1
|
Naik AR, Save SN, Sahoo SS, Yadav SS, Kumar A, Chugh J, Sharma S. Metabolic perturbations associated with hIAPP-induced insulin resistance in skeletal muscles: Implications to the development of type 2 diabetes. Int J Biochem Cell Biol 2024; 176:106665. [PMID: 39322038 DOI: 10.1016/j.biocel.2024.106665] [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: 04/23/2024] [Revised: 09/16/2024] [Accepted: 09/18/2024] [Indexed: 09/27/2024]
Abstract
The human islet amyloid polypeptide (hIAPP) tends to misfold and self-assemble to form amyloid fibrils, which has been associated with the loss of function and viability of pancreatic β-cells in type 2 diabetes mellitus (T2DM). The role of hIAPP in the development of insulin resistance (a hallmark of T2DM) in skeletal muscles - the major sites for glucose utilization - needs further investigation. Even though, insulin-resistant conditions have been known to stimulate hIAPP aggregation, the events that lead to the development of insulin resistance due to hIAPP aggregation in skeletal muscles remain unidentified. Here, we have attempted to identify metabolic perturbations in L6 myotubes that were exposed to increasing concentrations of recombinant hIAPP for different time durations. It was observed that hIAPP exposure was associated with increased mitochondrial and cellular ROS levels, loss in mitochondrial membrane potential and viability of the myotubes. Metabolomic investigations of hIAPP-treated myotubes revealed significant perturbations in o-phosphocholine, sn-glycero-3-phosphocholine and dimethylamine levels (p < 0.05). Therefore, we anticipate that defects in glycerophospholipid metabolism and the associated oxidative stress and membrane damage may play key roles in the development of insulin resistance due to protein misfolding in skeletal muscles. In summary, the perturbed metabolites and their pathways have not only the potential to be used as early biomarkers to predict the onset of insulin resistance and T2DM but also as therapeutic targets for the effective management of the same.
Collapse
Affiliation(s)
- Arya R Naik
- Department of Biotechnology, Savitribai Phule Pune University, Ganeshkhind Road, Pune 411007, India
| | - Shreyada N Save
- Department of Biotechnology, Savitribai Phule Pune University, Ganeshkhind Road, Pune 411007, India
| | - Soumya S Sahoo
- Department of Chemistry, Indian Institute of Science Education and Research (IISER), Dr. Homi Bhabha Road, Pashan, Pune 411008, India
| | - Saurabh S Yadav
- Department of Biotechnology, Savitribai Phule Pune University, Ganeshkhind Road, Pune 411007, India
| | - Ashutosh Kumar
- Department of Biosciences and Bioengineering, Indian institute of technology Bombay, Powai, Mumbai, Maharashtra 400076, India
| | - Jeetender Chugh
- Department of Chemistry, Indian Institute of Science Education and Research (IISER), Dr. Homi Bhabha Road, Pashan, Pune 411008, India
| | - Shilpy Sharma
- Department of Biotechnology, Savitribai Phule Pune University, Ganeshkhind Road, Pune 411007, India.
| |
Collapse
|
2
|
Osmond AD, Leija RG, Arevalo JA, Curl CC, Duong JJ, Huie MJ, Masharani U, Brooks GA. Aging delays the suppression of lipolysis and fatty acid oxidation in the postprandial period. J Appl Physiol (1985) 2024; 137:1200-1219. [PMID: 39236144 PMCID: PMC11563596 DOI: 10.1152/japplphysiol.00437.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 08/14/2024] [Accepted: 08/29/2024] [Indexed: 09/07/2024] Open
Abstract
Plasma glycerol and free fatty acid concentrations decrease following oral glucose consumption, but changes in the rate of lipolysis during an oral glucose tolerance test (OGTT) have not been documented in conjunction with changes in fatty acid (FA) oxidation or reesterification rates in healthy individuals. After a 12-h overnight fast, 15 young (21-35 yr; 7 men and 8 women) and 14 older (60-80 yr; 7 men and 7 women) participants had the forearm vein catheterized for primed continuous infusion of [1,1,2,3,3-2H]glycerol. A contralateral hand vein was catheterized for arterialized blood sampling. Indirect calorimetry was performed simultaneously to determine total FA and carbohydrate (CHO) oxidation rates (Rox). Total FA reesterification rates (Rs) were estimated from tracer-measured lipolytic and FA oxidation rates. After a 90-min equilibration period, participants underwent a 120-min, 75-g OGTT. Glycerol rate of appearance (Ra), an index of lipolysis, decreased significantly from baseline 5 min postchallenge in young participants and 30 min in older participants. At 60 min, FA Rox decreased in both groups, but was significantly higher in older participants. Between 5 and 90 min, CHO Rox was significantly lower in older participants. In addition, FA Rs was significantly lower in older participants at 60 and 90 min. The area under the curve (AUC) for FA Rox was greater than that for FA Rs in older, but not in young participants. Our results indicate that, in aging, the postprandial suppression of lipolysis and FA oxidation are delayed such that FA oxidation is favored over CHO oxidation and FA reesterification.NEW & NOTEWORTHY To our knowledge, our investigation is the first to demonstrate changes in lipolysis during an oral glucose tolerance test (OGTT) in healthy young and older individuals. Plasma glycerol and free fatty acid concentrations changed after glycerol rate of appearance (Ra), indicating that plasma concentrations are incomplete surrogates of the lipolytic rate. Moreover, simultaneous determinations of substrate oxidation rates are interpreted to indicate that metabolic inflexibility in aging is characterized by delayed changes in postprandial substrate utilization related to the lipolytic rate.
Collapse
Affiliation(s)
- Adam D Osmond
- Exercise Physiology Laboratory, Department of Integrative Biology, University of California, Berkeley, California, United States
| | - Robert G Leija
- Exercise Physiology Laboratory, Department of Integrative Biology, University of California, Berkeley, California, United States
| | - Jose A Arevalo
- Exercise Physiology Laboratory, Department of Integrative Biology, University of California, Berkeley, California, United States
| | - Casey C Curl
- Exercise Physiology Laboratory, Department of Integrative Biology, University of California, Berkeley, California, United States
| | - Justin J Duong
- Exercise Physiology Laboratory, Department of Integrative Biology, University of California, Berkeley, California, United States
| | - Melvin J Huie
- Exercise Physiology Laboratory, Department of Integrative Biology, University of California, Berkeley, California, United States
| | - Umesh Masharani
- Division of Endocrinology, Department of Medicine, University of California, San Francisco, California, United States
| | - George A Brooks
- Exercise Physiology Laboratory, Department of Integrative Biology, University of California, Berkeley, California, United States
| |
Collapse
|
3
|
Tseng CH. Metformin-associated lactic acidosis and mortality in type 2 diabetes patients hospitalized with heart failure and/or acute coronary syndrome: A neglected clinical scenario and the potential role of insulin. Int J Cardiol 2024; 414:132400. [PMID: 39089483 DOI: 10.1016/j.ijcard.2024.132400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Accepted: 07/26/2024] [Indexed: 08/04/2024]
Affiliation(s)
- Chin-Hsiao Tseng
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Far Eastern Polyclinic, Taipei 10043, Taiwan; Division of Endocrinology and Metabolism, Department of Internal Medicine, National Taiwan University Hospital, Taipei 10002, Taiwan; Department of Internal Medicine, National Taiwan University College of Medicine, Taipei 10051, Taiwan.
| |
Collapse
|
4
|
Shukla R, Lamichhane K, Pandey D, Gupta CK, Shukla S. Insulin in aluminum phosphide poisoning: A systematic review of the current literature. Medicine (Baltimore) 2024; 103:e40066. [PMID: 39432630 PMCID: PMC11495704 DOI: 10.1097/md.0000000000040066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 09/13/2024] [Indexed: 10/23/2024] Open
Abstract
BACKGROUND Aluminum phosphide (AlP) is a commonly used fumigant in agriculture and grain preservation because of its high potency and low cost. Due to the absence of a specific antidote and promising treatment modality, poisoning with this substance is deadly. Amid multiple studies in different parts of the world, each exploring options like lavage, inotropes, antioxidants, etc, we conducted a systematic review to find the possible role of exogenous insulin in treating symptomatic cases of AlP poisoning. METHODS Experimental studies released before February 15, 2024, that reported the use of exogenous insulin were systematically reviewed following the Preferred Reporting Items for Systematic Review and Meta-Analysis statement. The search was done on PubMed Central, Cochrane Library, and Google Scholar. RESULTS After applying the inclusion and exclusion criteria, we finalized a few scientific papers for the review. Studying data from 4 scientific papers (3 quasi-experimental studies and 1 randomized controlled trial), we could postulate the significant improvement in survivability after the inclusion of exogenous insulin in the treatment of poisoned cases of AlP. Studies showed divergent results for the blood pressure, blood gases, and need for mechanical ventilation. Hypoglycemia, hyperglycemia, and hypokalemia were the reported adverse effects of this therapy. CONCLUSION Our review found that the use of exogenous insulin in AlP poisoning reduced mortality rates, which was consistent across all studies. With available knowledge, its inclusion as a part of therapy might be beneficial in AlP poisoning, but to put it forward confidently, we still need high-quality randomized control trials. It is indeed a subject of interest for future research.
Collapse
Affiliation(s)
- Ravi Shukla
- Nepal Medical College and Teaching Hospital, Kathmandu, Nepal
| | | | - Dhritee Pandey
- Nepal Army Institute of Health Sciences, Kathmandu, Nepal
| | | | - Sashank Shukla
- Nepal Army Institute of Health Sciences, Kathmandu, Nepal
| |
Collapse
|
5
|
Brossaud J, Barat P, Moisan MP. Cognitive Disorders in Type 1 Diabetes: Role of Brain Glucose Variation, Insulin Activity, and Glucocorticoid Exposure. Neuroendocrinology 2024:1-15. [PMID: 39401497 DOI: 10.1159/000541989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Accepted: 10/09/2024] [Indexed: 11/19/2024]
Abstract
BACKGROUND The number of patients with type 2 diabetes (T2D) and type 1 diabetes (T1D) is on the rise, partly due to a global increase in new T1D cases among children. Beyond the well-documented microvascular and macrovascular complications, there is now substantial evidence indicating that diabetes also impacts the brain, leading to neuropsychological impairments. The risk of developing neuropsychiatric symptoms is notably higher in childhood due to the ongoing maturation of the brain, which makes it more susceptible to damage. Despite this awareness, the specific effects of diabetes on cognitive function remain poorly understood. SUMMARY This review synthesizes literature on the impact of diabetes on cognition and its relationship with brain structural changes. It presents data and hypotheses to explain how T1D contributes to cognitive dysfunction, with a particular focus on children and adolescents. The emphasis on the pediatric population is intentional, as young diabetic patients typically have fewer comorbidities, reducing confounding factors and simplifying the investigation of cognitive alterations. KEY MESSAGE We examine the roles of hypo- and hyperglycemia, as well as the emerging role of glucocorticoids in the development of neuropsychological disorders. When specific mechanisms related to T1D are available, they are highlighted; otherwise, data and hypotheses applicable to both T1D and T2D are discussed.
