1
|
Schleh MW, Ryan BJ, Ahn C, Ludzki AC, Van Pelt DW, Pitchford LM, Chugh OK, Luker AT, Luker KE, Samovski D, Abumrad NA, Burant CF, Horowitz JF. Impaired suppression of fatty acid release by insulin is a strong predictor of reduced whole-body insulin-mediated glucose uptake and skeletal muscle insulin receptor activation. Acta Physiol (Oxf) 2024:e14249. [PMID: 39487600 DOI: 10.1111/apha.14249] [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: 06/26/2024] [Revised: 09/06/2024] [Accepted: 09/24/2024] [Indexed: 11/04/2024]
Abstract
AIM To examine factors underlying why most, but not all, adults with obesity exhibit impaired insulin-mediated glucose uptake, we compared: (1) adipose tissue fatty acid (FA) release, (2) skeletal muscle lipid droplet (LD) characteristics, and (3) insulin signalling events, in skeletal muscle of adults with obesity with relatively high versus low insulin-mediated glucose uptake. METHODS Seventeen adults with obesity (BMI: 36 ± 3 kg/m2) completed a 2 h hyperinsulinemic-euglycemic clamp with stable isotope tracer infusions to measure glucose rate of disappearance (glucose Rd) and FA rate of appearance (FA Ra). Skeletal muscle biopsies were collected at baseline and 30 min into the insulin infusion. Participants were stratified into HIGH (n = 7) and LOW (n = 10) insulin sensitivity cohorts by their glucose Rd during the hyperinsulinemic clamp (LOW< 400; HIGH >550 nmol/kgFFM/min/[μU/mL]). RESULTS Insulin-mediated suppression of FA Ra was lower in LOW compared with HIGH (p < 0.01). In skeletal muscle, total intramyocellular lipid content did not differ between cohorts. However, the size of LDs in the subsarcolemmal region (SS) of type II muscle fibres was larger in LOW compared with HIGH (p = 0.01). Additionally, insulin receptor-β (IRβ) interactions with regulatory proteins CD36 and Fyn were lower in LOW versus HIGH (p < 0.01), which aligned with attenuated insulin-mediated Tyr phosphorylation of IRβ and downstream insulin-signalling proteins in LOW. CONCLUSION Collectively, reduced ability for insulin to suppress FA mobilization, with accompanying modifications in intramyocellular LD size and distribution, and diminished IRβ interaction with key regulatory proteins may be key contributors to impaired insulin-mediated glucose uptake commonly found in adults with obesity.
Collapse
Affiliation(s)
- Michael W Schleh
- Substrate Metabolism Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Benjamin J Ryan
- Substrate Metabolism Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Cheehoon Ahn
- Substrate Metabolism Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Alison C Ludzki
- Substrate Metabolism Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Douglas W Van Pelt
- Substrate Metabolism Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Lisa M Pitchford
- Substrate Metabolism Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Olivia K Chugh
- Substrate Metabolism Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Austin T Luker
- Substrate Metabolism Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Kathryn E Luker
- Center for Molecular Imaging, Department of Radiology, University of Michigan, Ann Arbor, Michigan, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, Michigan, USA
| | - Dmitri Samovski
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Nada A Abumrad
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Charles F Burant
- Division of Metabolism, Endocrinology, and Diabetes, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Jeffrey F Horowitz
- Substrate Metabolism Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
2
|
Fiorenza M, Onslev J, Henríquez-Olguín C, Persson KW, Hesselager SA, Jensen TE, Wojtaszewski JFP, Hostrup M, Bangsbo J. Reducing the mitochondrial oxidative burden alleviates lipid-induced muscle insulin resistance in humans. SCIENCE ADVANCES 2024; 10:eadq4461. [PMID: 39475607 PMCID: PMC11524190 DOI: 10.1126/sciadv.adq4461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 09/23/2024] [Indexed: 11/02/2024]
Abstract
Preclinical models suggest mitochondria-derived oxidative stress as an underlying cause of insulin resistance. However, it remains unknown whether this pathophysiological mechanism is conserved in humans. Here, we used an invasive in vivo mechanistic approach to interrogate muscle insulin action while selectively manipulating the mitochondrial redox state in humans. To this end, we conducted insulin clamp studies combining intravenous infusion of a lipid overload with intake of a mitochondria-targeted antioxidant (mitoquinone). Under lipid overload, selective modulation of mitochondrial redox state by mitoquinone enhanced insulin-stimulated glucose uptake in skeletal muscle. Mechanistically, mitoquinone did not affect canonical insulin signaling but augmented insulin-stimulated glucose transporter type 4 (GLUT4) translocation while reducing the mitochondrial oxidative burden under lipid oversupply. Complementary ex vivo studies in human muscle fibers exposed to high intracellular lipid levels revealed that mitoquinone improves features of mitochondrial bioenergetics, including diminished mitochondrial H2O2 emission. These findings provide translational and mechanistic evidence implicating mitochondrial oxidants in the development of lipid-induced muscle insulin resistance in humans.
Collapse
Affiliation(s)
- Matteo Fiorenza
- August Krogh Section for Human Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen 2100, Denmark
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
| | - Johan Onslev
- August Krogh Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen 2100, Denmark
| | - Carlos Henríquez-Olguín
- August Krogh Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen 2100, Denmark
- Exercise Science Laboratory, Faculty of Medicine, Universidad Finis Terrae, Santiago 1509, Chile
| | - Kaspar W. Persson
- August Krogh Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen 2100, Denmark
| | - Sofie A. Hesselager
- August Krogh Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen 2100, Denmark
| | - Thomas E. Jensen
- August Krogh Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen 2100, Denmark
| | - Jørgen F. P. Wojtaszewski
- August Krogh Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen 2100, Denmark
| | - Morten Hostrup
- August Krogh Section for Human Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen 2100, Denmark
| | - Jens Bangsbo
- August Krogh Section for Human Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen 2100, Denmark
| |
Collapse
|
3
|
Ansari P, Khan JT, Chowdhury S, Reberio AD, Kumar S, Seidel V, Abdel-Wahab YHA, Flatt PR. Plant-Based Diets and Phytochemicals in the Management of Diabetes Mellitus and Prevention of Its Complications: A Review. Nutrients 2024; 16:3709. [PMID: 39519546 PMCID: PMC11547802 DOI: 10.3390/nu16213709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 09/27/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024] Open
Abstract
Diabetes mellitus (DM) is currently regarded as a global public health crisis for which lifelong treatment with conventional drugs presents limitations in terms of side effects, accessibility, and cost. Type 2 diabetes (T2DM), usually associated with obesity, is characterized by elevated blood glucose levels, hyperlipidemia, chronic inflammation, impaired β-cell function, and insulin resistance. If left untreated or when poorly controlled, DM increases the risk of vascular complications such as hypertension, nephropathy, neuropathy, and retinopathy, which can be severely debilitating or life-threatening. Plant-based foods represent a promising natural approach for the management of T2DM due to the vast array of phytochemicals they contain. Numerous epidemiological studies have highlighted the importance of a diet rich in plant-based foods (vegetables, fruits, spices, and condiments) in the prevention and management of DM. Unlike conventional medications, such natural products are widely accessible, affordable, and generally free from adverse effects. Integrating plant-derived foods into the daily diet not only helps control the hyperglycemia observed in DM but also supports weight management in obese individuals and has broad health benefits. In this review, we provide an overview of the pathogenesis and current therapeutic management of DM, with a particular focus on the promising potential of plant-based foods.
