1
|
Yin R, Jing G, Tian Y, Ma M, Zhang M. The impact of lactate on diabetic cognitive dysfunction: Insights from energy metabolism to epigenetic modulation. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167749. [PMID: 40010229 DOI: 10.1016/j.bbadis.2025.167749] [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: 09/19/2024] [Revised: 02/15/2025] [Accepted: 02/20/2025] [Indexed: 02/28/2025]
Abstract
This manuscript elucidates the intricate roles of lactate in Diabetic Cognitive Dysfunction (DCD), extending beyond its conventional role as an energy substrate. The investigation centers on the participation of lactate in energy metabolism and epigenetic modulation, with a particular emphasis on its influence on cognitive faculties through histone lactylation. The discourse scrutinizes lactate's part in the metabolic equilibrium of the central nervous system, encompassing its fluctuating concentrations under various conditions and its pivotal function within the Astrocyte-Neuron Lactate Shuttle (ANLS) mechanism as an energy conduit. The involvement of lactate in DCD is multilayered, encompassing metabolic pathways, cellular signaling cascades, and the regulation of gene expression. Dysregulation in lactate metabolism and the histone lactylation process may modulate neuronal functionality by impacting genes integral to neuroplasticity and cognitive capabilities. These revelations offer novel insights into the molecular underpinnings of DCD and lay the groundwork for the discovery of potential therapeutic targets. Subsequent scholarly endeavors are poised to dissect the nuanced mechanisms by which lactate and its lactylation exert influence in DCD, pinpointing the critical genes modulated by lactylation and assessing their ramifications on neuronal function and signal transduction pathways. Given the intricate regulatory dynamics of lactate, contingent upon concentration, temporal factors, and disease etiology, a more profound elucidation of lactate's role in DCD necessitates an augmented cadre of animal experimentation and clinical observational research. Such investigative pursuits are anticipated to yield innovative approaches and methodologies for the comprehensive management of DCD, spanning prevention, diagnosis, and therapeutic intervention.
Collapse
Affiliation(s)
- Ruiying Yin
- Department of Traditional Chinese Medicine, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Guangchan Jing
- Department of Traditional Chinese Medicine, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Yue Tian
- Department of Traditional Chinese Medicine, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Mei Ma
- Department of Traditional Chinese Medicine, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Mengren Zhang
- Department of Traditional Chinese Medicine, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100730, China.
| |
Collapse
|
2
|
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
|
3
|
Wong A, Sun Q, Latif II, Karwi QG. Macrophage energy metabolism in cardiometabolic disease. Mol Cell Biochem 2025; 480:1763-1783. [PMID: 39198360 PMCID: PMC11842501 DOI: 10.1007/s11010-024-05099-6] [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: 04/27/2024] [Accepted: 08/19/2024] [Indexed: 09/01/2024]
Abstract
In a rapidly expanding body of literature, the major role of energy metabolism in determining the response and polarization status of macrophages has been examined, and it is currently a very active area of research. The metabolic flux through different metabolic pathways in the macrophage is interconnected and complex and could influence the polarization of macrophages. Earlier studies suggested glucose flux through cytosolic glycolysis is a prerequisite to trigger the pro-inflammatory phenotypes of macrophages while proposing that fatty acid oxidation is essential to support anti-inflammatory responses by macrophages. However, recent studies have shown that this understanding is oversimplified and that the metabolic control of macrophage polarization is highly complex and not fully defined yet. In this review, we systematically reviewed and summarized the literature regarding the role of energy metabolism in controlling macrophage activity and how that might be altered in cardiometabolic diseases, namely heart failure, obesity, and diabetes. We critically appraised the experimental studies and methodologies in the published studies. We also highlighted the challenging concepts in macrophage metabolism and identified several research questions yet to be addressed in future investigations.
Collapse
Affiliation(s)
- Angela Wong
- Division of BioMedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John's, Newfoundland and Labrador, A1B 3V6, Canada
- Cardiovascular Research Centre, University of Alberta, Edmonton, Alberta, Canada
| | - Qiuyu Sun
- Division of BioMedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John's, Newfoundland and Labrador, A1B 3V6, Canada
- Cardiovascular Research Centre, University of Alberta, Edmonton, Alberta, Canada
| | - Ismail I Latif
- Department of Microbiology, College of Medicine, University of Diyala, Baqubaa, Diyala, Iraq
| | - Qutuba G Karwi
- Division of BioMedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John's, Newfoundland and Labrador, A1B 3V6, Canada.
| |
Collapse
|
4
|
Martín-San Agustín R, Cuerda Del Pino A, Laguna Sanz AJ, Palanca A, Rossetti P, Marco Romero C, Bondia J, Ampudia-Blasco FJ. Impact of High-Intensity Interval Exercise With Elastic Bands Versus Continuous Moderate-Intensity Aerobic Exercise on Glycemic Control in People With Type 1 Diabetes. Sports Health 2025:19417381251316247. [PMID: 39905638 DOI: 10.1177/19417381251316247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2025] Open
Abstract
BACKGROUND Engaging in physical exercise is recommended to enhance cardiovascular health and manage blood sugar levels in people with type 1 diabetes (T1D). HYPOTHESIS The impact of high-intensity interval exercise with elastic bands (EB-HIIE) versus continuous moderate-intensity aerobic exercise (CONT) on glycemic control is different in men with T1D. STUDY DESIGN Crossover study design. LEVEL OF EVIDENCE Level 3. METHODS Participants (39 men with T1D) underwent either an EB-HIIE or a CONT session in randomized order, with a separation of ≥72 hours to avoid carry-over effects. Changes in glucose values during exercise were measured simultaneously from venous blood (YSI) and interstitial fluid (Dexcom G6 glucose sensor). Subsequent 24-hour glucose was monitored using the glucose sensor. RESULTS Blood glucose was lower in CONT vs EB-HIIE (P < .01). Post hoc analysis revealed clinically relevant differences during exercise (-35.1 mg/dl; P = .02), at its end (-49.5 mg/dl; P < .01), and at 10 and 20 minutes after completion (-51.2 mg/dl; P < .01 and -45.9 mg/dl; P < .01, respectively). Time-in-range 24 hours after exercise completion was significantly higher with EB-HIIE than with CONT (66.5% vs 59.3%), although both were significantly better than the previous 24 hours before exercise (50%). CONCLUSION Results suggest that EB-HIIE is a safe training method for male adults with diabetes, resulting in euglycemia during and immediately after exercise and improving glucose outcomes in the subsequent 24 hours. CLINICAL RELEVANCE This study provides new evidence and practical information on how to implement safe physical activity in daily life of patients with diabetes. EB-HIIT exhibited lower hypoglycemia risk during exercise and better glycemic control in the subsequent 24 hours. In contrast, practicing CONT exercise is associated with higher risk of hypoglycemia. Healthcare providers should take this information into account when prescribing exercise.
Collapse
Affiliation(s)
- Rodrigo Martín-San Agustín
- Clinimetry and Technological Development in Therapeutic Exercise Research Group (CLIDET), Department of Physiotherapy, University of Valencia, Valencia, Spain
| | - Alba Cuerda Del Pino
- Clinimetry and Technological Development in Therapeutic Exercise Research Group (CLIDET), Department of Physiotherapy, University of Valencia, Valencia, Spain
| | - Alejandro José Laguna Sanz
- Clinimetry and Technological Development in Therapeutic Exercise Research Group (CLIDET), Department of Physiotherapy, University of Valencia, Valencia, Spain, and Department of Electronic Engineering, University of Valencia, Valencia, Spain
| | - Ana Palanca
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain, and INCLIVA Biomedical Research Institute, Valencia, Spain, Department of Endocrinology & Nutrition, University and Polytechnic La Fe Hospital of Valencia, Spain
| | - Paolo Rossetti
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain, and Department of Endocrinology & Nutrition, University and Polytechnic La Fe Hospital of Valencia, Spain
| | - Cynthia Marco Romero
- Department of Endocrinology & Nutrition, Clinic University Hospital of Valencia, Spain
| | - Jorge Bondia
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain, and Instituto Universitario de Automática e Informática Industrial, Universitat Politècnica de València, València, Spain
| | - F Javier Ampudia-Blasco
- Clinimetry and Technological Development in Therapeutic Exercise Research Group (CLIDET), Department of Physiotherapy, University of Valencia, Valencia, Spain, Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain, INCLIVA Biomedical Research Institute, Valencia, Spain, Department of Endocrinology & Nutrition, Clinic University Hospital of Valencia, Spain, and Department of Medicine, University of Valencia, Valencia, Spain
| |
Collapse
|
5
|
Abbastabar M, Mohammadi-Pirouz Z, Omidvar S, Bakhtiari A, Crowe FL, Sepidarkish M. Dietary Acid Load and Human Health: A Systematic Review and Meta-analysis of Observational Studies. Nutr Rev 2025:nuae222. [PMID: 39899655 DOI: 10.1093/nutrit/nuae222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2025] Open
Abstract
CONTEXT Dietary acid load (DAL) plays an important role in different aspects of human health. OBJECTIVE The aim of this meta-analysis was to systematically synthesize the observational evidence reporting on the associations between the DAL (represented by a potential renal acid load [PRAL] and net endogenous acid production [NEAP]) and a range of health outcomes. DATA SOURCES A systematic search of Medline (via PubMed), EMBASE, Scopus, and Web of Science was performed to identify eligible studies. DATA EXTRACTION Study screening and risk-of-bias assessment were conducted by 2 independent reviewers. DATA ANALYSIS The credibility of each outcome was graded based on predefined criteria: pooled effect size with corresponding 95% CI, 95% prediction interval, heterogeneity, small-study effect, and excess significance bias. In total, 118 observational studies (case-control [n = 22], cross-sectional [n = 65], and cohort [n = 31]) on 38 outcomes were included, incorporating a total population of 1 014 081 participants. Overall, 21/37 and 20/38 of the outcomes reported statistically significant effect sizes for PRAL and NEAP, respectively. The credibility of the evidence for PRAL was rated convincing (class I) for type 2 diabetes (T2D), and as highly suggestive evidence (class II) for overall cancer and systolic blood pressure. By assessing the credibility of the evidence for NEAP, T2D and overall cancer presented convincing evidence (class I) and breast cancer presented highly suggestive evidence (class II). The remaining outcomes presented class III (suggestive) or lower evidence (weak or no association). CONCLUSION The higher acid-forming potential of diet was associated with a higher risk of T2D and overall cancer. Since this meta-analysis included observational studies and some of the associations were graded as weak, caution should be exercised in interpreting these associations. Further cohort studies are required with consideration of other factors that can cause biases. SYSTEMATIC REVIEW REGISTRATION PROSPERO registration no. CRD42022336050.
Collapse
Affiliation(s)
- Maryam Abbastabar
- Department of Clinical Biochemistry, School of Medicine, Babol University of Medical Sciences, Babol, Iran
| | - Zahra Mohammadi-Pirouz
- Department of Biostatistics and Epidemiology, School of Public Health, Babol University of Medical Sciences, Babol, Iran
| | - Shabnam Omidvar
- Social Determinants of Health Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Afsaneh Bakhtiari
- Social Determinants of Health Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Francesca L Crowe
- Institute of Applied Health Research, University of Birmingham, Birmingham, United Kingdom
| | - Mahdi Sepidarkish
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| |
Collapse
|
6
|
Chanoine JP, Thompson DM, Lehman A. Diabetes Associated With Maternally Inherited Diabetes and Deafness (MIDD): From Pathogenic Variant to Phenotype. Diabetes 2025; 74:153-163. [PMID: 39556456 PMCID: PMC11755681 DOI: 10.2337/db24-0515] [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: 06/24/2024] [Accepted: 11/13/2024] [Indexed: 11/20/2024]
Abstract
ARTICLE HIGHLIGHTS Maternally inherited diabetes and deafness (MIDD) is a mitochondrial disorder characterized primarily by hearing impairment and diabetes. m.3243A>G, the most common phenotypic variant, causes a complex rewiring of the cell with discontinuous remodeling of both mitochondrial and nuclear genome expressions. We propose that MIDD depends on a combination of insulin resistance and impaired β-cell function that occurs in the presence of high skeletal muscle heteroplasmy (approximately ≥60%) and more moderate cell heteroplasmy (∼25%-72%) for m.3243A>G. Understanding the complex mechanisms of MIDD is necessary to develop disease-specific management guidelines that are presently lacking.
Collapse
Affiliation(s)
- Jean-Pierre Chanoine
- Endocrinology and Diabetes Unit, Department of Pediatrics, BC Children’s Hospital and The University of British Columbia, Vancouver, British Columbia, Canada
| | - David M. Thompson
- Division of Endocrinology, Department of Medicine, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Anna Lehman
- Department of Medical Genetics, Faculty of Medicine, The University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
7
|
Romeres D, Yadav Y, Ruchi FNU, Carter R, Cobelli C, Basu R, Basu A. Hyperglycemia Suppresses Lactate Clearance During Exercise in Type 1 Diabetes. J Clin Endocrinol Metab 2024; 109:e1720-e1731. [PMID: 38174728 PMCID: PMC11318997 DOI: 10.1210/clinem/dgae005] [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: 09/28/2023] [Revised: 12/19/2023] [Accepted: 01/02/2024] [Indexed: 01/05/2024]
Abstract
CONTEXT Circulating lactate concentration is an important determinant of exercise tolerance. OBJECTIVE This work aimed to determine the role of hyperglycemia on lactate metabolism during exercise in individuals with type 1 diabetes (T1D). METHODS The protocol at the University of Virginia compared 7 T1D participants and 7 participants without diabetes (ND) at euglycemia (5.5 mM) or hyperglycemia (9.2 mM) in random order in T1D and at euglycemia in ND. Intervention included [1-13C] lactate infusion, exercise at 65% maximal oxygen uptake (VO2max), euglycemia, and hyperglycemia visits. The main outcome measure was lactate turnover before, during, and after 60 minutes of exercise at 65% VO2max. RESULTS A 2-compartment model with loss only from the peripheral compartment described lactate kinetics. Volume of distribution of the accessible compartment was similar between T1D and ND individuals (P = .76) and concordant with plasma volume (∼40 mL/kg). Circulating lactate concentrations were higher (P < .001) in T1D participants during exercise at hyperglycemia than euglycemia. Exercise-induced lactate appearance did not differ (P = .13) between hyperglycemia and euglycemia. However, lactate clearance (CL) was lower (P = .03) during hyperglycemia than euglycemia in T1D participants. There were no differences in any of the aforementioned parameters between T1D and ND participants during euglycemia. CONCLUSION Hyperglycemia modulates lactate metabolism during exercise by lowering CL, leading to higher circulating lactate concentrations in T1D individuals. This novel observation implies that exercise during hyperglycemia can lead to higher circulating lactate concentrations thus increasing the likelihood of reaching the lactate threshold sooner in T1D, and has high translational relevance both for providers and recreationally active people with T1D.
Collapse
Affiliation(s)
- Davide Romeres
- Division of Endocrinology, Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Yogesh Yadav
- Division of Endocrinology, Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - F N U Ruchi
- Division of Endocrinology, Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Rickey Carter
- Department of Health Sciences Research, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Claudio Cobelli
- Department of Woman and Child's Health, University of Padova, Padua 35122, Italy
| | - Rita Basu
- Division of Endocrinology, Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Ananda Basu
- Division of Endocrinology, Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| |
Collapse
|
8
|
Bartoloni B, Mannelli M, Gamberi T, Fiaschi T. The Multiple Roles of Lactate in the Skeletal Muscle. Cells 2024; 13:1177. [PMID: 39056759 PMCID: PMC11274880 DOI: 10.3390/cells13141177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 06/27/2024] [Accepted: 07/09/2024] [Indexed: 07/28/2024] Open
Abstract
Believed for a long time to be merely a waste product of cell metabolism, lactate is now considered a molecule with several roles, having metabolic and signalling functions together with a new, recently discovered role as an epigenetic modulator. Lactate produced by the skeletal muscle during physical exercise is conducted to the liver, which uses the metabolite as a gluconeogenic precursor, thus generating the well-known "Cori cycle". Moreover, the presence of lactate in the mitochondria associated with the lactate oxidation complex has become increasingly clear over the years. The signalling role of lactate occurs through binding with the GPR81 receptor, which triggers the typical signalling cascade of the G-protein-coupled receptors. Recently, it has been demonstrated that lactate regulates chromatin state and gene transcription by binding to histones. This review aims to describe the different roles of lactate in skeletal muscle, in both healthy and pathological conditions, and to highlight how lactate can influence muscle regeneration by acting directly on satellite cells.
Collapse
Affiliation(s)
| | | | | | - Tania Fiaschi
- Dipartimento di Scienze Biomediche, Sperimentali e Cliniche “M. Serio”, Università degli Studi di Firenze, 50134 Firenze, Italy; (B.B.); (M.M.); (T.G.)
| |
Collapse
|
9
|
Ting KK. John Yudkin's hypothesis: sugar is a major dietary culprit in the development of cardiovascular disease. Front Nutr 2024; 11:1407108. [PMID: 39027662 PMCID: PMC11257042 DOI: 10.3389/fnut.2024.1407108] [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: 03/26/2024] [Accepted: 06/24/2024] [Indexed: 07/20/2024] Open
Abstract
To date, the risk of developing atherosclerosis has extended beyond Western countries and now affecting individuals from various ethnic backgrounds and age groups. Traditional risk factors of atherosclerosis, such as hypercholesterolemia, has been better controlled than before due to highly effective and inexpensive therapies at lowering plasma cholesterol levels. However, the role of reducing dietary cholesterol intake, as a public healthy strategy, in preventing the occurrence of cardiovascular mortalities has been recently challenged. Indeed, despite our continuous decline of dietary cholesterol intake within the last 50 years, the incidence of cardiovascular mortalities has continued to rise, thus raising the possibility that other dietary factors, such as fructose-containing sugars, are the major culprit. In the 1970s, John Yudkin first proposed that sugar was the predominant dietary factor that underlies the majority of cardiovascular mortalities, yet his hypothesis was dismissed. However, over the last 25 years substantial scientific evidence has been accumulated to support Yudkin's hypothesis. The objectives of this review are to highlight Yudkin's significant contribution to nutritional science by reviewing his hypothesis and summarizing the recent advances in our understanding of fructose metabolism. The metabolic consequences of fructose metabolism, such as fructose-induced uricemia, insulin resistance, lipoprotein hyperproduction and chronic inflammation, and how they are linked to atherosclerosis as risk factors will be discussed. Finally, the review will explore areas that warrant future research and raise important considerations that we need to evaluate when designing future studies.