Collapse
Affiliation(s)
- Julie Brossaud
- Univ. Bordeaux, INRAE, Bordeaux INP, NutriNeurO, UMR 1286, Team NutriPsy, Bordeaux, France
- CHU Bordeaux, Nuclear Medicine, Pessac, France
| | - Pascal Barat
- Univ. Bordeaux, INRAE, Bordeaux INP, NutriNeurO, UMR 1286, Team NutriPsy, Bordeaux, France
- CHU Bordeaux, Pediatric Endocrinology and DiaBEA Unit, Hôpital des Enfants, Bordeaux, France
| | - Marie-Pierre Moisan
- Univ. Bordeaux, INRAE, Bordeaux INP, NutriNeurO, UMR 1286, Team NutriPsy, Bordeaux, France
| |
Collapse
|
6
|
Arevalo JA, Leija RG, Osmond AD, Curl CC, Duong JJ, Huie MJ, Masharani U, Brooks GA. Delayed and diminished postprandial lactate shuttling in healthy older men and women. Am J Physiol Endocrinol Metab 2024; 327:E430-E440. [PMID: 39110417 PMCID: PMC11482286 DOI: 10.1152/ajpendo.00183.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 07/22/2024] [Accepted: 07/30/2024] [Indexed: 09/19/2024]
Abstract
Lactate, a product of glycolysis, is formed under aerobic conditions. Extensive work has shown lactate flux in young and exercising humans; however, the effect of age is not known. We tested the hypothesis that postprandial lactate shuttling (PLS) would be diminished in older adults. We used [3-13C]lactate and [6,6-2H]glucose tracers, an oral glucose tolerance test (OGTT), and arterialized blood sampling to determine postprandial lactate rates of appearance (Ra), disappearance (Rd), and oxidation (Rox) in 15 young (28.1 ± 1.4 yr) and 13 older (70.6 ± 2.4 yr) healthy men and women. In young participants, fasting blood [lactate] (≈0.5 mM) rose after the glucose challenge, peaked at 15 min, dipped to a nadir at 30 min, and rose again peaking at 60 min (≈1.0 mM). Initial responses in lactate Ra of older participants were delayed and diminished until 90 min rising by 0.83 mg·kg-1·min-1. Lactate Rox was higher throughout the entire trial in young participants by a difference of ∼0.5 mg·kg-1·min-1. Initial peaks in lactate Ra and concentration in all volunteers demonstrated the presence of an enteric PLS following an OGTT. Notably, in the systemic, but not enteric, PLS phase, lactate Ra correlated highly with glucose Rd (r2 = 0.92). Correspondence of second peaks in lactate Ra and concentration and glucose Rd shows dependence of lactate Ra on glucose Rd. Although results show both enteric and systemic PLS phases in young and older study cohorts, metabolic responses were delayed and diminished in healthy older individuals.NEW & NOTEWORTHY We used isotope tracers, an oral glucose tolerance test, and arterialized blood sampling to determine postprandial lactate flux rates in healthy young and older men and women. Lactate rates of appearance and oxidation and the lactate-pyruvate exchange were delayed and diminished in both enteric and systemic postprandial lactate shuttle phases in older participants.
Collapse
Affiliation(s)
- Jose A. Arevalo
- Exercise Physiology Laboratory, Department of Integrative Biology, University of California, Berkeley, United States
| | - Robert G. Leija
- Exercise Physiology Laboratory, Department of Integrative Biology, University of California, Berkeley, United States
| | - Adam D. Osmond
- Exercise Physiology Laboratory, Department of Integrative Biology, University of California, Berkeley, United States
| | - Casey C. Curl
- Exercise Physiology Laboratory, Department of Integrative Biology, University of California, Berkeley, United States
| | - Justin J. Duong
- Exercise Physiology Laboratory, Department of Integrative Biology, University of California, Berkeley, United States
| | - Melvin J. Huie
- Exercise Physiology Laboratory, Department of Integrative Biology, University of California, Berkeley, United States
| | - Umesh Masharani
- Department of Medicine, University of California, San Francisco, United States
| | - George A. Brooks
- Exercise Physiology Laboratory, Department of Integrative Biology, University of California, Berkeley, United States
| |
Collapse
|
7
|
Pohl A, Schünemann F, Schaaf K, Yang W, Heck H, Heine O, Jacko D, Gehlert S. Increased resting lactate levels and reduced carbohydrate intake cause νLa.max underestimation by reducing net lactate accumulation-A pilot study in young adults. Physiol Rep 2024; 12:e70020. [PMID: 39187400 PMCID: PMC11347020 DOI: 10.14814/phy2.70020] [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: 05/18/2024] [Revised: 08/16/2024] [Accepted: 08/16/2024] [Indexed: 08/28/2024] Open
Abstract
Modulation of testing conditions such as resting lactate (Larest) levels or carbohydrate intake may affect the calculation of the maximal glycolytic rate (νLa.max). To evaluate the impact of elevated Larest as well as reduced and increased carbohydrate availability on νLa.max in running sprints (RST), twenty-one participants completed five 15-s RST tests on a running track under five different conditions: (I). baseline: Larest ≤1.5 mmol·L-1; (II). Lactate+: Larest ≥2.5 mmol·L-1; (III). CHO-: carbohydrate intake: ≤ 1 g·kg-1 BW d-1 for 3 days; (IV). CHO+: carbohydrate intake: ≥ 9 g·kg-1 BW d-1 for one day; and (V). acuteCHO: 500 mL glucose containing beverage consumed before RST. νLa.max was significantly reduced in lactate+ and CHO- conditions compared to the baseline RST, due to a reduction in the arithmetic mean delta (∆) between Lapeak and Larest lactate concentration (Lapeak, mmol · L-1). AcuteCHO led to an increase in Larest compared to baseline, CHO- and CHO+ with a high interindividual variability but did not significantly reduce νLa.max. Therefore, avoiding low carbohydrate nutrition before νLa.max testing, along with carefully adjusting Larest to below ≤1.5 mmol·L-1, is crucial to prevent the unintentional underestimation of νLa.max.
Collapse
Affiliation(s)
- Alexander Pohl
- Department for Biosciences of SportsInstitute of Sport Science, University of HildesheimHildesheimGermany
| | - Frederik Schünemann
- Department for Biosciences of SportsInstitute of Sport Science, University of HildesheimHildesheimGermany
| | - Kirill Schaaf
- Institute of Cardiovascular Research and Sports Medicine, German Sport University CologneCologneGermany
- Olympic Base Center, North Rhine‐Westphalia/RhinelandCologneGermany
| | - Woo‐Hwi Yang
- Graduate School of Sports MedicineCHA UniversityPocheonRepublic of Korea
| | - Hermann Heck
- Faculty for Sports SciencesRuhr Universität BochumBochumGermany
| | - Oliver Heine
- Olympic Base Center, North Rhine‐Westphalia/RhinelandCologneGermany
| | - Daniel Jacko
- Institute of Cardiovascular Research and Sports Medicine, German Sport University CologneCologneGermany
- Olympic Base Center, North Rhine‐Westphalia/RhinelandCologneGermany
| | - Sebastian Gehlert
- Department for Biosciences of SportsInstitute of Sport Science, University of HildesheimHildesheimGermany
- Institute of Cardiovascular Research and Sports Medicine, German Sport University CologneCologneGermany
| |
Collapse
|
8
|
Manning JE, Harris E, Mathieson H, Sorensen L, Luqmani R, McGettrick HM, Morgan AW, Young SP, Mackie SL. Polymyalgia rheumatica shows metabolomic alterations that are further altered by glucocorticoid treatment: Identification of metabolic correlates of fatigue. J Autoimmun 2024; 147:103260. [PMID: 38797046 DOI: 10.1016/j.jaut.2024.103260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 04/17/2024] [Accepted: 05/21/2024] [Indexed: 05/29/2024]
Abstract
OBJECTIVE In polymyalgia rheumatica (PMR), glucocorticoids (GCs) relieve pain and stiffness, but fatigue may persist. We aimed to explore the effect of disease, GCs and PMR symptoms in the metabolite signatures of peripheral blood from patients with PMR or the related disease, giant cell arteritis (GCA). METHODS Nuclear magnetic resonance spectroscopy was performed on serum from 40 patients with untreated PMR, 84 with new-onset confirmed GCA, and 53 with suspected GCA who later were clinically confirmed non-GCA, and 39 age-matched controls. Further samples from PMR patients were taken one and six months into glucocorticoid therapy to explore relationship of metabolites to persistent fatigue. 100 metabolites were identified using Chenomx and statistical analysis performed in SIMCA-P to examine the relationship between metabolic profiles and, disease, GC treatment or symptoms. RESULTS The metabolite signature of patients with PMR and GCA differed from that of age-matched non-inflammatory controls (R2 > 0.7). There was a smaller separation between patients with clinically confirmed GCA and those with suspected GCA who later were clinically confirmed non-GCA (R2 = 0.135). In PMR, metabolite signatures were further altered with glucocorticoid treatment (R2 = 0.42) but did not return to that seen in controls. Metabolites correlated with CRP, pain, stiffness, and fatigue (R2 ≥ 0.39). CRP, pain, and stiffness declined with treatment and were associated with 3-hydroxybutyrate and acetoacetate, but fatigue did not. Metabolites differentiated patients with high and low fatigue both before and after treatment (R2 > 0.9). Low serum glutamine was predictive of high fatigue at both time points (0.79-fold change). CONCLUSION PMR and GCA alter the metabolite signature. In PMR, this is further altered by glucocorticoid therapy. Treatment-induced metabolite changes were linked to measures of inflammation (CRP, pain and stiffness), but not to fatigue. Furthermore, metabolite signatures distinguished patients with high or low fatigue.
Collapse
Affiliation(s)
- Julia E Manning
- Institute for Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Birmingham, B15-2TT, UK.
| | - Emma Harris
- School of Medicine, University of Leeds, Leeds, LS7 4SA, UK and School of Human and Health Sciences, University of Huddersfield, Huddersfield, UK.
| | - Hannah Mathieson
- School of Medicine, University of Leeds, Leeds, LS7 4SA, UK and Leeds NIHR Biomedical Research Centre, Leeds Teaching Hospitals NHS Trust, Leeds, UK.
| | - Louise Sorensen
- School of Medicine, University of Leeds, Leeds, LS7 4SA, UK.
| | - Raashid Luqmani
- NIHR Musculoskeletal Biomedical Research Unit, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Science, University of Oxford, Oxford, UK.
| | - Helen M McGettrick
- Institute for Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Birmingham, B15-2TT, UK.
| | - Ann W Morgan
- School of Medicine, University of Leeds, Leeds, School of Human and Health Sciences, University of Huddersfield, Huddersfield, And Leeds NIHR Medtech and in Vitro Diagnostics Co-operative, Leeds Teaching Hospitals NHS Trust, Leeds, LS7 4SA, UK.
| | - Stephen P Young
- Institute for Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Birmingham, B15-2TT, UK.
| | - Sarah L Mackie
- School of Medicine, University of Leeds, Leeds, LS7 4SA, UK and School of Human and Health Sciences, University of Huddersfield, Huddersfield, UK.
| |
Collapse
|
9
|
Huang X, Yip K, Nie H, Chen R, Wang X, Wang Y, Lin W, Li R. ChIP-seq and RNA-seq Reveal the Involvement of Histone Lactylation Modification in Gestational Diabetes Mellitus. J Proteome Res 2024. [PMID: 38776154 DOI: 10.1021/acs.jproteome.3c00727] [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: 05/24/2024]
Abstract
Lactylation is a novel post-translational modification of proteins. Although the histone lactylation modification has been reported to be involved in glucose metabolism, its role and molecular pathways in gestational diabetes mellitus (GDM) are still unclear. This study aims to elucidate the histone lactylation modification landscapes of GDM patients and explore lactylation-modification-related genes involved in GDM. We employed a combination of RNA-seq analysis and chromatin immunoprecipitation sequencing (ChIP-seq) analysis to identify upregulated differentially expressed genes (DEGs) with hyperhistone lactylation modification in GDM. We demonstrated that the levels of lactate and histone lactylation were significantly elevated in GDM patients. DEGs were involved in diabetes-related pathways, such as the PI3K-Akt signaling pathway, Jak-STAT signaling pathway, and mTOR signaling pathway. ChIP-seq analysis indicated that histone lactylation modification in the promoter regions of the GDM group was significantly changed. By integrating the results of RNA-seq and ChIP-seq analysis, we found that CACNA2D1 is a key gene for histone lactylation modification and is involved in the progression of GDM by promoting cell vitality and proliferation. In conclusion, we identified the key gene CACNA2D1, which upregulated and exhibited hypermodification of histone lactylation in GDM. These findings establish a theoretical groundwork for the targeted therapy of GDM.
