, INRAE, CRNH Auvergne, Clermont-Ferrand, France
| | - Mathieu Rambeau
- UMR1019 Unité de Nutrition Humaine, Université Clermont Auvergne, INRAE, CRNH Auvergne, Clermont-Ferrand, France
| | - Jean-Paul Rigaudière
- UMR1019 Unité de Nutrition Humaine, Université Clermont Auvergne, INRAE, CRNH Auvergne, Clermont-Ferrand, France
| | - Chrystèle Jouve
- UMR1019 Unité de Nutrition Humaine, Université Clermont Auvergne, INRAE, CRNH Auvergne, Clermont-Ferrand, France
| | - Vera Mazurak
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada
| | - Frédéric Capel
- UMR1019 Unité de Nutrition Humaine, Université Clermont Auvergne, INRAE, CRNH Auvergne, Clermont-Ferrand, France.
| |
Collapse
|
34
|
Quiriarte H, Noland RC, Stampley JE, Davis G, Li Z, Cho E, Kim Y, Doiron J, Spielmann G, Ghosh S, Shah SJ, Irving BA, Lefer DJ, Allerton TD. Exercise Therapy Rescues Skeletal Muscle Dysfunction and Exercise Intolerance in Cardiometabolic HFpEF. JACC Basic Transl Sci 2024; 9:1409-1425. [PMID: 39822600 PMCID: PMC11733766 DOI: 10.1016/j.jacbts.2024.07.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 07/23/2024] [Accepted: 07/23/2024] [Indexed: 01/19/2025]
Abstract
Exercise intolerance, a hallmark of heart failure with preserved ejection fraction (HFpEF) exacerbated by obesity, involves unclear mechanisms related to skeletal muscle metabolism. In a "2-hit" model of HFpEF, we investigated the ability of exercise therapy (voluntary wheel running) to reverse skeletal muscle dysfunction and exercise intolerance. Using state-of-the-art metabolic cages and a multiomic approach, we demonstrate exercise can rescue dysfunctional skeletal muscle lipid and branched-chain amino acid oxidation and restore exercise capacity in mice with cardiometabolic HFpEF. These results underscore the importance of skeletal muscle metabolism to improve exercise intolerance in HFpEF.
Collapse
Affiliation(s)
- Heather Quiriarte
- Vascular Metabolism Laboratory, Pennington Biomedical Research Center, Baton Rouge, Louisiana, USA
- Department of Kinesiology, Louisiana State University, Baton Rouge, Louisiana, USA
| | - Robert C. Noland
- Skeletal Muscle Metabolism Laboratory, Pennington Biomedical Research Center, Baton Rouge, Louisiana, USA
| | - James E. Stampley
- Department of Kinesiology, Louisiana State University, Baton Rouge, Louisiana, USA
| | - Gregory Davis
- Department of Kinesiology, Louisiana State University, Baton Rouge, Louisiana, USA
| | - Zhen Li
- Department of Cardiac Surgery, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Eunhan Cho
- Department of Kinesiology, Louisiana State University, Baton Rouge, Louisiana, USA
| | - Youyoung Kim
- Department of Kinesiology, Louisiana State University, Baton Rouge, Louisiana, USA
| | - Jake Doiron
- Vascular Metabolism Laboratory, Pennington Biomedical Research Center, Baton Rouge, Louisiana, USA
- Department of Pharmacology and Experimental Therapeutics, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA
| | - Guillaume Spielmann
- Department of Kinesiology, Louisiana State University, Baton Rouge, Louisiana, USA
| | - Sujoy Ghosh
- Bioinformatics and Computational Biology Core, Pennington Biomedical Research Center, Baton Rouge, Louisiana, USA
| | - Sanjiv J. Shah
- Division of Cardiology, Department of Medicine, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
| | - Brian A. Irving
- Department of Kinesiology, Louisiana State University, Baton Rouge, Louisiana, USA
| | - David J. Lefer
- Department of Cardiac Surgery, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Timothy D. Allerton
- Vascular Metabolism Laboratory, Pennington Biomedical Research Center, Baton Rouge, Louisiana, USA
- Louisiana State University Cardiovascular Center of Excellence, New Orleans, Louisiana, USA
| |
Collapse
|
35
|
Sammut MJ, Dotzert MS, Melling CWJ. Mechanisms of insulin resistance in type 1 diabetes mellitus: A case of glucolipotoxicity in skeletal muscle. J Cell Physiol 2024; 239:e31419. [PMID: 39192756 PMCID: PMC11649966 DOI: 10.1002/jcp.31419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 07/16/2024] [Accepted: 08/09/2024] [Indexed: 08/29/2024]
Abstract
Insulin resistance (IR), a hallmark of type 2 diabetes mellitus, develops in a significant number of patients with type 1 diabetes mellitus (T1DM) despite the use of insulin therapy to control glycemia. However, little is currently understood regarding the underlying mechanisms of IR in T1DM, especially within the context of chronic insulin treatment. Recent evidence suggests an important influence of glucolipotoxicity in skeletal muscle on insulin sensitivity in T1DM. Thus, this review summarizes our current knowledge regarding impairments in skeletal muscle lipid, glucose, and oxidative metabolism in the development of IR in insulin-treated T1DM.
Collapse
Affiliation(s)
- Mitchell J. Sammut
- School of Kinesiology, Faculty of Health SciencesWestern UniversityLondonOntarioCanada
| | - Michelle S. Dotzert
- School of Kinesiology, Faculty of Health SciencesWestern UniversityLondonOntarioCanada
| | - C. W. James Melling
- School of Kinesiology, Faculty of Health SciencesWestern UniversityLondonOntarioCanada
- Department of Physiology & Pharmacology, Schulich School of Medicine & DentistryWestern UniversityLondonOntarioCanada
| |
Collapse
|
36
|
Perry AS, Piaggi P, Huang S, Nayor M, Freedman J, North KE, Below JE, Clish CB, Murthy VL, Krakoff J, Shah RV. Human metabolic chambers reveal a coordinated metabolic-physiologic response to nutrition. JCI Insight 2024; 9:e184279. [PMID: 39576013 PMCID: PMC11601946 DOI: 10.1172/jci.insight.184279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 09/25/2024] [Indexed: 11/27/2024] Open
Abstract
Human studies linking metabolism with organism-wide physiologic function have been challenged by confounding, adherence, and precisionHere, we united physiologic and molecular phenotypes of metabolism during controlled dietary intervention to understand integrated metabolic-physiologic responses to nutrition. In an inpatient study of individuals who underwent serial 24-hour metabolic chamber experiments (indirect calorimetry) and metabolite profiling, we mapped a human metabolome onto substrate oxidation rates and energy expenditure across up to 7 dietary conditions (energy balance, fasting, multiple 200% caloric excess overfeeding of varying fat, protein, and carbohydrate composition). Diets exhibiting greater fat oxidation (e.g., fasting, high-fat) were associated with changes in metabolites within pathways of mitochondrial β-oxidation, ketogenesis, adipose tissue fatty acid liberation, and/or multiple anapleurotic substrates for tricarboxylic acid cycle flux, with inverse associations for diets with greater carbohydrate availability. Changes in each of these metabolite classes were strongly related to 24-hour respiratory quotient (RQ) and substrate oxidation rates (e.g., acylcarnitines related to lower 24-hour RQ and higher 24-hour lipid oxidation), underscoring links between substrate availability, physiology, and metabolism in humans. Physiologic responses to diet determined by gold-standard human metabolic chambers are strongly coordinated with biologically consistent, interconnected metabolic pathways encoded in the metabolome.