Collapse
Affiliation(s)
- Kenneth K.Y. Ting
- Department of Immunology, University of Toronto, Toronto, ON, Canada
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
| |
Collapse
|
10
|
Nhau PT, Gamede M, Sibiya N. COVID-19-Induced Diabetes Mellitus: Comprehensive Cellular and Molecular Mechanistic Insights. PATHOPHYSIOLOGY 2024; 31:197-209. [PMID: 38651404 PMCID: PMC11036300 DOI: 10.3390/pathophysiology31020016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 04/06/2024] [Accepted: 04/07/2024] [Indexed: 04/25/2024] Open
Abstract
Despite evidence demonstrating the risks of developing diabetes mellitus because of SARS-CoV-2, there is, however, insufficient scientific data available to elucidate the relationship between diabetes mellitus and COVID-19. Research indicates that SARS-CoV-2 infection is associated with persistent damage to organ systems due to the systemic inflammatory response. Since COVID-19 is known to induce these conditions, further investigation is necessary to fully understand its long-term effects on human health. Consequently, it is essential to consider the effect of the COVID-19 pandemic when predicting the prevalence of diabetes mellitus in the future, especially since the incidence of diabetes mellitus was already on the rise before the pandemic. Additional research is required to fully comprehend the impact of SARS-CoV-2 infection on glucose tolerance and insulin sensitivity. Therefore, this article delves deeper into the current literature and links the perceived relationship between SARS-CoV-2 and diabetes. In addition, the article highlights the necessity for further research to fully grasp the mechanisms that SARS-CoV-2 utilises to induce new-onset diabetes. Where understanding and consensus are reached, therapeutic interventions to prevent the onset of diabetes could be proposed. Lastly, we propose advocating for the regular screening of diabetes and pre-diabetes, particularly for the high-risk population with a history of COVID-19 infection.
Collapse
Affiliation(s)
- Praise Tatenda Nhau
- Pharmacology Division, Faculty of Pharmacy, Rhodes University, Makhanda 6139, South Africa;
| | - Mlindeli Gamede
- Human Physiology Department, University of Pretoria, Pretoria 0028, South Africa;
| | - Ntethelelo Sibiya
- Pharmacology Division, Faculty of Pharmacy, Rhodes University, Makhanda 6139, South Africa;
| |
Collapse
|
11
|
Bosso M, Haddad D, Al Madhoun A, Al-Mulla F. Targeting the Metabolic Paradigms in Cancer and Diabetes. Biomedicines 2024; 12:211. [PMID: 38255314 PMCID: PMC10813379 DOI: 10.3390/biomedicines12010211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 01/09/2024] [Accepted: 01/11/2024] [Indexed: 01/24/2024] Open
Abstract
Dysregulated metabolic dynamics are evident in both cancer and diabetes, with metabolic alterations representing a facet of the myriad changes observed in these conditions. This review delves into the commonalities in metabolism between cancer and type 2 diabetes (T2D), focusing specifically on the contrasting roles of oxidative phosphorylation (OXPHOS) and glycolysis as primary energy-generating pathways within cells. Building on earlier research, we explore how a shift towards one pathway over the other serves as a foundational aspect in the development of cancer and T2D. Unlike previous reviews, we posit that this shift may occur in seemingly opposing yet complementary directions, akin to the Yin and Yang concept. These metabolic fluctuations reveal an intricate network of underlying defective signaling pathways, orchestrating the pathogenesis and progression of each disease. The Warburg phenomenon, characterized by the prevalence of aerobic glycolysis over minimal to no OXPHOS, emerges as the predominant metabolic phenotype in cancer. Conversely, in T2D, the prevailing metabolic paradigm has traditionally been perceived in terms of discrete irregularities rather than an OXPHOS-to-glycolysis shift. Throughout T2D pathogenesis, OXPHOS remains consistently heightened due to chronic hyperglycemia or hyperinsulinemia. In advanced insulin resistance and T2D, the metabolic landscape becomes more complex, featuring differential tissue-specific alterations that affect OXPHOS. Recent findings suggest that addressing the metabolic imbalance in both cancer and diabetes could offer an effective treatment strategy. Numerous pharmaceutical and nutritional modalities exhibiting therapeutic effects in both conditions ultimately modulate the OXPHOS-glycolysis axis. Noteworthy nutritional adjuncts, such as alpha-lipoic acid, flavonoids, and glutamine, demonstrate the ability to reprogram metabolism, exerting anti-tumor and anti-diabetic effects. Similarly, pharmacological agents like metformin exhibit therapeutic efficacy in both T2D and cancer. This review discusses the molecular mechanisms underlying these metabolic shifts and explores promising therapeutic strategies aimed at reversing the metabolic imbalance in both disease scenarios.
Collapse
Affiliation(s)
- Mira Bosso
- Department of Pathology, Faculty of Medicine, Health Science Center, Kuwait University, Safat 13110, Kuwait
| | - Dania Haddad
- Department of Genetics and Bioinformatics, Dasman Diabetes Institute, Dasman 15462, Kuwait; (D.H.); (A.A.M.)
| | - Ashraf Al Madhoun
- Department of Genetics and Bioinformatics, Dasman Diabetes Institute, Dasman 15462, Kuwait; (D.H.); (A.A.M.)
- Department of Animal and Imaging Core Facilities, Dasman Diabetes Institute, Dasman 15462, Kuwait
| | - Fahd Al-Mulla
- Department of Pathology, Faculty of Medicine, Health Science Center, Kuwait University, Safat 13110, Kuwait
- Department of Genetics and Bioinformatics, Dasman Diabetes Institute, Dasman 15462, Kuwait; (D.H.); (A.A.M.)
| |
Collapse
|
12
|
Pirani H, Soltany A, Hossein Rezaei M, Khodabakhshi Fard A, Nikooie R, Khoramipoor K, Chamari K, Khoramipour K. Lactate-induced autophagy activation: unraveling the therapeutic impact of high-intensity interval training on insulin resistance in type 2 diabetic rats. Sci Rep 2024; 14:1108. [PMID: 38212600 PMCID: PMC10784291 DOI: 10.1038/s41598-023-50589-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 12/21/2023] [Indexed: 01/13/2024] Open
Abstract
Impaired autophagy is a hallmark of diabetes. The current study proposed to investigate if high intensity interval training (HIIT) induced lactate accumulation could stimulate autophagy in type 2 diabetic male rats. 28 male Wistar rats were randomly assigned into four groups: Healthy Control (CO), Diabetes Control (T2D), Exercise (EX), and Diabetes + Exercise (T2D + EX). Diabetes was induced by feeding high-fat diet and administrating single dose of streptozotocin (35 mg/kg). After becoming diabetic, the animals in the exercise groups (EX and T2D + EX) performed an eight-week HIIT (4-10 interval, 80-100% Vmax, 5 days per week). Serum levels of lactate, glucose and insulin as well as the levels of lactate, pyruvate, lactate transporter monocarboxylate transporter 1 (MCT1), phosphorylated mitogen-activated protein kinases (p-MAP 1 and 2), phosphorylated extracellular signal-regulated protein kinases 1 and 2 (p-ERK 1 and 2), mammalian target of rapamycin (p-mTOR), ribosomal protein S6 kinase beta-1 (p-70S6k), p90 ribosomal S6 kinases (p-90RSK), autophagy related 7 (ATG7), Beclin-1, microtubule-associated protein 1A/1B, and 2A/2B -light chain 3 levels (LC3-I), (LC3- II), (LC3I/LC3II) in soleus muscle were measured. Homeostatic Model Assessment for Insulin Resistance (HOMA-IR) and serum glucose was lower in T2D + EX compared to T2D group (P < 0.0001). While serum and soleus muscle levels of lactate was not different between T2D and T2D + Ex, the levels of Pyruvate (P < 0.01), MCT1, p-ERK1/2, p-mTOR, p70S6k, P-90RSK, ATG7, LC3-II, and LC3-II/LC3I ratios were higher in T2D + EX compared to T2D group (P < 0.0001). We concluded that eight weeks of high-intensity interval training could activated ERK/P90SRK while inhibiting mTOR/P70S6K signaling pathway in lactate dependent manner. It means increased autophagy which resulted in improve insulin resistance (IR) and reduce blood glucose.
Collapse
Affiliation(s)
- Hossein Pirani
- Department of Basic Sciences, Chabahar Maritime University, Chabahar, Iran
| | - Afsaneh Soltany
- Department of Biology, Faculty of Science, University of Shiraz, Shiraz, Iran
| | - Maryam Hossein Rezaei
- Department of Exercise Physiology, Faculty of Physical Education and Sport Sciences, Shahid Bahonar University, Kerman, Iran
| | - Adeleh Khodabakhshi Fard
- Department on Nutrition, Faculty of Public Health, Kerman University of Medical Sciences, Kerman, Iran
| | - Rohollah Nikooie
- Department of Exercise Physiology, Faculty of Physical Education and Sport Sciences, Shahid Bahonar University, Kerman, Iran
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Kimya Khoramipoor
- Department of Nursing, Faculty of Nursing and Midwifery, Kurdistan University of Medical Sciences, Kurdistan, Iran
| | - Karim Chamari
- Higher Institute of Sport and Physical Education, ISSEP Ksar Said, Manouba University, Manouba, Tunisia
| | - Kayvan Khoramipour
- Neuroscience Research Center, Institute of Neuropharmacology and Department of Physiology and Pharmacology, Afzalipour School of Medicine, Kerman University of Medical Sciences, Kerman, Iran.
| |
Collapse
|
13
|
Yang N, Ma K, Liu W, Zhang N, Shi Z, Ren J, Xu W, Li Y, Su R, Liang Y, Wang S, Li X. Serum metabolomics probes the molecular mechanism of action of acupuncture on metabolic pathways related to glucose metabolism in patients with polycystic ovary syndrome-related obesity. Biomed Chromatogr 2023; 37:e5710. [PMID: 37593801 DOI: 10.1002/bmc.5710] [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: 04/03/2023] [Revised: 07/03/2023] [Accepted: 07/17/2023] [Indexed: 08/19/2023]
Abstract
Polycystic ovary syndrome (PCOS) is a common endocrine syndrome, and obesity is the most common clinical manifestation. Acupuncture is effective in treating PCOS, but the differences in the biological mechanisms of acupuncture therapy and Western medicine treatment have not been determined. Thus, the purpose of this study was to find glucose metabolism-related pathways in acupuncture treatment and differentiate them from Western medical treatment. Sixty patients with PCOS-related obesity were randomly distributed into three groups: patients receiving (1) acupuncture treatment alone, (2) conventional Western medicine treatment, and (3) acupuncture combined with Western medicine treatment. A targeted metabolomics approach was used to identify small molecules and metabolites related to glucose metabolism in the serum of each group, and ultra-high-performance liquid chromatography-tandem mass spectrometry was used to analyze different metabolic fractions. The results showed acupuncture treatment modulates the activity of citric and succinic acids in the tricarboxylic acid cycle, regulates glycolytic and gluconeogenesis pathways, and improves the levels of sex hormones and energy metabolism. The intervention effects on the metabolic pathways were different between patients receiving combination therapy and patients receiving acupuncture therapy alone, suggesting that the dominant modulatory effect of Western drugs may largely conceal the efficacy of acupuncture intervention.
Collapse
Affiliation(s)
- Nan Yang
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Ke Ma
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Weidong Liu
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Ning Zhang
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Zhouhua Shi
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Jian Ren
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Wanli Xu
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Yuqiu Li
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Riliang Su
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Yanbo Liang
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Shijun Wang
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Xiuyang Li
- Postdoctoral Station, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| |
Collapse
|
14
|
Keijer J, Escoté X, Galmés S, Palou-March A, Serra F, Aldubayan MA, Pigsborg K, Magkos F, Baker EJ, Calder PC, Góralska J, Razny U, Malczewska-Malec M, Suñol D, Galofré M, Rodríguez MA, Canela N, Malcic RG, Bosch M, Favari C, Mena P, Del Rio D, Caimari A, Gutierrez B, Del Bas JM. Omics biomarkers and an approach for their practical implementation to delineate health status for personalized nutrition strategies. Crit Rev Food Sci Nutr 2023; 64:8279-8307. [PMID: 37077157 DOI: 10.1080/10408398.2023.2198605] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/21/2023]
Abstract
Personalized nutrition (PN) has gained much attention as a tool for empowerment of consumers to promote changes in dietary behavior, optimizing health status and preventing diet related diseases. Generalized implementation of PN faces different obstacles, one of the most relevant being metabolic characterization of the individual. Although omics technologies allow for assessment the dynamics of metabolism with unprecedented detail, its translatability as affordable and simple PN protocols is still difficult due to the complexity of metabolic regulation and to different technical and economical constrains. In this work, we propose a conceptual framework that considers the dysregulation of a few overarching processes, namely Carbohydrate metabolism, lipid metabolism, inflammation, oxidative stress and microbiota-derived metabolites, as the basis of the onset of several non-communicable diseases. These processes can be assessed and characterized by specific sets of proteomic, metabolomic and genetic markers that minimize operational constrains and maximize the information obtained at the individual level. Current machine learning and data analysis methodologies allow the development of algorithms to integrate omics and genetic markers. Reduction of dimensionality of variables facilitates the implementation of omics and genetic information in digital tools. This framework is exemplified by presenting the EU-Funded project PREVENTOMICS as a use case.
Collapse
Affiliation(s)
- Jaap Keijer
- Human and Animal Physiology, Wageningen University, Wageningen, the Netherlands
| | - Xavier Escoté
- EURECAT, Centre Tecnològic de Catalunya, Nutrition and Health, Reus, Spain
| | - Sebastià Galmés
- Laboratory of Molecular Biology, Nutrition and Biotechnology (Group of Nutrigenomics, Biomarkers and Risk Evaluation - NuBE), University of the Balearic Islands, Palma, Spain
- Health Research Institute of the Balearic Islands (IdISBa), Palma, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
- Spin-off n.1 of the University of the Balearic Islands, Alimentómica S.L, Palma, Spain
| | - Andreu Palou-March
- Laboratory of Molecular Biology, Nutrition and Biotechnology (Group of Nutrigenomics, Biomarkers and Risk Evaluation - NuBE), University of the Balearic Islands, Palma, Spain
- Health Research Institute of the Balearic Islands (IdISBa), Palma, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
- Spin-off n.1 of the University of the Balearic Islands, Alimentómica S.L, Palma, Spain
| | - Francisca Serra
- Laboratory of Molecular Biology, Nutrition and Biotechnology (Group of Nutrigenomics, Biomarkers and Risk Evaluation - NuBE), University of the Balearic Islands, Palma, Spain
- Health Research Institute of the Balearic Islands (IdISBa), Palma, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
- Spin-off n.1 of the University of the Balearic Islands, Alimentómica S.L, Palma, Spain
| | - Mona Adnan Aldubayan
- Department of Nutrition, Exercise, and Sports, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Nutrition, College of Applied Medical Sciences, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Kristina Pigsborg
- Department of Nutrition, Exercise, and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Faidon Magkos
- Department of Nutrition, Exercise, and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Ella J Baker
- School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Philip C Calder
- School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust and University of Southampton, Southampton, UK
| | - Joanna Góralska
- Department of Clinical Biochemistry, Jagiellonian University Medical College, Krakow, Poland
| | - Urszula Razny
- Department of Clinical Biochemistry, Jagiellonian University Medical College, Krakow, Poland
| | | | - David Suñol
- Digital Health, Eurecat, Centre Tecnològic de Catalunya, Barcelona, Spain
| | - Mar Galofré
- Digital Health, Eurecat, Centre Tecnològic de Catalunya, Barcelona, Spain
| | - Miguel A Rodríguez
- Centre for Omic Sciences (COS), Joint Unit URV-EURECAT, Unique Scientific and Technical Infrastructures (ICTS), Eurecat, Centre Tecnològic de Catalunya, Reus, Spain
| | - Núria Canela
- Centre for Omic Sciences (COS), Joint Unit URV-EURECAT, Unique Scientific and Technical Infrastructures (ICTS), Eurecat, Centre Tecnològic de Catalunya, Reus, Spain
| | - Radu G Malcic
- Health and Biomedicine, LEITAT Technological Centre, Barcelona, Spain
| | - Montserrat Bosch
- Applied Microbiology and Biotechnologies, LEITAT Technological Centre, Terrassa, Spain
| | - Claudia Favari
- Human Nutrition Unit, Department of Food & Drug, University of Parma, Parma, Italy
| | - Pedro Mena
- Human Nutrition Unit, Department of Food & Drug, University of Parma, Parma, Italy
| | - Daniele Del Rio
- Human Nutrition Unit, Department of Food & Drug, University of Parma, Parma, Italy
| | - Antoni Caimari
- Eurecat, Centre Tecnològic de Catalunya, Biotechnology area, Reus, Spain
| | | | - Josep M Del Bas
- Eurecat, Centre Tecnològic de Catalunya, Biotechnology area, Reus, Spain
| |
Collapse
|
15
|
Janssen L, Hopman MTE, Swaans GJA, Allard NAE, Boss M, Lobeek D, Gotthardt M, Schirris TJJ, Blijlevens NMA, Timmers S. Impact of tyrosine kinase inhibitors on glucose control and insulin regulation in patients with chronic myeloid leukemia. Am J Physiol Endocrinol Metab 2023; 324:E209-E216. [PMID: 36696600 DOI: 10.1152/ajpendo.00163.2022] [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] [Indexed: 01/26/2023]
Abstract
Treatment with tyrosine kinase inhibitors (TKIs), especially nilotinib, often results in hyperglycemia, which may further increase cardiovascular disease risk in patients with chronic myeloid leukemia (CML). The mechanism underlying the TKI-induced glucose dysregulation is not clear. TKIs are suggested to affect insulin secretion but also insulin sensitivity of peripheral tissue has been proposed to play a role in the pathogenesis of TKI-induced hyperglycemia. Here, we aimed to assess whether skeletal muscle glucose uptake and insulin responses are altered in nondiabetic patients with CML receiving TKI treatment. After a glycogen-depleted exercise bout, an intravenous glucose bolus (0.3 g/kg body weight) was administered to monitor 2-h glucose tolerance and insulin response in 14 patients with CML receiving nilotinib, 14 patients with CML receiving imatinib, and 14 non-CML age- and gender-matched controls. A dynamic [18F]-FDG PET scan during a hyperinsulinemic-euglycemic clamp was performed in a subgroup of 12 male patients with CML to assess m. quadriceps glucose uptake. We showed that patients with CML treated with nilotinib have an increased insulin response to intravenous glucose administration after muscle glycogen-depleted exercise. Despite the increased insulin response to glucose administration in patients with CML receiving nilotinib, glucose disappearance rates were significantly slower in nilotinib-treated patients when compared with controls in the first 15 min after glucose administration. Although [18F]-FDG uptake in m. quadriceps was not different, patients receiving nilotinib showed a trend toward decreased glucose infusion rates during euglycemic clamping when compared with patients receiving imatinib. Together, these findings indicate disturbed skeletal muscle glucose handling in patients with CML receiving nilotinib therapy.NEW & NOTEWORTHY In this study, we have shown that non-diabetic patients with CML receiving nilotinib therapy show early signs of disturbed skeletal muscle glucose handling, which was not observed in imatinib-treated patients. These observations in nilotinib users may reflect decreased muscle insulin sensitivity, which could serve as a potential target to counteract glycemic dysregulation, and is of clinical importance since these patients have an increased cardiovascular disease risk.