Collapse
Affiliation(s)
- Xiaman Huang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - KaCheuk Yip
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Hanhui Nie
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Ruiping Chen
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Xiufang Wang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Yun Wang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Weizhao Lin
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Ruiman Li
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| |
Collapse
|
10
|
Bima A, Eldakhakhny B, Alamoudi AA, Awan Z, Alnami A, Abo-Elkhair SM, Sakr H, Ghoneim FM, Elsamanoudy A. Molecular Study of the Protective Effect of a Low-Carbohydrate, High-Fat Diet against Brain Insulin Resistance in an Animal Model of Metabolic Syndrome. Brain Sci 2023; 13:1383. [PMID: 37891752 PMCID: PMC10605073 DOI: 10.3390/brainsci13101383] [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: 09/04/2023] [Revised: 09/25/2023] [Accepted: 09/27/2023] [Indexed: 10/29/2023] Open
Abstract
Brain insulin resistance is linked to metabolic syndrome (MetS). A low-carbohydrate, high-fat (LCHF) diet has been proposed to have a protective effect. Therefore, this study aimed to investigate the brain insulin resistance markers in a rat animal model of MetS and the protective effects of the LCHF diet. Four groups of male rats (10/group) were created. Group I (Control) was fed a regular diet. Groups II-IV were injected with dexamethasone (DEX) to induce MetS. Group II received DEX with a regular diet. Group III (DEX + LCHF) rates were fed a low-carbohydrate, high-fat diet, while Group IV (DEX + HCLF) rats were fed a high-carbohydrate, low-fat (HCLF) diet. At the end of the four-week experiment, HOMA-IR was calculated. Moreover, cerebral gene expression analysis of S-100B, BDNF, TNF-α, IGF-1, IGF-1 R, IGFBP-2, IGFBP-5, Bax, Bcl-2, and caspase-3 was carried out. In the DEX group, rats showed a significant increase in the HOMA-IR and a decrease in the gene expression of IGF-1, IGF-1 R, IGFBP-2, IGFBP-5, BDNF, and Bcl2, with a concomitant rise in S100B, TNF-α, Bax, and caspase-3. The LCHF diet group showed a significantly opposite effect on all parameters. In conclusion, MetS is associated with dysregulated cerebral gene expression of BDNF, S100B, and TNF-α and disturbed IGF-1 signaling, with increased apoptosis and neuroinflammation. Moreover, the LCHF diet showed a protective effect, as evidenced by preservation of the investigated biochemical and molecular parameters.
Collapse
Affiliation(s)
- Abdulhadi Bima
- Clinical Biochemistry Department, Faculty of Medicine, King Abdulaziz University, Jeddah 21465, Saudi Arabia; (A.B.); (B.E.); (A.A.A.); (Z.A.); (A.A.)
| | - Basmah Eldakhakhny
- Clinical Biochemistry Department, Faculty of Medicine, King Abdulaziz University, Jeddah 21465, Saudi Arabia; (A.B.); (B.E.); (A.A.A.); (Z.A.); (A.A.)
- Food, Nutrition, and Lifestyle Research Unit, King Fahd for Medical Research Centre, King Abdulaziz University, Jeddah 21465, Saudi Arabia
| | - Aliaa A. Alamoudi
- Clinical Biochemistry Department, Faculty of Medicine, King Abdulaziz University, Jeddah 21465, Saudi Arabia; (A.B.); (B.E.); (A.A.A.); (Z.A.); (A.A.)
| | - Zuhier Awan
- Clinical Biochemistry Department, Faculty of Medicine, King Abdulaziz University, Jeddah 21465, Saudi Arabia; (A.B.); (B.E.); (A.A.A.); (Z.A.); (A.A.)
| | - Abrar Alnami
- Clinical Biochemistry Department, Faculty of Medicine, King Abdulaziz University, Jeddah 21465, Saudi Arabia; (A.B.); (B.E.); (A.A.A.); (Z.A.); (A.A.)
| | - Salwa Mohamed Abo-Elkhair
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt;
| | - Hussein Sakr
- Physiology Department, College of Medicine and Health Sciences, Sultan Qaboos University, Muscat 123, Oman;
- Medical Physiology Department, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt
| | - Fatma Mohamed Ghoneim
- Faculty Development Unit, Physiological Science and Medical Education Department, Fakeeh College for Medical Sciences, Jeddah 23323, Saudi Arabia;
| | - Ayman Elsamanoudy
- Clinical Biochemistry Department, Faculty of Medicine, King Abdulaziz University, Jeddah 21465, Saudi Arabia; (A.B.); (B.E.); (A.A.A.); (Z.A.); (A.A.)
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt;
| |
Collapse
|
11
|
Racil G, Russo L, Migliaccio GM, Signorelli P, Larion A, Padulo J, Jlid MC. High-Intensity Interval Training in Female Adolescents with Moderate or Severe Obesity. CHILDREN (BASEL, SWITZERLAND) 2023; 10:1495. [PMID: 37761456 PMCID: PMC10528164 DOI: 10.3390/children10091495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 08/25/2023] [Accepted: 08/28/2023] [Indexed: 09/29/2023]
Abstract
This study aimed to investigate the impact of moderate- or high-intensity interval training (MIIT or HIIT) on anthropometric and biological measurements in four groups of females with obesity. Fifty-seven participants were divided into a moderate obesity group (MOG, n = 29) and a severe obesity group (SOG, n = 28). Two sub-groups were established to practice HIIT and MIIT programs (SOGHI, n = 14; SOGMI, n = 14; MOGHI, n = 14; MOGMI, n = 15). During the training sessions, each group performed two sets of 4 × 1 min intervals on a cycle ergometer. The intervals were conducted at 65% and 85% of the heart rate reserve (HRR) for MIIT and HIIT, respectively. Between each repetition, there was an active recovery phase at 50% HRR, and, between sets, there was a 4 min period of free pedaling. All groups significantly improved their anthropometric data, while only MOGHI and SOGHI significantly improved their lean body mass (LBM) and blood lactate (BL), with p ˂ 0.05; the higher percentage of change in blood insulin levels (-25.49 and -25.34) and the homeostasis model assessment of the insulin resistance index (-31.42 and -28.88) were noted. Only MOGHI showed improvements in growth hormone (GH) and blood glucose (p < 0.05), which were negatively correlated with body fat percentage (r = -0.76 and r = -0.72) and waist circumference (r = -0.77 and r = -0.82), respectively. We may conclude that HIIT was an effective method of managing anthropometric and biological parameters, as confirmed by the pronounced body fat reduction in the moderate obesity group.
Collapse
Affiliation(s)
- Ghazi Racil
- Research Unit (UR 17JS01) “Sport Performance, Health & Society” Higher Institute of Sport and Physical Education of Ksar Said, Tunis 1000, Tunisia; (G.R.); (M.C.J.)
- Department of Biological Sciences Applied for Physical Activities and Sport, Higher Institute of Sport and Physical Education of Ksar Said, University of La Manouba, Manouba 2010, Tunisia
| | - Luca Russo
- Department of Human Sciences, Università Telematica degli Studi IUL, 50122 Florence, Italy;
| | - Gian Mario Migliaccio
- Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Rome Open University, 00166 Rome, Italy;
| | - Paola Signorelli
- Institute for Molecular and Translational Cardiology (IMTC), San Donato Milanese, 20097 Milan, Italy;
- Aldo Ravelli Center for Neurotechnology and Experimental Brain Therapeutics, Department of Health Sciences, University of Milan, Via Antonio di Rudinì 8, 20142 Milan, Italy
| | - Alin Larion
- Faculty of Physical Education and Sport, Ovidius University of Constanta, 900029 Constanta, Romania;
| | - Johnny Padulo
- Department of Biomedical Sciences for Health, Università degli Studi di Milano, 20133 Milan, Italy
| | - Mohamed Chedly Jlid
- Research Unit (UR 17JS01) “Sport Performance, Health & Society” Higher Institute of Sport and Physical Education of Ksar Said, Tunis 1000, Tunisia; (G.R.); (M.C.J.)
- Department of Biological Sciences Applied for Physical Activities and Sport, Higher Institute of Sport and Physical Education of Ksar Said, University of La Manouba, Manouba 2010, Tunisia
| |
Collapse
|
12
|
Flockhart M, Tischer D, Nilsson LC, Blackwood SJ, Ekblom B, Katz A, Apró W, Larsen FJ. Reduced glucose tolerance and insulin sensitivity after prolonged exercise in endurance athletes. Acta Physiol (Oxf) 2023; 238:e13972. [PMID: 37017615 DOI: 10.1111/apha.13972] [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] [Received: 12/21/2022] [Revised: 03/30/2023] [Accepted: 03/31/2023] [Indexed: 04/06/2023]
Abstract
AIM The purpose of this study was to 1. investigate if glucose tolerance is affected after one acute bout of different types of exercise; 2. assess if potential differences between two exercise paradigms are related to changes in mitochondrial function; and 3. determine if endurance athletes differ from nonendurance-trained controls in their metabolic responses to the exercise paradigms. METHODS Nine endurance athletes (END) and eight healthy nonendurance-trained controls (CON) were studied. Oral glucose tolerance tests (OGTT) and mitochondrial function were assessed on three occasions: in the morning, 14 h after an overnight fast without prior exercise (RE), as well as after 3 h of prolonged continuous exercise at 65% of VO2 max (PE) or 5 × 4 min at ~95% of VO2 max (HIIT) on a cycle ergometer. RESULTS Glucose tolerance was markedly reduced in END after PE compared with RE. END also exhibited elevated fasting serum FFA and ketones levels, reduced insulin sensitivity and glucose oxidation, and increased fat oxidation during the OGTT. CON showed insignificant changes in glucose tolerance and the aforementioned measurements compared with RE. HIIT did not alter glucose tolerance in either group. Neither PE nor HIIT affected mitochondrial function in either group. END also exhibited increased activity of 3-hydroxyacyl-CoA dehydrogenase activity in muscle extracts vs. CON. CONCLUSION Prolonged exercise reduces glucose tolerance and increases insulin resistance in endurance athletes the following day. These findings are associated with an increased lipid load, a high capacity to oxidize lipids, and increased fat oxidation.
Collapse
Affiliation(s)
- Mikael Flockhart
- Department of Physiology, Nutrition and Biomechanics, The Swedish School of Sport and Health Sciences, GIH, Stockholm, Sweden
| | - Dominik Tischer
- Department of Physiology, Nutrition and Biomechanics, The Swedish School of Sport and Health Sciences, GIH, Stockholm, Sweden
| | - Lina C Nilsson
- Department of Physiology, Nutrition and Biomechanics, The Swedish School of Sport and Health Sciences, GIH, Stockholm, Sweden
| | - Sarah J Blackwood
- Department of Physiology, Nutrition and Biomechanics, The Swedish School of Sport and Health Sciences, GIH, Stockholm, Sweden
| | - Björn Ekblom
- Department of Physiology, Nutrition and Biomechanics, The Swedish School of Sport and Health Sciences, GIH, Stockholm, Sweden
| | - Abram Katz
- Department of Physiology, Nutrition and Biomechanics, The Swedish School of Sport and Health Sciences, GIH, Stockholm, Sweden
| | - William Apró
- Department of Physiology, Nutrition and Biomechanics, The Swedish School of Sport and Health Sciences, GIH, Stockholm, Sweden
- Department of Clinical Sciences, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Filip J Larsen
- Department of Physiology, Nutrition and Biomechanics, The Swedish School of Sport and Health Sciences, GIH, Stockholm, Sweden
| |
Collapse
|
13
|
Racil G, Chelly MS, Coquart J, Padulo J, Teodor DF, Russo L. Long- and Short-Term High-Intensity Interval Training on Lipid Profile and Cardiovascular Disorders in Obese Male Adolescents. CHILDREN (BASEL, SWITZERLAND) 2023; 10:1180. [PMID: 37508677 PMCID: PMC10378083 DOI: 10.3390/children10071180] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 06/28/2023] [Accepted: 07/03/2023] [Indexed: 07/30/2023]
Abstract
This study investigated the effects of short-term and long-term periods (8 and 16 weeks) of high-intensity interval training (HIIT) on cardiovascular components, blood lipids, and 6-min walking test performance in obese young boys (age = 16.2 ± 0.7) with >34% body fat. The participants were split into two groups: severe obesity (SOG; n = 17) and moderate obesity (MOG; n = 16). All participants performed on a cycle ergometer for 16 weeks (3 times per week) of HIIT at 100% peak power output at the ventilatory threshold and recovered at 50% of peak power. Except for BMI, both groups improved all body composition measures after 16 weeks, with a higher percentage of change (Δ) in SOG. The 6-min walking test increased in both groups (p < 0.001). Furthermore, cardiovascular variables, blood lactate concentration at rest and after 5-min post-exercise, blood lipids, and insulin concentrations improved significantly in both groups. After 16 weeks, MOG significantly improved in HRpeak, blood glucose concentration, and rating of perceived exertion (RPE), but the percentage of change (Δ) was higher in SOG for all the other variables. SOG showed a higher (Δ) waist-to-hip ratio, maximum heart rate, resting heart rate, systolic blood pressure, blood lactate at 5-min post-exercise, and triglyceride concentrations after 8 and 16 weeks of training. In conclusion, a long-term HIIT program appears to be an appropriate training approach for obese boys with extra body fat. However, considering the RPE values, short-duration training sessions should be planned.