Collapse
Affiliation(s)
- Prawej Ansari
- Comprehensive Diabetes Center, Heersink School of Medicine, University of Alabama, Birmingham (UAB), Birmingham, AL 35233, USA
- School of Pharmacy and Public Health, Department of Pharmacy, Independent University, Bangladesh (IUB), Dhaka 1229, Bangladesh
- Centre for Diabetes Research, School of Biomedical Sciences, Ulster University, Coleraine BT52 1SA, UK; (Y.H.A.A.-W.); (P.R.F.)
| | - Joyeeta T. Khan
- School of Pharmacy and Public Health, Department of Pharmacy, Independent University, Bangladesh (IUB), Dhaka 1229, Bangladesh
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences (UAMS), Little Rock, AR 72205, USA
| | - Suraiya Chowdhury
- School of Pharmacy and Public Health, Department of Pharmacy, Independent University, Bangladesh (IUB), Dhaka 1229, Bangladesh
| | - Alexa D. Reberio
- School of Pharmacy and Public Health, Department of Pharmacy, Independent University, Bangladesh (IUB), Dhaka 1229, Bangladesh
| | - Sandeep Kumar
- Comprehensive Diabetes Center, Heersink School of Medicine, University of Alabama, Birmingham (UAB), Birmingham, AL 35233, USA
| | - Veronique Seidel
- Natural Products Research Laboratory, Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow G4 0RE, UK;
| | - Yasser H. A. Abdel-Wahab
- Centre for Diabetes Research, School of Biomedical Sciences, Ulster University, Coleraine BT52 1SA, UK; (Y.H.A.A.-W.); (P.R.F.)
| | - Peter R. Flatt
- Centre for Diabetes Research, School of Biomedical Sciences, Ulster University, Coleraine BT52 1SA, UK; (Y.H.A.A.-W.); (P.R.F.)
| |
Collapse
|
4
|
Shabestari M, Azizi R, Ghadiri-Anari A. Type 2 diabetes and susceptibility to COVID-19: a machine learning analysis. BMC Endocr Disord 2024; 24:221. [PMID: 39434075 PMCID: PMC11492751 DOI: 10.1186/s12902-024-01758-3] [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: 07/23/2024] [Accepted: 10/16/2024] [Indexed: 10/23/2024] Open
Abstract
BACKGROUND Type 2 diabetes mellitus (T2DM) was one of the most prevalent comorbidities among patients with coronavirus disease 2019 (COVID-19). Interactions between different metabolic parameters contribute to the susceptibility to the virus; thereby, this study aimed to rank the importance of clinical and laboratory variables as risk factors for COVID-19 or as protective factors against it by applying machine learning methods. METHOD This study is a retrospective cohort conducted at a single center, focusing on a population with T2DM. The patients attended the Yazd Diabetes Research Center in Yazd, Iran, from February 20, 2020, to October 21, 2020. Clinical and laboratory data were collected within three months before the onset of the COVID-19 pandemic in Iran. 59 patients were infected with COVID-19, while 59 were not. The dataset was split into 70% training and 30% test sets. Principal Component Analysis (PCA) was applied to the data. The most important components were selected using a 'sequential feature selector' and scored by a Linear Discriminant Analysis model. PCA loadings were then multiplied by the PCs' scores to determine the importance of the original variables in contracting COVID-19. RESULTS HDL-C, followed by eGFR, showed a strong negative correlation with the risk of contracting the virus. Higher levels of HDL-C and eGFR offer protection against COVID-19 in the T2DM population. But, the ratio of BUN to creatinine did not show any correlation. Conversely, the AIP, TyG index and TG showed the most positive correlation with susceptibility to COVID-19 in such a way that higher levels of these factors increase the risk of contracting the virus. The positive correlation of diastolic BP, TyG-BMI index, MAP, BMI, weight, TC, FPG, HbA1C, Cr, systolic BP, BUN, and LDL-C with the risk of COVID-19 decreased, respectively. CONCLUSION The atherogenic index of plasma, triglyceride glucose index, and triglyceride levels are the most significant risk factors for COVID-19 contracting in individuals with T2DM. Meanwhile, high-density lipoprotein cholesterol is the most protective factor.
Collapse
Affiliation(s)
| | - Reyhaneh Azizi
- Diabetes Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Akram Ghadiri-Anari
- Diabetes Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.
| |
Collapse
|
5
|
Zhong F, Chen X, Li J. The burden of type 2 diabetes attributable to dietary risks in China: Insights from the global burden of disease study 2021. Public Health 2024; 237:122-129. [PMID: 39368403 DOI: 10.1016/j.puhe.2024.09.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Revised: 09/25/2024] [Accepted: 09/30/2024] [Indexed: 10/07/2024]
Abstract
OBJECTIVES This study aims to comprehensively assess how dietary risk factors have influenced the prevalence of Type 2 Diabetes Mellitus (T2DM) in China from 1990 to 2021. The study seeks to provide robust data and scientific evidence essential for formulating effective preventive and control strategies to combat T2DM in China. STUDY DESIGN This cross-sectional study conducted secondary analyses using data from the Global Burden of Disease 2021 (GBD 2021) to assess the burden of T2DM in China attributable to dietary risks. METHODS The study analyzed age-adjusted metrics related to T2DM, including death counts, Disability-Adjusted Life Years (DALYs), and Age-Standardized Rates (ASRs), using GBD 2021 data, stratified by age and sex. Additionally, Estimated Annual Percentage Changes (EAPCs) were employed to track trends over time. RESULTS In 2021, the results show that 21.43 % of T2DM-related deaths and 23.51 % of DALYs were attributable to dietary risk factors, notably a diet low in whole grains and high in red and processed meats. Over the period from 1990 to 2021, there has been an increasing trend in the EAPCs of death rates and DALYs associated with dietary risks in China, suggesting a substantial impact of dietary factors on the burden of T2DM in the country. CONCLUSION This study highlights the urgent need for targeted public health interventions to promote dietary changes and reduce the burden of T2DM in China.