Collapse
Affiliation(s)
- Andrew S. Perry
- Vanderbilt Translational and Clinical Cardiovascular Research Center, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Paolo Piaggi
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Phoenix, Arizona, USA
| | - Shi Huang
- Department of Biostatistics, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Matthew Nayor
- Sections of Cardiovascular Medicine and Preventive Medicine and Epidemiology, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Jane Freedman
- Vanderbilt Translational and Clinical Cardiovascular Research Center, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Kari E. North
- CVD Genetic Epidemiology Computational Laboratory, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Jennifer E. Below
- Vanderbilt Genetics Institute, Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Clary B. Clish
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts USA
| | | | - Jonathan Krakoff
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Phoenix, Arizona, USA
| | - Ravi V. Shah
- Vanderbilt Translational and Clinical Cardiovascular Research Center, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| |
Collapse
|
37
|
Ortlund E, Hou Z, Chen CY, Gaul D, Zhang T, Moore S, Liu X, Ivanova A, Maner-Smith K, Newgard C, Bodine S, Savage E, Bennett A, Fernandez F. Endurance Exercise Training Alters Lipidomic Profiles of Plasma and Eight Tissues in Rats: a MoTrPAC study. RESEARCH SQUARE 2024:rs.3.rs-5263273. [PMID: 39606465 PMCID: PMC11601870 DOI: 10.21203/rs.3.rs-5263273/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Endurance exercise training (ExT) induces metabolic, structural, and functional adaptations via lipidomic modifications, yet the systematic elucidation of lipidome alterations in response to ExT remains incomplete. As a part of the Molecular Transducers of Physical Activity Consortium (MoTrPAC), we leveraged non-targeted and targeted lipidomics for the systematic discovery of lipid alterations in the brown adipose tissue, heart, hippocampus, kidney, liver, lung, skeletal muscle gastrocnemius, subcutaneous white adipose tissue, and plasma in response to 1, 2, 4 or 8 weeks of ExT in 6-month-old male and female Fischer-344 rats. This study demonstrates that these tissues, each with distinct lipidomic features, underwent dynamic, sexually dimorphic lipid remodeling. Exercise trained animals showed reduced whole-body adiposity and improved cardiorespiratory fitness, along with enhanced utilization of lipid stores and dynamic triacylglycerol remodeling compared to sedentary controls in all tissues except hippocampus. They also showed modifications in phospholipids, lysophospholipids, oxylipins, and ceramides in several tissues. Coordinated changes across tissues reflect systemic tissue communication, with liver-plasma-heart connection potentially playing a key role in systemic lipid metabolism during ExT. These data will improve our understanding of lipid-associated biological processes underlying the health-promoting benefits of ExT.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Anna Ivanova
- Centers for Disease Control and Prevention (CDC)
| | | | | | | | | | | | | |
Collapse
|
38
|
Tan Y, Liu M, Zhou X, Gao T, Fang J, Wang S, Chen S. Mapping the mitochondrial landscape in T2DM: key findings from 2003-2023. Front Endocrinol (Lausanne) 2024; 15:1474232. [PMID: 39634184 PMCID: PMC11614640 DOI: 10.3389/fendo.2024.1474232] [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: 08/01/2024] [Accepted: 11/01/2024] [Indexed: 12/07/2024] Open
Abstract
Backgound T2DM, a chronic metabolic disorder, poses a significant threat to global public health. Mitochondria play a crucial role in the pathogenesis of T2DM. This study intends to investigate the correlation between mitochondria and T2DM over the past two decades (2003-2023) through bibliometric analysis. Its objectives are to pinpoint trends, emphasize research priorities, and establish a foundation for future investigations. Methods A literature search was conducted using the SCI-E database. All recorded results were downloaded in plain text format for further analysis. The following terms were analyzed using Vosviewer 1.6.18, citespace 6.3r1, bibliometrix in RStudio (v.4.4.1), and Microsoft Excel 2021: country, institution, author, journal, references, and keywords. Results From January 1, 2003 to December 31, 2023, a total of 2,732 articles were retrieved. The United States, China, and Italy contributed most of the records. UNIVERSITY OF CALIFORNIA SYSTEM, INSTITUT NATIONAL DE LA SANTE ET DE LA RECHERCHE MEDICAL INSERM, and US DEPARTMENT OF VETERANS AFFAIRS were the top 3 most productive institutions. rocha milagros, victor victor m had the most publications, followed by roden michael, and petersen kf had the most citations together. DIABETES published the most articles on research on this topic, followed by AMERICAN JOURNAL OF PHYSIOLOGY-ENDOCRINOLOGY AND METABOLISM, DIABETOLOGIA. The key points of this topic are the relationship between mitochondria and T2DM, the skeletal muscle mitochondrial changes observed in T2DM, and the impact of mitochondrial dysfunction on T2DM. Over the past five years, particle dynamics, mitochondrial dysfunction, and mechanism research have emerged as significant focal points in this field. Conclude This paper successfully identified the key areas and emerging trends in the relationship between mitochondria and T2DM, thereby offering valuable insights for future research.