Collapse
Affiliation(s)
- Ghazi Racil
- Research Laboratory (LR23JS01) "Sport Performance, Health & Society", Higher Institute of Sport and Physical Education of Ksar Said, University of Manouba, Tunis 1000, Tunisia
- Department of Biological Sciences Applied for Physical Activities and Sport, Higher Institute of Sport and Physical Education of Ksar Said, University of Manouba, Manouba 2010, Tunisia
| | - Mohamed-Souhaiel Chelly
- Research Laboratory (LR23JS01) "Sport Performance, Health & Society", Higher Institute of Sport and Physical Education of Ksar Said, University of Manouba, Tunis 1000, Tunisia
- Department of Biological Sciences Applied for Physical Activities and Sport, Higher Institute of Sport and Physical Education of Ksar Said, University of Manouba, Manouba 2010, Tunisia
| | - Jeremy Coquart
- Univ. Lille, Univ. Artois, Univ. Littoral Côte d'Opale, ULR 7369-URePSSS-Unité de Recherche Pluridisciplinaire Sport Santé Société, 59000 Lille, France
| | - Johnny Padulo
- Department of Biomedical Sciences for Health, Università degli Studi di Milano, 20133 Milan, Italy
| | - Dragos Florin Teodor
- Faculty of Physical Education and Sport, Ovidius University of Constanta, 900029 Constanta, Romania
| | - Luca Russo
- Department of Human Sciences, Università Telematica Degli Studi IUL, 50122 Florence, Italy
| |
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
|
Steck DT, Jelacic S, Mostofi N, Wu D, Wells L, Fong CT, Cain KC, Sheu RD, Togashi K. The Association Between Hypophosphatemia and Lactic Acidosis After Cardiac Surgery With Cardiopulmonary Bypass: A Retrospective Cohort Study. J Cardiothorac Vasc Anesth 2023; 37:374-381. [PMID: 36528501 DOI: 10.1053/j.jvca.2022.11.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Revised: 11/19/2022] [Accepted: 11/21/2022] [Indexed: 11/27/2022]
Abstract
OBJECTIVES The clinical significance of hypophosphatemia in cardiac surgery has not been investigated extensively. The aim of this study was to evaluate the association of postoperative hypophosphatemia and lactic acidosis in cardiac surgery patients at the time of intensive care unit (ICU) admission. DESIGN A retrospective cohort study. SETTING At a single academic center. PARTICIPANTS Patients who underwent nontransplant cardiac surgery with cardiopulmonary bypass between August 2009 and December 2020. INTERVENTIONS None. MEASUREMENTS AND MAIN RESULTS Serum phosphate and lactate levels were measured upon ICU admission in patients undergoing nontransplant cardiac surgery with cardiopulmonary bypass. There were 681 patients in the low-phosphate (<2.5 mg/dL) group and 2,579 patients in the normal phosphate group (2.5-4.5 mg/dL). A higher proportion of patients in the low phosphate group (26%; 179 of 681; 95% CI: 23-30) had severe lactic acidosis compared to patients in the normal phosphate group (16%; 417 of 2,579; 95% CI: 15-18). In an unadjusted logistic regression model, patients in the low phosphate group had 1.9-times the odds of having severe lactic acidosis (serum lactate ≥4.0 mmol/L) when compared to patients in the normal phosphate group (95% CI: 1.5-2.3), and still 1.4-times the odds (95% CI: 1.1-1.7) after adjusting for several possible confounders. CONCLUSIONS Hypophosphatemia is associated with lactic acidosis in the immediate postoperative period in cardiac surgery patients. Future studies will need to investigate it as a potential treatment target for lactic acidosis.
Collapse
Affiliation(s)
- Dominik T Steck
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA.
| | - Srdjan Jelacic
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA
| | - Nicki Mostofi
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA
| | - David Wu
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University, Stanford, CA
| | - Lauren Wells
- Section of Emergency Medicine, University of Chicago, Chicago, IL
| | - Christine T Fong
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA
| | - Kevin C Cain
- Office of Nursing Research and Department of Biostatistics, University of Washington, Seattle, WA
| | - Richard D Sheu
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA
| | - Kei Togashi
- Department of Anesthesiology and Perioperative Care, University of California, Irvine, Orange, CA
| |
Collapse
|
16
|
Kim JH, An JH, Lee JH, Park SM, Lim GH, Oh YI, Seo KW, Youn HY. Changes in Lactate-related Fecal Microbiome in Hyperlactatemia Diabetic Dogs. In Vivo 2023; 37:696-701. [PMID: 36881052 PMCID: PMC10026652 DOI: 10.21873/invivo.13130] [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: 12/27/2022] [Revised: 01/12/2023] [Accepted: 01/13/2023] [Indexed: 03/08/2023]
Abstract
BACKGROUND/AIM The correlation between the intestinal microbiome and endocrine disorders has recently been drawing attention as an important key for determining their pathology and clinical assessment. In this study, we evaluated the microbiome of dogs with insulin-dependent diabetes mellitus (IDDM) with respect to blood lactate. MATERIALS AND METHODS Fecal samples were obtained from 17 subjects and real-time quantitative polymerase chain reaction determinations were performed to quantify the gene expression levels of lactate-producing and dysbiosis index-related bacteria. RESULTS Expression levels of the lactate-producing bacteria Lactobacillus spp., Enterococcus spp., and Bifidobacterium spp., were confirmed in patients with high concentrations of lactate in the blood. The abundance of Enterococcus and Bifidobacterium was higher in diabetic dogs compared to that of non-diabetic dogs. When blood lactate concentrations were high, the abundance of Bifidobacterium also increased. CONCLUSION Blood lactate levels influence the gut microbiome in dogs with IDDM. This study will help understand the gut microbiota in the context of diabetes in human and veterinary medicine.
Collapse
Affiliation(s)
- Ji-Hyeon Kim
- Laboratory of Veterinary Internal Medicine, Department of Veterinary Clinical Science, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Ju-Hyun An
- Department of Veterinary Emergency and Critical Care Medicine and Institute of Veterinary Science, College of Veterinary Medicine, Kangwon National University, Chuncheon-si, Republic of Korea
| | - Jeong-Hwa Lee
- Laboratory of Veterinary Internal Medicine, Department of Veterinary Clinical Science, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Su-Min Park
- Laboratory of Veterinary Internal Medicine, Department of Veterinary Clinical Science, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Ga-Hyun Lim
- Laboratory of Veterinary Internal Medicine, Department of Veterinary Clinical Science, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Ye-In Oh
- Department of Veterinary Internal Medicine, College of Veterinary Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Kyeong Won Seo
- Laboratory of Veterinary Internal Medicine, Department of Veterinary Clinical Science, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Hwa-Young Youn
- Laboratory of Veterinary Internal Medicine, Department of Veterinary Clinical Science, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea;
| |
Collapse
|
17
|
Făgărășan I, Rusu A, Cristea M, Bala CG, Vulturar DM, Cristea C, Todea DA. Predictors of New-Onset Diabetes in Hospitalized Patients with SARS-CoV-2 Infection. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:13230. [PMID: 36293811 PMCID: PMC9603418 DOI: 10.3390/ijerph192013230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 10/10/2022] [Accepted: 10/11/2022] [Indexed: 06/16/2023]
Abstract
The pandemic caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection is one of the world's most disruptive health crises. The presence of diabetes plays an important role in the severity of the infection, and a rise in newly diagnosed diabetes cases has been identified. The aim of this retrospective study was to determine the incidence of new-onset diabetes (NOD) and predictive factors with their cut-off values for patients hospitalized with COVID-19. All patients (n = 219) hospitalized for COVID-19 during three consecutive months were included. NOD was diagnosed in 26.48% of patients. The severity of the infection, hospital admission values for fasting plasma glucose, lactate dehydrogenase (LDH), PaO2/FiO2 ratio, the peak values for leucocytes, neutrophils, C-reactive protein, triglycerides, and the need for care in the intensive care unit were predictors for the occurrence of NOD in univariate analysis, while only LDH level remained a significant predictor in the multivariable analysis. In conclusion, the results of the study showed a high incidence of NOD in patients hospitalized with COVID-19 and identified LDH levels at hospital admission as a significant predictor of NOD during SARS-CoV-2 infection. However, the persistence of NOD after the COVID-19 infection is not known, therefore, the results must be interpreted with caution.
Collapse
Affiliation(s)
- Iulia Făgărășan
- Department of Pneumology, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400332 Cluj-Napoca, Romania
| | - Adriana Rusu
- Department of Diabetes and Nutrition Diseases, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400006 Cluj-Napoca, Romania
| | - Maria Cristea
- Faculty of Electrical Engineering, Technical University of Cluj-Napoca, 26–28 G. Barițiu Street, 400027 Cluj-Napoca, Romania
| | - Cornelia-Gabriela Bala
- Department of Diabetes and Nutrition Diseases, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400006 Cluj-Napoca, Romania
| | - Damiana-Maria Vulturar
- Department of Pneumology, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400332 Cluj-Napoca, Romania
| | - Ciprian Cristea
- Faculty of Electrical Engineering, Technical University of Cluj-Napoca, 26–28 G. Barițiu Street, 400027 Cluj-Napoca, Romania
| | - Doina-Adina Todea
- Department of Pneumology, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400332 Cluj-Napoca, Romania
| |
Collapse
|
18
|
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
|
19
|
Barbosa P, Landes RD, Graw S, Byrum SD, Bennuri S, Delhey L, Randolph C, MacLeod S, Reis A, Børsheim E, Rose S, Carvalho E. Effect of excess weight and insulin resistance on DNA methylation in prepubertal children. Sci Rep 2022; 12:8430. [PMID: 35589784 PMCID: PMC9120504 DOI: 10.1038/s41598-022-12325-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Accepted: 05/03/2022] [Indexed: 11/13/2022] Open
Abstract
Epigenetic mechanisms, such as DNA methylation, regulate gene expression and play a role in the development of insulin resistance. This study evaluates how the BMI z-score (BMIz) and the homeostatic model assessment of insulin resistance (HOMA-IR), alone or in combination, relate to clinical outcomes and DNA methylation patterns in prepubertal children. DNA methylation in peripheral blood mononuclear cells (PBMCs) and clinical outcomes were measured in a cohort of 41 prepubertal children. Children with higher HOMA-IR had higher blood pressure and plasma lactate levels while children with higher BMIz had higher triglycerides levels. Moreover, the DNA methylation analysis demonstrated that a 1 unit increase in the BMIz was associated with a 0.41 (95% CI: 0.29, 0.53) increase in methylation of a CpG near the PPP6R2 gene. This gene is important in the regulation of NF-kB expression. However, there was no strong evidence that the BMIz and the HOMA-IR were synergistically related to any clinical or DNA methylation outcomes. In summary, the results suggest that obesity and insulin resistance may impact metabolic health both independently in prepubertal children. In addition, obesity also has an impact on the DNA methylation of the PPP6R2 gene. This may be a novel underlying starting point for the systemic inflammation associated with obesity and insulin resistance, in this population.