Collapse
Affiliation(s)
- Feifei Zhong
- School of Public Health and Healthcare Management, Gannan Medical University, Ganzhou, Jiangxi, 341000, China.
| | - Xiaochen Chen
- School of Public Health and Healthcare Management, Gannan Medical University, Ganzhou, Jiangxi, 341000, China.
| | - Juan Li
- School of Public Health and Healthcare Management, Gannan Medical University, Ganzhou, Jiangxi, 341000, China.
| |
Collapse
|
6
|
Abolfazli S, Butler AE, Kesharwani P, Sahebkar A. The beneficial impact of curcumin on cardiac lipotoxicity. J Pharm Pharmacol 2024; 76:1269-1283. [PMID: 39180454 DOI: 10.1093/jpp/rgae102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 07/02/2024] [Indexed: 08/26/2024]
Abstract
Lipotoxicity is defined as a prolonged metabolic imbalance of lipids that results in ectopic fat distribution in peripheral organs such as the liver, heart, and kidney. The harmful consequences of excessive lipid accumulation in cardiomyocytes cause cardiac lipotoxicity, which alters the structure and function of the heart. Obesity and diabetes are linked to lipotoxic cardiomyopathy. These anomalies might be caused by a harmful metabolic shift that accumulates toxic lipids and shifts glucose oxidation to less fatty acid oxidation. Research has linked fatty acids, fatty acyl coenzyme A, diacylglycerol, and ceramide to lipotoxic stress in cells. This stress can be brought on by apoptosis, impaired insulin signaling, endoplasmic reticulum stress, protein kinase C activation, p38 Ras-mitogen-activated protein kinase (MAPK) activation, or modification of peroxisome proliferator-activated receptors (PPARs) family members. Curcuma longa is used to extract curcumin, a hydrophobic polyphenol derivative with a variety of pharmacological characteristics. Throughout the years, curcumin has been utilized as an anti-inflammatory, antioxidant, anticancer, hepatoprotective, cardioprotective, anti-diabetic, and anti-obesity drug. Curcumin reduces cardiac lipotoxicity by inhibiting apoptosis and decreasing the expression of apoptosis-related proteins, reducing the expression of inflammatory cytokines, activating the autophagy signaling pathway, and inhibiting the expression of endoplasmic reticulum stress marker proteins.
Collapse
Affiliation(s)
- Sajad Abolfazli
- Student Research Committee, School of Pharmacy, Mazandaran University Medical Science, Sari, Iran
| | - Alexandra E Butler
- Research Department, Royal College of Surgeons in Ireland, Bahrain, Adliya, Bahrain
| | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Amirhossein Sahebkar
- Center for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
7
|
Ma S, Morris MC, Hubal MJ, Ross LM, Huffman KM, Vann CG, Moore N, Hauser ER, Bareja A, Jiang R, Kummerfeld E, Barberio MD, Houmard JA, Bennett WB, Johnson JL, Timmons JA, Broderick G, Kraus VB, Aliferis CF, Kraus WE. Sex-Specific Skeletal Muscle Gene Expression Responses to Exercise Reveal Novel Direct Mediators of Insulin Sensitivity Change. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.09.07.24313236. [PMID: 39281755 PMCID: PMC11398589 DOI: 10.1101/2024.09.07.24313236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/18/2024]
Abstract
BACKGROUND Understanding the causal pathways, systems, and mechanisms through which exercise impacts human health is complex. This study explores molecular signaling related to whole-body insulin sensitivity (Si) by examining changes in skeletal muscle gene expression. The analysis considers differences by biological sex, exercise amount, and exercise intensity to identify potential molecular targets for developing pharmacologic agents that replicate the health benefits of exercise. METHODS The study involved 53 participants from the STRRIDE I and II trials who completed eight months of aerobic training. Skeletal muscle gene expression was measured using Affymetrix and Illumina technologies, while pre- and post-training Si was assessed via an intravenous glucose tolerance test. A novel gene discovery protocol, integrating three literature-derived and data-driven modeling strategies, was employed to identify causal pathways and direct causal factors based on differentially expressed transcripts associated with exercise intensity and amount. RESULTS In women, the transcription factor targets identified were primarily influenced by exercise amount and were generally inhibitory. In contrast, in men, these targets were driven by exercise intensity and were generally activating. Transcription factors such as ATF1, CEBPA, BACH2, and STAT1 were commonly activating in both sexes. Specific transcriptional targets related to exercise-induced Si improvements included TACR3 and TMC7 for intensity-driven effects, and GRIN3B and EIF3B for amount-driven effects. Two key signaling pathways mediating aerobic exercise-induced Si improvements were identified: one centered on estrogen signaling and the other on phorbol ester (PKC) signaling, both converging on the epidermal growth factor receptor (EGFR) and other relevant targets. CONCLUSIONS The signaling pathways mediating Si improvements from aerobic exercise differed by sex and were further distinguished by exercise intensity and amount. Transcriptional adaptations in skeletal muscle related to Si improvements appear to be causally linked to estrogen and PKC signaling, with EGFR and other identified targets emerging as potential skeletal muscle-specific drug targets to mimic the beneficial effects of exercise on Si.