Collapse
Affiliation(s)
- Yi Tan
- Departments of Acupuncture and Massage, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Mingjun Liu
- Departments of Acupuncture and Massage, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Xinfeng Zhou
- Departments of Acupuncture and Massage, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Tianjiao Gao
- The Affliated Hospital of Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Jinxu Fang
- Departments of Acupuncture and Massage, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Sixian Wang
- Departments of Acupuncture and Massage, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Shaotao Chen
- Departments of Acupuncture and Massage, Changchun University of Chinese Medicine, Changchun, Jilin, China
| |
Collapse
|
39
|
Estevao IL, Kazman JB, Bramer LM, Nicora C, Ren MQ, Sambuughin N, Munoz N, Kim YM, Bloodsworth K, Richert M, Teeguarden J, Burnum-Johnson K, Deuster PA, Nakayasu ES, Many G. The human plasma lipidome response to exertional heat tolerance testing. Lipids Health Dis 2024; 23:380. [PMID: 39548465 PMCID: PMC11566608 DOI: 10.1186/s12944-024-02322-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 10/01/2024] [Indexed: 11/18/2024] Open
Abstract
BACKGROUND The year of 2023 displayed the highest average global temperatures since it has been recorded-the duration and severity of extreme heat are projected to increase. Rising global temperatures represent a major public health threat, especially to occupations exposed to hot environments, such as construction and agricultural workers, and first responders. Despite efforts of the scientific community, there is still a need to characterize the pathophysiological processes leading to heat related illness and develop biomarkers that can predict its onset. METHODS Liquid chromatography-tandem mass spectrometry (LC-MS/MS)-based lipidomics analysis was performed on plasma from male and female subjects who underwent exertional heat tolerance testing (HTT), consisting of a 2-h treadmill walk at 5 km/h with 2.0% incline at a controlled temperature of 40ºC. From HTT, heat tolerance was calculated using the physiological strain index (PSI). RESULTS Nearly half of all 995 detected lipids from 27 classes were responsive to HTT. Lipid classes related to substrate utilization were predominantly affected by HTT, with a downregulation of triacylglycerols and upregulation of free fatty acids and acyl-carnitines (CARs). Even chain CAR 4:0, 14:0 and 16:1, suggested by-products of incomplete beta oxidation, and diacylglycerols displayed the highest correlation to PSI. PSI did not correlate with plasma lactate levels, suggesting that correlations between even chain CARs and PSI are related to metabolic efficiency versus physical exertion. CONCLUSIONS Overall, HTT displays a strong impact on the human plasma lipidome and lipid metabolic inefficiencies may underlie reduced heat tolerance.
Collapse
Affiliation(s)
| | - Josh B Kazman
- Consortium for Health and Military Performance, Department of Military and Emergency Medicine, F. Edward Hébert School of Medicine, Uniformed Services University, Bethesda, MD, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | - Lisa M Bramer
- Biological Sciences Division, Richland, WA, 99352, USA
| | - Carrie Nicora
- Biological Sciences Division, Richland, WA, 99352, USA
| | - Ming Qiang Ren
- Consortium for Health and Military Performance, Department of Military and Emergency Medicine, F. Edward Hébert School of Medicine, Uniformed Services University, Bethesda, MD, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | - Nyamkhishig Sambuughin
- Consortium for Health and Military Performance, Department of Military and Emergency Medicine, F. Edward Hébert School of Medicine, Uniformed Services University, Bethesda, MD, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | - Nathalie Munoz
- Environmental and Molecular Sciences Division, Pacific Northwest National Laboratory, Richland, WA, 99352, USA
| | - Young-Mo Kim
- Biological Sciences Division, Richland, WA, 99352, USA
| | | | - Maile Richert
- Consortium for Health and Military Performance, Department of Military and Emergency Medicine, F. Edward Hébert School of Medicine, Uniformed Services University, Bethesda, MD, USA
| | - Justin Teeguarden
- Environmental and Molecular Sciences Division, Pacific Northwest National Laboratory, Richland, WA, 99352, USA
| | - Kristin Burnum-Johnson
- Environmental and Molecular Sciences Division, Pacific Northwest National Laboratory, Richland, WA, 99352, USA
| | - Patricia A Deuster
- Consortium for Health and Military Performance, Department of Military and Emergency Medicine, F. Edward Hébert School of Medicine, Uniformed Services University, Bethesda, MD, USA
| | | | - Gina Many
- Biological Sciences Division, Richland, WA, 99352, USA.
| |
Collapse
|
40
|
Fiorenza M, Onslev J, Henríquez-Olguín C, Persson KW, Hesselager SA, Jensen TE, Wojtaszewski JFP, Hostrup M, Bangsbo J. Reducing the mitochondrial oxidative burden alleviates lipid-induced muscle insulin resistance in humans. SCIENCE ADVANCES 2024; 10:eadq4461. [PMID: 39475607 PMCID: PMC11524190 DOI: 10.1126/sciadv.adq4461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 09/23/2024] [Indexed: 11/02/2024]
Abstract
Preclinical models suggest mitochondria-derived oxidative stress as an underlying cause of insulin resistance. However, it remains unknown whether this pathophysiological mechanism is conserved in humans. Here, we used an invasive in vivo mechanistic approach to interrogate muscle insulin action while selectively manipulating the mitochondrial redox state in humans. To this end, we conducted insulin clamp studies combining intravenous infusion of a lipid overload with intake of a mitochondria-targeted antioxidant (mitoquinone). Under lipid overload, selective modulation of mitochondrial redox state by mitoquinone enhanced insulin-stimulated glucose uptake in skeletal muscle. Mechanistically, mitoquinone did not affect canonical insulin signaling but augmented insulin-stimulated glucose transporter type 4 (GLUT4) translocation while reducing the mitochondrial oxidative burden under lipid oversupply. Complementary ex vivo studies in human muscle fibers exposed to high intracellular lipid levels revealed that mitoquinone improves features of mitochondrial bioenergetics, including diminished mitochondrial H2O2 emission. These findings provide translational and mechanistic evidence implicating mitochondrial oxidants in the development of lipid-induced muscle insulin resistance in humans.