Collapse
Affiliation(s)
- Pedro Barbosa
- PhD Programme in Experimental Biology and Biomedicine, Institute for Interdisciplinary Research (IIIUC), University of Coimbra, Coimbra, Portugal.,Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.,Institute for Interdisciplinary Research (IIIUC), University of Coimbra, Coimbra, Portugal
| | - Reid D Landes
- Department of Biostatistics, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Stefan Graw
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR, USA.,Arkansas Children's Research Institute, Little Rock, AR, USA.,Everest Clinical Research Corporation, Markham, ON, Canada
| | - Stephanie D Byrum
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR, USA.,Arkansas Children's Research Institute, Little Rock, AR, USA
| | - Sirish Bennuri
- Arkansas Children's Research Institute, Little Rock, AR, USA
| | - Leanna Delhey
- Arkansas Children's Research Institute, Little Rock, AR, USA.,Department of Epidemiology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Chris Randolph
- Arkansas Children's Research Institute, Little Rock, AR, USA
| | - Stewart MacLeod
- Arkansas Children's Research Institute, Little Rock, AR, USA
| | - Andreia Reis
- Department of Medical Sciences (DCM), Institute for Research in Biomedicine (iBiMED), University of Aveiro, Aveiro, Portugal
| | - Elisabet Børsheim
- Arkansas Children's Research Institute, Little Rock, AR, USA.,Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, USA.,Arkansas Children's Nutrition Center, Little Rock, AR, USA.,Department of Geriatrics, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Shannon Rose
- Arkansas Children's Research Institute, Little Rock, AR, USA.,Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Eugenia Carvalho
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal. .,Institute for Interdisciplinary Research (IIIUC), University of Coimbra, Coimbra, Portugal. .,Arkansas Children's Research Institute, Little Rock, AR, USA. .,Department of Geriatrics, University of Arkansas for Medical Sciences, Little Rock, AR, USA.
| |
Collapse
|
20
|
Cisbani G, Koppel A, Metherel AH, Smith ME, Aji KN, Andreazza AC, Mizrahi R, Bazinet RP. Serum lipid analysis and isotopic enrichment is suggestive of greater lipogenesis in young long-term cannabis users: A secondary analysis of a case-control study. Lipids 2022; 57:125-140. [PMID: 35075659 PMCID: PMC8923992 DOI: 10.1002/lipd.12336] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 12/27/2021] [Accepted: 12/28/2021] [Indexed: 01/15/2023]
Abstract
Cannabis is now legal in many countries and while numerous studies have reported on its impact on cognition and appetite regulation, none have examined fatty acid metabolism in young cannabis users. We conducted an exploratory analysis to evaluate cannabis impact on fatty acid metabolism in cannabis users (n = 21) and non-cannabis users (n = 16). Serum levels of some saturated and monounsaturated fatty acids, including palmitic, palmitoleic, and oleic acids were higher in cannabis users compared to nonusers. As palmitic acid can be derived from diet or lipogenesis from sugars, we evaluated lipogenesis using a de novo lipogenesis index (palmitate/linoleic acid) and carbon-specific isotope analysis, which allows for the determination of fatty acid 13 C signature. The significantly higher de novo lipogenesis index in the cannabis users group along with a more enriched 13 C signature of palmitic acid suggested an increase in lipogenesis. In addition, while serum glucose concentration did not differ between groups, pyruvate and lactate were lower in the cannabis user group, with pyruvate negatively correlating with palmitic acid. Furthermore, the endocannabinoid 2-arachidonoylglycerol was elevated in cannabis users and could contribute to lipogenesis by activating the cannabinoid receptor 1. Because palmitic acid has been suggested to increase inflammation, we measured peripheral cytokines and observed no changes in inflammatory cytokines. Finally, an anti-inflammatory metabolite of palmitic acid, palmitoylethanolamide was elevated in cannabis users. Our results suggest that lipogenic activity is increased in cannabis users; however, future studies, including prospective studies that control dietary intake are required.
Collapse
Affiliation(s)
- Giulia Cisbani
- Department of Nutritional Sciences, Temerty Faculty of Medicine, University of Toronto, Canada
| | - Alex Koppel
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada; Department of Psychiatry, University of Toronto, Toronto, Ontario
| | - Adam H. Metherel
- Department of Nutritional Sciences, Temerty Faculty of Medicine, University of Toronto, Canada
| | - Mackenzie E. Smith
- Department of Nutritional Sciences, Temerty Faculty of Medicine, University of Toronto, Canada
| | - Kankana N. Aji
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada; Department of Psychiatry, University of Toronto, Toronto, Ontario
| | - Ana C. Andreazza
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada; Department of Psychiatry, University of Toronto, Toronto, Ontario
| | - Romina Mizrahi
- Department of Psychiatry, McGill University, Montreal, Canada,Douglas Research Center, Montreal, Canada,Corresponding author: Richard P. Bazinet, Department of Nutritional Sciences, Temerty Faculty of Medicine, University of Toronto, Canada, Medical Sciences Building, 5th Floor, Room 5358, 1 King’s College Circle, Toronto, ON, M5S 1A8, , Phone number: (416) 946-8276, Romina Mizrahi, Department of Psychiatry, McGill University, 6875 Boulevard Lasalle, Montréal, QC H4H 1R3,
| | - Richard P. Bazinet
- Department of Nutritional Sciences, Temerty Faculty of Medicine, University of Toronto, Canada,Corresponding author: Richard P. Bazinet, Department of Nutritional Sciences, Temerty Faculty of Medicine, University of Toronto, Canada, Medical Sciences Building, 5th Floor, Room 5358, 1 King’s College Circle, Toronto, ON, M5S 1A8, , Phone number: (416) 946-8276, Romina Mizrahi, Department of Psychiatry, McGill University, 6875 Boulevard Lasalle, Montréal, QC H4H 1R3,
| |
Collapse
|
21
|
Redox Imbalance and Methylation Disturbances in Early Childhood Obesity. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:2207125. [PMID: 34457110 PMCID: PMC8387800 DOI: 10.1155/2021/2207125] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Revised: 07/13/2021] [Accepted: 08/02/2021] [Indexed: 11/29/2022]
Abstract
Obesity is increasing worldwide in prepubertal children, reducing the age of onset of associated comorbidities, including type 2 diabetes. Sulfur-containing amino acids, methionine, cysteine, and their derivatives play important roles in the transmethylation and transsulfuration pathways. Dysregulation of these pathways leads to alterations in the cellular methylation patterns and an imbalanced redox state. Therefore, we tested the hypothesis that one-carbon metabolism is already dysregulated in prepubertal children with obesity. Peripheral blood was collected from 64 children, and the plasma metabolites from transmethylation and transsulfuration pathways were quantified by HPLC. The cohort was stratified by BMI z-scores and HOMA-IR indices into healthy lean (HL), healthy obese (HO), and unhealthy obese (UHO). Fasting insulin levels were higher in the HO group compared to the HL, while the UHO had the highest. All groups presented normal fasting glycemia. Furthermore, high-density lipoprotein (HDL) was lower while triglycerides and lactate levels were higher in the UHO compared to HO subjects. S-adenosylhomocysteine (SAH) and total homocysteine levels were increased in the HO group compared to HL. Additionally, glutathione metabolism was also altered. Free cystine and oxidized glutathione (GSSG) were increased in the HO as compared to HL subjects. Importantly, the adipocyte secretory function was already compromised at this young age. Elevated circulating leptin and decreased adiponectin levels were observed in the UHO as compared to the HO subjects. Some of these alterations were concomitant with alterations in the DNA methylation patterns in the obese group, independent of the impaired insulin levels. In conclusion, our study informs on novel and important metabolic alterations in the transmethylation and the transsulfuration pathways in the early stages of obesity. Moreover, the altered secretory function of the adipocyte very early in life may be relevant in identifying early metabolic markers of disease that may inform on the increased risk for specific future comorbidities in this population.
Collapse
|
22
|
Allen MO, Salman TM, Alada ARA, Odetayo AF, Patrick EB, Salami SA. Effect of the beta-adrenergic blockade on intestinal lactate production and glycogen concentration in dogs infused with hexoses. JOURNAL OF COMPLEMENTARY & INTEGRATIVE MEDICINE 2021; 19:287-296. [PMID: 34323061 DOI: 10.1515/jcim-2021-0062] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 07/05/2021] [Indexed: 11/15/2022]
Abstract
OBJECTIVES To investigate effect of beta adrenergic blockade on intestinal lactate production and glycogen concentration in dogs infused with hexoses. METHODS Experiments were carried out on 35 fasted male anaesthetized dogs weighing between 9 and 16 kg. The animals were divided into 7 (5 dogs per group) groups. Group I dogs served as control and infused with normal saline, groups II-IV were intravenously infused with glucose (1.1 mg/kg/min), fructose (1.1 mg/kg/min) and galactose (1.1 mg/kg/min) respectively while groups V-VII animals were pretreated with propranolol (0.5 mg/kg) and were infused with glucose, fructose or galactose respectively. A vein draining the proximal segment of the jejunum was cannulated along with right and left femoral arteries and veins. Glucose uptake was calculated as the product of jejunal blood flow and the difference between arterial and venous glucose levels (A-V glucose), part of the jejunum tissue was homogenized for estimation of glycogen concentration, and plasma lactate was assayed using lactate colorimetric kit. RESULTS The result showed significant increase in venous lactate production in response to glucose (78.30 ± 4.57 mg/dl), fructose (60.72 ± 1.82 mg/dl) and galactose (71.70 ± 1.30 mg/dl) when compared with the control group (51.75 ± 1.32 mg/dl) at (p<0.05) with no significant difference in animals pretreated with propranolol. There was no significant difference in glycogen concentration (p>0.05) in animals infused with hexoses only compared with propanolol pretreated group. CONCLUSIONS Results suggests that one of the possible fates of the enormous amount of glucose taken up by the intestine is conversion to lactate and not glycogen and β-adrenergic receptor does not affect it.
Collapse
Affiliation(s)
- Michael O Allen
- Department of physiology, Lagos State University College of Medicine, Ikeja, Lagos, Nigeria
| | - Toyin M Salman
- Department of physiology, University of Ilorin, Ilorin, Nigeria
| | | | | | - Eli B Patrick
- Department of physiology, University of Ilorin, Ilorin, Nigeria
| | - Shakiru A Salami
- Department of physiology, Lagos State University College of Medicine, Ikeja, Lagos, Nigeria
| |
Collapse
|
23
|
El-Sikaily A, Helal M. Environmental pollution and diabetes mellitus. World J Meta-Anal 2021; 9:234-256. [DOI: 10.13105/wjma.v9.i3.234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 03/17/2021] [Accepted: 06/03/2021] [Indexed: 02/06/2023] Open
Abstract
Diabetes mellitus (DM) is a chromic metabolic disease that affects a large segment of the population worldwide. Physical inactivity, poor nutrition, and genetic predisposition are main risk factors for disease development. In the last decade, it was clear to the scientific community that DM development is linked to a novel disease inducer that was later defined as diabetogenic factors of pollution and endocrine disrupting agents. Environmental pollution is exponentially increasing in uncontrolled manner in several countries. Environmental pollutants are of diverse nature and toxicities, including polyaromatic hydrocarbons (PAHs), pesticides, and heavy metals. In the current review, we shed light on the impact of each class of these pollutants and the underlined molecular mechanism of diabetes induction and biological toxicities. Finally, a brief overview about the connection between coronavirus disease 2019 and diabetes pandemics is presented.
Collapse
Affiliation(s)
- Amany El-Sikaily
- National Institute of Oceanography and Fisheries (NIOF), Cairo 21513, Egypt
| | - Mohamed Helal
- National Institute of Oceanography and Fisheries (NIOF), Cairo 21513, Egypt
| |
Collapse
|
24
|
Dunkel B, Knowles EJ, Chang YM, Menzies-Gow NJ. Influence of endocrine disease on l-lactate concentrations in blood of ponies. J Vet Intern Med 2021; 35:1582-1588. [PMID: 34043845 PMCID: PMC8162605 DOI: 10.1111/jvim.16079] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 02/05/2021] [Accepted: 02/05/2021] [Indexed: 12/12/2022] Open
Abstract
Background Blood l‐lactate concentrations are higher in people with developing or established diabetes mellitus and insulin resistance. Objectives To investigate whether blood l‐lactate concentrations are positively correlated with measures of insulin dysregulation (ID) or increased autumnal ACTH concentrations in ponies. Animals Systemically healthy client‐owned ponies (n = 101). Methods Prospective case‐control study. Blood samples were obtained from 101 clinically healthy ponies. Breed, weight, height, and subjective and objective measures of body condition were recorded. Blood l‐lactate, glucose, triglyceride, total adiponectin, and ACTH concentrations were measured and an oral sugar test (OST) was carried out. Correlations between blood l‐lactate and variables of endocrine health were determined. Results Using a seasonal cutoff point of ACTH concentrations ≥47 pg/mL, 55 ponies had increased autumnal ACTH concentrations and 45 did not. Using a basal insulin concentration of >50 μiU/mL, 42 ponies were diagnosed with ID and 58 were not. Using a 60 minutes after OST cutoff point of >45 μiU/mL, 57 ponies had ID and 37 did not. Blood l‐lactate concentrations were significantly lower in obese (average body condition score ≥ 7/9) compared to nonobese ponies (0.6 mmol/L; range, 0.0‐1.9 mmol/L vs 0.8 mmol/L; range, 0.3‐2.7 mmol/L; P = .01). No other significant correlations were detected. No differences were detected between ponies with and without increased autumnal ACTH concentrations (0.7 mmol/L; range, 0.0‐2.7 mmol/L vs 0.7 mmol/L; range, 0.3‐1.8 mmol/L; P = .84) and with and without ID (0.7 mmol/L; range, 0.3‐2.7 mmol/L vs 0.8 mmol/L; range, 0.0‐1.6 mmol/L; P = .63). Conclusions and Clinical Importance Results do not support an effect of endocrine status on l‐lactate concentrations in blood of ponies.