Collapse
Affiliation(s)
- Lando Janssen
- Radboud Institute for Health Sciences, Department of Hematology, Radboud University Medical Center, Nijmegen, The Netherlands
- Radboud Institute for Health Sciences, Department of Physiology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Maria T E Hopman
- Radboud Institute for Health Sciences, Department of Physiology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Greetje J A Swaans
- Radboud Institute for Health Sciences, Department of Physiology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Neeltje A E Allard
- Radboud Institute for Health Sciences, Department of Physiology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Marti Boss
- Radboud Institute for Health Sciences, Department of Medical Imaging, Nuclear Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Daphne Lobeek
- Radboud Institute for Health Sciences, Department of Medical Imaging, Nuclear Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Martin Gotthardt
- Radboud Institute for Health Sciences, Department of Medical Imaging, Nuclear Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Tom J J Schirris
- Radboud Institute for Molecular Life Sciences, Department of Pharmacology and Toxicology, Radboud Centre for Mitochondrial Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Nicole M A Blijlevens
- Radboud Institute for Health Sciences, Department of Hematology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Silvie Timmers
- Human and Animal Physiology, Wageningen University, Wageningen, The Netherlands
| |
Collapse
|
16
|
Kunasegaran T, Balasubramaniam VRMT, Arasoo VJT, Palanisamy UD, Ramadas A. Diet Gut Microbiota Axis in Pregnancy: A Systematic Review of Recent Evidence. Curr Nutr Rep 2023; 12:203-214. [PMID: 36810808 PMCID: PMC9974723 DOI: 10.1007/s13668-023-00453-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/27/2022] [Indexed: 02/23/2023]
Abstract
PURPOSE OF REVIEW Although gut microbiota have been associated with the etiology of some diseases, the influence of foods on gut microbiota, especially among pregnant women, remains unclear. Hence, a systematic review was performed to investigate the association between diet and gut microbiota and their influence on metabolic health in pregnant women. RECENT FINDINGS We performed the systematic review using the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) 2020 protocol to investigate the association between diet and gut microbiota and their influence on metabolic role in pregnant women. Five databases were searched for relevant peer-reviewed articles published in English since 2011. Two-staged screening of 659 retrieved records resulted in the inclusion of 10 studies. The collated findings suggested associations between nutrient intakes and four key microbes: Collinsella, Lachnospira, Sutterella, Faecalibacterium, and the Firmicutes/Bacteroidetes ratio in pregnant women. Dietary intakes in pregnancy were found to modify the gut microbiota and positively influence the cell metabolism in pregnant women. This review, however, emphasizes the importance of conducting well-designed prospective cohorts to investigate the role of changes in dietary intakes within the pregnancy and the influence of such changes on gut microbiota.
Collapse
Affiliation(s)
- Thubasni Kunasegaran
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, 47500 Bandar Sunway, Malaysia
| | | | | | - Uma Devi Palanisamy
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, 47500 Bandar Sunway, Malaysia
| | - Amutha Ramadas
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, 47500 Bandar Sunway, Malaysia
| |
Collapse
|
17
|
Stacpoole PW, McCall CE. The pyruvate dehydrogenase complex: Life's essential, vulnerable and druggable energy homeostat. Mitochondrion 2023; 70:59-102. [PMID: 36863425 DOI: 10.1016/j.mito.2023.02.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 01/30/2023] [Accepted: 02/13/2023] [Indexed: 03/04/2023]
Abstract
Found in all organisms, pyruvate dehydrogenase complexes (PDC) are the keystones of prokaryotic and eukaryotic energy metabolism. In eukaryotic organisms these multi-component megacomplexes provide a crucial mechanistic link between cytoplasmic glycolysis and the mitochondrial tricarboxylic acid (TCA) cycle. As a consequence, PDCs also influence the metabolism of branched chain amino acids, lipids and, ultimately, oxidative phosphorylation (OXPHOS). PDC activity is an essential determinant of the metabolic and bioenergetic flexibility of metazoan organisms in adapting to changes in development, nutrient availability and various stresses that challenge maintenance of homeostasis. This canonical role of the PDC has been extensively probed over the past decades by multidisciplinary investigations into its causal association with diverse physiological and pathological conditions, the latter making the PDC an increasingly viable therapeutic target. Here we review the biology of the remarkable PDC and its emerging importance in the pathobiology and treatment of diverse congenital and acquired disorders of metabolic integration.
Collapse
Affiliation(s)
- Peter W Stacpoole
- Department of Medicine (Division of Endocrinology, Metabolism and Diabetes), and Department of Biochemistry and Molecular Biology, University of Florida, College of Medicine, Gainesville, FL, United States.
| | - Charles E McCall
- Department of Internal Medicine and Translational Sciences, and Department of Microbiology and Immunology, Wake Forest University School of Medicine, Winston-Salem, NC, United States
| |
Collapse
|
18
|
Shen HC, Chen ZQ, Liu XC, Guan JF, Xie DZ, Li YY, Xu C. Sodium oxamate reduces lactate production to improve the glucose homeostasis of Micropterus salmoides fed high-carbohydrate diets. Am J Physiol Regul Integr Comp Physiol 2023; 324:R227-R241. [PMID: 36572554 DOI: 10.1152/ajpregu.00226.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The study was performed to evaluate the effects of the reduced lactate production by sodium oxamate (SO) on growth performance, lactate and glucose and lipid metabolism, and glucose tolerance of Micropterus salmoides fed high-carbohydrate (CHO) diets. In in vitro study, primary hepatocytes were incubated for 48 h in a control medium (5.5 mM glucose), a high-glucose medium (25 mM glucose, HG), or a SO-containing high-glucose medium (25 mM glucose + 50 mM SO, HG-SO). Results indicated lactate and triglyceride (TG) levels, and lactate dehydrogenase a (LDH-a) expression in the HG-SO group were remarkably lower than those of the HG group. In in vivo study, M. salmoides (5.23 ± 0.03 g) were fed four diets containing a control diet (10% CHO, C) and three SO contents [0 (HC), 100 (HC-SO1), and 200 (HC-SO2) mg·kg-1, respectively] of high-CHO diets (20% CHO) for 11 wk. High-CHO diets significantly reduced weight gain rate (WGR), specific growth rate (SGR), p-AMPK-to-t-AMPK ratio, and expression of insulin receptor substrate 1 (IRS1), insulin-like growth factor I (IGF-I), insulin-like growth factor I receptor (IGF-IR), fructose-1,6-biphosphatase (FBPase), peroxisome proliferator-activated receptor α (PPARα), and carnitine palmitoyl transferase 1α (CPT1α) compared with the C group, whereas the opposite was true for plasma levels of glucose, TG, lactate, tissue glycogen, and lipid contents, and expression of LDH-a, monocarboxylate transporter 1 and 4 (MCT1 and MCT4), insulin, glucokinase (GK), pyruvate dehydrogenase E1 subunit (PDH), sterol-regulatory element-binding protein 1 (SREBP1), fatty acid synthase (FAS). The HC-SO2 diets remarkably increased WGR, SGR, p-AMPK-to-t-AMPK ratio, and expression of IRS1, IGF-I, IGF-IR, GK, PDHα, PDHβ, FAS, acetyl-CoA carboxylase 1 (ACC1), PPARα, and CPT1α compared with the HC group. Besides, HC-SO2 diets also enhanced glucose tolerance of fish after a glucose loading. Overall, the reduced lactate production by SO benefits growth performance and glucose homeostasis of high-CHO-fed M. salmoides through the enhancement of glycolysis, lipogenesis, and fatty acid β-oxidation coupled with the suppression of glycogenesis and gluconeogenesis.
Collapse
Affiliation(s)
- Hui-Chao Shen
- College of Marine Sciences, South China Agricultural University, Guangzhou, China.,University Joint Laboratory of Guangdong Province, Hong Kong and Macao Region on Marine Bioresource Conservation and Exploitation, Guangzhou, China
| | - Zhi-Qiang Chen
- College of Marine Sciences, South China Agricultural University, Guangzhou, China
| | - Xiao-Cheng Liu
- College of Marine Sciences, South China Agricultural University, Guangzhou, China
| | - Jun-Feng Guan
- College of Marine Sciences, South China Agricultural University, Guangzhou, China.,University Joint Laboratory of Guangdong Province, Hong Kong and Macao Region on Marine Bioresource Conservation and Exploitation, Guangzhou, China
| | - Di-Zhi Xie
- College of Marine Sciences, South China Agricultural University, Guangzhou, China.,University Joint Laboratory of Guangdong Province, Hong Kong and Macao Region on Marine Bioresource Conservation and Exploitation, Guangzhou, China
| | - Yuan-You Li
- College of Marine Sciences, South China Agricultural University, Guangzhou, China.,University Joint Laboratory of Guangdong Province, Hong Kong and Macao Region on Marine Bioresource Conservation and Exploitation, Guangzhou, China
| | - Chao Xu
- College of Marine Sciences, South China Agricultural University, Guangzhou, China.,University Joint Laboratory of Guangdong Province, Hong Kong and Macao Region on Marine Bioresource Conservation and Exploitation, Guangzhou, China
| |
Collapse
|
19
|
Le S, Schumann M, Lei SM, Yao W, Cheng S. Exercise precision medicine for type 2 diabetics: Targeted benefit or risk? SPORTS MEDICINE AND HEALTH SCIENCE 2023. [DOI: 10.1016/j.smhs.2023.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/22/2023] Open
|
20
|
Kuretu A, Arineitwe C, Mothibe M, Ngubane P, Khathi A, Sibiya N. Drug-induced mitochondrial toxicity: Risks of developing glucose handling impairments. Front Endocrinol (Lausanne) 2023; 14:1123928. [PMID: 36860368 PMCID: PMC9969099 DOI: 10.3389/fendo.2023.1123928] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 01/31/2023] [Indexed: 02/15/2023] Open
Abstract
Mitochondrial impairment has been associated with the development of insulin resistance, the hallmark of type 2 diabetes mellitus (T2DM). However, the relationship between mitochondrial impairment and insulin resistance is not fully elucidated due to insufficient evidence to support the hypothesis. Insulin resistance and insulin deficiency are both characterised by excessive production of reactive oxygen species and mitochondrial coupling. Compelling evidence states that improving the function of the mitochondria may provide a positive therapeutic tool for improving insulin sensitivity. There has been a rapid increase in reports of the toxic effects of drugs and pollutants on the mitochondria in recent decades, interestingly correlating with an increase in insulin resistance prevalence. A variety of drug classes have been reported to potentially induce toxicity in the mitochondria leading to skeletal muscle, liver, central nervous system, and kidney injury. With the increase in diabetes prevalence and mitochondrial toxicity, it is therefore imperative to understand how mitochondrial toxicological agents can potentially compromise insulin sensitivity. This review article aims to explore and summarise the correlation between potential mitochondrial dysfunction caused by selected pharmacological agents and its effect on insulin signalling and glucose handling. Additionally, this review highlights the necessity for further studies aimed to understand drug-induced mitochondrial toxicity and the development of insulin resistance.
Collapse
Affiliation(s)
- Auxiliare Kuretu
- Pharmacology Division, Faculty of Pharmacy, Rhodes University, Makhanda, South Africa
| | - Charles Arineitwe
- Pharmacology Division, Faculty of Pharmacy, Rhodes University, Makhanda, South Africa
| | - Mamosheledi Mothibe
- Pharmacology Division, Faculty of Pharmacy, Rhodes University, Makhanda, South Africa
| | - Phikelelani Ngubane
- School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Andile Khathi
- School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Ntethelelo Sibiya
- Pharmacology Division, Faculty of Pharmacy, Rhodes University, Makhanda, South Africa
- *Correspondence: Ntethelelo Sibiya,
| |
Collapse
|
21
|
Calderón-DuPont D, Torre-Villalvazo I, Díaz-Villaseñor A. Is insulin resistance tissue-dependent and substrate-specific? The role of white adipose tissue and skeletal muscle. Biochimie 2023; 204:48-68. [PMID: 36099940 DOI: 10.1016/j.biochi.2022.08.021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 08/19/2022] [Accepted: 08/31/2022] [Indexed: 01/12/2023]
Abstract
Insulin resistance (IR) refers to a reduction in the ability of insulin to exert its metabolic effects in organs such as adipose tissue (AT) and skeletal muscle (SM), leading to chronic diseases such as type 2 diabetes, hepatic steatosis, and cardiovascular diseases. Obesity is the main cause of IR, however not all subjects with obesity develop clinical insulin resistance, and not all clinically insulin-resistant people have obesity. Recent evidence implies that IR onset is tissue-dependent (AT or SM) and/or substrate-specific (glucometabolic or lipometabolic). Therefore, the aims of the present review are 1) to describe the glucometabolic and lipometabolic activities of insulin in AT and SM in the maintenance of whole-body metabolic homeostasis, 2) to discuss the pathophysiology of substrate-specific IR in AT and SM, and 3) to highlight novel validated tests to assess tissue and substrate-specific IR that are easy to perform in clinical practice. In AT, glucometabolic IR reduces glucose availability for glycerol and fatty acid synthesis, thus decreasing the esterification and synthesis of signaling bioactive lipids. Lipometabolic IR in AT impairs the antilipolytic effect of insulin and lipogenesis, leading to an increase in circulating FFAs and generating lipotoxicity in peripheral tissues. In SM, glucometabolic IR reduces glucose uptake, whereas lipometabolic IR impairs mitochondrial lipid oxidation, increasing oxidative stress and inflammation, all of which lead to metabolic inflexibility. Understanding tissue-dependent and substrate-specific IR is of paramount importance for early detection before clinical manifestations and for the development of more specific treatments or direct interventions to prevent chronic life-threatening diseases.
Collapse
Affiliation(s)
- Diana Calderón-DuPont
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), Mexico City, 04510, Mexico; Doctorado en Ciencias Biomédicas, Universidad Nacional Autónoma de México (UNAM), Mexico City, 04510, Mexico
| | - Ivan Torre-Villalvazo
- Departamento de Fisiología de la Nutrición, Instituto Nacional en Ciencias Médicas y Nutricíon Salvador Zubirán, Mexico City, 14000, Mexico
| | - Andrea Díaz-Villaseñor
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), Mexico City, 04510, Mexico.
| |
Collapse
|
22
|
Das S, Taylor K, Kozubek J, Sardell J, Gardner S. Genetic risk factors for ME/CFS identified using combinatorial analysis. J Transl Med 2022; 20:598. [PMID: 36517845 PMCID: PMC9749644 DOI: 10.1186/s12967-022-03815-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Accepted: 12/07/2022] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) is a debilitating chronic disease that lacks known pathogenesis, distinctive diagnostic criteria, and effective treatment options. Understanding the genetic (and other) risk factors associated with the disease would begin to help to alleviate some of these issues for patients. METHODS We applied both GWAS and the PrecisionLife combinatorial analytics platform to analyze ME/CFS cohorts from UK Biobank, including the Pain Questionnaire cohort, in a case-control design with 1000 cycles of fully random permutation. Results from this study were supported by a series of replication and cohort comparison experiments, including use of disjoint Verbal Interview CFS, post-viral fatigue syndrome and fibromyalgia cohorts also derived from UK Biobank, and compared results for overlap and reproducibility. RESULTS Combinatorial analysis revealed 199 SNPs mapping to 14 genes that were significantly associated with 91% of the cases in the ME/CFS population. These SNPs were found to stratify by shared cases into 15 clusters (communities) made up of 84 high-order combinations of between 3 and 5 SNPs. p-values for these communities range from 2.3 × 10-10 to 1.6 × 10-72. Many of the genes identified are linked to the key cellular mechanisms hypothesized to underpin ME/CFS, including vulnerabilities to stress and/or infection, mitochondrial dysfunction, sleep disturbance and autoimmune development. We identified 3 of the critical SNPs replicated in the post-viral fatigue syndrome cohort and 2 SNPs replicated in the fibromyalgia cohort. We also noted similarities with genes associated with multiple sclerosis and long COVID, which share some symptoms and potentially a viral infection trigger with ME/CFS. CONCLUSIONS This study provides the first detailed genetic insights into the pathophysiological mechanisms underpinning ME/CFS and offers new approaches for better diagnosis and treatment of patients.