Collapse
Affiliation(s)
- S Ma
- Institute for Health Informatics (IHI), Academic Health Center, University of Minnesota, Minneapolis, MN 55455
| | - M C Morris
- Center for Clinical Systems Biology, Rochester General Hospital, Rochester, NY 14621
| | - M J Hubal
- Department of Kinesiology, Indiana University - Indianapolis, Indianapolis IN 46202
| | - L M Ross
- Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC 27701
| | - K M Huffman
- Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC 27701
| | - C G Vann
- Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC 27701
| | - N Moore
- Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC 27701
| | - E R Hauser
- Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC 27701
- Department of Head and Neck Surgery & Communication Sciences, Duke University School of Medicine, Durham, NC 27701
| | - A Bareja
- Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC 27701
| | - R Jiang
- Department of Head and Neck Surgery & Communication Sciences, Duke University School of Medicine, Durham, NC 27701
| | - E Kummerfeld
- Institute for Health Informatics (IHI), Academic Health Center, University of Minnesota, Minneapolis, MN 55455
| | - M D Barberio
- Department of Exercise and Nutrition Sciences, George Washington University, Washington DC 20052
| | - J A Houmard
- Department of Kinesiology, ECU, Greenville, NC 27858
| | - W B Bennett
- Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC 27701
| | - J L Johnson
- Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC 27701
| | - J A Timmons
- School of Medicine and Dentistry, Queen Mary University of London, UK
| | - G Broderick
- Center for Clinical Systems Biology, Rochester General Hospital, Rochester, NY 14621
| | - V B Kraus
- Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC 27701
| | - C F Aliferis
- Institute for Health Informatics (IHI), Academic Health Center, University of Minnesota, Minneapolis, MN 55455
| | - W E Kraus
- Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC 27701
| |
Collapse
|
8
|
Qiu X, Lan X, Li L, Chen H, Zhang N, Zheng X, Xie X. The role of perirenal adipose tissue deposition in chronic kidney disease progression: Mechanisms and therapeutic implications. Life Sci 2024; 352:122866. [PMID: 38936605 DOI: 10.1016/j.lfs.2024.122866] [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: 04/24/2024] [Revised: 06/11/2024] [Accepted: 06/20/2024] [Indexed: 06/29/2024]
Abstract
Chronic kidney disease (CKD) represents a significant and escalating global health challenge, with morbidity and mortality rates rising steadily. Evidence increasingly implicates perirenal adipose tissue (PRAT) deposition as a contributing factor in the pathogenesis of CKD. This review explores how PRAT deposition may exert deleterious effects on renal structure and function. The anatomical proximity of PRAT to the kidneys not only potentially causes mechanical compression but also leads to the dysregulated secretion of adipokines and inflammatory mediators, such as adiponectin, leptin, visfatin, tumor necrosis factor-alpha (TNF-α), interleukin-6 (IL-6), and exosomes. Additionally, PRAT deposition may contribute to renal lipotoxicity through elevated levels of free fatty acids (FFA), triglycerides (TAG), diacylglycerol (DAG), and ceramides (Cer). PRAT deposition is also linked to the hyperactivation of the renin-angiotensin-aldosterone system (RAAS), which further exacerbates CKD progression. Recognizing PRAT deposition as an independent risk factor for CKD underscores the potential of targeting PRAT as a novel strategy for the prevention and management of CKD. This review further discusses interventions that could include measuring PRAT thickness to establish a baseline, managing metabolic risk factors that promote its deposition, and inhibiting key PRAT-induced signaling pathways.
Collapse
Affiliation(s)
- Xiang Qiu
- The School of Basic Medical Sciences, Southwest Medical University, Luzhou, China; Public Center of Experimental Technology, Model Animal and Human Disease Research of Luzhou Key Laboratory, Southwest Medical University, Luzhou, China
| | - Xin Lan
- The School of Basic Medical Sciences, Southwest Medical University, Luzhou, China; Public Center of Experimental Technology, Model Animal and Human Disease Research of Luzhou Key Laboratory, Southwest Medical University, Luzhou, China
| | - Langhui Li
- The School of Basic Medical Sciences, Southwest Medical University, Luzhou, China; Public Center of Experimental Technology, Model Animal and Human Disease Research of Luzhou Key Laboratory, Southwest Medical University, Luzhou, China
| | - Huan Chen
- The School of Basic Medical Sciences, Southwest Medical University, Luzhou, China; Public Center of Experimental Technology, Model Animal and Human Disease Research of Luzhou Key Laboratory, Southwest Medical University, Luzhou, China; Nucleic Acid Medicine of Luzhou Key Laboratory, Southwest Medical University, Luzhou, China
| | - Ningjuan Zhang
- The School of Clinical Medical Sciences, Southwest Medical University, Luzhou, China
| | - Xiaoli Zheng
- The School of Basic Medical Sciences, Southwest Medical University, Luzhou, China.
| | - Xiang Xie
- The School of Basic Medical Sciences, Southwest Medical University, Luzhou, China; Public Center of Experimental Technology, Model Animal and Human Disease Research of Luzhou Key Laboratory, Southwest Medical University, Luzhou, China.
| |
Collapse
|
9
|
Gupta MK, Gouda G, Vadde R. Relation Between Obesity and Type 2 Diabetes: Evolutionary Insights, Perspectives and Controversies. Curr Obes Rep 2024; 13:475-495. [PMID: 38850502 DOI: 10.1007/s13679-024-00572-1] [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] [Accepted: 05/13/2024] [Indexed: 06/10/2024]
Abstract
PURPOSE OF REVIEW Since the mid-twentieth century, obesity and its related comorbidities, notably insulin resistance (IR) and type 2 diabetes (T2D), have surged. Nevertheless, their underlying mechanisms remain elusive. Evolutionary medicine (EM) sheds light on these issues by examining how evolutionary processes shape traits and diseases, offering insights for medical practice. This review summarizes the pathogenesis and genetics of obesity-related IR and T2D. Subsequently, delving into their evolutionary connections. Addressing limitations and proposing future research directions aims to enhance our understanding of these conditions, paving the way for improved treatments and prevention strategies. RECENT FINDINGS Several evolutionary hypotheses have been proposed to unmask the origin of obesity-related IR and T2D, e.g., the "thrifty genotype" hypothesis suggests that certain "thrifty genes" that helped hunter-gatherer populations efficiently store energy as fat during feast-famine cycles are now maladaptive in our modern obesogenic environment. The "drifty genotype" theory suggests that if thrifty genes were advantageous, they would have spread widely, but proposes genetic drift instead. The "behavioral switch" and "carnivore connection" hypotheses propose insulin resistance as an adaptation for a brain-dependent, low-carbohydrate lifestyle. The thrifty phenotype theory suggests various metabolic outcomes shaped by genes and environment during development. However, the majority of these hypotheses lack experimental validation. Understanding why ancestral advantages now predispose us to diseases may aid in drug development and prevention of disease. EM helps us to understand the evolutionary relation between obesity-related IR and T2D. But still gaps and contradictions persist. Further interdisciplinary research is required to elucidate complete mechanisms.