Collapse
Affiliation(s)
- Bettina Dunkel
- Department of Clinical Science and Services, The Royal Veterinary College, North Mymms, United Kingdom
| | - Edward J Knowles
- Department of Clinical Science and Services, The Royal Veterinary College, North Mymms, United Kingdom
| | - Yu-Mei Chang
- Research Support Office, The Royal Veterinary College, North Mymms, United Kingdom
| | - Nicola J Menzies-Gow
- Department of Clinical Science and Services, The Royal Veterinary College, North Mymms, United Kingdom
| |
Collapse
|
25
|
Dotson BL, Heiston EM, Miller SL, Malin SK. Insulin stimulation reduces aortic wave reflection in adults with metabolic syndrome. Am J Physiol Heart Circ Physiol 2021; 320:H2305-H2312. [PMID: 33861146 DOI: 10.1152/ajpheart.00975.2020] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Adults with metabolic syndrome (MetS) have increased fasting arterial stiffness and altered central hemodynamics that contribute, partly, to increased cardiovascular disease (CVD) risk. Although insulin affects aortic wave reflections in healthy adults, the effects in individuals with MetS are unclear. We hypothesized that insulin stimulation would reduce measures of pressure waveforms and hemodynamics in people with MetS. Thirty-five adults with obesity (27 women; 54.2 ± 6.0 yr; 37.1 ± 4.8 kg/m2) were selected for MetS (ATP III criteria) following an overnight fast. Pulse wave analysis was assessed using applanation tonometry before and after a 2-h euglycemic-hyperinsulinemic clamp (90 mg/dL, 40 mU/m2/min). Deconvolution analysis was used to decompose the aortic waveform [augmentation index corrected to heart rate of 75 beats/min (AIx@75); augmentation pressure (AP)] into backward and forward pressure components. Aerobic fitness (V̇o2max), body composition (DXA), and blood biochemistries were also assessed. Insulin significantly reduced augmentation index (AIx@75, 28.0 ± 9.6 vs. 23.0 ± 9.9%, P < 0.01), augmentation pressure (14.8 ± 6.4 vs. 12.0 ± 5.7 mmHg, P < 0.01), pulse pressure amplification (1.26 ± 0.01 vs. 0.03 ± 0.01, P = 0.01), and inflammation [high-sensitivity C-reactive protein (hsCRP): P = 0.02; matrix metallopeptidase 7 (MMP-7): P = 0.03] compared to fasting. In subgroup analyses to understand HTN influence, there were no insulin stimulation differences on any outcome. V̇o2max, visceral fat, and blood potassium correlated with fasting AIx@75 (r = -0.39, P = 0.02; r = 0.41, P = 0.03; r = -0.53, P = 0.002). Potassium levels were also associated with insulin-mediated reductions in AP (r = 0.52, P = 0.002). Our results suggest insulin stimulation improves indices of aortic reflection in adults with MetS.NEW & NOTEWORTHY This study is one of the first to investigate the effects of insulin on central and peripheral hemodynamics in adults with metabolic syndrome. We provide evidence that insulin infusion reduces aortic wave reflection, potentially through a reduction in inflammation and/or via a potassium-mediated vascular response.
Collapse
Affiliation(s)
- Brielle L Dotson
- Department of Kinesiology, University of Virginia, Charlottesville, Virginia
| | - Emily M Heiston
- Department of Kinesiology, University of Virginia, Charlottesville, Virginia.,Division of Cardiology, Pauley Heart Center, Virginia Commonwealth University, Richmond, Virginia
| | - Stephanie L Miller
- Department of Kinesiology, University of Virginia, Charlottesville, Virginia
| | - Steven K Malin
- Department of Kinesiology, University of Virginia, Charlottesville, Virginia.,Department of Kinesiology and Health, Rutgers University, New Brunswick, New Jersey.,Division of Endocrinology, Metabolism and Nutrition; Rutgers University, New Brunswick, New Jersey.,New Jersey Institute for Food, Nutrition and Health, Rutgers University, New Brunswick, New Jersey.,Institute of Translational Medicine and Science, Rutgers University, New Brunswick, New Jersey
| |
Collapse
|
26
|
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
|
27
|
Berlanga-Acosta J, Guillén-Nieto G, Rodríguez-Rodríguez N, Bringas-Vega ML, García-del-Barco-Herrera D, Berlanga-Saez JO, García-Ojalvo A, Valdés-Sosa MJ, Valdés-Sosa PA. Insulin Resistance at the Crossroad of Alzheimer Disease Pathology: A Review. Front Endocrinol (Lausanne) 2020; 11:560375. [PMID: 33224105 PMCID: PMC7674493 DOI: 10.3389/fendo.2020.560375] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 08/13/2020] [Indexed: 12/16/2022] Open
Abstract
Insulin plays a major neuroprotective and trophic function for cerebral cell population, thus countering apoptosis, beta-amyloid toxicity, and oxidative stress; favoring neuronal survival; and enhancing memory and learning processes. Insulin resistance and impaired cerebral glucose metabolism are invariantly reported in Alzheimer's disease (AD) and other neurodegenerative processes. AD is a fatal neurodegenerative disorder in which progressive glucose hypometabolism parallels to cognitive impairment. Although AD may appear and progress in virtue of multifactorial nosogenic ingredients, multiple interperpetuative and interconnected vicious circles appear to drive disease pathophysiology. The disease is primarily a metabolic/energetic disorder in which amyloid accumulation may appear as a by-product of more proximal events, especially in the late-onset form. As a bridge between AD and type 2 diabetes, activation of c-Jun N-terminal kinase (JNK) pathway with the ensued serine phosphorylation of the insulin response substrate (IRS)-1/2 may be at the crossroads of insulin resistance and its subsequent dysmetabolic consequences. Central insulin axis bankruptcy translates in neuronal vulnerability and demise. As a link in the chain of pathogenic vicious circles, mitochondrial dysfunction, oxidative stress, and peripheral/central immune-inflammation are increasingly advocated as major pathology drivers. Pharmacological interventions addressed to preserve insulin axis physiology, mitochondrial biogenesis-integral functionality, and mitophagy of diseased organelles may attenuate the adjacent spillover of free radicals that further perpetuate mitochondrial damages and catalyze inflammation. Central and/or peripheral inflammation may account for a local flood of proinflammatory cytokines that along with astrogliosis amplify insulin resistance, mitochondrial dysfunction, and oxidative stress. All these elements are endogenous stressor, pro-senescent factors that contribute to JNK activation. Taken together, these evidences incite to identify novel multi-mechanistic approaches to succeed in ameliorating this pandemic affliction.
Collapse
Affiliation(s)
- Jorge Berlanga-Acosta
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Lab for Neuroinformation, University of Electronic Science and Technology of China, Chengdu, China
- Tissue Repair and Cytoprotection Research Group, Center for Genetic Engineering and Biotechnology, Havana, Cuba
| | - Gerardo Guillén-Nieto
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Lab for Neuroinformation, University of Electronic Science and Technology of China, Chengdu, China
- Tissue Repair and Cytoprotection Research Group, Center for Genetic Engineering and Biotechnology, Havana, Cuba
| | - Nadia Rodríguez-Rodríguez
- Tissue Repair and Cytoprotection Research Group, Center for Genetic Engineering and Biotechnology, Havana, Cuba
| | - Maria Luisa Bringas-Vega
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Lab for Neuroinformation, University of Electronic Science and Technology of China, Chengdu, China
- Cuban Neurosciences Center, Cubanacan, Havana, Cuba
| | | | - Jorge O. Berlanga-Saez
- Applied Mathematics Department, Institute of Mathematics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Ariana García-Ojalvo
- Tissue Repair and Cytoprotection Research Group, Center for Genetic Engineering and Biotechnology, Havana, Cuba
| | - Mitchell Joseph Valdés-Sosa
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Lab for Neuroinformation, University of Electronic Science and Technology of China, Chengdu, China
- Cuban Neurosciences Center, Cubanacan, Havana, Cuba
| | - Pedro A. Valdés-Sosa
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Lab for Neuroinformation, University of Electronic Science and Technology of China, Chengdu, China
- Cuban Neurosciences Center, Cubanacan, Havana, Cuba
| |
Collapse
|
28
|
Zaborska KE, Dadi PK, Dickerson MT, Nakhe AY, Thorson AS, Schaub CM, Graff SM, Stanley JE, Kondapavuluru RS, Denton JS, Jacobson DA. Lactate activation of α-cell K ATP channels inhibits glucagon secretion by hyperpolarizing the membrane potential and reducing Ca 2+ entry. Mol Metab 2020; 42:101056. [PMID: 32736089 PMCID: PMC7479281 DOI: 10.1016/j.molmet.2020.101056] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 07/17/2020] [Accepted: 07/24/2020] [Indexed: 12/20/2022] Open
Abstract
Objective Elevations in pancreatic α-cell intracellular Ca2+ ([Ca2+]i) lead to glucagon (GCG) secretion. Although glucose inhibits GCG secretion, how lactate and pyruvate control α-cell Ca2+ handling is unknown. Lactate enters cells through monocarboxylate transporters (MCTs) and is also produced during glycolysis by lactate dehydrogenase A (LDHA), an enzyme expressed in α-cells. As lactate activates ATP-sensitive K+ (KATP) channels in cardiomyocytes, lactate may also modulate α-cell KATP. Therefore, this study investigated how lactate signaling controls α-cell Ca2+ handling and GCG secretion. Methods Mouse and human islets were used in combination with confocal microscopy, electrophysiology, GCG immunoassays, and fluorescent thallium flux assays to assess α-cell Ca2+ handling, Vm, KATP currents, and GCG secretion. Results Lactate-inhibited mouse (75 ± 25%) and human (47 ± 9%) α-cell [Ca2+]i fluctuations only under low-glucose conditions (1 mM) but had no effect on β- or δ-cells [Ca2+]i. Glyburide inhibition of KATP channels restored α-cell [Ca2+]i fluctuations in the presence of lactate. Lactate transport into α-cells via MCTs hyperpolarized mouse (14 ± 1 mV) and human (12 ± 1 mV) α-cell Vm and activated KATP channels. Interestingly, pyruvate showed a similar KATP activation profile and α-cell [Ca2+]i inhibition as lactate. Lactate-induced inhibition of α-cell [Ca2+]i influx resulted in reduced GCG secretion in mouse (62 ± 6%) and human (43 ± 13%) islets. Conclusions These data demonstrate for the first time that lactate entry into α-cells through MCTs results in KATP activation, Vm hyperpolarization, reduced [Ca2+]i, and inhibition of GCG secretion. Thus, taken together, these data indicate that lactate either within α-cells and/or elevated in serum could serve as important modulators of α-cell function. Lactate reduces islet α-cell Ca2+ entry under low glucose conditions. Lactate does not alter β- or δ-cell Ca2+ handling under low glucose conditions. Lactate enters islet α-cells through monocarboxylate transporters. Lactate hyperpolarizes islet α-cell membrane potential by activating KATP channels. Lactate reduces mouse and human islet glucagon secretion.