Collapse
Affiliation(s)
- Matteo Fiorenza
- August Krogh Section for Human Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen 2100, Denmark
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
| | - Johan Onslev
- August Krogh Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen 2100, Denmark
| | - Carlos Henríquez-Olguín
- August Krogh Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen 2100, Denmark
- Exercise Science Laboratory, Faculty of Medicine, Universidad Finis Terrae, Santiago 1509, Chile
| | - Kaspar W. Persson
- August Krogh Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen 2100, Denmark
| | - Sofie A. Hesselager
- August Krogh Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen 2100, Denmark
| | - Thomas E. Jensen
- August Krogh Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen 2100, Denmark
| | - Jørgen F. P. Wojtaszewski
- August Krogh Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen 2100, Denmark
| | - Morten Hostrup
- August Krogh Section for Human Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen 2100, Denmark
| | - Jens Bangsbo
- August Krogh Section for Human Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen 2100, Denmark
| |
Collapse
|
41
|
Pereyra AS, Fernandez RF, Amorese A, Castro JN, Lin CT, Spangenburg EE, Ellis JM. Loss of mitochondria long-chain fatty acid oxidation impairs skeletal muscle contractility by disrupting myofibril structure and calcium homeostasis. Mol Metab 2024; 89:102015. [PMID: 39182841 PMCID: PMC11408158 DOI: 10.1016/j.molmet.2024.102015] [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: 06/06/2024] [Revised: 08/05/2024] [Accepted: 08/19/2024] [Indexed: 08/27/2024] Open
Abstract
OBJECTIVE Abnormal lipid metabolism in mammalian tissues can be highly deleterious, leading to organ failure. Carnitine Palmitoyltransferase 2 (CPT2) deficiency is an inherited metabolic disorder affecting the liver, heart, and skeletal muscle due to impaired mitochondrial oxidation of long-chain fatty acids (mLCFAO) for energy production. METHODS However, the basis of tissue damage in mLCFAO disorders is not fully understood. Mice lacking CPT2 in skeletal muscle (Cpt2Sk-/-) were generated to investigate the nexus between mFAO deficiency and myopathy. RESULTS Compared to controls, ex-vivo contractile force was reduced by 70% in Cpt2Sk-/- oxidative soleus muscle despite the preserved capacity to couple ATP synthesis to mitochondrial respiration on alternative substrates to long-chain fatty acids. Increased mitochondrial biogenesis, lipid accumulation, and the downregulation of 80% of dystrophin-related and contraction-related proteins severely compromised the structure and function of Cpt2Sk-/- soleus. CPT2 deficiency affected oxidative muscles more than glycolytic ones. Exposing isolated sarcoplasmic reticulum to long-chain acylcarnitines (LCACs) inhibited calcium uptake. In agreement, Cpt2Sk-/- soleus had decreased calcium uptake and significant accumulation of palmitoyl-carnitine, suggesting that LCACs and calcium dyshomeostasis are linked in skeletal muscle. CONCLUSIONS Our data demonstrate that loss of CPT2 and mLCFAO compromise muscle structure and function due to excessive mitochondrial biogenesis, downregulation of the contractile proteome, and disruption of calcium homeostasis.
Collapse
Affiliation(s)
- Andrea S Pereyra
- Brody School of Medicine at East Carolina University, Department of Physiology and East Carolina Diabetes and Obesity Institute, Greenville, NC, 27834, USA.
| | - Regina F Fernandez
- Brody School of Medicine at East Carolina University, Department of Physiology and East Carolina Diabetes and Obesity Institute, Greenville, NC, 27834, USA
| | - Adam Amorese
- Brody School of Medicine at East Carolina University, Department of Physiology and East Carolina Diabetes and Obesity Institute, Greenville, NC, 27834, USA
| | - Jasmine N Castro
- Brody School of Medicine at East Carolina University, Department of Physiology and East Carolina Diabetes and Obesity Institute, Greenville, NC, 27834, USA
| | - Chien-Te Lin
- Brody School of Medicine at East Carolina University, Department of Physiology and East Carolina Diabetes and Obesity Institute, Greenville, NC, 27834, USA
| | - Espen E Spangenburg
- Brody School of Medicine at East Carolina University, Department of Physiology and East Carolina Diabetes and Obesity Institute, Greenville, NC, 27834, USA
| | - Jessica M Ellis
- Brody School of Medicine at East Carolina University, Department of Physiology and East Carolina Diabetes and Obesity Institute, Greenville, NC, 27834, USA.
| |
Collapse
|
42
|
Abolfazli S, Butler AE, Kesharwani P, Sahebkar A. The beneficial impact of curcumin on cardiac lipotoxicity. J Pharm Pharmacol 2024; 76:1269-1283. [PMID: 39180454 DOI: 10.1093/jpp/rgae102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 07/02/2024] [Indexed: 08/26/2024]
Abstract
Lipotoxicity is defined as a prolonged metabolic imbalance of lipids that results in ectopic fat distribution in peripheral organs such as the liver, heart, and kidney. The harmful consequences of excessive lipid accumulation in cardiomyocytes cause cardiac lipotoxicity, which alters the structure and function of the heart. Obesity and diabetes are linked to lipotoxic cardiomyopathy. These anomalies might be caused by a harmful metabolic shift that accumulates toxic lipids and shifts glucose oxidation to less fatty acid oxidation. Research has linked fatty acids, fatty acyl coenzyme A, diacylglycerol, and ceramide to lipotoxic stress in cells. This stress can be brought on by apoptosis, impaired insulin signaling, endoplasmic reticulum stress, protein kinase C activation, p38 Ras-mitogen-activated protein kinase (MAPK) activation, or modification of peroxisome proliferator-activated receptors (PPARs) family members. Curcuma longa is used to extract curcumin, a hydrophobic polyphenol derivative with a variety of pharmacological characteristics. Throughout the years, curcumin has been utilized as an anti-inflammatory, antioxidant, anticancer, hepatoprotective, cardioprotective, anti-diabetic, and anti-obesity drug. Curcumin reduces cardiac lipotoxicity by inhibiting apoptosis and decreasing the expression of apoptosis-related proteins, reducing the expression of inflammatory cytokines, activating the autophagy signaling pathway, and inhibiting the expression of endoplasmic reticulum stress marker proteins.
Collapse
Affiliation(s)
- Sajad Abolfazli
- Student Research Committee, School of Pharmacy, Mazandaran University Medical Science, Sari, Iran
| | - Alexandra E Butler
- Research Department, Royal College of Surgeons in Ireland, Bahrain, Adliya, Bahrain
| | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Amirhossein Sahebkar
- Center for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
43
|
Cabral-García GA, Cruz-Muñoz JR, Valdez-Morales EE, Barajas-Espinosa A, Liñán-Rico A, Guerrero-Alba R. Pharmacology of P2X Receptors and Their Possible Therapeutic Potential in Obesity and Diabetes. Pharmaceuticals (Basel) 2024; 17:1291. [PMID: 39458933 PMCID: PMC11509955 DOI: 10.3390/ph17101291] [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: 08/08/2024] [Revised: 09/21/2024] [Accepted: 09/26/2024] [Indexed: 10/28/2024] Open
Abstract
The role of P2X ionotropic receptors in the behavior of purinergic signaling on pathophysiological processes has been widely studied. In recent years, the important participation of P2X receptors in physiological and pathological processes, such as energy metabolism, characteristic inflammatory responses of the immune system, and nociceptive activity in response to pain stimuli, has been noted. Here, we explore the molecular characteristics of the P2X receptors and the use of the different agonist and antagonist agents recently described, focusing on their potential as new therapeutic targets in the treatment of diseases with emphasis on obesity, diabetes, and some of the complications derived from these pathologies.