Collapse
Affiliation(s)
- Manoj Kumar Gupta
- Department of Biotechnology & Bioinformatics, Yogi Vemana University, Kadapa, 516005, Andhra Pradesh, India.
| | - Gayatri Gouda
- ICAR-National Rice Research Institute, Cuttack, 753 006, Odisha, India
| | - Ramakrishna Vadde
- Department of Biotechnology & Bioinformatics, Yogi Vemana University, Kadapa, 516005, Andhra Pradesh, India
| |
Collapse
|
10
|
Araujo SL, Martins PL, Pereira THDS, Sampaio TL, de Menezes RRPPB, da Costa MDR, Martins AMC, da Silva ING, de Morais GB, Evangelista JSAM. Evidence of obesity-induced inflammatory changes in client-owned cats. Vet World 2024; 17:1685-1692. [PMID: 39328456 PMCID: PMC11422647 DOI: 10.14202/vetworld.2024.1685-1692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Accepted: 07/08/2024] [Indexed: 09/28/2024] Open
Abstract
Background and Aim Insulin resistance and type 2 diabetes mellitus are common health issues in obese (OB) cats. In humans, obesity leads to alterations in adipokine and proinflammatory cytokine secretion, causing persistent inflammation. The inflammatory impact of obesity in cats remains unproven. This study investigated associations between obesity and inflammatory and metabolic changes in three groups of client-owned Brazilian domestic shorthair cats: naturally lean, overweight (OW), and OB. Materials and Methods Cats from the Veterinary Hospital of Professor Sylvio Barbosa e Cardoso (FAVET/UECE) were clinically evaluated. Blood samples were collected for hematological and biochemical profile measurements, and part of the serum was used for measuring adipokine and inflammatory cytokines using sandwich enzyme-linked immunosorbent assay. Results In both the OW and OB groups, serum cholesterol and insulin concentrations increased, while triglyceride concentrations were notably elevated in the OB group. In the OW and OB groups, serum adiponectin, tumor necrosis factor-α, and interleukin-1β levels were elevated, and leptin levels were significantly higher in the OB group. Conclusion This study is the first in Brazil to reveal increased serum levels of inflammatory markers in OW and OB client-owned felines. OW cats exhibited higher proinflammatory marker levels, implying obesity-induced inflammation.
Collapse
Affiliation(s)
- Steffi L. Araujo
- Laboratory of Comparative Experimental Morphology, Faculty of Veterinary, State University of Ceará, Fortaleza, 60714-903, Ceará, Brazil
| | - Patricia L. Martins
- Laboratory of Comparative Experimental Morphology, Faculty of Veterinary, State University of Ceará, Fortaleza, 60714-903, Ceará, Brazil
| | | | - Tiago L. Sampaio
- Department of Clinical and Toxicological Analysis, Faculty of Pharmacy, Dentistry and Nursing, Federal University of Ceará, Fortaleza, 60430-275, Ceará, Brazil
| | | | - Mac D. Rodrigues da Costa
- Department of Clinical and Toxicological Analysis, Faculty of Pharmacy, Dentistry and Nursing, Federal University of Ceará, Fortaleza, 60430-275, Ceará, Brazil
| | - Alice M. Costa Martins
- Department of Clinical and Toxicological Analysis, Faculty of Pharmacy, Dentistry and Nursing, Federal University of Ceará, Fortaleza, 60430-275, Ceará, Brazil
| | - Isaac Neto Goes da Silva
- Laboratoy of Veterinary Clinical Pathology, Faculty of Veterinary, State University of Ceará, Fortaleza, 60714-903, Ceará, Brazil
| | - Glayciane Bezerra de Morais
- Laboratory of Comparative Experimental Morphology, Faculty of Veterinary, State University of Ceará, Fortaleza, 60714-903, Ceará, Brazil
| | | |
Collapse
|
11
|
Carl CS, Jensen MM, Sjøberg KA, Constantin-Teodosiu D, Hill IR, Kjøbsted R, Greenhaff PL, Wojtaszewski JFP, Richter EA, Fritzen AM, Kiens B. Pharmacological Activation of PDC Flux Reverses Lipid-Induced Inhibition of Insulin Action in Muscle During Recovery From Exercise. Diabetes 2024; 73:1072-1083. [PMID: 38608261 DOI: 10.2337/db23-0879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Accepted: 04/02/2024] [Indexed: 04/14/2024]
Abstract
Insulin resistance is a risk factor for type 2 diabetes, and exercise can improve insulin sensitivity. However, following exercise, high circulating fatty acid (FA) levels might counteract this. We hypothesized that such inhibition would be reduced by forcibly increasing carbohydrate oxidation through pharmacological activation of the pyruvate dehydrogenase complex (PDC). Insulin-stimulated glucose uptake was examined with a crossover design in healthy young men (n = 8) in a previously exercised and a rested leg during a hyperinsulinemic-euglycemic clamp 5 h after one-legged exercise with 1) infusion of saline, 2) infusion of intralipid imitating circulating FA levels during recovery from whole-body exercise, and 3) infusion of intralipid + oral PDC activator, dichloroacetate (DCA). Intralipid infusion reduced insulin-stimulated glucose uptake by 19% in the previously exercised leg, which was not observed in the contralateral rested leg. Interestingly, this effect of intralipid in the exercised leg was abolished by DCA, which increased muscle PDC activity (130%) and flux (acetylcarnitine 130%) and decreased inhibitory phosphorylation of PDC on Ser293 (∼40%) and Ser300 (∼80%). Novel insight is provided into the regulatory interaction between glucose and lipid metabolism during exercise recovery. Coupling exercise and PDC flux activation upregulated the capacity for both glucose transport (exercise) and oxidation (DCA), which seems necessary to fully stimulate insulin-stimulated glucose uptake during recovery. ARTICLE HIGHLIGHTS
Collapse
Affiliation(s)
- Christian S Carl
- The August Krogh Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Marie M Jensen
- Clinical Research, Copenhagen University Hospital-Steno Diabetes Center Copenhagen, Herlev, Denmark
| | - Kim A Sjøberg
- The August Krogh Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Dumitru Constantin-Teodosiu
- David Greenfield Human Physiology Laboratory, National Institute for Health and Care Research Nottingham Biomedical Research Centre, Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, The Medical School, Queen's Medical Centre, University of Nottingham, Nottingham, U.K
| | - Ian R Hill
- David Greenfield Human Physiology Laboratory, National Institute for Health and Care Research Nottingham Biomedical Research Centre, Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, The Medical School, Queen's Medical Centre, University of Nottingham, Nottingham, U.K
| | - Rasmus Kjøbsted
- The August Krogh Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Paul L Greenhaff
- David Greenfield Human Physiology Laboratory, National Institute for Health and Care Research Nottingham Biomedical Research Centre, Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, The Medical School, Queen's Medical Centre, University of Nottingham, Nottingham, U.K
| | - Jørgen F P Wojtaszewski
- The August Krogh Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Erik A Richter
- The August Krogh Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Andreas M Fritzen
- The August Krogh Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Bente Kiens
- The August Krogh Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
12
|
Del Carmen Fernández-Fígares Jiménez M. Plant foods, healthy plant-based diets, and type 2 diabetes: a review of the evidence. Nutr Rev 2024; 82:929-948. [PMID: 37550262 DOI: 10.1093/nutrit/nuad099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/09/2023] Open
Abstract
Type 2 diabetes (T2D) is a metabolic chronic disease in which insulin resistance and insufficient insulin production lead to elevated blood glucose levels. The prevalence of T2D is growing worldwide, mainly due to obesity and the adoption of Western diets. Replacing animal foods with healthy plant foods is associated with a lower risk of T2D in prospective studies. In randomized controlled trials, the consumption of healthy plant foods in place of animal foods led to cardiometabolic improvements in patients with T2D or who were at high risk of the disease. Dietary patterns that limit or exclude animal foods and focus on healthy plant foods (eg, fruits, vegetables, whole grains, nuts, legumes), known as healthy, plant-based diets, are consistently associated with a lower risk of T2D in cohort studies. The aim of this review is to examine the differential effects of plant foods and animal foods on T2D risk and to describe the existing literature about the role of healthy, plant-based diets, particularly healthy vegan diets, in T2D prevention and management. The evidence from cohort studies and randomized controlled trials will be reported, in addition to the potential biological mechanisms that seem to be involved.