Collapse
Affiliation(s)
- Sayoni Das
- PrecisionLife Ltd, Long Hanborough, Oxford, UK
| | | | | | | | | |
Collapse
|
23
|
Eckstein ML, Erlmann MP, Aberer F, Haupt S, Zimmermann P, Wachsmuth NB, Schierbauer J, Zimmer RT, Herz D, Obermayer-Pietsch B, Moser O. Glucose and Fructose Supplementation and Their Acute Effects on Anaerobic Endurance and Resistance Exercise Performance in Healthy Individuals: A Double-Blind Randomized Placebo-Controlled Crossover Trial. Nutrients 2022; 14:nu14235128. [PMID: 36501158 PMCID: PMC9736485 DOI: 10.3390/nu14235128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Revised: 11/25/2022] [Accepted: 11/29/2022] [Indexed: 12/05/2022] Open
Abstract
BACKGROUND The effects of glucose, fructose and a combination of these on physical performance have been subject of investigation, resulting in diverse findings. OBJECTIVE The aim of this study was to investigate how an individualized amount of glucose, fructose, and a combination of these compared to placebo (sucralose) alter endurance performance on a cycle ergometer, lower and upper body resistance exercise performance at individualized thresholds in healthy young individuals. METHODS A total of 16 healthy adults (9 females) with an age of 23.8 ± 1.6 years and a BMI of 22.6 ± 1.8 kg/m2 (body mass (BM) 70.9 ± 10.8 kg, height 1.76 ± 0.08 m) participated in this study. During the screening visit, the lactate turn point 2 (LTP2) was defined and the weights for chest-press and leg-press were determined. Furthermore, 30 min prior to each exercise session, participants received either 1 g/kg BM of glucose (Glu), 1 g/kg BM of fructose (Fru), 0.5 g/kg BM of glucose/fructose (GluFru) (each), or 0.2 g sucralose (placebo), respectively, which were dissolved in 300 mL of water. All exercises were performed until volitional exhaustion. Time until exhaustion (TTE) and cardio-pulmonary variables were determined for all cycling visits; during resistance exercise, repetitions until muscular failure were counted and time was measured. During all visits, capillary blood glucose and blood lactate concentrations as well as venous insulin levels were measured. RESULTS TTE in cycling was 449 ± 163 s (s) (Glu), 443 ± 156 s (Fru), 429 ± 160 s (GluFru) and 466 ± 162 s (Pla) (p = 0.48). TTE during chest-press sessions was 180 ± 95 s (Glu), 180 ± 92 s (Fru), 172 ± 78 s (GluFru) and 162 ± 66 s (Pla) (p = 0.25), respectively. CONCLUSIONS Pre-exercise supplementation of Glu, Fru and a combination of these did not have an ergogenic effect on high-intensity anaerobic endurance performance and on upper and lower body moderate resistance exercise in comparison to placebo.
Collapse
Affiliation(s)
- Max L. Eckstein
- BaySpo—Bayreuth Center of Sport Science, Division of Exercise Physiology and Metabolism, University of Bayreuth, 95440 Bayreuth, Germany
| | - Maximilian P. Erlmann
- BaySpo—Bayreuth Center of Sport Science, Division of Exercise Physiology and Metabolism, University of Bayreuth, 95440 Bayreuth, Germany
| | - Felix Aberer
- Department of Endocrinology and Diabetology, Medical University of Graz, 8036 Graz, Austria
| | - Sandra Haupt
- BaySpo—Bayreuth Center of Sport Science, Division of Exercise Physiology and Metabolism, University of Bayreuth, 95440 Bayreuth, Germany
| | - Paul Zimmermann
- BaySpo—Bayreuth Center of Sport Science, Division of Exercise Physiology and Metabolism, University of Bayreuth, 95440 Bayreuth, Germany
- Department of Cardiology, Klinikum Bamberg, 96049 Bamberg, Germany
| | - Nadine B. Wachsmuth
- BaySpo—Bayreuth Center of Sport Science, Division of Exercise Physiology and Metabolism, University of Bayreuth, 95440 Bayreuth, Germany
| | - Janis Schierbauer
- BaySpo—Bayreuth Center of Sport Science, Division of Exercise Physiology and Metabolism, University of Bayreuth, 95440 Bayreuth, Germany
| | - Rebecca T. Zimmer
- BaySpo—Bayreuth Center of Sport Science, Division of Exercise Physiology and Metabolism, University of Bayreuth, 95440 Bayreuth, Germany
| | - Daniel Herz
- BaySpo—Bayreuth Center of Sport Science, Division of Exercise Physiology and Metabolism, University of Bayreuth, 95440 Bayreuth, Germany
| | - Barbara Obermayer-Pietsch
- Department of Internal Medicine, Division of Endocrinology and Diabetology, Endocrinology Lab Platform, Medical University of Graz, 8036 Graz, Austria
| | - Othmar Moser
- BaySpo—Bayreuth Center of Sport Science, Division of Exercise Physiology and Metabolism, University of Bayreuth, 95440 Bayreuth, Germany
- Cardiovascular Diabetology Research Group, Division of Endocrinology and Diabetology, Department of Internal Medicine, Medical University of Graz, 8036 Graz, Austria
- Correspondence: ; Tel.: +49-(0)9-2155-3465
| |
Collapse
|
24
|
Zhang Y, Shen T, Wang S. Progression from prediabetes to type 2 diabetes mellitus induced by overnutrition. Hormones (Athens) 2022; 21:591-597. [PMID: 36197636 DOI: 10.1007/s42000-022-00399-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 09/19/2022] [Indexed: 12/31/2022]
Abstract
Prediabetes has developed into a global pandemic, its prevalence increasing year by year. Although lifestyle changes are advocated as the basis for prediabetes treatment, some patients fail to choose or adhere to appropriate interventions. The basis for selecting an appropriate intervention is determining the stage and cause of the disease. In this review, we aimed to examine the various types and disease processes of prediabetes caused by overnutrition, the present review supporting the hypothesis that overnutrition-induced hyperinsulinemia precedes insulin resistance (IR) and independently causes β-cell dysfunction. Tissue insulin resistance is the main feature of prediabetes with the crosstalk between tissues promoting the formation of systemic insulin resistance. Finally, both β-cell dysfunction induced by hyperinsulinemia or IR and reduced β-cell mass can lead to abnormal insulin secretion and contribute to development of type 2 diabetes mellitus (T2DM). Hence, overnutrition can cause multiple prediabetes phenotypes resulting in development of T2DM through different trajectories. Future diagnosis and treatment should therefore more carefully consider the disease phenotype and stage of development in patients with prediabetes to reduce the incidence of T2DM.
Collapse
Affiliation(s)
- Yuli Zhang
- School of Physical Education & Sports Science, South China Normal University, No.55, West of Zhongshan Ave., Tianhe District, Guangzhou City, 510006, Guangdong Province, China
| | - Tuming Shen
- School of Physical Education & Sports Science, South China Normal University, No.55, West of Zhongshan Ave., Tianhe District, Guangzhou City, 510006, Guangdong Province, China
| | - Songtao Wang
- School of Physical Education & Sports Science, South China Normal University, No.55, West of Zhongshan Ave., Tianhe District, Guangzhou City, 510006, Guangdong Province, China.
| |
Collapse
|
25
|
Memon B, Elsayed AK, Bettahi I, Suleiman N, Younis I, Wehedy E, Abou-Samra AB, Abdelalim EM. iPSCs derived from insulin resistant offspring of type 2 diabetic patients show increased oxidative stress and lactate secretion. Stem Cell Res Ther 2022; 13:428. [PMID: 35987697 PMCID: PMC9392338 DOI: 10.1186/s13287-022-03123-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 08/05/2022] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
The genetic factors associated with insulin resistance (IR) are not well understood. Clinical studies on first-degree relatives of type 2 diabetic (T2D) patients, which have the highest genetic predisposition to T2D, have given insights into the role of IR in T2D pathogenesis. Induced pluripotent stem cells (iPSCs) are excellent tools for disease modeling as they can retain the genetic imprint of the disease. Therefore, in this study, we aimed to investigate the genetic perturbations associated with insulin resistance (IR) in the offspring of T2D parents using patient-specific iPSCs.
Methods
We generated iPSCs from IR individuals (IR-iPSCs) that were offspring of T2D parents as well as from insulin-sensitive (IS-iPSCs) individuals. We then performed transcriptomics to identify key dysregulated gene networks in the IR-iPSCs in comparison to IS-iPSCs and functionally validated them.
Results
Transcriptomics on IR-iPSCs revealed dysregulated gene networks and biological processes indicating that they carry the genetic defects associated with IR that may lead to T2D. The IR-iPSCs had increased lactate secretion and a higher phosphorylation of AKT upon stimulation with insulin. IR-iPSCs have increased cellular oxidative stress indicated by a high production of reactive oxygen species and higher susceptibility to H2O2 -induced apoptosis.
Conclusions
IR-iPSCs generated from offspring of diabetic patients confirm that oxidative stress and increased lactate secretion, associated with IR, are inherited in this population, and may place them at a high risk of T2D. Overall, our IR-iPSC model can be employed for T2D modeling and drug screening studies that target genetic perturbations associated with IR in individuals with a high risk for T2D.
Collapse
|
26
|
Scheffschick A, Babel J, Sperling S, Nerusch J, Herzog N, Seehofer D, Damm G. Primary-like Human Hepatocytes Genetically Engineered to Obtain Proliferation Competence as a Capable Application for Energy Metabolism Experiments in In Vitro Oncologic Liver Models. BIOLOGY 2022; 11:biology11081195. [PMID: 36009822 PMCID: PMC9405410 DOI: 10.3390/biology11081195] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 07/21/2022] [Accepted: 08/03/2022] [Indexed: 11/17/2022]
Abstract
Simple Summary Fatty liver disease is an increasing health concern in Westernized countries. A fatty liver can lead to hepatocellular carcinoma (HCC), a type of liver cancer arising from hepatocytes, the major cells of the liver. How HCC may develop from the fatty liver is not known, and good cellular systems to investigate this are lacking. Recently, hepatocytes that can multiply continuously have been generated and suggested for hepatocyte research. In this study, we compared these continuously multiplying human hepatocytes to normal human hepatocytes and liver cancer cells, both within the state of fatty liver or not. We identified that these multiplying hepatocytes displayed many similarities to the liver cancer cells in terms of energy metabolism and concluded that these hepatocytes could be a pre-cancer model for liver cancer research and would be a valuable tool for HCC research. Abstract Non-alcoholic fatty liver disease (NAFLD), characterized by lipid accumulation in the liver, is the most common cause of liver diseases in Western countries. NAFLD is a major risk factor for developing hepatocellular carcinoma (HCC); however, in vitro evaluation of hepatic cancerogenesis fails due to a lack of liver models displaying a proliferation of hepatocytes. Originally designed to overcome primary human hepatocyte (PHH) shortages, upcyte hepatocytes were engineered to obtain continuous proliferation and, therefore, could be a suitable tool for HCC research. We generated upcyte hepatocytes, termed HepaFH3 cells, and compared their metabolic characteristics to HepG2 hepatoma cells and PHHs isolated from resected livers. For displaying NAFLD-related HCCs, we induced steatosis in all liver models. Lipid accumulation, lipotoxicity and energy metabolism were characterized using biochemical assays and Western blot analysis. We showed that proliferating HepaFH3 cells resemble HepG2, both showing a higher glucose uptake rate, lactate levels and metabolic rate compared to PHHs. Confluent HepaFH3 cells displayed some similarities to PHHs, including higher levels of the transaminases AST and ALT compared to proliferating HepaFH3 cells. We recommend proliferating HepaFH3 cells as a pre-malignant cellular model for HCC research, while confluent HepaFH3 cells could serve as PHH surrogates for energy metabolism studies.
Collapse
Affiliation(s)
- Andrea Scheffschick
- Department of Hepatobiliary Surgery and Visceral Transplantation, University Hospital, Leipzig University, 04103 Leipzig, Germany
- Saxonian Incubator for Clinical Translation (SIKT), Leipzig University, 04103 Leipzig, Germany
| | - Jonas Babel
- Department of Hepatobiliary Surgery and Visceral Transplantation, University Hospital, Leipzig University, 04103 Leipzig, Germany
| | - Sebastian Sperling
- Department of General, Visceral and Transplantation Surgery, Charité University Medicine Berlin, 13353 Berlin, Germany
| | - Julia Nerusch
- Department of Hepatobiliary Surgery and Visceral Transplantation, University Hospital, Leipzig University, 04103 Leipzig, Germany
- Saxonian Incubator for Clinical Translation (SIKT), Leipzig University, 04103 Leipzig, Germany
| | - Natalie Herzog
- Faculty of Science, Brandenburg University of Technology Cottbus-Senftenberg, 01968 Senftenberg, Germany
| | - Daniel Seehofer
- Department of Hepatobiliary Surgery and Visceral Transplantation, University Hospital, Leipzig University, 04103 Leipzig, Germany
- Department of General, Visceral and Transplantation Surgery, Charité University Medicine Berlin, 13353 Berlin, Germany
| | - Georg Damm
- Department of Hepatobiliary Surgery and Visceral Transplantation, University Hospital, Leipzig University, 04103 Leipzig, Germany
- Saxonian Incubator for Clinical Translation (SIKT), Leipzig University, 04103 Leipzig, Germany
- Department of General, Visceral and Transplantation Surgery, Charité University Medicine Berlin, 13353 Berlin, Germany
- Correspondence: ; Tel.: +49-341-97-39656
| |
Collapse
|
27
|
Bakar MHA, Shahril NSN, Khalid MSFM, Mohammad S, Shariff KA, Karunakaran T, Salleh RM, Rosdi MN. Celastrol alleviates high-fat diet-induced obesity via enhanced muscle glucose utilization and mitochondrial oxidative metabolism-mediated upregulation of pyruvate dehydrogenase complex. Toxicol Appl Pharmacol 2022; 449:116099. [DOI: 10.1016/j.taap.2022.116099] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 05/23/2022] [Accepted: 06/01/2022] [Indexed: 12/25/2022]
|
28
|
Giussani M, Lieti G, Orlando A, Parati G, Genovesi S. Fructose Intake, Hypertension and Cardiometabolic Risk Factors in Children and Adolescents: From Pathophysiology to Clinical Aspects. A Narrative Review. Front Med (Lausanne) 2022; 9:792949. [PMID: 35492316 PMCID: PMC9039289 DOI: 10.3389/fmed.2022.792949] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 03/21/2022] [Indexed: 01/09/2023] Open
Abstract
Arterial hypertension, dyslipidemia, alterations in glucose metabolism and fatty liver, either alone or in association, are frequently observed in obese children and may seriously jeopardize their health. For obesity to develop, an excessive intake of energy-bearing macronutrients is required; however, ample evidence suggests that fructose may promote the development of obesity and/or metabolic alterations, independently of its energy intake. Fructose consumption is particularly high among children, because they do not have the perception, and more importantly, neither do their parents, that high fructose intake is potentially dangerous. In fact, while this sugar is erroneously viewed favorably as a natural nutrient, its excessive intake can actually cause adverse cardio-metabolic alterations. Fructose induces the release of pro-inflammatory cytokines, and reduces the production of anti-atherosclerotic cytokines, such as adiponectin. Furthermore, by interacting with hunger and satiety control systems, particularly by inducing leptin resistance, it leads to increased caloric intake. Fructose, directly or through its metabolites, promotes the development of obesity, arterial hypertension, dyslipidemia, glucose intolerance and fatty liver. This review aims to highlight the mechanisms by which the early and excessive consumption of fructose may contribute to the development of a variety of cardiometabolic risk factors in children, thus representing a potential danger to their health. It will also describe the main clinical trials performed in children and adolescents that have evaluated the clinical effects of excessive intake of fructose-containing drinks and food, with particular attention to the effects on blood pressure. Finally, we will discuss the effectiveness of measures that can be taken to reduce the intake of this sugar.