Collapse
Affiliation(s)
- Guillermo A. Cabral-García
- Departamento de Fisiología y Farmacología, Centro de Ciencias Básicas, Universidad Autónoma de Aguascalientes, Aguascalientes 20100, Mexico; (G.A.C.-G.); (J.R.C.-M.); (E.E.V.-M.)
| | - José R. Cruz-Muñoz
- Departamento de Fisiología y Farmacología, Centro de Ciencias Básicas, Universidad Autónoma de Aguascalientes, Aguascalientes 20100, Mexico; (G.A.C.-G.); (J.R.C.-M.); (E.E.V.-M.)
| | - Eduardo E. Valdez-Morales
- Departamento de Fisiología y Farmacología, Centro de Ciencias Básicas, Universidad Autónoma de Aguascalientes, Aguascalientes 20100, Mexico; (G.A.C.-G.); (J.R.C.-M.); (E.E.V.-M.)
- Consejo Nacional de Humanidades, Ciencias y Tecnologías (CONAHCyT), Ciudad de México 03940, Mexico;
| | - Alma Barajas-Espinosa
- Escuela Superior de Huejutla, Universidad Autónoma del Estado de Hidalgo, Huejutla de Reyes 43000, Hidalgo, Mexico;
| | - Andrómeda Liñán-Rico
- Consejo Nacional de Humanidades, Ciencias y Tecnologías (CONAHCyT), Ciudad de México 03940, Mexico;
- Centro Universitario de Investigaciones Biomédicas, Universidad de Colima, Colima 28045, Mexico
| | - Raquel Guerrero-Alba
- Departamento de Fisiología y Farmacología, Centro de Ciencias Básicas, Universidad Autónoma de Aguascalientes, Aguascalientes 20100, Mexico; (G.A.C.-G.); (J.R.C.-M.); (E.E.V.-M.)
| |
Collapse
|
44
|
Ostrom EL, Stuppard R, Mattson-Hughes A, Marcinek DJ. Inducible and reversible SOD2 knockdown in mouse skeletal muscle drives impaired pyruvate oxidation and reduced metabolic flexibility. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.23.614547. [PMID: 39386714 PMCID: PMC11463494 DOI: 10.1101/2024.09.23.614547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Introduction Skeletal muscle mitochondrial dysfunction is a key characteristic of aging muscle and contributes to age related diseases such as sarcopenia, frailty, and type 2 diabetes. Mitochondrial oxidative distress has been implicated as a driving factor in these age-related diseases, however whether it is a cause, or a consequence of mitochondrial dysfunction remains to be determined. The development of more flexible genetic models is an important tool to test the mechanistic role of mitochondrial oxidative stress on skeletal muscle metabolic dysfunction. We characterize a new model of inducible and reversible mitochondrial redox stress using a tetracycline controlled skeletal muscle specific short hairpin RNA targeted to superoxide dismutase 2 (iSOD2). Methods iSOD2 KD and control (CON) animals were administered doxycycline for 3- or 12- weeks and followed for up to 24 weeks and mitochondrial respiration and muscle contraction were measured to define the time course of SOD2 KD and muscle functional changes and recovery. Results Maximum knockdown of SOD2 protein occurred by 6 weeks and recovered by 24 weeks after DOX treatment. Mitochondrial aconitase activity and maximum mitochondrial respiration declined in KD muscle by 12 weeks and recovered by 24 weeks. There were minimal changes in gene expression between KD and CON muscle. Twelve-week KD showed a small, but significant decrease in muscle fatigue resistance. The primary phenotype was reduced metabolic flexibility characterized by impaired pyruvate driven respiration when other substrates are present. The pyruvate dehydrogenase kinase inhibitor dichloroacetate partially restored pyruvate driven respiration, while the thiol reductant DTT did not. Conclusion We use a model of inducible and reversible skeletal muscle SOD2 knockdown to demonstrate that elevated matrix superoxide reversibly impairs mitochondrial substrate flexibility characterized by impaired pyruvate oxidation. Despite the bioenergetic effect, the limited change in gene expression suggests that the elevated redox stress in this model is confined to the mitochondrial matrix.