Collapse
Affiliation(s)
- Karolina E Zaborska
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37232, USA
| | - Prasanna K Dadi
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37232, USA
| | - Matthew T Dickerson
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37232, USA
| | - Arya Y Nakhe
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37232, USA
| | - Ariel S Thorson
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37232, USA
| | - Charles M Schaub
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37232, USA
| | - Sarah M Graff
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37232, USA
| | - Jade E Stanley
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37232, USA
| | - Roy S Kondapavuluru
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37232, USA
| | - Jerod S Denton
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA
| | - David A Jacobson
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37232, USA.
| |
Collapse
|
29
|
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: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [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,Address correspondence to MR-C (e-mail: )
| |
Collapse
|
30
|
Hernandez-Baixauli J, Quesada-Vázquez S, Mariné-Casadó R, Gil Cardoso K, Caimari A, Del Bas JM, Escoté X, Baselga-Escudero L. Detection of Early Disease Risk Factors Associated with Metabolic Syndrome: A New Era with the NMR Metabolomics Assessment. Nutrients 2020; 12:E806. [PMID: 32197513 PMCID: PMC7146483 DOI: 10.3390/nu12030806] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 03/11/2020] [Accepted: 03/17/2020] [Indexed: 02/07/2023] Open
Abstract
The metabolic syndrome is a multifactorial disease developed due to accumulation and chronification of several risk factors associated with disrupted metabolism. The early detection of the biomarkers by NMR spectroscopy could be helpful to prevent multifactorial diseases. The exposure of each risk factor can be detected by traditional molecular markers but the current biomarkers have not been enough precise to detect the primary stages of disease. Thus, there is a need to obtain novel molecular markers of pre-disease stages. A promising source of new molecular markers are metabolomics standing out the research of biomarkers in NMR approaches. An increasing number of nutritionists integrate metabolomics into their study design, making nutrimetabolomics one of the most promising avenues for improving personalized nutrition. This review highlight the major five risk factors associated with metabolic syndrome and related diseases including carbohydrate dysfunction, dyslipidemia, oxidative stress, inflammation, and gut microbiota dysbiosis. Together, it is proposed a profile of metabolites of each risk factor obtained from NMR approaches to target them using personalized nutrition, which will improve the quality of life for these patients.
Collapse
Affiliation(s)
- Julia Hernandez-Baixauli
- Eurecat, Centre Tecnològic de Catalunya, Unitat de Nutrició i Salut, 43204 Reus, Spain; (J.H.-B.); (S.Q.-V.); (R.M.-C.); (K.G.C.); (A.C.); (J.M.D.B.)
| | - Sergio Quesada-Vázquez
- Eurecat, Centre Tecnològic de Catalunya, Unitat de Nutrició i Salut, 43204 Reus, Spain; (J.H.-B.); (S.Q.-V.); (R.M.-C.); (K.G.C.); (A.C.); (J.M.D.B.)
| | - Roger Mariné-Casadó
- Eurecat, Centre Tecnològic de Catalunya, Unitat de Nutrició i Salut, 43204 Reus, Spain; (J.H.-B.); (S.Q.-V.); (R.M.-C.); (K.G.C.); (A.C.); (J.M.D.B.)
- Universitat Rovira i Virgili; Department of Biochemistry and Biotechnology, Ctra. De Valls, s/n, 43007 Tarragona, Spain
| | - Katherine Gil Cardoso
- Eurecat, Centre Tecnològic de Catalunya, Unitat de Nutrició i Salut, 43204 Reus, Spain; (J.H.-B.); (S.Q.-V.); (R.M.-C.); (K.G.C.); (A.C.); (J.M.D.B.)
- Universitat Rovira i Virgili; Department of Biochemistry and Biotechnology, Ctra. De Valls, s/n, 43007 Tarragona, Spain
| | - Antoni Caimari
- Eurecat, Centre Tecnològic de Catalunya, Unitat de Nutrició i Salut, 43204 Reus, Spain; (J.H.-B.); (S.Q.-V.); (R.M.-C.); (K.G.C.); (A.C.); (J.M.D.B.)
| | - Josep M Del Bas
- Eurecat, Centre Tecnològic de Catalunya, Unitat de Nutrició i Salut, 43204 Reus, Spain; (J.H.-B.); (S.Q.-V.); (R.M.-C.); (K.G.C.); (A.C.); (J.M.D.B.)
| | - Xavier Escoté
- Eurecat, Centre Tecnològic de Catalunya, Unitat de Nutrició i Salut, 43204 Reus, Spain; (J.H.-B.); (S.Q.-V.); (R.M.-C.); (K.G.C.); (A.C.); (J.M.D.B.)
| | - Laura Baselga-Escudero
- Eurecat, Centre Tecnològic de Catalunya, Unitat de Nutrició i Salut, 43204 Reus, Spain; (J.H.-B.); (S.Q.-V.); (R.M.-C.); (K.G.C.); (A.C.); (J.M.D.B.)
| |
Collapse
|
31
|
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
|
32
|
Dunkel B, Knowles EJ, Chang YM, Menzies-Gow NJ. Correlation between l-lactate and glucose concentrations and body condition score in healthy horses and ponies. J Vet Intern Med 2019; 33:2267-2271. [PMID: 31390098 PMCID: PMC6766520 DOI: 10.1111/jvim.15587] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 07/22/2019] [Indexed: 12/19/2022] Open
Abstract
Background Blood l‐lactate and glucose concentrations were higher in ponies with gastrointestinal disease than in horses, possibly because of differences in body condition (BC). Objectives To investigate whether l‐lactate and glucose concentrations correlate with BC and differ between healthy horses and ponies. Animals Systemically healthy client‐owned ponies (n = 101) and horses (n = 51). Methods Prospective observational study. Breed, weight, height, and subjective and objective measures of BC were recorded and l‐lactate and glucose concentrations were measured. Correlations between l‐lactate and glucose concentrations and BC were established. The association between l‐lactate concentrations, equid type (pony or horse), BC, age, and glucose concentrations was investigated using a multivariable model. Results Weak but significant (P = .001) negative correlations were detected between l‐lactate concentration and average BC score (r = −0.29), heart girth:height ratio (r = −0.27), and age (r = −0.27). Glucose concentrations were significantly (P < .001) positively correlated with neck length:heart girth ratio (r = 0.37) and heart girth:height ratio (r = 0.31). l‐lactate and glucose concentrations were weakly correlated (r = 0.15; P = .04). In the final multivariable model, age (−0.02 ± 0.006; P = .001) and heart girth:height ratio (−1.74 ± 0.53; P = .001) were significantly associated with the natural logarithm of l‐lactate concentration (Lnl‐Lactate). This represents a 2% decrease in l‐lactate concentration per year increase in age and 10% decrease in l‐lactate concentration per 0.06 unit increase in heart girth:height ratio. Conclusions and Clinical Importance In healthy horses and ponies, age and BC significantly influence l‐lactate concentrations.
Collapse
Affiliation(s)
- Bettina Dunkel
- Department of Clinical Science and Services, The Royal Veterinary College, North Mymms, United Kingdom
| | - Edward J Knowles
- Department of Clinical Science and Services, The Royal Veterinary College, North Mymms, United Kingdom
| | - Yu-Mei Chang
- Research Support Office, The Royal Veterinary College, North Mymms, United Kingdom
| | - Nicola J Menzies-Gow
- Department of Clinical Science and Services, The Royal Veterinary College, North Mymms, United Kingdom
| |
Collapse
|
33
|
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
|
34
|
Zou K, Hinkley JM, Park S, Zheng D, Jones TE, Pories WJ, Hornby PJ, Lenhard J, Dohm GL, Houmard JA. Altered tricarboxylic acid cycle flux in primary myotubes from severely obese humans. Int J Obes (Lond) 2019; 43:895-905. [PMID: 29892037 DOI: 10.1038/s41366-018-0137-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Revised: 04/27/2018] [Accepted: 05/10/2018] [Indexed: 11/08/2022]
Abstract
BACKGROUND/OBJECTIVE The partitioning of glucose toward glycolytic end products rather than glucose oxidation and glycogen storage is evident in skeletal muscle with severe obesity and type 2 diabetes. The purpose of the present study was to determine the possible mechanism by which severe obesity alters insulin-mediated glucose partitioning in human skeletal muscle. SUBJECTS/METHODS Primary human skeletal muscle cells (HSkMC) were isolated from lean (BMI = 23.6 ± 2.6 kg/m2, n = 9) and severely obese (BMI = 48.8 ± 1.9 kg/m2, n = 8) female subjects. Glucose oxidation, glycogen synthesis, non-oxidized glycolysis, pyruvate oxidation, and targeted TCA cycle metabolomics were examined in differentiated myotubes under basal and insulin-stimulated conditions. RESULTS Myotubes derived from severely obese subjects exhibited attenuated response of glycogen synthesis (20.3%; 95% CI [4.7, 28.8]; P = 0.017) and glucose oxidation (5.6%; 95% CI [0.3, 8.6]; P = 0.046) with a concomitant greater increase (23.8%; 95% CI [5.7, 47.8]; P = 0.004) in non-oxidized glycolytic end products with insulin stimulation in comparison to the lean group (34.2% [24.9, 45.1]; 13.1% [8.6, 16.4], and 2.9% [-4.1, 12.2], respectively). These obesity-related alterations in glucose partitioning appeared to be linked with reduced TCA cycle flux, as 2-[14C]-pyruvate oxidation (358.4 pmol/mg protein/min [303.7, 432.9] vs. lean 439.2 pmol/mg protein/min [393.6, 463.1]; P = 0.013) along with several TCA cycle intermediates, were suppressed in the skeletal muscle of severely obese individuals. CONCLUSIONS These data suggest that with severe obesity the partitioning of glucose toward anaerobic glycolysis in response to insulin is a resilient characteristic of human skeletal muscle. This altered glucose partitioning appeared to be due, at least in part, to a reduction in TCA cycle flux.
Collapse
Affiliation(s)
- Kai Zou
- Department of Exercise and Health Sciences, University of Massachusetts Boston, Boston, MA, USA.
- Department of Kinesiology, East Carolina University, Greenville, NC, USA.
- Human Performance Laboratory, East Carolina University, Greenville, NC, USA.
- East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC, USA.
| | - J Matthew Hinkley
- Department of Kinesiology, East Carolina University, Greenville, NC, USA
- Human Performance Laboratory, East Carolina University, Greenville, NC, USA
- East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC, USA
- Translational Research Institute for Metabolism and Diabetes, Florida Hospital, Orlando, FL, USA
| | - Sanghee Park
- Department of Kinesiology, East Carolina University, Greenville, NC, USA
- Human Performance Laboratory, East Carolina University, Greenville, NC, USA
- East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC, USA
| | - Donghai Zheng
- Department of Kinesiology, East Carolina University, Greenville, NC, USA
- Human Performance Laboratory, East Carolina University, Greenville, NC, USA
- East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC, USA
| | - Terry E Jones
- Department of Physical Therapy, East Carolina University, Greenville, NC, USA
| | - Walter J Pories
- East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC, USA
- Department of Surgery, East Carolina University, Greenville, NC, USA
| | | | - James Lenhard
- Janssen Research & Development LLC, Spring House, PA, USA
| | - G Lynis Dohm
- East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC, USA
- Department of Physiology, East Carolina University, Greenville, NC, USA
| | - Joseph A Houmard
- Department of Kinesiology, East Carolina University, Greenville, NC, USA
- Human Performance Laboratory, East Carolina University, Greenville, NC, USA
- East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC, USA
| |
Collapse
|
35
|
Santos JL, Cataldo LR, Cortés-Rivera C, Bravo C, Díaz-Casanova L, Martínez JA, Milagro FI, Galgani J. Plasma lactate and leukocyte mitochondrial DNA copy number as biomarkers of insulin sensitivity in non-diabetic women. J Physiol Biochem 2019; 75:285-297. [DOI: 10.1007/s13105-019-00672-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Accepted: 02/28/2019] [Indexed: 11/29/2022]
|
36
|
Tardelli LP, Breda L, Marques LF, Gomes Carvalho Lima NC, Furtado de Camargo T, Scherer BR, Moreira NF, Dias JF, Dalia RA, Thomazini BF, Corezolla do Amaral ME, Alves AA. High lipid and low carbohydrate content diet, immediately after weaning, causes hepatic injury, systemic oxidative stress and diminishment of lipids in white adipose tissue. JOURNAL OF NUTRITION & INTERMEDIARY METABOLISM 2018. [DOI: 10.1016/j.jnim.2018.08.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022] Open
|
37
|
Brain insulin resistance in type 2 diabetes and Alzheimer disease: concepts and conundrums. Nat Rev Neurol 2018; 14:168-181. [PMID: 29377010 DOI: 10.1038/nrneurol.2017.185] [Citation(s) in RCA: 922] [Impact Index Per Article: 131.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Considerable overlap has been identified in the risk factors, comorbidities and putative pathophysiological mechanisms of Alzheimer disease and related dementias (ADRDs) and type 2 diabetes mellitus (T2DM), two of the most pressing epidemics of our time. Much is known about the biology of each condition, but whether T2DM and ADRDs are parallel phenomena arising from coincidental roots in ageing or synergistic diseases linked by vicious pathophysiological cycles remains unclear. Insulin resistance is a core feature of T2DM and is emerging as a potentially important feature of ADRDs. Here, we review key observations and experimental data on insulin signalling in the brain, highlighting its actions in neurons and glia. In addition, we define the concept of 'brain insulin resistance' and review the growing, although still inconsistent, literature concerning cognitive impairment and neuropathological abnormalities in T2DM, obesity and insulin resistance. Lastly, we review evidence of intrinsic brain insulin resistance in ADRDs. By expanding our understanding of the overlapping mechanisms of these conditions, we hope to accelerate the rational development of preventive, disease-modifying and symptomatic treatments for cognitive dysfunction in T2DM and ADRDs alike.