Collapse
Affiliation(s)
- Marco Giussani
- Cardiologic Unit, Istituto Auxologico Italiano, Istituto Ricovero Cura Carattere Scientifico (IRCCS), Milan, Italy
| | - Giulia Lieti
- School of Medicine and Surgery, University of Milano-Bicocca, Milan, Italy
| | - Antonina Orlando
- Cardiologic Unit, Istituto Auxologico Italiano, Istituto Ricovero Cura Carattere Scientifico (IRCCS), Milan, Italy
| | - Gianfranco Parati
- Cardiologic Unit, Istituto Auxologico Italiano, Istituto Ricovero Cura Carattere Scientifico (IRCCS), Milan, Italy.,School of Medicine and Surgery, University of Milano-Bicocca, Milan, Italy
| | - Simonetta Genovesi
- Cardiologic Unit, Istituto Auxologico Italiano, Istituto Ricovero Cura Carattere Scientifico (IRCCS), Milan, Italy.,School of Medicine and Surgery, University of Milano-Bicocca, Milan, Italy
| |
Collapse
|
29
|
Sigala DM, Hieronimus B, Medici V, Lee V, Nunez MV, Bremer AA, Cox CL, Price CA, Benyam Y, Abdelhafez Y, McGahan JP, Keim NL, Goran MI, Pacini G, Tura A, Sirlin CB, Chaudhari AJ, Havel PJ, Stanhope KL. The Dose-Response Effects of Consuming High Fructose Corn Syrup-Sweetened Beverages on Hepatic Lipid Content and Insulin Sensitivity in Young Adults. Nutrients 2022; 14:1648. [PMID: 35458210 PMCID: PMC9030734 DOI: 10.3390/nu14081648] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 04/08/2022] [Accepted: 04/10/2022] [Indexed: 01/27/2023] Open
Abstract
Increased hepatic lipid content and decreased insulin sensitivity have critical roles in the development of cardiometabolic diseases. Therefore, our objective was to investigate the dose-response effects of consuming high fructose corn syrup (HFCS)-sweetened beverages for two weeks on hepatic lipid content and insulin sensitivity in young (18-40 years) adults (BMI 18-35 kg/m2). In a parallel, double-blinded study, participants consumed three beverages/day providing 0% (aspartame: n = 23), 10% (n = 18), 17.5% (n = 16), or 25% (n = 28) daily energy requirements from HFCS. Magnetic resonance imaging for hepatic lipid content and oral glucose tolerance tests (OGTT) were conducted during 3.5-day inpatient visits at baseline and again at the end of a 15-day intervention. During the 12 intervening outpatient days participants consumed their usual diets with their assigned beverages. Significant linear dose-response effects were observed for increases of hepatic lipid content (p = 0.015) and glucose and insulin AUCs during OGTT (both p = 0.0004), and for decreases in the Matsuda (p = 0.0087) and Predicted M (p = 0.0027) indices of insulin sensitivity. These dose-response effects strengthen the mechanistic evidence implicating consumption of HFCS-sweetened beverages as a contributor to the metabolic dysregulation that increases risk for nonalcoholic fatty liver disease and type 2 diabetes.
Collapse
Affiliation(s)
- Desiree M. Sigala
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California—Davis, Sacramento, CA 95616, USA; (D.M.S.); (B.H.); (V.L.); (M.V.N.); (C.A.P.); (Y.B.); (P.J.H.)
- Department of Nutrition, University of California—Davis, Sacramento, CA 95616, USA
| | - Bettina Hieronimus
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California—Davis, Sacramento, CA 95616, USA; (D.M.S.); (B.H.); (V.L.); (M.V.N.); (C.A.P.); (Y.B.); (P.J.H.)
- Department of Nutrition, University of California—Davis, Sacramento, CA 95616, USA
- Institute for Physiology and Biochemistry of Nutrition, Max Rubner-Institut, 76131 Karlsruhe, Germany
| | - Valentina Medici
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of California—Davis, Sacramento, CA 95817, USA;
| | - Vivien Lee
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California—Davis, Sacramento, CA 95616, USA; (D.M.S.); (B.H.); (V.L.); (M.V.N.); (C.A.P.); (Y.B.); (P.J.H.)
- Department of Nutrition, University of California—Davis, Sacramento, CA 95616, USA
| | - Marinelle V. Nunez
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California—Davis, Sacramento, CA 95616, USA; (D.M.S.); (B.H.); (V.L.); (M.V.N.); (C.A.P.); (Y.B.); (P.J.H.)
- Department of Nutrition, University of California—Davis, Sacramento, CA 95616, USA
| | - Andrew A. Bremer
- Department of Pediatrics, School of Medicine, University of California—Davis, Sacramento, CA 95817, USA;
| | - Chad L. Cox
- Department of Chemistry, California State University, Sacramento, CA 95819, USA;
- Department of Family and Consumer Sciences, California State University, Sacramento, CA 95819, USA
| | - Candice A. Price
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California—Davis, Sacramento, CA 95616, USA; (D.M.S.); (B.H.); (V.L.); (M.V.N.); (C.A.P.); (Y.B.); (P.J.H.)
- Department of Nutrition, University of California—Davis, Sacramento, CA 95616, USA
| | - Yanet Benyam
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California—Davis, Sacramento, CA 95616, USA; (D.M.S.); (B.H.); (V.L.); (M.V.N.); (C.A.P.); (Y.B.); (P.J.H.)
- Department of Nutrition, University of California—Davis, Sacramento, CA 95616, USA
| | - Yasser Abdelhafez
- Department of Radiology, School of Medicine, University of California—Davis, Sacramento, CA 95817, USA; (Y.A.); (J.P.M.); (A.J.C.)
| | - John P. McGahan
- Department of Radiology, School of Medicine, University of California—Davis, Sacramento, CA 95817, USA; (Y.A.); (J.P.M.); (A.J.C.)
| | - Nancy L. Keim
- Western Human Nutrition Research Center, United States Department of Agriculture, Davis, CA 95616, USA;
| | - Michael I. Goran
- The Saban Research Institute, Children’s Hospital Los Angeles, Los Angeles, CA 90027, USA;
| | - Giovanni Pacini
- Metabolic Unit, Institute of Neuroscience, National Research Council (CNR), 35127 Padova, Italy;
| | - Andrea Tura
- Liver Imaging Group, Department of Radiology, University of California—San Diego, La Jolla, CA 92093, USA; (A.T.); (C.B.S.)
| | - Claude B. Sirlin
- Liver Imaging Group, Department of Radiology, University of California—San Diego, La Jolla, CA 92093, USA; (A.T.); (C.B.S.)
| | - Abhijit J. Chaudhari
- Department of Radiology, School of Medicine, University of California—Davis, Sacramento, CA 95817, USA; (Y.A.); (J.P.M.); (A.J.C.)
| | - Peter J. Havel
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California—Davis, Sacramento, CA 95616, USA; (D.M.S.); (B.H.); (V.L.); (M.V.N.); (C.A.P.); (Y.B.); (P.J.H.)
- Department of Nutrition, University of California—Davis, Sacramento, CA 95616, USA
| | - Kimber L. Stanhope
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California—Davis, Sacramento, CA 95616, USA; (D.M.S.); (B.H.); (V.L.); (M.V.N.); (C.A.P.); (Y.B.); (P.J.H.)
- Department of Nutrition, University of California—Davis, Sacramento, CA 95616, USA
| |
Collapse
|
30
|
Cai H, Wang X, Zhang Z, Chen J, Wang F, Wang L, Liu J. Moderate l-lactate administration suppresses adipose tissue macrophage M1 polarization to alleviate obesity-associated insulin resistance. J Biol Chem 2022; 298:101768. [PMID: 35218776 PMCID: PMC8941214 DOI: 10.1016/j.jbc.2022.101768] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Revised: 02/16/2022] [Accepted: 02/17/2022] [Indexed: 12/11/2022] Open
Abstract
As a crucial metabolic intermediate, l-lactate is involved in redox balance, energy balance, and acid-base balance in organisms. Moderate exercise training transiently elevates plasma l-lactate levels and ameliorates obesity-associated type 2 diabetes. However, whether moderate l-lactate administration improves obesity-associated insulin resistance remains unclear. In this study, we defined 800 mg/kg/day as the dose of moderate l-lactate administration. In mice fed with a high-fat diet (HFD), moderate l-lactate administration for 12 weeks was shown to alleviate weight gain, fat accumulation, and insulin resistance. Along with the phenotype alterations, white adipose tissue thermogenesis was also found to be elevated in HFD-fed mice. Meanwhile, moderate l-lactate administration suppressed the infiltration and proinflammatory M1 polarization of adipose tissue macrophages (ATMs) in HFD-fed mice. Furthermore, l-lactate treatment suppressed the lipopolysaccharide-induced M1 polarization of bone marrow-derived macrophages (BMDMs). l-lactate can bind to the surface receptor GPR132, which typically drives the downstream cAMP-PKA signaling. As a nutrient sensor, AMP-activated protein kinase (AMPK) critically controls macrophage inflammatory signaling and phenotype. Thus, utilizing inhibitors of the kinases PKA and AMPK as well as siRNA against GPR132, we demonstrated that GPR132-PKA-AMPKα1 signaling mediated the suppression caused by l-lactate treatment on BMDM M1 polarization. Finally, l-lactate addition remarkably resisted the impairment of lipopolysaccharide-treated BMDM conditional media on adipocyte insulin sensitivity. In summary, moderate l-lactate administration suppresses ATM proinflammatory M1 polarization through activation of the GPR132-PKA-AMPKα1 signaling pathway to improve insulin resistance in HFD-fed mice, suggesting a new therapeutic and interventional approach to obesity-associated type 2 diabetes.
Collapse
Affiliation(s)
- Hao Cai
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui, China
| | - Xin Wang
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui, China
| | - Zhixin Zhang
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui, China
| | - Juan Chen
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui, China
| | - Fangbin Wang
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui, China
| | - Lu Wang
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui, China
| | - Jian Liu
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui, China; Engineering Research Center of Bioprocess, Ministry of Education, Hefei University of Technology, Hefei, Anhui, China.
| |
Collapse
|
31
|
Lagarde D, Jeanson Y, Portais JC, Galinier A, Ader I, Casteilla L, Carrière A. Lactate Fluxes and Plasticity of Adipose Tissues: A Redox Perspective. Front Physiol 2021; 12:689747. [PMID: 34276410 PMCID: PMC8278056 DOI: 10.3389/fphys.2021.689747] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 06/09/2021] [Indexed: 12/18/2022] Open
Abstract
Lactate, a metabolite produced when the glycolytic flux exceeds mitochondrial oxidative capacities, is now viewed as a critical regulator of metabolism by acting as both a carbon and electron carrier and a signaling molecule between cells and tissues. In recent years, increasing evidence report its key role in white, beige, and brown adipose tissue biology, and highlights new mechanisms by which lactate participates in the maintenance of whole-body energy homeostasis. Lactate displays a wide range of biological effects in adipose cells not only through its binding to the membrane receptor but also through its transport and the subsequent effect on intracellular metabolism notably on redox balance. This study explores how lactate regulates adipocyte metabolism and plasticity by balancing intracellular redox state and by regulating specific signaling pathways. We also emphasized the contribution of adipose tissues to the regulation of systemic lactate metabolism, their roles in redox homeostasis, and related putative physiopathological repercussions associated with their decline in metabolic diseases and aging.
Collapse
Affiliation(s)
- Damien Lagarde
- Goodman Cancer Research Center, McGill University, Montreal, QC, Canada.,Department of Biochemistry, McGill University, Montreal, QC, Canada.,Institut RESTORE, UMR 1301 INSERM, 5070 CNRS, Université Paul Sabatier, Toulouse, France
| | - Yannick Jeanson
- Institut RESTORE, UMR 1301 INSERM, 5070 CNRS, Université Paul Sabatier, Toulouse, France
| | - Jean-Charles Portais
- Institut RESTORE, UMR 1301 INSERM, 5070 CNRS, Université Paul Sabatier, Toulouse, France.,MetaboHUB-MetaToul, National Infrastructure of Metabolomics and Fluxomics, Toulouse, France
| | - Anne Galinier
- Institut RESTORE, UMR 1301 INSERM, 5070 CNRS, Université Paul Sabatier, Toulouse, France.,Institut Fédératif de Biologie, CHU Purpan, Toulouse, France
| | - Isabelle Ader
- Institut RESTORE, UMR 1301 INSERM, 5070 CNRS, Université Paul Sabatier, Toulouse, France
| | - Louis Casteilla
- Institut RESTORE, UMR 1301 INSERM, 5070 CNRS, Université Paul Sabatier, Toulouse, France
| | - Audrey Carrière
- Institut RESTORE, UMR 1301 INSERM, 5070 CNRS, Université Paul Sabatier, Toulouse, France
| |
Collapse
|
32
|
Zhao T, Le S, Freitag N, Schumann M, Wang X, Cheng S. Effect of Chronic Exercise Training on Blood Lactate Metabolism Among Patients With Type 2 Diabetes Mellitus: A Systematic Review and Meta-Analysis. Front Physiol 2021; 12:652023. [PMID: 33776804 PMCID: PMC7992008 DOI: 10.3389/fphys.2021.652023] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 02/17/2021] [Indexed: 12/29/2022] Open
Abstract
Purpose: To assess the effect of chronic exercise training on blood lactate metabolism at rest (i.e., basal lactate concentrations) and during exercise (i.e., blood lactate concentration at a fixed load, load at a fixed blood lactate concentration, and load at the individual blood lactate threshold) among patients with type 2 diabetes mellitus (T2DM). Methods: PubMed (MedLine), Embase, Web of Science, and Scopus were searched. Randomized controlled trials, non-randomized controlled trials, and case-control studies using chronic exercise training (i.e., 4 weeks) and that assessed blood lactate concentrations at rest and during exercise in T2DM patients were included. Results: Thirteen studies were eligible for the systematic review, while 12 studies with 312 participants were included into the meta-analysis. In the pre-to-post intervention meta-analysis, chronic exercise training had no significant effect on changes in basal blood lactate concentrations (standardized mean difference (SMD) = -0.20; 95% CI, -0.55 to 0.16; p = 0.28), and the results were similar when comparing the effect of intervention and control groups. Furthermore, blood lactate concentration at a fixed load significantly decreased (SMD = -0.73; 95% CI, -1.17 to -0.29; p = 0.001), while load at a fixed blood lactate concentration increased (SMD = 0.40; 95% CI, 0.07 to 0.72; p = 0.02) after chronic exercise training. No change was observed in load at the individual blood lactate threshold (SMD = 0.28; 95% CI, -0.14 to 0.71; p = 0.20). Conclusion: Chronic exercise training does not statistically affect basal blood lactate concentrations; however, it may decrease the blood lactate concentrations during exercise, indicating improvements of physical performance capacity which is beneficial for T2DM patients' health in general. Why chronic exercise training did not affect basal blood lactate concentrations needs further investigation.
Collapse
Affiliation(s)
- Tong Zhao
- Exercise, Health and Technology Centre, Department of Physical Education, Shanghai Jiao Tong University, Shanghai, China
| | - Shenglong Le
- Exercise, Health and Technology Centre, Department of Physical Education, Shanghai Jiao Tong University, Shanghai, China.,Faculty of Sport and Health Science, University of Jyväskylä, Jyväskylä, Finland.,Exercise Translational Medicine Centre, Shanghai Centre for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, China.,School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Nils Freitag
- Department of Molecular and Cellular Sport Medicine, Institute of Cardiovascular Research and Sport Medicine, German Sport University, Cologne, Germany.,Olympic Training Center Berlin, Berlin, Germany
| | - Moritz Schumann
- Exercise Translational Medicine Centre, Shanghai Centre for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, China.,Department of Molecular and Cellular Sport Medicine, Institute of Cardiovascular Research and Sport Medicine, German Sport University, Cologne, Germany
| | - Xiuqiang Wang
- Exercise, Health and Technology Centre, Department of Physical Education, Shanghai Jiao Tong University, Shanghai, China.,Exercise Translational Medicine Centre, Shanghai Centre for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, China
| | - Sulin Cheng
- Exercise, Health and Technology Centre, Department of Physical Education, Shanghai Jiao Tong University, Shanghai, China.,Faculty of Sport and Health Science, University of Jyväskylä, Jyväskylä, Finland.,Exercise Translational Medicine Centre, Shanghai Centre for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, China.,School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China.,Department of Molecular and Cellular Sport Medicine, Institute of Cardiovascular Research and Sport Medicine, German Sport University, Cologne, Germany
| |
Collapse
|
33
|
Joseph A, Parvathy S, Varma KK. Hyperinsulinemia Induced Altered Insulin Signaling Pathway in Muscle of High Fat- and Carbohydrate-Fed Rats: Effect of Exercise. J Diabetes Res 2021; 2021:5123241. [PMID: 33708999 PMCID: PMC7929694 DOI: 10.1155/2021/5123241] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 12/08/2020] [Accepted: 02/01/2021] [Indexed: 12/18/2022] Open
Abstract
Insulin resistance is a state of impaired responsiveness to insulin action. This condition not only results in deficient glucose uptake but increases the risk for cardiovascular diseases (CVD), stroke, and obesity. The present work investigates the molecular mechanisms of high carbohydrate and fat diet in inducing prediabetic hyperinsulinemia and effect of exercise on InsR signaling events, muscular AChE, and lactate dehydrogenase activity. Adult male Wistar rats were divided into the control (C) diet group, high-carbohydrate diet (HCD) group, high-fat diet (HFD) group, and HCD and HFD groups with exercise (HCD Ex and HFD Ex, respectively). Acetyl choline esterase activity, lactate dehydrogenase activity, total lactate levels, IRS1 phosphorylations, and Glut4 expression patterns were studied in the muscle tissue among these groups. High carbohydrate and fat feeding led to hyperinsulinemic status with reduced acetylcholine esterase (AChE) activity and impaired phosphorylation of IRS1 along with increased lactate concentrations in the muscle. Exercise significantly upregulated phosphoinositide 3 kinase (PI3K) docking site phosphorylation and downregulated the negative IRS1 phosphorylations thereby increasing the glucose transporter (GLUT) expressions and reducing the lactate accumulation. Also, the levels of second messengers like IP3 and cAMP were increased with exercise. Increased second messenger levels induce calcium release thereby activating the downstream pathway promoting the translocation of GLUT4 to the plasma membrane. Our results showed that the metabolic and signaling pathway dysregulations seen during diet-induced hyperinsulinemia, a metabolic condition seen during the early stages in the development of prediabetes, were improved with vigorous physical exercise. Thus, exercise can be considered as an excellent management approach over drug therapy for diabetes and its complications.