Collapse
Affiliation(s)
- Ethan L Ostrom
- Department of Radiology, University of Washington School of Medicine, Seattle, WA, USA
| | - Rudy Stuppard
- Department of Radiology, University of Washington School of Medicine, Seattle, WA, USA
| | - Aurora Mattson-Hughes
- Department of Radiology, University of Washington School of Medicine, Seattle, WA, USA
| | - David J Marcinek
- Department of Radiology, University of Washington School of Medicine, Seattle, WA, USA
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, WA, USA
| |
Collapse
|
45
|
An J, Astapova I, Zhang G, Cangelosi AL, Ilkayeva O, Marchuk H, Muehlbauer MJ, George T, Brozinick J, Herman MA, Newgard CB. Integration of metabolomic and transcriptomic analyses reveals novel regulatory functions of the ChREBP transcription factor in energy metabolism. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.17.613577. [PMID: 39345566 PMCID: PMC11429843 DOI: 10.1101/2024.09.17.613577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Carbohydrate Response Element-Binding Protein (ChREBP) is a transcription factor that activates key genes involved in glucose, fructose, and lipid metabolism in response to carbohydrate feeding, but its other potential roles in metabolic homeostasis have not been as well studied. We used liver-selective GalNAc-siRNA technology to suppress expression of ChREBP in rats fed a high fat/high sucrose diet and characterized hepatic and systemic responses by integrating transcriptomic and metabolomic analyses. GalNAc-siChREBP-treated rats had lower levels of multiple short-chain acyl CoA metabolites compared to rats treated with GalNAc-siCtrl containing a non-targeting siRNA sequence. These changes were related to a sharp decrease in free CoA levels in GalNAc-siChREBP treated-rats, accompanied by lower expression of transcripts encoding enzymes and transporters involved in CoA biosynthesis. These activities of ChREBP likely contribute to its complex effects on hepatic lipid and energy metabolism. While core enzymes of fatty acid (FA) oxidation are induced by ChREBP knockdown, accumulation of liver acylcarnitines and circulating ketones indicate diversion of acetyl CoA to ketone production rather than complete oxidation in the TCA cycle. Despite strong suppression of pyruvate kinase and activation of pyruvate dehydrogenase, pyruvate levels were maintained, likely via increased expression of pyruvate transporters, and decreased expression of lactate dehydrogenase and alanine transaminase. GalNAc-siChREBP treatment increased hepatic citrate and isocitrate levels while decreasing levels of distal TCA cycle intermediates. The drop in free CoA levels, needed for the 2-ketoglutarate dehydrogenase reaction, as well as a decrease in transcripts encoding the anaplerotic enzymes pyruvate carboxylase, glutamate dehydrogenase, and aspartate transaminase likely contributed to these effects. GalNAc-siChREBP treatment caused striking increases in PRPP and ZMP/AICAR levels, and decreases in GMP, IMP, AMP, NaNM, NAD(P), and NAD(P)H levels, accompanied by reduced expression of enzymes that catalyze late steps in purine and NAD synthesis. ChREBP suppression also increased expression of a set of plasma membrane amino acid transporters, possibly as an attempt to replenish TCA cycle intermediates. In sum, combining transcriptomic and metabolomic analyses has revealed regulatory functions of ChREBP that go well beyond its canonical roles in control of carbohydrate and lipid metabolism to now include mitochondrial metabolism and cellular energy balance.
Collapse
Affiliation(s)
- Jie An
- Sarah W. Stedman Nutrition and Metabolism Center & Duke Molecular Physiology Institute, Duke University Medical Center
| | - Inna Astapova
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Baylor College of Medicine
| | - Guofang Zhang
- Sarah W. Stedman Nutrition and Metabolism Center & Duke Molecular Physiology Institute, Duke University Medical Center
- Division of Endocrinology, Metabolism and Nutrition, Department of Medicine, Duke University Medical Center
| | - Andrew L. Cangelosi
- Sarah W. Stedman Nutrition and Metabolism Center & Duke Molecular Physiology Institute, Duke University Medical Center
| | - Olga Ilkayeva
- Sarah W. Stedman Nutrition and Metabolism Center & Duke Molecular Physiology Institute, Duke University Medical Center
- Division of Endocrinology, Metabolism and Nutrition, Department of Medicine, Duke University Medical Center
| | - Hannah Marchuk
- Sarah W. Stedman Nutrition and Metabolism Center & Duke Molecular Physiology Institute, Duke University Medical Center
| | - Michael J. Muehlbauer
- Sarah W. Stedman Nutrition and Metabolism Center & Duke Molecular Physiology Institute, Duke University Medical Center
| | - Tabitha George
- Sarah W. Stedman Nutrition and Metabolism Center & Duke Molecular Physiology Institute, Duke University Medical Center
| | | | - Mark A. Herman
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Baylor College of Medicine
| | - Christopher B. Newgard
- Sarah W. Stedman Nutrition and Metabolism Center & Duke Molecular Physiology Institute, Duke University Medical Center
- Division of Endocrinology, Metabolism and Nutrition, Department of Medicine, Duke University Medical Center
- Department of Pharmacology & Cancer Biology, Duke University Medical Center
| |
Collapse
|
46
|
Watson H, Nilsson JÅ, Smith E, Ottosson F, Melander O, Hegemann A, Urhan U, Isaksson C. Urbanisation-associated shifts in the avian metabolome within the annual cycle. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 944:173624. [PMID: 38821291 DOI: 10.1016/j.scitotenv.2024.173624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 05/07/2024] [Accepted: 05/27/2024] [Indexed: 06/02/2024]
Abstract
While organisms have evolved to cope with predictable changes in the environment, the rapid rate of current global change presents numerous novel and unpredictable stressors to which organisms have had less time to adapt. To persist in the urban environment, organisms must modify their physiology, morphology and behaviour accordingly. Metabolomics offers great potential for characterising organismal responses to natural and anthropogenic stressors at the systems level and can be applied to any species, even without genomic knowledge. Using metabolomic profiling of blood, we investigated how two closely related species of passerine bird respond to the urban environment. Great tits Parus major and blue tits Cyanistes caeruleus residing in urban and forest habitats were sampled during the breeding (spring) and non-breeding (winter) seasons across replicated sites in southern Sweden. During breeding, differences in the plasma metabolome between urban and forest birds were characterised by higher levels of amino acids in urban-dwelling tits and higher levels of fatty acyls in forest-dwelling tits. The suggested higher rates of fatty acid oxidation in forest tits could be driven by habitat-associated differences in diet and could explain the higher reproductive investment and success of forest tits. High levels of amino acids in breeding urban tits could reflect the lack of lipid-rich caterpillars in the urban environment and a dietary switch to protein-rich spiders, which could be of benefit for tackling inflammation and oxidative stress associated with pollution. In winter, metabolomic profiles indicated lower overall levels of amino acids and fatty acyls in urban tits, which could reflect relaxed energetic demands in the urban environment. Our metabolomic profiling of two urban-adapted species suggests that their metabolism is modified by urban living, though whether these changes represent adaptative or non-adaptive mechanisms to cope with anthropogenic challenges remains to be determined.