Collapse
|
13
|
Kovacevic I, Schmidt PH, Kowalski A, Helms BJ, Lest CHAVD, Kluttig A, Posern G. ER stress inhibition enhances formation of triacylglcerols and protects endothelial cells from lipotoxicity. Cell Commun Signal 2024; 22:304. [PMID: 38831326 PMCID: PMC11145897 DOI: 10.1186/s12964-024-01682-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 05/27/2024] [Indexed: 06/05/2024] Open
Abstract
Elevated concentrations of palmitate in serum of obese individuals can impair endothelial function, contributing to development of cardiovascular disease. Although several molecular mechanisms of palmitate-induced endothelial dysfunction have been proposed, there is no consensus on what signaling event is the initial trigger of detrimental palmitate effects. Here we report that inhibitors of ER stress or ceramid synthesis can rescue palmitate-induced autophagy impairment in macro- and microvascular endothelial cells. Furthermore, palmitate-induced cholesterol synthesis was reverted using these inhibitors. Similar to cell culture data, autophagy markers were increased in serum of obese individuals. Subsequent lipidomic analysis revealed that palmitate changed the composition of membrane phospholipids in endothelial cells and that these effects were not reverted upon application of above-mentioned inhibitors. However, ER stress inhibition in palmitate-treated cells enhanced the synthesis of trilglycerides and restored ceramide levels to control condition. Our results suggest that palmitate induces ER-stress presumably by shift in membrane architecture, leading to impaired synthesis of triglycerides and enhanced production of ceramides and cholesterol, which altogether enhances lipotoxicity of palmitate in endothelial cells.
Collapse
Affiliation(s)
- Igor Kovacevic
- Institute of Physiological Chemistry, Medical Faculty, Martin Luther University Halle-Wittenberg, 06114, Halle (Saale), Germany.
| | - Paula Henriette Schmidt
- Institute of Physiological Chemistry, Medical Faculty, Martin Luther University Halle-Wittenberg, 06114, Halle (Saale), Germany
| | - Annkatrin Kowalski
- Institute of Physiological Chemistry, Medical Faculty, Martin Luther University Halle-Wittenberg, 06114, Halle (Saale), Germany
| | - Bernd J Helms
- Department Biomolecular Health Sciences, Veterinary Medicine, Utrecht University, Utrecht, 3584CM, The Netherlands
| | - Chris H A van de Lest
- Department Biomolecular Health Sciences, Veterinary Medicine, Utrecht University, Utrecht, 3584CM, The Netherlands
| | - Alexander Kluttig
- Institute of Medical Epidemiology, Biostatistics, and Informatics, Medical Faculty, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Guido Posern
- Institute of Physiological Chemistry, Medical Faculty, Martin Luther University Halle-Wittenberg, 06114, Halle (Saale), Germany
| |
Collapse
|
14
|
El-Araby RE, Tu Q, Xie Y, Aboushousha T, Li Z, Xu X, Zhu ZX, Dong LQ, Chen J. Adiponectin mRNA Conjugated with Lipid Nanoparticles Specifically Targets the Pathogenesis of Type 2 Diabetes. Aging Dis 2024:AD.2024.0162. [PMID: 38916734 DOI: 10.14336/ad.2024.0162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Accepted: 05/15/2024] [Indexed: 06/26/2024] Open
Abstract
Type 2 diabetes (T2D) is a widespread health condition both in the United States and around the world, with insulin resistance playing a critical role in its development. Effective treatment strategies are essential for managing T2D and mitigating associated risks. Adiponectin (APN), secreted by adipocytes, exhibits an inverse correlation with obesity-related adiposity, and its levels are negatively associated with insulin resistance and body mass index. This study aimed to enhance endogenous APN levels in a diet-induced obese (DIO) mouse model using lipid nanoparticles (LNP) as safe delivery agents for APN mRNA conjugates. The results indicate that APN-mRNA-LNP administration successfully induced APN synthesis in various tissues, including muscle, liver, kidney, pancreas, and adipose cells. This induction was associated with several positive outcomes, such as preventing diet-induced body weight gain, improving hyperglycemia by promoting Glut-4 expression, alleviating diabetic nephropathy symptoms by blocking the EGFR pathway, and reducing pro-inflammatory cytokine production. In addition, the treatment demonstrated enhanced insulin sensitivity by activating DGKd and inhibiting PKCε. This resulted in reactivation of insulin receptors in insulin target tissues and stimulation of insulin secretion from pancreatic beta cells. The findings of the present study highlight the potential of APN-mRNA-LNP-based nucleic acid therapy as a treatment for type 2 diabetes, offering a comprehensive approach to addressing its complexities.