Collapse
|
38
|
Singh V, Sharma RK, Athilingam T, Sinha P, Sinha N, Thakur AK. NMR Spectroscopy-based Metabolomics of Drosophila Model of Huntington's Disease Suggests Altered Cell Energetics. J Proteome Res 2017; 16:3863-3872. [PMID: 28871787 DOI: 10.1021/acs.jproteome.7b00491] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Huntington's disease (HD) is a neurodegenerative disorder induced by aggregation of the pathological form of Huntingtin protein that has expanded polyglutamine (polyQ) repeats. In the Drosophila model, for instance, expression of transgenes with polyQ repeats induces HD-like pathologies, progressively correlating with the increasing lengths of these repeats. Previous studies on both animal models and clinical samples have revealed metabolite imbalances during HD progression. To further explore the physiological processes linked to metabolite imbalances during HD, we have investigated the 1D 1H NMR spectroscopy-based metabolomics profile of Drosophila HD model. Using multivariate analysis (PCA and PLS-DA) of metabolites obtained from methanolic extracts of fly heads displaying retinal deformations due to polyQ overexpression, we show that the metabolite imbalance during HD is likely to affect cell energetics. Six out of the 35 metabolites analyzed, namely, nicotinamide adenine dinucleotide (NAD), lactate, pyruvate, succinate, sarcosine, and acetoin, displayed segregation with progressive severity of HD. Specifically, HD progression was seen to be associated with reduction in NAD and increase in lactate-to-pyruvate ratio. Furthermore, comparative analysis of fly HD metabolome with those of mouse HD model and HD human patients revealed comparable metabolite imbalances, suggesting altered cellular energy homeostasis. These findings thus raise the possibility of therapeutic interventions for HD via modulation of cellular energetics.
Collapse
Affiliation(s)
- Virender Singh
- Biological Science and Bioengineering, Indian Institute of Technology Kanpur , Kanpur 208016, India
| | - Raj Kumar Sharma
- Centre of Biomedical Research, SGPGIMS Campus , Lucknow 226014, India
| | | | - Pradip Sinha
- Biological Science and Bioengineering, Indian Institute of Technology Kanpur , Kanpur 208016, India
| | - Neeraj Sinha
- Centre of Biomedical Research, SGPGIMS Campus , Lucknow 226014, India
| | - Ashwani Kumar Thakur
- Biological Science and Bioengineering, Indian Institute of Technology Kanpur , Kanpur 208016, India
| |
Collapse
|
39
|
The Effects of Peripheral and Central High Insulin on Brain Insulin Signaling and Amyloid-β in Young and Old APP/PS1 Mice. J Neurosci 2017; 36:11704-11715. [PMID: 27852778 DOI: 10.1523/jneurosci.2119-16.2016] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2016] [Revised: 09/13/2016] [Accepted: 09/25/2016] [Indexed: 12/19/2022] Open
Abstract
Hyperinsulinemia is a risk factor for late-onset Alzheimer's disease (AD). In vitro experiments describe potential connections between insulin, insulin signaling, and amyloid-β (Aβ), but in vivo experiments are needed to validate these relationships under physiological conditions. First, we performed hyperinsulinemic-euglycemic clamps with concurrent hippocampal microdialysis in young, awake, behaving APPswe/PS1dE9 transgenic mice. Both a postprandial and supraphysiological insulin clamp significantly increased interstitial fluid (ISF) and plasma Aβ compared with controls. We could detect no increase in brain, ISF, or CSF insulin or brain insulin signaling in response to peripheral hyperinsulinemia, despite detecting increased signaling in the muscle. Next, we delivered insulin directly into the hippocampus of young APP/PS1 mice via reverse microdialysis. Brain tissue insulin and insulin signaling was dose-dependently increased, but ISF Aβ was unchanged by central insulin administration. Finally, to determine whether peripheral and central high insulin has differential effects in the presence of significant amyloid pathology, we repeated these experiments in older APP/PS1 mice with significant amyloid plaque burden. Postprandial insulin clamps increased ISF and plasma Aβ, whereas direct delivery of insulin to the hippocampus significantly increased tissue insulin and insulin signaling, with no effect on Aβ in old mice. These results suggest that the brain is still responsive to insulin in the presence of amyloid pathology but increased insulin signaling does not acutely modulate Aβ in vivo before or after the onset of amyloid pathology. Peripheral hyperinsulinemia modestly increases ISF and plasma Aβ in young and old mice, independent of neuronal insulin signaling. SIGNIFICANCE STATEMENT The transportation of insulin from blood to brain is a saturable process relevant to understanding the link between hyperinsulinemia and AD. In vitro experiments have found direct connections between high insulin and extracellular Aβ, but these mechanisms presume that peripheral high insulin elevates brain insulin significantly. We found that physiological hyperinsulinemia in awake, behaving mice does not increase CNS insulin to an appreciable level yet modestly increases extracellular Aβ. We also found that the brain of aged APP/PS1 mice was not insulin resistant, contrary to the current state of the literature. These results further elucidate the relationship between insulin, the brain, and AD and its conflicting roles as both a risk factor and potential treatment.
Collapse
|
40
|
Heden TD, Liu Y, Kanaley JA. Exercise timing and blood lactate concentrations in individuals with type 2 diabetes. Appl Physiol Nutr Metab 2017; 42:732-737. [DOI: 10.1139/apnm-2016-0382] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The purpose of this study was to characterize how resistance exercise prior to or after a meal alters fasting and postprandial blood lactate concentrations in individuals with type 2 diabetes. Obese individuals with type 2 diabetes (N = 12) completed three 2-day trials, including (i) no exercise (NoEx), (ii) resistance exercise prior to dinner (Ex-M), and (iii) resistance exercise beginning at 45 min postdinner (M-Ex). During day 1 of each trial, fasting and postprandial blood lactate concentrations, perceived exertion, and substrate oxidation were measured, and subsequently on day 2 the following morning fasting blood lactate was measured. The premeal lactate incremental area under the curve (iAUC) during Ex-M (109 ± 66 mmol·L−1·1.6 h−1) was over 100-fold greater (P < 0.01) compared with NoEx (−15 ± 24 mmol·L−1·1.6 h−1) and M-Ex (−2 ± 18 mmol·L−1·1.6 h−1). The postprandial lactate iAUC during M-Ex (304 ± 116 mmol·L−1·4 h−1) was over 2-fold greater (P < 0.01) compared with NoEx (149 ± 74 mmol·L−1·4 h−1) and Ex-M (−140 ± 196 mmol·L−1·4 h−1). Average lactate during exercise was ∼45% greater (P = 0.03) during M-Ex (3.2 ± 0.9 mmol/L) compared with Ex-M (2.2 ± 0.9 mmol/L), but the change in lactate during Ex-M (2.4 ± 1.6 mmol/L) or M-Ex (2.3 ± 1.3 mmol/L) was not different (P > 0.05). Perceived exertion, substrate oxidation, or fasting blood lactate concentrations the day after testing were not different between trials. Blood lactate concentrations during acute resistance exercise are greater when exercise is performed in the postprandial period. Acute resistance exercise performed the night prior does not alter fasting blood lactate concentrations the following morning.
Collapse
Affiliation(s)
- Timothy D. Heden
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO 65211, USA
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO 65211, USA
| | - Ying Liu
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO 65211, USA
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO 65211, USA
| | - Jill A. Kanaley
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO 65211, USA
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO 65211, USA
| |
Collapse
|
41
|
Lactate, a Neglected Factor for Diabetes and Cancer Interaction. Mediators Inflamm 2016; 2016:6456018. [PMID: 28077918 PMCID: PMC5203906 DOI: 10.1155/2016/6456018] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Revised: 11/17/2016] [Accepted: 11/23/2016] [Indexed: 12/23/2022] Open
Abstract
Increasing body of evidence suggests that there exists a connection between diabetes and cancer. Nevertheless, to date, the potential reasons for this association are still poorly understood and currently there is no clinical evidence available to direct the proper management of patients presenting with these two diseases concomitantly. Both cancer and diabetes have been associated with abnormal lactate metabolism and high level of lactate production is the key biological property of these diseases. Conversely, high lactate contribute to a higher insulin resistant status and a more malignant phenotype of cancer cells, promoting diabetes and cancer development and progression. In view of associations between diabetes and cancers, the role of high lactate production in diabetes and cancer interaction should not be neglected. Here, we review the available evidence of lactate's role in different biological characteristics of diabetes and cancer and interactive relationship between them. Understanding the molecular mechanisms behind metabolic remodeling of diabetes- and cancer-related signaling would endow novel preventive and therapeutic approaches for diabetes and cancer treatment.
Collapse
|
42
|
Se CH, Chuah KA, Mishra A, Wickneswari R, Karupaiah T. Evaluating Crossbred Red Rice Variants for Postprandial Glucometabolic Responses: A Comparison with Commercial Varieties. Nutrients 2016; 8:nu8050308. [PMID: 27213446 PMCID: PMC4882720 DOI: 10.3390/nu8050308] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Revised: 05/09/2016] [Accepted: 05/10/2016] [Indexed: 12/17/2022] Open
Abstract
Consumption of white rice predisposes some Asian populations to increased risk of type 2 diabetes. We compared the postprandial glucometabolic responses to three newly-developed crossbred red rice variants (UKMRC9, UKMRC10, UKMRC11) against three selected commercial rice types (Thai red, Basmati white, Jasmine white) using 50-g carbohydrate equivalents provided to 12 normoglycaemic adults in a crossover design. Venous blood was drawn fasted and postprandially for three hours. Glycaemic (GI) and insulin (II) indices, incremental areas-under-the-curves for glucose and insulin (IAUCins), indices of insulin sensitivity and secretion, lactate and peptide hormones (motilin, neuropeptide-Y, orexin-A) were analyzed. The lowest to highest trends for GI and II were similar i.e., UKMRC9 < Basmati < Thai red < UKMRC10 < UKMRC11 < Jasmine. Postprandial insulinaemia and IAUCins of only UKMRC9 were significantly the lowest compared to Jasmine. Crude protein and fiber content correlated negatively with the GI values of the test rice. Although peptide hormones were not associated with GI and II characteristics of test rice, early and late phases of prandial neuropeptide-Y changes were negatively correlated with postprandial insulinaemia. This study indicated that only UKMRC9 among the new rice crossbreeds could serve as an alternative cereal option to improve diet quality of Asians with its lowest glycaemic and insulinaemic burden.
Collapse
Affiliation(s)
- Chee-Hee Se
- School of Healthcare Sciences, Faculty of Health Sciences, National University of Malaysia (UKM), Jalan Raja Muda Abdul Aziz, Kuala Lumpur 50300, Malaysia.
| | - Khun-Aik Chuah
- School of Healthcare Sciences, Faculty of Health Sciences, National University of Malaysia (UKM), Jalan Raja Muda Abdul Aziz, Kuala Lumpur 50300, Malaysia.
| | - Ankitta Mishra
- School of Environmental and Natural Resource Sciences, Faculty of Science and Technology, National University of Malaysia (UKM), Bangi, Selangor 43600, Malaysia.
| | - Ratnam Wickneswari
- School of Environmental and Natural Resource Sciences, Faculty of Science and Technology, National University of Malaysia (UKM), Bangi, Selangor 43600, Malaysia.
| | - Tilakavati Karupaiah
- School of Healthcare Sciences, Faculty of Health Sciences, National University of Malaysia (UKM), Jalan Raja Muda Abdul Aziz, Kuala Lumpur 50300, Malaysia.
| |
Collapse
|