Collapse
Affiliation(s)
- Anu Joseph
- MIMS Research Foundation, Mankavu P.O., Calicut, Kerala 673007, India
| | - S. Parvathy
- MIMS Research Foundation, Mankavu P.O., Calicut, Kerala 673007, India
| | | |
Collapse
|
34
|
Quan W, Jiao Y, Xue C, Li Y, Liu G, He Z, Qin F, Zeng M, Chen J. The Effect of Exogenous Free Nε-(Carboxymethyl)Lysine on Diabetic-Model Goto-Kakizaki Rats: Metabolomics Analysis in Serum and Urine. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2021; 69:783-793. [PMID: 33401897 DOI: 10.1021/acs.jafc.0c06445] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
The current study investigated the effects of exogenous free Nε-(carboxymethyl) lysine (CML) from daily diet on diabetic-model Goto-Kakizaki rats. Rats were fed with free CML (2 mg/kg body weight) for 8 weeks, then metabolomics evaluation was performed on serum and urine, and biochemical and histopathologic examinations were conducted to verify metabolic results. Diabetic rats fed with free CML showed significantly increased (P < 0.05) fasting blood glucose (11.1 ± 1.07 mmol/L) and homeostasis model assessment values (homeostatic model assessment of insulin resistance: 16.0 ± 4.24; homeostatic model assessment of beta cell function: 6.66 ± 2.01; and modified beta cell function index: 11.5 ± 2.66) and a significantly altered (P < 0.05) oxidative stress level when compared to the control group. Serum and urine metabolomics showed a significantly altered (P < 0.05) level of aminomalonic acid, 2-oxoadipic acid, l-malic acid, β-alanine, 2-oxoglutaric acid, d-threitol, N-acetyl-leucine, methylmalonic acid, l-cysteine, thymine, glycine, l-alanine, 4-hydroxyproline, hexadecane, succinic acid, l-ornithine, gluconolactone, maleic acid, l-lactate, tryptophan, 5-methoxyindoleacetate, γ-aminobutyric acid, homoserine, maltose, and quinolinic acid. Our results indicated that these metabolites altered by exposure to exogenous free CML were mapped to the citric acid cycle and amino acid and carbohydrate metabolism, which might be related to increased progression of diabetes and some other diabetic complications, including diabetic brain and neurological diseases, retinopathy, nephropathy, and impaired wound healing.
Collapse
Affiliation(s)
- Wei Quan
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Ye Jiao
- School of Chemistry and Food Engineering, Changsha University of Science & Technology, Changsha 410114, China
| | - Chaoyi Xue
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Yong Li
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Guoping Liu
- Wuxi People's Hospital, Nanjing Medical University, Wuxi, Jiangsu 214023, China
| | - Zhiyong He
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Fang Qin
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Maomao Zeng
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Jie Chen
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
- International Joint Laboratory on Food Safety, Jiangnan University, Wuxi, Jiangsu 214122, China
| |
Collapse
|
35
|
Mazori AY, Bass IR, Chan L, Mathews KS, Altman DR, Saha A, Soh H, Wen HH, Bose S, Leven E, Wang JG, Mosoyan G, Pattharanitima P, Greco G, Gallagher EJ. Hyperglycemia is Associated With Increased Mortality in Critically Ill Patients With COVID-19. Endocr Pract 2021; 27:95-100. [PMID: 33551315 PMCID: PMC7796656 DOI: 10.1016/j.eprac.2020.12.015] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 12/24/2020] [Accepted: 12/30/2020] [Indexed: 01/08/2023]
Abstract
Objective To explore the relationship between hyperglycemia in the presence and absence of diabetes mellitus (DM) and adverse outcomes in critically ill patients with coronavirus disease 2019 (COVID-19). Methods The study included 133 patients with COVID-19 admitted to an intensive care unit (ICU) at an urban academic quaternary-care center between March 10 and April 8, 2020. Patients were categorized based on the presence or absence of DM and early-onset hyperglycemia (EHG), defined as a blood glucose >180 mg/dL during the first 2 days after ICU admission. The primary outcome was 14-day all-cause in-hospital mortality; also examined were 60-day all-cause in-hospital mortality and the levels of C-reactive protein, interleukin 6, procalcitonin, and lactate. Results Compared to non-DM patients without EHG, non-DM patients with EHG exhibited higher adjusted hazard ratios (HRs) for mortality at 14 days (HR 7.51, CI 1.70-33.24) and 60 days (HR 6.97, CI 1.86-26.13). Non-DM patients with EHG also featured higher levels of median C-reactive protein (306.3 mg/L, P = .036), procalcitonin (1.26 ng/mL, P = .028), and lactate (2.2 mmol/L, P = .023). Conclusion Among critically ill COVID-19 patients, those without DM with EHG were at greatest risk of 14-day and 60-day in-hospital mortality. Our study was limited by its retrospective design and relatively small cohort. However, our results suggest the combination of elevated glucose and lactate may identify a specific cohort of individuals at high risk for mortality from COVID-19. Glucose testing and control are important in individuals with COVID-19, even those without preexisting diabetes.
Collapse
Affiliation(s)
- Alon Y Mazori
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Ilana Ramer Bass
- Division of Endocrinology, Diabetes and Bone Disease, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Lili Chan
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Kusum S Mathews
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York; Department of Emergency Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Deena R Altman
- Division of Infectious Disease, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Aparna Saha
- Institute of Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Howard Soh
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Huei Hsun Wen
- Institute of Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Sonali Bose
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Emily Leven
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Jing Gennie Wang
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York; Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Gohar Mosoyan
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Pattharawin Pattharanitima
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Giampaolo Greco
- Department of Population Health Science and Policy, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Emily J Gallagher
- Division of Endocrinology, Diabetes and Bone Disease, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York; Tisch Cancer Institute at Mount Sinai, Icahn School of Medicine at Mount Sinai, New York, New York.
| |
Collapse
|
36
|
Posa DK, Baba SP. Intracellular pH Regulation of Skeletal Muscle in the Milieu of Insulin Signaling. Nutrients 2020; 12:nu12102910. [PMID: 32977552 PMCID: PMC7598285 DOI: 10.3390/nu12102910] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 08/24/2020] [Accepted: 08/31/2020] [Indexed: 12/18/2022] Open
Abstract
Type 2 diabetes (T2D), along with obesity, is one of the leading health problems in the world which causes other systemic diseases, such as cardiovascular diseases and kidney failure. Impairments in glycemic control and insulin resistance plays a pivotal role in the development of diabetes and its complications. Since skeletal muscle constitutes a significant tissue mass of the body, insulin resistance within the muscle is considered to initiate the onset of diet-induced metabolic syndrome. Insulin resistance is associated with impaired glucose uptake, resulting from defective post-receptor insulin responses, decreased glucose transport, impaired glucose phosphorylation, oxidation and glycogen synthesis in the muscle. Although defects in the insulin signaling pathway have been widely studied, the effects of cellular mechanisms activated during metabolic syndrome that cross-talk with insulin responses are not fully elucidated. Numerous reports suggest that pathways such as inflammation, lipid peroxidation products, acidosis and autophagy could cross-talk with insulin-signaling pathway and contribute to diminished insulin responses. Here, we review and discuss the literature about the defects in glycolytic pathway, shift in glucose utilization toward anaerobic glycolysis and change in intracellular pH [pH]i within the skeletal muscle and their contribution towards insulin resistance. We will discuss whether the derangements in pathways, which maintain [pH]i within the skeletal muscle, such as transporters (monocarboxylate transporters 1 and 4) and depletion of intracellular buffers, such as histidyl dipeptides, could lead to decrease in [pH]i and the onset of insulin resistance. Further we will discuss, whether the changes in [pH]i within the skeletal muscle of patients with T2D, could enhance the formation of protein aggregates and activate autophagy. Understanding the mechanisms by which changes in the glycolytic pathway and [pH]i within the muscle, contribute to insulin resistance might help explain the onset of obesity-linked metabolic syndrome. Finally, we will conclude whether correcting the pathways which maintain [pH]i within the skeletal muscle could, in turn, be effective to maintain or restore insulin responses during metabolic syndrome.
Collapse
Affiliation(s)
- Dheeraj Kumar Posa
- Diabetes and Obesity Center, University of Louisville, Louisville, KY 40202, USA
- Christina Lee Brown Envirome Institute, University of Louisville, Louisville, KY 40202, USA
| | - Shahid P Baba
- Diabetes and Obesity Center, University of Louisville, Louisville, KY 40202, USA
- Christina Lee Brown Envirome Institute, University of Louisville, Louisville, KY 40202, USA
| |
Collapse
|
37
|
Hosseini Khorami SA, Mutalib MSA, Feili Shiraz M, Abdullah JA, Rejali Z, Ali RM, Khaza'ai H. Genetic determinants of obesity heterogeneity in type II diabetes. Nutr Metab (Lond) 2020; 17:55. [PMID: 32670384 PMCID: PMC7346329 DOI: 10.1186/s12986-020-00476-6] [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: 03/21/2020] [Accepted: 07/01/2020] [Indexed: 11/23/2022] Open
Abstract
Background Although obesity is considered as the main cause of Type II diabetes (T2DM), non-obese individuals may still develop T2DM and obese individuals may not. Method The mRNA expression of PI3K/AKT axis from 100 non-obese and obese participants with insulin sensitivity and insulin resistance states were compared in this study toward the understanding of obesity heterogeneity molecular mechanism. Result In present study, there was no statistically significant difference in gene expression levels of IRS1 and PTEN between groups, whereas PI3K, AKT2 and GLUT4 genes were expressed at a lower level in obese diabetic group compared to other groups and were statistically significant. PDK1 gene was expressed at a higher level in non-obese diabetic group compared to obese diabetic and non-obese non-diabetics groups. No statistically significant difference was identified in gene expression pattern of PI3K/AKT pathway between obese non-diabetics and non-obese non-diabetics. Conclusion The components of PI3K/AKT pathway which is related to the fasting state, showed reduced expression in obese diabetic group due to the chronic over-nutrition which may induced insensitivity and reduced gene expression. The pathogenesis of insulin resistance in the absence of obesity in non-obese diabetic group could be due to disturbance in another pathway related to the non-fasting state like gluconeogenesis. Therefore, the molecular mechanism of insulin signalling in non-obese diabetic individuals is different from obese diabetics which more investigations are required to study insulin signalling pathways in greater depth, in order to assess nutritional factors, contribute to insulin resistance in obese diabetic and non-obese diabetic individuals.
Collapse
Affiliation(s)
| | - Mohd Sokhini Abd Mutalib
- Department of Nutrition and Dietetic, University Putra Malaysia, 43400 Serdang, Selangor Malaysia
| | - Mohammad Feili Shiraz
- Department of Artificial Intelligence and Computer Engineering, Faculty of Electrical Engineering, Computer and IT, Qazvin Branch, Islamic Azad University, Qazvin, Iran
| | | | - Zulida Rejali
- Department of Obstetrics and Gynaecology, University Putra Malaysia, 43400 Serdang, Selangor Malaysia
| | - Razana Mohd Ali
- Department of Pathology, University Putra Malaysia, 43400 Serdang, Selangor Malaysia
| | - Huzwah Khaza'ai
- Department of Biomedical Science, University Putra Malaysia, 43400 Serdang, Selangor Malaysia
| |
Collapse
|
38
|
Guasch-Ferré M, Santos JL, Martínez-González MA, Clish CB, Razquin C, Wang D, Liang L, Li J, Dennis C, Corella D, Muñoz-Bravo C, Romaguera D, Estruch R, Santos-Lozano JM, Castañer O, Alonso-Gómez A, Serra-Majem L, Ros E, Canudas S, Asensio EM, Fitó M, Pierce K, Martínez JA, Salas-Salvadó J, Toledo E, Hu FB, Ruiz-Canela M. Glycolysis/gluconeogenesis- and tricarboxylic acid cycle-related metabolites, Mediterranean diet, and type 2 diabetes. Am J Clin Nutr 2020; 111:835-844. [PMID: 32060497 PMCID: PMC7138680 DOI: 10.1093/ajcn/nqaa016] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Accepted: 01/23/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Glycolysis/gluconeogenesis and tricarboxylic acid (TCA) cycle metabolites have been associated with type 2 diabetes (T2D). However, the associations of these metabolites with T2D incidence and the potential effect of dietary interventions remain unclear. OBJECTIVES We aimed to evaluate the association of baseline and 1-y changes in glycolysis/gluconeogenesis and TCA cycle metabolites with insulin resistance and T2D incidence, and the potential modifying effect of Mediterranean diet (MedDiet) interventions. METHODS We included 251 incident T2D cases and 638 noncases in a nested case-cohort study within the PREDIMED Study during median follow-up of 3.8 y. Participants were allocated to MedDiet + extra-virgin olive oil, MedDiet + nuts, or control diet. Plasma metabolites were measured using a targeted approach by LC-tandem MS. We tested the associations of baseline and 1-y changes in glycolysis/gluconeogenesis and TCA cycle metabolites with subsequent T2D risk using weighted Cox regression models and adjusting for potential confounders. We designed a weighted score combining all these metabolites and applying the leave-one-out cross-validation approach. RESULTS Baseline circulating concentrations of hexose monophosphate, pyruvate, lactate, alanine, glycerol-3 phosphate, and isocitrate were significantly associated with higher T2D risk (17-44% higher risk for each 1-SD increment). The weighted score including all metabolites was associated with a 30% (95% CI: 1.12, 1.51) higher relative risk of T2D for each 1-SD increment. Baseline lactate and alanine were associated with baseline and 1-y changes of homeostasis model assessment of insulin resistance. One-year increases in most metabolites and in the weighted score were associated with higher relative risk of T2D after 1 y of follow-up. Lower risks were observed in the MedDiet groups than in the control group although no significant interactions were found after adjusting for multiple comparisons. CONCLUSIONS We identified a panel of glycolysis/gluconeogenesis-related metabolites that was significantly associated with T2D risk in a Mediterranean population at high cardiovascular disease risk. A MedDiet could counteract the detrimental effects of these metabolites.This trial was registered at controlled-trials.com as ISRCTN35739639.