Collapse
Affiliation(s)
- Rady E El-Araby
- Division of Oral Biology, Tufts University School of Dental Medicine, Boston, MA 02111, USA
- Theodor Bilharz Research Institute, Ministry of scientific Research, Cairo, Egypt
| | - Qisheng Tu
- Division of Oral Biology, Tufts University School of Dental Medicine, Boston, MA 02111, USA
| | - Ying Xie
- Division of Oral Biology, Tufts University School of Dental Medicine, Boston, MA 02111, USA
| | - Tarek Aboushousha
- Theodor Bilharz Research Institute, Ministry of scientific Research, Cairo, Egypt
| | - Zhongyu Li
- Department of Chemical and Materials Engineering, New Jersey Institute of Technology, Newark, NJ, USA
| | - Xiaoyang Xu
- Department of Chemical and Materials Engineering, New Jersey Institute of Technology, Newark, NJ, USA
| | - Zoe X Zhu
- Division of Oral Biology, Tufts University School of Dental Medicine, Boston, MA 02111, USA
| | - Lily Q Dong
- Department of Cell Systems and Anatomy, The University of Texas Health San Antonio, San Antonio, Texas 78229, USA
| | - Jake Chen
- Division of Oral Biology, Tufts University School of Dental Medicine, Boston, MA 02111, USA
- Department of Genetics, Molecular and Cellular Biology, Tufts University School of Medicine, and Graduate School of Biomedical Sciences. 136 Harrison Ave, M&;ampV 811, Boston, MA 02111, USA
| |
Collapse
|
15
|
Ji H, Tang Z, Jiang K, Lyu S, Zhao Y, Feng J, Dai R, Liang H. Investigating potential biomarkers of acute pancreatitis in patients with a BMI>30 using Mendelian randomization and transcriptomic analysis. Lipids Health Dis 2024; 23:119. [PMID: 38649912 PMCID: PMC11034057 DOI: 10.1186/s12944-024-02102-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Accepted: 04/04/2024] [Indexed: 04/25/2024] Open
Abstract
BACKGROUND Acute pancreatitis (AP) has become a significant global health concern, and a high body mass index (BMI) has been identified as a key risk factor exacerbating this condition. Within this context, lipid metabolism assumes a critical role. The complex relationship between elevated BMI and AP, mediated by lipid metabolism, markedly increases the risk of complications and mortality. This study aimed to accurately define the correlation between BMI and AP, incorporating a comprehensive analysis of the interactions between individuals with high BMI and AP. METHODS Mendelian randomization (MR) analysis was first applied to determine the causal relationship between BMI and the risk of AP. Subsequently, three microarray datasets were obtained from the GEO database. This was followed by an analysis of differentially expressed genes and the application of weighted gene coexpression network analysis (WGCNA) to identify key modular genes associated with AP and elevated BMI. Functional enrichment analysis was then performed to shed light on disease pathogenesis. To identify the most informative genes, machine learning algorithms, including Random Forest (RF), Support Vector Machine-Recursive Feature Elimination (SVM-RFE), and Least Absolute Shrinkage and Selection Operator (LASSO), were employed. Subsequent analysis focused on the colocalization of the Quantitative Trait Loci (eQTL) data associated with the selected genes and Genome-Wide Association Studies (GWAS) data related to the disease. Preliminary verification of gene expression trends was conducted using external GEO datasets. Ultimately, the diagnostic potential of these genes was further confirmed through the development of an AP model in mice with a high BMI. RESULTS A total of 21 intersecting genes related to BMI>30, AP, and lipid metabolism were identified from the datasets. These genes were primarily enriched in pathways related to cytosolic DNA sensing, cytokine‒cytokine receptor interactions, and various immune and inflammatory responses. Next, three machine learning techniques were utilized to identify HADH as the most prevalent diagnostic gene. Colocalization analysis revealed that HADH significantly influenced the risk factors associated with BMI and AP. Furthermore, the trend in HADH expression within the external validation dataset aligned with the trend in the experimental data, thus providing a preliminary validation of the experimental findings.The changes in its expression were further validated using external datasets and quantitative real-time polymerase chain reaction (qPCR). CONCLUSION This study systematically identified HADH as a potential lipid metabolism-grounded biomarker for AP in patients with a BMI>30.
Collapse
Affiliation(s)
- Hua Ji
- Department of Hepatobilialy Surgery, General Surgery Center, General Hospital of Western Theater Command, Chengdu, 610083, China
- Department of General Surgery, Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Zheng Tang
- Department of Hepatobilialy Surgery, General Surgery Center, General Hospital of Western Theater Command, Chengdu, 610083, China
- Department of General Surgery, Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Kexin Jiang
- Department of Hepatobilialy Surgery, General Surgery Center, General Hospital of Western Theater Command, Chengdu, 610083, China
- College of Medicine, Affiliated Hospital of Southwest Jiaotong University, College of Medicine, Southwest Jiaotong University, Chengdu, 610031, China
| | - Shuang Lyu
- Department of Hepatobilialy Surgery, General Surgery Center, General Hospital of Western Theater Command, Chengdu, 610083, China
- College of Medicine, Affiliated Hospital of Southwest Jiaotong University, College of Medicine, Southwest Jiaotong University, Chengdu, 610031, China
| | - Yiwen Zhao
- Department of Hepatobilialy Surgery, General Surgery Center, General Hospital of Western Theater Command, Chengdu, 610083, China
- Department of General Surgery, Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Jiajie Feng
- Department of Hepatobilialy Surgery, General Surgery Center, General Hospital of Western Theater Command, Chengdu, 610083, China
- Department of General Surgery, Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Ruiwu Dai
- Department of Hepatobilialy Surgery, General Surgery Center, General Hospital of Western Theater Command, Chengdu, 610083, China.
- Department of General Surgery, Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China.
- College of Medicine, Affiliated Hospital of Southwest Jiaotong University, College of Medicine, Southwest Jiaotong University, Chengdu, 610031, China.
| | - Hongyin Liang
- Department of Hepatobilialy Surgery, General Surgery Center, General Hospital of Western Theater Command, Chengdu, 610083, China.