Collapse
Affiliation(s)
- Hannah Watson
- Department of Biology, Lund University, 223 62 Lund, Sweden.
| | | | - Einar Smith
- Department of Clinical Sciences, Lund University, 214 28 Malmö, Sweden
| | - Filip Ottosson
- Department of Clinical Sciences, Lund University, 214 28 Malmö, Sweden
| | - Olle Melander
- Department of Clinical Sciences, Lund University, 214 28 Malmö, Sweden
| | - Arne Hegemann
- Department of Biology, Lund University, 223 62 Lund, Sweden
| | - Utku Urhan
- Department of Biology, Lund University, 223 62 Lund, Sweden
| | | |
Collapse
|
47
|
Gyllenhammer LE, Zaegel V, Duensing AM, Lixandrao ME, Dabelea D, Bergman BC, Boyle KE. Lipidomics of infant mesenchymal stem cells associate with the maternal milieu and child adiposity. JCI Insight 2024; 9:e180016. [PMID: 39226911 PMCID: PMC11466181 DOI: 10.1172/jci.insight.180016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 08/21/2024] [Indexed: 09/05/2024] Open
Abstract
Our objective was to interrogate mesenchymal stem cell (MSC) lipid metabolism and gestational exposures beyond maternal body mass that may contribute to child obesity risk. MSCs were cultured from term infants of mothers with obesity (n = 16) or normal weight (n = 15). In MSCs undergoing myogenesis in vitro, we used lipidomics to distinguish phenotypes by unbiased cluster analysis and lipid challenge (24-hour excess fatty acid [24hFA]). We measured MSC AMP-activated protein kinase (AMPK) activity and fatty acid oxidation (FAO), and a composite index of maternal glucose, insulin, triglycerides, free fatty acids, TNF-α, and high-density lipoprotein and total cholesterol in fasting blood from mid and late gestation (~17 and ~27 weeks, respectively). We measured child adiposity at birth (n = 29), 4-6 months (n = 29), and 4-6 years (n = 13). Three MSC clusters were distinguished by triacylglycerol (TAG) stores, with greatest TAGs in Cluster 2. All clusters increased acylcarnitines and TAGs with 24hFA, although Cluster 2 was more pronounced and corresponded to AMPK activation and FAO. Maternal metabolic markers predicted MSC clusters and child adiposity at 4-6 years (both highest in Cluster 3). Our data support the notion that MSC phenotypes are predicted by comprehensive maternal metabolic milieu exposures, independent of maternal BMI, and suggest utility as an at-birth predictor for child adiposity, although validation with larger longitudinal samples is warranted.
Collapse
Affiliation(s)
- Lauren E. Gyllenhammer
- Department of Pediatrics, University of California, Irvine, School of Medicine, Irvine, California, USA
| | - Vincent Zaegel
- Department of Pediatrics, Section of Nutrition, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Allison M. Duensing
- Department of Pediatrics, Section of Nutrition, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Manoel E. Lixandrao
- Department of Pediatrics, Section of Nutrition, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Dana Dabelea
- The Lifecourse Epidemiology of Adiposity and Diabetes (LEAD) Center, Aurora, Colorado, USA
- Department of Epidemiology, Colorado School of Public Health
- Department of Pediatrics, and
| | - Bryan C. Bergman
- Department of Endocrinology, Metabolism, and Diabetes, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Kristen E. Boyle
- Department of Pediatrics, Section of Nutrition, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- The Lifecourse Epidemiology of Adiposity and Diabetes (LEAD) Center, Aurora, Colorado, USA
| |
Collapse
|
48
|
Park SY, Jung SR, Kim JY, Kim YW, Sung HK, Park SY, Doh KO, Koh JH. Lactate promotes fatty acid oxidation by the tricarboxylic acid cycle and mitochondrial respiration in muscles of obese mice. Am J Physiol Cell Physiol 2024; 327:C619-C633. [PMID: 38981606 DOI: 10.1152/ajpcell.00060.2024] [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: 01/28/2024] [Revised: 06/04/2024] [Accepted: 06/20/2024] [Indexed: 07/11/2024]
Abstract
Lower oxidative capacity in skeletal muscles (SKMs) is a prevailing cause of metabolic diseases. Exercise not only enhances the fatty acid oxidation (FAO) capacity of SKMs but also increases lactate levels. Given that lactate may contribute to tricarboxylic acid cycle (TCA) flux and impact monocarboxylate transporter 1 in the SKMs, we hypothesize that lactate can influence glucose and fatty acid (FA) metabolism. To test this hypothesis, we investigated the mechanism underlying lactate-driven FAO regulation in the SKM of mice with diet-induced obesity (DIO). Lactate was administered to DIO mice immediately after exercise for over 3 wk. We found that increased lactate levels enhanced energy expenditure mediated by fat metabolism during exercise recovery and decreased triglyceride levels in DIO mice SKMs. To determine the lactate-specific effects without exercise, we administered lactate to mice on a high-fat diet (HFD) for 8 wk. Similar to our exercise conditions, lactate increased FAO, TCA cycle activity, and mitochondrial respiration in the SKMs of HFD-fed mice. In addition, under sufficient FA conditions, lactate increased uncoupling protein-3 abundance via the NADH-NAD+ shuttle. Conversely, ATP synthase abundance decreased in the SKMs of HFD mice. Taken together, our results suggest that lactate amplifies the adaptive increase in FAO capacity mediated by the TCA cycle and mitochondrial respiration in SKMs under sufficient FA abundance.NEW & NOTEWORTHY Lactate administration post-exercise promotes triglyceride content loss in skeletal muscles (SKMs) and reduced body weight. Lactate enhances fatty acid oxidation in the SKMs of high-fat diet (HFD)-fed mice due to enhanced mitochondrial oxygen consumption. In addition, lactate restores the malate-aspartate shuttle, which is reduced by a HFD, and activates the tricarboxylic acid cycle (TCA) cycle in SKMs. Interestingly, supraphysiological lactate facilitates uncoupling protein-3 expression through NADH/NAD+, which is enhanced under high-fat levels in SKMs.