Collapse
Affiliation(s)
- Marta Guasch-Ferré
- Department of Nutrition, Harvard TH Chan School of Public Health, Boston, MA, USA
- Human Nutrition Unit, Faculty of Medicine and Health Sciences, Pere Virgili Health Research Institute, Rovira i Virgili University, Reus, Spain
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - José L Santos
- Department of Nutrition, Diabetes and Metabolism, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Miguel A Martínez-González
- Department of Nutrition, Harvard TH Chan School of Public Health, Boston, MA, USA
- Department of Preventive Medicine and Public Health, IdiSNA (Health Research Institute of Navarra), University of Navarra, Pamplona, Spain
- The Spanish Biomedical Research Center in Physiopathology of Obesity and Nutrition, Health Institute Carlos III, Madrid, Spain
| | - Clary B Clish
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Cristina Razquin
- Department of Preventive Medicine and Public Health, IdiSNA (Health Research Institute of Navarra), University of Navarra, Pamplona, Spain
- The Spanish Biomedical Research Center in Physiopathology of Obesity and Nutrition, Health Institute Carlos III, Madrid, Spain
| | - Dong Wang
- Department of Nutrition, Harvard TH Chan School of Public Health, Boston, MA, USA
| | - Liming Liang
- Department of Biostatistics, Harvard TH Chan School of Public Health, Boston, MA, USA
| | - Jun Li
- Department of Nutrition, Harvard TH Chan School of Public Health, Boston, MA, USA
| | - Courtney Dennis
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Dolores Corella
- The Spanish Biomedical Research Center in Physiopathology of Obesity and Nutrition, Health Institute Carlos III, Madrid, Spain
- Department of Preventive Medicine, University of Valencia, Valencia, Spain
| | - Carlos Muñoz-Bravo
- Department of Public Health and Psychiatry, University of Málaga, Málaga, Spain
| | - Dora Romaguera
- The Spanish Biomedical Research Center in Physiopathology of Obesity and Nutrition, Health Institute Carlos III, Madrid, Spain
- Health Research Institute of the Balearic Islands (IdISBa), University Hospital Son Espases, Mallorca, Spain
| | - Ramón Estruch
- The Spanish Biomedical Research Center in Physiopathology of Obesity and Nutrition, Health Institute Carlos III, Madrid, Spain
- Department of Internal Medicine, Department of Endocrinology and Nutrition Biomedical Research Institute August Pi Sunyer (IDI-BAPS), Hospital Clinic, University of Barcelona, Barcelona, Spain
| | - José Manuel Santos-Lozano
- The Spanish Biomedical Research Center in Physiopathology of Obesity and Nutrition, Health Institute Carlos III, Madrid, Spain
- Department of Family Medicine, Primary Care Division of Sevilla, San Pablo Health Center, Sevilla, Spain
| | - Olga Castañer
- The Spanish Biomedical Research Center in Physiopathology of Obesity and Nutrition, Health Institute Carlos III, Madrid, Spain
- Cardiovascular and Nutrition Research Group, Hospital del Mar Research Institute (IMIM), Barcelona, Spain
| | - Angel Alonso-Gómez
- The Spanish Biomedical Research Center in Physiopathology of Obesity and Nutrition, Health Institute Carlos III, Madrid, Spain
- Bioaraba Health Research Institute; Osakidetza Baseque Health Service, Araba University Hospital; Unibersity of the Basque Country UPV/EHU; Vitoria-Gasteiz, Spain
| | - Luis Serra-Majem
- The Spanish Biomedical Research Center in Physiopathology of Obesity and Nutrition, Health Institute Carlos III, Madrid, Spain
- Research Institute of Biomedical and Health Sciences (IUIBS), University of Las Palmas de Gran Canaria and Service of Preventive Medicine, Complejo Hospitalario Universitario Insular Materno Infantil (CHUIMI), Canary Health Service, Las Palmas de Gran Canaria, Spain
| | - Emilio Ros
- The Spanish Biomedical Research Center in Physiopathology of Obesity and Nutrition, Health Institute Carlos III, Madrid, Spain
- Lipid Clinic, Department of Endocrinology and Nutrition Biomedical Research Institute August Pi Sunyer (IDI-BAPS), Hospital Clinic, University of Barcelona, Barcelona, Spain
| | - Sílvia Canudas
- Human Nutrition Unit, Faculty of Medicine and Health Sciences, Pere Virgili Health Research Institute, Rovira i Virgili University, Reus, Spain
- The Spanish Biomedical Research Center in Physiopathology of Obesity and Nutrition, Health Institute Carlos III, Madrid, Spain
| | - Eva M Asensio
- Department of Preventive Medicine, University of Valencia, Valencia, Spain
| | - Montserrat Fitó
- The Spanish Biomedical Research Center in Physiopathology of Obesity and Nutrition, Health Institute Carlos III, Madrid, Spain
- Cardiovascular and Nutrition Research Group, Hospital del Mar Research Institute (IMIM), Barcelona, Spain
| | - Kerry Pierce
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - J Alfredo Martínez
- The Spanish Biomedical Research Center in Physiopathology of Obesity and Nutrition, Health Institute Carlos III, Madrid, Spain
- Department of Nutrition, Food Sciences, and Physiology, Center for Nutrition Research, University of Navarra, Pamplona, IMDEA Food, Madrid, Spain
| | - Jordi Salas-Salvadó
- Human Nutrition Unit, Faculty of Medicine and Health Sciences, Pere Virgili Health Research Institute, Rovira i Virgili University, Reus, Spain
- The Spanish Biomedical Research Center in Physiopathology of Obesity and Nutrition, Health Institute Carlos III, Madrid, Spain
| | - Estefanía Toledo
- Department of Preventive Medicine and Public Health, IdiSNA (Health Research Institute of Navarra), University of Navarra, Pamplona, Spain
- The Spanish Biomedical Research Center in Physiopathology of Obesity and Nutrition, Health Institute Carlos III, Madrid, Spain
| | - Frank B Hu
- Department of Nutrition, Harvard TH Chan School of Public Health, Boston, MA, USA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Miguel Ruiz-Canela
- Department of Preventive Medicine and Public Health, IdiSNA (Health Research Institute of Navarra), University of Navarra, Pamplona, Spain
- The Spanish Biomedical Research Center in Physiopathology of Obesity and Nutrition, Health Institute Carlos III, Madrid, Spain
| |
Collapse
|
39
|
Stokes SM, Bertin FR, Stefanovski D, Belknap JK, Medina-Torres CE, Pollitt CC, van Eps AW. Lamellar energy metabolism and perfusion in the euglycaemic hyperinsulinaemic clamp model of equine laminitis. Equine Vet J 2020; 52:577-584. [PMID: 31845378 DOI: 10.1111/evj.13224] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 10/29/2019] [Accepted: 12/04/2019] [Indexed: 01/19/2023]
Abstract
BACKGROUND Hyperinsulinaemia is associated with the development of endocrinopathic laminitis; however, the mechanisms remain unclear. OBJECTIVES Evaluate the effects of hyperinsulinaemia on lamellar energy metabolism and perfusion during laminitis development. STUDY DESIGN In vivo experiment. METHODS Eight Standardbred horses were instrumented with a microdialysis probe in the lamellae of a forelimb. A 24 hours baseline period (BASELINE) was followed by 48 hours of a continuous euglycaemic hyperinsulinaemic clamp (EHC) from 24 to 72 hours (CLAMP). Microdialysate was collected every 6 hours and analysed for glucose, lactate and pyruvate concentrations and lactate-to-pyruvate ratio (L:P). Microdialysis urea clearance was used to estimate lamellar tissue perfusion. Archived microdialysis samples from six identically instrumented Standardbred horses served as controls (CON). Variables were compared over time and between EHC and CON horses using a mixed-effects linear regression model. RESULTS Glucose concentration decreased during the CLAMP period in CON and EHC horses (P < .001), but there was no difference between CON and EHC (P > .9). Lactate concentration increased during the CLAMP period in CON and EHC horses (P < .001), however, the rate of increase was significantly higher in EHC horses relative to CON (P = .014). There was a relative increase in pyruvate concentration in EHC horses compared with CON during the CLAMP period (P = .03). L:P increased significantly in CON horses during the CLAMP period (P < .001) but not in EHC (P = .1). Urea clearance did not change in CON (P = .9) or EHC (P = .05) during the CLAMP, but did increase in EHC relative to CON (P = .02). MAIN LIMITATIONS The effects of microdialysis probe implantation on perfusion and metabolism remain unclear. The EHC model may not mimic natural endocrinopathic laminitis. CONCLUSIONS Laminitis developed without evidence of lamellar hypoperfusion or energy stress. Therapies to improve perfusion are unlikely to affect the initial development of endocrinopathic laminitis.
Collapse
Affiliation(s)
- Simon M Stokes
- Australian Equine Laminitis Research Unit, School of Veterinary Science, The University of Queensland, Gatton, Qld, Australia
| | - Francois R Bertin
- Australian Equine Laminitis Research Unit, School of Veterinary Science, The University of Queensland, Gatton, Qld, Australia
| | - Darko Stefanovski
- New Bolton Center, Department of Clinical Studies, School of Veterinary Medicine, University of Pennsylvania, Kennett Square, Pennsylvania
| | - James K Belknap
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio
| | - Carlos E Medina-Torres
- Australian Equine Laminitis Research Unit, School of Veterinary Science, The University of Queensland, Gatton, Qld, Australia
| | - Christopher C Pollitt
- Australian Equine Laminitis Research Unit, School of Veterinary Science, The University of Queensland, Gatton, Qld, Australia
| | - Andrew W van Eps
- Australian Equine Laminitis Research Unit, School of Veterinary Science, The University of Queensland, Gatton, Qld, Australia.,New Bolton Center, Department of Clinical Studies, School of Veterinary Medicine, University of Pennsylvania, Kennett Square, Pennsylvania
| |
Collapse
|
40
|
Couselo-Seijas M, Agra-Bermejo RM, Fernández AL, Martínez-Cereijo JM, Sierra J, Soto-Pérez M, Rozados-Luis A, González-Juanatey JR, Eiras S. High released lactate by epicardial fat from coronary artery disease patients is reduced by dapagliflozin treatment. Atherosclerosis 2020; 292:60-69. [PMID: 31783199 DOI: 10.1016/j.atherosclerosis.2019.11.016] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 10/31/2019] [Accepted: 11/13/2019] [Indexed: 02/06/2023]
Abstract
BACKGROUND AND AIMS Dapagliflozin, a sodium-glucose co-transporter 2 inhibitor, improves glucose uptake by epicardial adipose tissue (EAT). However, its metabolism might raise the lactate production and acidosis under hypoxia conditions, i.e. coronary artery disease (CAD), or lipogenesis and, in consequence, expand adipose tissue. Since lactate secreted by adipose tissue is correlated with tissue stress and inflammation, our aim was to study glucose metabolism by epicardial fat in CAD and its regulation by dapagliflozin. METHODS Paired EAT and subcutaneous adipose tissue (SAT) biopsies from 49 patients who underwent open-heart surgery were cultured and split into three equal pieces, some treated with and others without dapagliflozin at 10 or 100 μM for 6 h. Anaerobic glucose metabolites were measured in supernatants of fat pads, and acidosis on adipogenesis-induced primary culture cells was analysed by colorimetric or fluorescence assays. Gene expression levels were assessed by real-time polymerase chain reaction. RESULTS Our results showed that dapagliflozin reduced the released lactate and acidosis in epicardial fat (p < 0.05) without changes in lipid storage-involved genes. In addition, this drug induced gene expression levels of peroxisome proliferator-activated receptor gamma coactivator 1 alpha (PGC1α), a mitochondrial biogenesis-involved gene in both EAT and SAT (p < 0.05). After splitting the population regarding the presence of CAD, we observed higher lactate production in EAT from these patients (2.46 [1.75-3.47] mM), which was reduced after treatment with dapagliflozin 100 μM (1.99 [1.08-2.99] mM, p < 0.01). CONCLUSIONS Dapagliflozin improved glucose metabolism without lipogenesis-involved gene regulation or lactate production, mainly in patients with CAD. These results suggest an improvement of glucose oxidation metabolism that can contribute to cardiovascular benefits.
Collapse
Affiliation(s)
| | - Rosa María Agra-Bermejo
- Cardiovascular Area and Coronary Unit, University Clinical Hospital of Santiago de Compostela, Spain; CIBERCV, Madrid, Spain; Cardiology Group, Health Research Institute of Santiago de Compostela, Spain
| | - Angel Luis Fernández
- CIBERCV, Madrid, Spain; Heart Surgery Department University Clinical Hospital of Santiago de Compostela, Spain
| | | | - Juan Sierra
- Heart Surgery Department University Clinical Hospital of Santiago de Compostela, Spain
| | - Maeve Soto-Pérez
- Cardiovascular Area and Coronary Unit, University Clinical Hospital of Santiago de Compostela, Spain
| | - Adriana Rozados-Luis
- Translational Cardiology Group, Health Research Institute of Santiago de Compostela, Spain
| | - José Ramón González-Juanatey
- Cardiovascular Area and Coronary Unit, University Clinical Hospital of Santiago de Compostela, Spain; CIBERCV, Madrid, Spain; Cardiology Group, Health Research Institute of Santiago de Compostela, Spain
| | - Sonia Eiras
- Translational Cardiology Group, Health Research Institute of Santiago de Compostela, Spain; CIBERCV, Madrid, Spain.
| |
Collapse
|
41
|
Borghi F, Morais CL, Silva C, da Silva PC, Ishizu LY, Costa GT, Grassi-Kassisse DM. A new perspective of lactatogenesis by isolated adipocytes. Mol Cell Endocrinol 2019; 498:110560. [PMID: 31442545 DOI: 10.1016/j.mce.2019.110560] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 07/02/2019] [Accepted: 08/19/2019] [Indexed: 11/24/2022]
Abstract
Increased adipose tissue mass exhibited greater capacity of glucose transformation in lactate, highlighting lactatogenesis as a crucial factor in body size. Classically, lactate produced by isolated adipocytes are expressed per million of cells and were never correlated with their size. Spontaneously hypertensive rats (SHR) have a lower body weight and smaller adipocytes when compared to Wistar-Kyoto. We evaluated basal lactate by epididymal 15-weeks-old isolated adipocytes of SHR, Wistar-Kyoto and Wistar. Basal lactate was similar when expressed by one million cells. However, SHR adipocytes were smaller, so we adjusted the results by cell volume and SHR showed higher basal lactate production which was directly endorsed by hyperlactatemia in the presented conditions. Thereby, we suggest a new perspective on lactatogenesis analysis by adipocytes, which could be linked to the receptors density and associate enzymes. Moreover, we showed that the thin and hypertensive rats can be hyperlactemic in fasting conditions.
Collapse
Affiliation(s)
- Filipy Borghi
- LABEEST - Laboratory of Stress Study, Department of Structural and Functional Biology, Institute of Biology, University of Campinas - UNICAMP, 13083-862, Campinas, SP, Brazil
| | - Camila L Morais
- LABEEST - Laboratory of Stress Study, Department of Structural and Functional Biology, Institute of Biology, University of Campinas - UNICAMP, 13083-862, Campinas, SP, Brazil
| | - Carolina Silva
- LABEEST - Laboratory of Stress Study, Department of Structural and Functional Biology, Institute of Biology, University of Campinas - UNICAMP, 13083-862, Campinas, SP, Brazil
| | - Priscila C da Silva
- LABEEST - Laboratory of Stress Study, Department of Structural and Functional Biology, Institute of Biology, University of Campinas - UNICAMP, 13083-862, Campinas, SP, Brazil
| | - Larissa Y Ishizu
- LABEEST - Laboratory of Stress Study, Department of Structural and Functional Biology, Institute of Biology, University of Campinas - UNICAMP, 13083-862, Campinas, SP, Brazil
| | - Gustavo T Costa
- LABEEST - Laboratory of Stress Study, Department of Structural and Functional Biology, Institute of Biology, University of Campinas - UNICAMP, 13083-862, Campinas, SP, Brazil
| | - Dora M Grassi-Kassisse
- LABEEST - Laboratory of Stress Study, Department of Structural and Functional Biology, Institute of Biology, University of Campinas - UNICAMP, 13083-862, Campinas, SP, Brazil.
| |
Collapse
|
42
|
Takahashi N, Nakada TA, Sakai T, Kato Y, Moriyama K, Nishida O, Oda S. A CO 2 removal system using extracorporeal lung and renal assist device with an acid and alkaline infusion. J Artif Organs 2019; 23:54-61. [PMID: 31584110 DOI: 10.1007/s10047-019-01136-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Accepted: 09/22/2019] [Indexed: 01/04/2023]
Abstract
The patients with respiratory failure need high tidal volume by mechanical ventilation, which lead to the ventilator-induced lung injury. We developed an extracorporeal lung and renal assist device (ELRAD), comprising acid infusion, membrane lung, continuous hemodiafiltration and alkaline infusion. To evaluate this system, we conducted in vivo studies using experimental swine which were connected to the new system. In vivo experiments consist of four protocols; baseline = hemodiafiltration only (no O2 gas flow to membrane lung); membrane lung = "Baseline" plus O2 gas flow to membrane lung; "Acid infusion" = "Membrane lung" plus continuous acid infusion; ELRAD = "Acid infusion" plus continuous alkaline infusion. We changed the ventilatory rate of the mechanical ventilation to maintain PCO2 at 50-55 mmHg during the four protocols. The results showed that there was statistically no significant difference in the levels of pH, HCO3-, and base excess when each study protocol was initiated. The amount of CO2 eliminated by the membrane lung significantly increased by 1.6 times in the acid infusion protocol and the ELRAD protocol compared to the conventional membrane lung protocol. Minute ventilation in the ELRAD protocol significantly decreased by 0.5 times compared with the hemodiafiltration only protocol (P < 0.0001), the membrane lung (P = 0.0006) and acid infusion protocol (P = 0.0017), respectively. In conclusion, a developed CO2 removal system efficiently removed CO2 at low blood flow and reduced minute ventilation, while maintaining acid-base balance within the normal range.
Collapse
Affiliation(s)
- Nozomi Takahashi
- Department of Emergency and Critical Care Medicine, Chiba University Graduate School of Medicine, 1-8-1 Inohana, Chuo, Chiba, 260-8677, Japan
| | - Taka-Aki Nakada
- Department of Emergency and Critical Care Medicine, Chiba University Graduate School of Medicine, 1-8-1 Inohana, Chuo, Chiba, 260-8677, Japan.
| | - Toshikazu Sakai
- Department of Anesthesiology and Critical Care Medicine, Fujita Health University School of Medicine, 1-98 Dengakugakubo, Kutsukake, Toyoake, Aichi, 470-1192, Japan
| | - Yu Kato
- Department of Anesthesiology and Critical Care Medicine, Fujita Health University School of Medicine, 1-98 Dengakugakubo, Kutsukake, Toyoake, Aichi, 470-1192, Japan
| | - Kazuhiro Moriyama
- Laboratory for Immune Response and Regulatory Medicine, Fujita Health University School of Medicine, 1-98 Dengakugakubo, Kutsukake, Toyoake, Aichi, 470-1192, Japan
| | - Osamu Nishida
- Department of Anesthesiology and Critical Care Medicine, Fujita Health University School of Medicine, 1-98 Dengakugakubo, Kutsukake, Toyoake, Aichi, 470-1192, Japan
| | - Shigeto Oda
- Department of Emergency and Critical Care Medicine, Chiba University Graduate School of Medicine, 1-8-1 Inohana, Chuo, Chiba, 260-8677, Japan
| |
Collapse
|
43
|
Langdahl JH, Frederiksen AL, Vissing J, Frost M, Yderstræde KB, Andersen PH. Mitochondrial mutation m.3243A>G associates with insulin resistance in non-diabetic carriers. Endocr Connect 2019; 8:829-837. [PMID: 31146262 PMCID: PMC6590205 DOI: 10.1530/ec-19-0118] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Accepted: 05/30/2019] [Indexed: 01/08/2023]
Abstract
AIM This case-control study aimed to examine impairments in glucose metabolism in non-diabetic carriers of the mitochondrial mutation m.3243A>G by evaluating insulin secretion capacity and sensitivity. METHODS Glucose metabolism was investigated in 23 non-diabetic m.3243A>G carriers and age-, sex- and BMI-matched healthy controls with an extended 4-h oral glucose tolerance test (OGTT). Insulin sensitivity index and acute insulin response were estimated on the basis of the OGTT. This was accompanied by examination of body composition by dual-energy X-ray absorptiometry (DXA), maximum aerobic capacity and a Recent Physical Activity Questionnaire (RPAQ). RESULTS Fasting p-glucose, s-insulin and s-c-peptide levels did not differ between m.3243A>G carriers and controls. Insulin sensitivity index (BIGTT-S1) was significantly lower in the m.3243A>G carriers, but there was no difference in the acute insulin response between groups. P-lactate levels were higher in carriers throughout the OGTT. VO2max, but not BMI, waist and hip circumferences, lean and fat body mass%, MET or grip strength, was lower in mutation carriers. BIGTT-S1 remained lower in mutation carriers after adjustment for multiple confounding factors including VO2max in regression analyses. CONCLUSIONS Glucose metabolism in m.3243A>G carriers was characterized by reduced insulin sensitivity, which could represent the earliest phase in the pathogenesis of m.3243A>G-associated diabetes.