Collapse
Affiliation(s)
- Sol-Yi Park
- Department of Physiology, Yeungnam University College of Medicine, Daegu, Republic of Korea
| | - Su-Ryun Jung
- Department of Physiology, Yeungnam University College of Medicine, Daegu, Republic of Korea
| | - Jong-Yeon Kim
- Department of Physiology, Yeungnam University College of Medicine, Daegu, Republic of Korea
| | - Yong-Woon Kim
- Department of Physiology, Yeungnam University College of Medicine, Daegu, Republic of Korea
| | - Hoon-Ki Sung
- Translational Medicine Program, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - So-Young Park
- Department of Physiology, Yeungnam University College of Medicine, Daegu, Republic of Korea
| | - Kyung-Oh Doh
- Department of Physiology, Yeungnam University College of Medicine, Daegu, Republic of Korea
| | - Jin-Ho Koh
- Department of Convergence Medicine, Yonsei University Wonju College of Medicine, Wonju, Republic of Korea
- Department of Global Medical Science, Yonsei University Wonju College of Medicine, Wonju, Republic of Korea
| |
Collapse
|
49
|
Schipper MC, Blaauwendraad SM, Koletzko B, Oei EHG, Jaddoe VWV, Gaillard R. Associations of childhood BMI, general and visceral fat mass with metabolite profiles at school-age. Int J Obes (Lond) 2024; 48:1307-1317. [PMID: 38851839 DOI: 10.1038/s41366-024-01558-8] [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] [Received: 01/23/2024] [Revised: 05/22/2024] [Accepted: 05/30/2024] [Indexed: 06/10/2024]
Abstract
BACKGROUND Childhood obesity increases metabolic disease risk. Underlying mechanisms remain unknown. We examined associations of body mass index (BMI), total body fat mass, and visceral fat mass with serum metabolites at school-age, and explored whether identified metabolites improved the identification of children at risk of a metabolically unhealthy phenotype. METHODS We performed a cross-sectional analysis among 497 children with a mean age of 9.8 (95% range 9.1, 10.6) years, participating in a population-based cohort study. We measured BMI, total body fat mass using DXA, and visceral fat mass using MRI. Serum concentrations of amino-acids, non-esterified-fatty-acids, phospholipids, and carnitines were determined using LC-MS/MS. Children were categorized as metabolically healthy or metabolically unhealthy, according to BMI, blood pressure, lipids, glucose, and insulin levels. RESULTS Higher BMI and total body fat mass were associated with altered concentrations of branched-chain amino-acids, essential amino-acids, and free carnitines. Higher BMI was also associated with higher concentrations of aromatic amino-acids and alkyl-lysophosphatidylcholines (FDR-corrected p-values < 0.05). The strongest associations were present for Lyso.PC.a.C14.0 and SM.a.C32.2 (FDR-corrected p-values < 0.01). Higher visceral fat mass was only associated with higher concentrations of 6 individual metabolites, particularly Lyso.PC.a.C14.0, PC.aa.C32.1, and SM.a.C32.2. We selected 15 metabolites that improved the prediction of a metabolically unhealthy phenotype, compared to BMI only (AUC: BMI: 0.59 [95% CI 0.47,0.71], BMI + Metabolites: 0.91 [95% CI 0.85,0.97]). CONCLUSIONS An adverse childhood body fat profile, characterized by higher BMI and total body fat mass, is associated with metabolic alterations, particularly in amino acids, phospholipids, and carnitines. Fewer associations were present for visceral fat mass. We identified a metabolite profile that improved the identification of impaired cardiometabolic health in children, compared to BMI only.
Collapse
Affiliation(s)
- Mireille C Schipper
- The Generation R Study Group Erasmus MC, University Medical Center, Rotterdam, the Netherlands
- Department of Pediatrics, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
| | - Sophia M Blaauwendraad
- The Generation R Study Group Erasmus MC, University Medical Center, Rotterdam, the Netherlands
- Department of Pediatrics, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
| | - Berthold Koletzko
- LMU - Ludwig Maximilians Universität Munich, Department of Pediatrics, Dr. von Hauner Children's Hospital, LMU University Hospitals, Munich, Germany
| | - Edwin H G Oei
- Department of Radiology & Nuclear Medicine, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Vincent W V Jaddoe
- The Generation R Study Group Erasmus MC, University Medical Center, Rotterdam, the Netherlands
- Department of Pediatrics, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
| | - Romy Gaillard
- The Generation R Study Group Erasmus MC, University Medical Center, Rotterdam, the Netherlands.
- Department of Pediatrics, Erasmus MC, University Medical Center, Rotterdam, the Netherlands.
| |
Collapse
|
50
|
Roszczyc-Owsiejczuk K, Imierska M, Sokołowska E, Kuźmicki M, Pogodzińska K, Błachnio-Zabielska A, Zabielski P. shRNA-mediated down-regulation of Acsl1 reverses skeletal muscle insulin resistance in obese C57BL6/J mice. PLoS One 2024; 19:e0307802. [PMID: 39178212 PMCID: PMC11343424 DOI: 10.1371/journal.pone.0307802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 07/12/2024] [Indexed: 08/25/2024] Open
Abstract
Prolonged consumption of diet rich in fats is regarded as the major factor leading to the insulin resistance (IR) and type 2 diabetes (T2D). Emerging evidence link excessive accumulation of bioactive lipids such as diacylglycerol (DAG) and ceramide (Cer), with impairment of insulin signaling in skeletal muscle. Until recently, little has been known about the involvement of long-chain acyl-CoAs synthetases in the above mechanism. To examine possible role of long-chain acyl-coenzyme A synthetase 1 (Acsl1) (a major muscular ACSL isoform) in mediating HFD-induced IR we locally silenced Acsl1 in gastrocnemius of high-fat diet (HFD)-fed C57BL/6J mice through electroporation-delivered shRNA and compared it to non-silenced tissue within the same animal. Acsl1 down-regulation decreased the content of muscular long-chain acyl-CoA (LCACoA) and both the Cer (C18:1-Cer and C24:1-Cer) and DAG (C16:0/18:0-DAG, C16:0/18:2-DAG, C18:0/18:0-DAG) and simultaneously improved insulin sensitivity and glucose uptake as compared with non-silenced tissue. Acsl1 down-regulation decreased expression of mitochondrial β-oxidation enzymes, and the content of both the short-chain acylcarnitine (SCA-Car) and short-chain acyl-CoA (SCACoA) in muscle, pointing towards reduction of mitochondrial FA oxidation. The results indicate, that beneficial effects of Acsl1 partial ablation on muscular insulin sensitivity are connected with inhibition of Cer and DAG accumulation, and outweigh detrimental impact of decreased mitochondrial fatty acids metabolism in skeletal muscle of obese HFD-fed mice.
Collapse
Affiliation(s)
- Kamila Roszczyc-Owsiejczuk
- Department of Hygiene, Epidemiology and Metabolic Disorders, Medical University of Bialystok, Bialystok, Poland
| | - Monika Imierska
- Department of Hygiene, Epidemiology and Metabolic Disorders, Medical University of Bialystok, Bialystok, Poland
| | - Emilia Sokołowska
- Department of Hygiene, Epidemiology and Metabolic Disorders, Medical University of Bialystok, Bialystok, Poland
| | - Mariusz Kuźmicki
- Department of Gynecology and Gynecological Oncology, Medical University of Bialystok, Bialystok, Poland
| | - Karolina Pogodzińska
- Department of Hygiene, Epidemiology and Metabolic Disorders, Medical University of Bialystok, Bialystok, Poland
| | | | - Piotr Zabielski
- Department of Medical Biology, Medical University of Bialystok, Bialystok, Poland
| |
Collapse
|