Collapse
Affiliation(s)
- Jakob Høgild Langdahl
- Department of Clinical Genetics, Odense University Hospital, Odense, Denmark
- Institute of Clinical Research, University of Southern Denmark, Odense, Denmark
- Department of Endocrinology, Hospital of Southwest Jutland, Esbjerg, Denmark
- Correspondence should be addressed to J H Langdahl:
| | - Anja Lisbeth Frederiksen
- Department of Clinical Genetics, Odense University Hospital, Odense, Denmark
- Institute of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - John Vissing
- Copenhagen Neuromuscular Center, Department of Neurology, Rigshospitalet, Copenhagen, Denmark
| | - Morten Frost
- Institute of Clinical Research, University of Southern Denmark, Odense, Denmark
- Steno Diabetes Center Odense, Odense University Hospital, Odense, Denmark
| | - Knud Bonnet Yderstræde
- Institute of Clinical Research, University of Southern Denmark, Odense, Denmark
- Steno Diabetes Center Odense, Odense University Hospital, Odense, Denmark
| | - Per Heden Andersen
- Department of Endocrinology, Hospital of Southwest Jutland, Esbjerg, Denmark
| |
Collapse
|
44
|
Bar-Or D, Rael LT, Madayag RM, Banton KL, Tanner A, Acuna DL, Lieser MJ, Marshall GT, Mains CW, Brody E. Stress Hyperglycemia in Critically Ill Patients: Insight Into Possible Molecular Pathways. Front Med (Lausanne) 2019; 6:54. [PMID: 30972338 PMCID: PMC6445875 DOI: 10.3389/fmed.2019.00054] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 03/05/2019] [Indexed: 01/08/2023] Open
Abstract
Severe sepsis, systemic inflammatory response syndrome (SIRS), and traumatic brain injury are frequently associated with hyperglycemia in non-diabetic patients. In patients suffering from any of these conditions, hyperglycemia at admission to an intensive care unit (ICU) is directly correlated with increased mortality or morbidity. Although there was initial enthusiasm for insulin treatment to blood glucose levels below 110 mg/dL in these patients, recent understanding suggests that the potential for hypoglycemic complications make this approach potentially dangerous. More moderate glucose control seems to be more beneficial than the aggressive glucose lowering initially suggested. An important publication has shown that hyperlactatemia accompanying hyperglycemia could be the real culprit in bad outcomes. This suggests that coupling moderate glucose lowering with therapeutic agents which might treat the underlying metabolic disturbances in these conditions may be a better strategy. The key metabolic disturbance in these three conditions seems to be persistent glycolysis as an energy source even in the presence of adequate tissue oxygenation (the Warburg Effect). We look at recent advances in understanding aerobic glycolysis and possibly the action of DPP4 on incretins resulting in insulin dysregulation and suggest key metabolic pathways involved in hyperglycemia regulation.
Collapse
Affiliation(s)
- David Bar-Or
- Trauma Research Laboratory, Swedish Medical Center, Englewood, CO, United States.,St. Anthony Hospital, Lakewood, CO, United States.,Penrose Hospital, Colorado Springs, CO, United States.,Wesley Medical Center, Wichita, KS, United States.,Research Medical Center, Kansas City, MO, United States.,Medical City Plano Hospital, Plano, TX, United States
| | - Leonard T Rael
- Trauma Research Laboratory, Swedish Medical Center, Englewood, CO, United States
| | | | - Kaysie L Banton
- Trauma Research Laboratory, Swedish Medical Center, Englewood, CO, United States
| | - Allen Tanner
- Penrose Hospital, Colorado Springs, CO, United States
| | | | - Mark J Lieser
- Research Medical Center, Kansas City, MO, United States
| | | | | | | |
Collapse
|
45
|
Kaur P, Choudhury D. Insulin Promotes Wound Healing by Inactivating NFkβP50/P65 and Activating Protein and Lipid Biosynthesis and alternating Pro/Anti-inflammatory Cytokines Dynamics. Biomol Concepts 2019; 10:11-24. [DOI: 10.1515/bmc-2019-0002] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Accepted: 01/04/2019] [Indexed: 02/07/2023] Open
Abstract
AbstractFour hundred and twenty-two million people have diabetes due to excess free body glucose in their body fluids. Diabetes leads to various problems including retinopathy, neuropathy, arthritis, damage blood vessels etc; it also causes a delay in wound healing. Insufficiency of insulin is the main reason for diabetes-I and systemic insulin treatment is a remedy. The perspective of the potential use of insulin/insulin based drugs to treat chronic wounds in diabetic conditions is focused on in this review. At the site of the wound, TNF-ɑ, IFN-ϒ, IL-1β and IL-6 pro-inflammatory cytokines cause the generation of free radicals, leading to inflammation which becomes persistent in diabetes. Insulin induces expression of IL-4/IL-13, IL-10 anti-inflammatory cytokines etc which further down-regulates NFkβP50/P65 assembly. Insulin shifts the equilibrium towards NFkβP50/P50 which leads to down-regulation of inflammatory cytokines such as IL-6, IL-10 etc through STAT6, STAT3 and c-Maf activation causing nullification of an inflammatory condition. Insulin also promotes protein and lipid biosynthesis which indeed promotes wound recovery. Here, in this article, the contributions of insulin in controlling wound tissue microenvironments and remodulation of tissue have been summarised, which may be helpful to develop novel insulin-based formulation(s) for effective treatment of wounds in diabetic conditions.
Collapse
Affiliation(s)
- Pawandeep Kaur
- School of Chemistry and Biochemistry, Thapar Institute of Engineering and Technology, Patiala-147004, Punjab, India
| | - Diptiman Choudhury
- School of Chemistry and Biochemistry, Thapar Institute of Engineering and Technology, Patiala-147004, Punjab, India
| |
Collapse
|
46
|
Riddell MC, Pooni R, Yavelberg L, Li Z, Kollman C, Brown RE, Li A, Aronson R. Reproducibility in the cardiometabolic responses to high-intensity interval exercise in adults with type 1 diabetes. Diabetes Res Clin Pract 2019; 148:137-143. [PMID: 30641168 DOI: 10.1016/j.diabres.2019.01.003] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 11/30/2018] [Accepted: 01/04/2019] [Indexed: 12/21/2022]
Abstract
AIMS Patients with type 1 diabetes (T1D) often report a rise in their blood glucose level following brief intense exercise. We sought to determine the reproducibility of the cardiometabolic responses to high-intensity interval training (HIIT). METHODS Sixteen adults with T1D, using an optimized multiple daily injection with basal insulin glargine 300 U/mL (Gla-300), performed four fasted HIIT sessions over a 4-6-week period. Exercise consisted of high-intensity interval cycling and multimodal training over 25 min. RESULTS Heart rate and rating of perceived exertion rose similarly in all sessions, as did lactate, catecholamine and growth hormone levels. Plasma glucose increased in response to HIIT in 62 of 64 visits (97%), with an overall increase of 3.7 ± 1.6 mmol/L (Mean ± SD) (P < 0.001). In within-patient comparisons, the change in plasma glucose among the four HIIT sessions was significantly correlated with a composite correlation of 0.58 ([r2 = 0.34]; 95% CI 0.35-0.80; P < 0.01). CONCLUSIONS Intersession observations of four separate HIIT sessions showed high intrasubject reproducibility in the cardiometabolic responses to exercise, including the rise in plasma glucose, when adults with T1D perform the activity in a fasted state.
Collapse
Affiliation(s)
- Michael C Riddell
- LMC Diabetes and Endocrinology, 1929 Bayview Ave., Toronto, Ontario, Canada; School of Kinesiology and Health Science, York University, 4700 Keele St., Toronto, Ontario, Canada.
| | - Rubin Pooni
- School of Kinesiology and Health Science, York University, 4700 Keele St., Toronto, Ontario, Canada.
| | - Loren Yavelberg
- School of Kinesiology and Health Science, York University, 4700 Keele St., Toronto, Ontario, Canada.
| | - Zoey Li
- JAEB Center for Health Research, 15310 Amberly Dr., Tampa, FL, USA.
| | - Craig Kollman
- JAEB Center for Health Research, 15310 Amberly Dr., Tampa, FL, USA.
| | - Ruth E Brown
- LMC Diabetes and Endocrinology, 1929 Bayview Ave., Toronto, Ontario, Canada.
| | - Aihua Li
- LMC Diabetes and Endocrinology, 1929 Bayview Ave., Toronto, Ontario, Canada.
| | - Ronnie Aronson
- LMC Diabetes and Endocrinology, 1929 Bayview Ave., Toronto, Ontario, Canada.
| |
Collapse
|
47
|
Aronson R, Brown RE, Li A, Riddell MC. Optimal Insulin Correction Factor in Post-High-Intensity Exercise Hyperglycemia in Adults With Type 1 Diabetes: The FIT Study. Diabetes Care 2019; 42:10-16. [PMID: 30455336 DOI: 10.2337/dc18-1475] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Accepted: 10/18/2018] [Indexed: 02/03/2023]
Abstract
OBJECTIVE Postexercise hyperglycemia, following high-intensity interval training (HIIT) in patients with type 1 diabetes (T1D), is largely underrecognized by the clinical community and generally undertreated. The aim of this study was to compare four multipliers of an individual's insulin correction factor (ICF) to treat post-HIIT hyperglycemia. RESEARCH DESIGN AND METHODS The FIT study had a randomized, crossover design in physically active subjects with T1D (mean ± SD age 34.9 ± 10.1 years, BMI 25.5 ± 2.5 kg/m2, and HbA1c 7.2 ± 0.9%) using multiple daily injections. Following an 8-week optimization period, with 300 units/mL insulin glargine used as the basal insulin, subjects performed four weekly sessions of 25 min of HIIT. If hyperglycemia (>8.0 mmol/L) resulted, subjects received a bolus insulin correction 15 min post-HIIT, based on their own ICF, adjusted by one of four multipliers: 0, 50, 100, or 150%. RESULTS Seventeen subjects completed 71 exercise trials, of which 64 (90%) resulted in hyperglycemia. At 40 min postexercise, plasma glucose (PG) increased from mean ± SD 8.8 ± 1.0 mmol/L at baseline to 12.7 ± 2.4 mmol/L (increase of 3.8 ± 1.5 mmol/L). After correction, adjusted mean ± SE PG was significantly reduced for the 50% (-2.3 ± 0.8 mmol/L, P < 0.01), 100% (-4.7 ± 0.8 mmol/L, P < 0.001), and 150% (-5.3 ± 0.8 mmol/L, P < 0.001) arms but had increased further in the 0% correction arm. Both the 100 and 150% corrections were more effective than the 50% correction (P < 0.01 and P < 0.001, respectively) but were not different from each other. Hypoglycemia was rare. CONCLUSIONS In post-HIIT hyperglycemia, correction based on a patient's usual ICF is safe and effective. Optimal PG reduction, with minimal hypoglycemia, occurred in the 100 and 150% correction arms.
Collapse
Affiliation(s)
| | - Ruth E Brown
- LMC Diabetes & Endocrinology, Toronto, Ontario, Canada
| | - Aihua Li
- LMC Diabetes & Endocrinology, Toronto, Ontario, Canada
| | - Michael C Riddell
- LMC Diabetes & Endocrinology, Toronto, Ontario, Canada.,School of Kinesiology and Health Science, York University, Toronto, Ontario, Canada
| |
Collapse
|
48
|
Rempel LA, Vallet JL, Nonneman DJ. Characterization of plasma metabolites at late gestation and lactation in early parity sows on production and post-weaning reproductive performance. J Anim Sci 2018; 96:521-531. [PMID: 29385465 DOI: 10.1093/jas/skx066] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Accepted: 12/19/2017] [Indexed: 11/12/2022] Open
Abstract
Lactation is a very energy demanding period for sows. The current study provides a better understanding of the biochemical response of first- (n = 246) or second-parity (n = 127) sows during late gestation through lactation and assesses relationships with piglet production and dam reproductive performance. Plasma samples were collected from first- or second-parity dams at late gestation (110 d gestation [d110G]), d 1 post-farrowing (d1PF), and weaning (WN) then analyzed for various stress and protein metabolism compounds, including; creatine, creatine phosphokinase (CPK) activity, creatinine, urea nitrogen, albumin, and lactate. Litter performance was measured as number of piglets nursed and piglet ADG. Post-weaning reproductive performance was assessed by measuring weaning-to-estrus interval (WEI) and subsequent ovulation rate collected at time of harvest. Plasma creatine and CPK activity increased (P < 0.05) between d110G and d1PF. Plasma creatinine decreased (P < 0.05) from d110G through WN in first-parity dams, but remained similar between d110G and d1PF before declining (P < 0.05) at WN in second-parity dams. Plasma urea nitrogen increased (P < 0.05) over the course of the study and was negatively (P < 0.05) associated with piglet ADG at d110G and d1PF and with ovulation rate at d110G (P < 0.05). Similarly, plasma albumin increased (P < 0.05) in first-parity dams over the course of the study, whereas it plateaued (P < 0.05) at d1PF and remained similar (P > 0.10) through WN in second-parity dams. First-parity dams had less (P < 0.05) plasma lactate at d110G than at d1PF or WN. However, second-parity dams had increased (P < 0.05) plasma lactate at d110G and d1PF, then decreased (P < 0.05) levels at WN. Plasma lactate at WN was positively (P < 0.05) associated with WEI in first-parity dams, but negatively (P < 0.05) related to WEI at d1PF in second-parity dams. Plasma lactate levels at all time points were positively (P < 0.05) associated with ovulation rate in second-parity dams. The biochemical profile of these dams differed by parity and merits further investigations into these differences to identify methods to improve physiological response to lactation for improved animal welfare, production, and reproductive performance.
Collapse
Affiliation(s)
- Lea A Rempel
- USDA-ARS, U.S. Meat Animal Research Center, Clay Center, NE
| | | | - Dan J Nonneman
- USDA-ARS, U.S. Meat Animal Research Center, Clay Center, NE
| |
Collapse
|
49
|
Onyango AN. Cellular Stresses and Stress Responses in the Pathogenesis of Insulin Resistance. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:4321714. [PMID: 30116482 PMCID: PMC6079365 DOI: 10.1155/2018/4321714] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/01/2018] [Accepted: 02/18/2018] [Indexed: 12/14/2022]
Abstract
Insulin resistance (IR), a key component of the metabolic syndrome, precedes the development of diabetes, cardiovascular disease, and Alzheimer's disease. Its etiological pathways are not well defined, although many contributory mechanisms have been established. This article summarizes such mechanisms into the hypothesis that factors like nutrient overload, physical inactivity, hypoxia, psychological stress, and environmental pollutants induce a network of cellular stresses, stress responses, and stress response dysregulations that jointly inhibit insulin signaling in insulin target cells including endothelial cells, hepatocytes, myocytes, hypothalamic neurons, and adipocytes. The insulin resistance-inducing cellular stresses include oxidative, nitrosative, carbonyl/electrophilic, genotoxic, and endoplasmic reticulum stresses; the stress responses include the ubiquitin-proteasome pathway, the DNA damage response, the unfolded protein response, apoptosis, inflammasome activation, and pyroptosis, while the dysregulated responses include the heat shock response, autophagy, and nuclear factor erythroid-2-related factor 2 signaling. Insulin target cells also produce metabolites that exacerbate cellular stress generation both locally and systemically, partly through recruitment and activation of myeloid cells which sustain a state of chronic inflammation. Thus, insulin resistance may be prevented or attenuated by multiple approaches targeting the different cellular stresses and stress responses.
Collapse
Affiliation(s)
- Arnold N. Onyango
- Department of Food Science and Technology, Jomo Kenyatta University of Agriculture and Technology, P.O. Box 62000, Nairobi 00200, Kenya
| |
Collapse
|
50
|
Della Guardia L, Thomas MA, Cena H. Insulin Sensitivity and Glucose Homeostasis Can Be Influenced by Metabolic Acid Load. Nutrients 2018; 10:E618. [PMID: 29762478 PMCID: PMC5986498 DOI: 10.3390/nu10050618] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Revised: 05/08/2018] [Accepted: 05/10/2018] [Indexed: 01/04/2023] Open
Abstract
Recent epidemiological findings suggest that high levels of dietary acid load can affect insulin sensitivity and glucose metabolism. Consumption of high protein diets results in the over-production of metabolic acids which has been associated with the development of chronic metabolic disturbances. Mild metabolic acidosis has been shown to impair peripheral insulin action and several epidemiological findings suggest that metabolic acid load markers are associated with insulin resistance and impaired glycemic control through an interference intracellular insulin signaling pathways and translocation. In addition, higher incidence of diabetes, insulin resistance, or impaired glucose control have been found in subjects with elevated metabolic acid load markers. Hence, lowering dietary acid load may be relevant for improving glucose homeostasis and prevention of type 2 diabetes development on a long-term basis. However, limitations related to patient acid load estimation, nutritional determinants, and metabolic status considerably flaws available findings, and the lack of solid data on the background physiopathology contributes to the questionability of results. Furthermore, evidence from interventional studies is very limited and the trials carried out report no beneficial results following alkali supplementation. Available literature suggests that poor acid load control may contribute to impaired insulin sensitivity and glucose homeostasis, but it is not sufficiently supportive to fully elucidate the issue and additional well-designed studies are clearly needed.
Collapse
Affiliation(s)
- Lucio Della Guardia
- Laboratory of Dietetics and Clinical Nutrition Department of Public Health, Experimental and Forensic Medicine, University of Pavia, 27100 Pavia, Italy.
| | - Michael Alex Thomas
- Department of Biology, Center for Obesity Reversal, Georgia State University, Atlanta, GA 30302, USA.
| | - Hellas Cena
- Laboratory of Dietetics and Clinical Nutrition Department of Public Health, Experimental and Forensic Medicine, University of Pavia, 27100 Pavia, Italy.
| |
Collapse
|