| |
Collapse
|
16
|
Petersen MC, Smith GI, Palacios HH, Farabi SS, Yoshino M, Yoshino J, Cho K, Davila-Roman VG, Shankaran M, Barve RA, Yu J, Stern JH, Patterson BW, Hellerstein MK, Shulman GI, Patti GJ, Klein S. Cardiometabolic characteristics of people with metabolically healthy and unhealthy obesity. Cell Metab 2024; 36:745-761.e5. [PMID: 38569471 PMCID: PMC11025492 DOI: 10.1016/j.cmet.2024.03.002] [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: 11/09/2023] [Revised: 01/06/2024] [Accepted: 03/06/2024] [Indexed: 04/05/2024]
Abstract
There is considerable heterogeneity in the cardiometabolic abnormalities associated with obesity. We evaluated multi-organ system metabolic function in 20 adults with metabolically healthy obesity (MHO; normal fasting glucose and triglycerides, oral glucose tolerance, intrahepatic triglyceride content, and whole-body insulin sensitivity), 20 adults with metabolically unhealthy obesity (MUO; prediabetes, hepatic steatosis, and whole-body insulin resistance), and 15 adults who were metabolically healthy lean. Compared with MUO, people with MHO had (1) altered skeletal muscle biology (decreased ceramide content and increased expression of genes involved in BCAA catabolism and mitochondrial structure/function); (2) altered adipose tissue biology (decreased expression of genes involved in inflammation and extracellular matrix remodeling and increased expression of genes involved in lipogenesis); (3) lower 24-h plasma glucose, insulin, non-esterified fatty acids, and triglycerides; (4) higher plasma adiponectin and lower plasma PAI-1 concentrations; and (5) decreased oxidative stress. These findings provide a framework of potential mechanisms responsible for MHO and the metabolic heterogeneity of obesity. This study was registered at ClinicalTrials.gov (NCT02706262).
Collapse
Affiliation(s)
- Max C Petersen
- Center for Human Nutrition, Washington University in St. Louis, St. Louis, MO, USA; Division of Endocrinology, Metabolism, and Lipid Research, Washington University in St. Louis, St. Louis, MO, USA
| | - Gordon I Smith
- Center for Human Nutrition, Washington University in St. Louis, St. Louis, MO, USA
| | - Hector H Palacios
- Center for Human Nutrition, Washington University in St. Louis, St. Louis, MO, USA
| | - Sarah S Farabi
- Center for Human Nutrition, Washington University in St. Louis, St. Louis, MO, USA; Goldfarb School of Nursing at Barnes-Jewish College, St. Louis, MO, USA
| | - Mihoko Yoshino
- Center for Human Nutrition, Washington University in St. Louis, St. Louis, MO, USA
| | - Jun Yoshino
- Center for Human Nutrition, Washington University in St. Louis, St. Louis, MO, USA; Division of Nephrology, Endocrinology and Metabolism, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Kevin Cho
- Department of Chemistry, Washington University in St. Louis, St. Louis, MO, USA
| | - Victor G Davila-Roman
- Cardiovascular Imaging and Clinical Research Core Laboratory, Cardiovascular Division, Washington University in St. Louis, St. Louis, MO, USA
| | | | - Ruteja A Barve
- Department of Genetics, Washington University in St. Louis, St. Louis, MO, USA
| | - Jinsheng Yu
- Department of Genetics, Washington University in St. Louis, St. Louis, MO, USA
| | - Jennifer H Stern
- Division of Endocrinology, Department of Medicine, University of Arizona College of Medicine, Tucson, AZ, USA
| | - Bruce W Patterson
- Center for Human Nutrition, Washington University in St. Louis, St. Louis, MO, USA
| | | | - Gerald I Shulman
- Departments of Internal Medicine and Cellular & Molecular Physiology, Yale School of Medicine, New Haven, CT, USA
| | - Gary J Patti
- Department of Chemistry, Washington University in St. Louis, St. Louis, MO, USA
| | - Samuel Klein
- Center for Human Nutrition, Washington University in St. Louis, St. Louis, MO, USA.
| |
Collapse
|
17
|
Barrett JS, Strauss JA, Chow LS, Shepherd SO, Wagenmakers AJM, Wang Y. GLUT4 localisation with the plasma membrane is unaffected by an increase in plasma free fatty acid availability. Lipids Health Dis 2024; 23:94. [PMID: 38566151 PMCID: PMC10986142 DOI: 10.1186/s12944-024-02079-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Accepted: 03/13/2024] [Indexed: 04/04/2024] Open
Abstract
BACKGROUND Insulin-stimulated glucose uptake into skeletal muscle occurs via translocation of GLUT4 from intracellular storage vesicles to the plasma membrane. Elevated free fatty acid (FFA) availability via a lipid infusion reduces glucose disposal, but this occurs in the absence of impaired proximal insulin signalling. Whether GLUT4 localisation to the plasma membrane is subsequently affected by elevated FFA availability is not known. METHODS Trained (n = 11) and sedentary (n = 10) individuals, matched for age, sex and body mass index, received either a 6 h lipid or glycerol infusion in the setting of a concurrent hyperinsulinaemic-euglycaemic clamp. Sequential muscle biopsies (0, 2 and 6 h) were analysed for GLUT4 membrane localisation and microvesicle size and distribution using immunofluorescence microscopy. RESULTS At baseline, trained individuals had more small GLUT4 spots at the plasma membrane, whereas sedentary individuals had larger GLUT4 spots. GLUT4 localisation with the plasma membrane increased at 2 h (P = 0.04) of the hyperinsulinemic-euglycemic clamp, and remained elevated until 6 h, with no differences between groups or infusion type. The number of GLUT4 spots was unchanged at 2 h of infusion. However, from 2 to 6 h there was a decrease in the number of small GLUT4 spots at the plasma membrane (P = 0.047), with no differences between groups or infusion type. CONCLUSION GLUT4 localisation with the plasma membrane increases during a hyperinsulinemic-euglycemic clamp, but this is not altered by elevated FFA availability. GLUT4 appears to disperse from small GLUT4 clusters located at the plasma membrane to support glucose uptake during a hyperinsulinaemic-euglycaemic clamp.
Collapse
Affiliation(s)
- J S Barrett
- Research Institute for Sport & Exercise Sciences, Liverpool John Moores University, Tom Reilly Building, Byrom Street, Liverpool, L3 3AF, UK
| | - J A Strauss
- Research Institute for Sport & Exercise Sciences, Liverpool John Moores University, Tom Reilly Building, Byrom Street, Liverpool, L3 3AF, UK
| | - L S Chow
- Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - S O Shepherd
- Research Institute for Sport & Exercise Sciences, Liverpool John Moores University, Tom Reilly Building, Byrom Street, Liverpool, L3 3AF, UK.
| | - A J M Wagenmakers
- Research Institute for Sport & Exercise Sciences, Liverpool John Moores University, Tom Reilly Building, Byrom Street, Liverpool, L3 3AF, UK
| | - Y Wang
- Discovery Sciences, AstraZeneca R&D, Cambridge Science Park, Milton Road, Cambridge, CB4 0WG, UK
| |
Collapse
|
18
|
Park JB, Moon GH, Cho A, Kwon M, Park JW, Yi EC, Kim H, Fukuda J, Kwak C, Ko YG, Chun YS. Neddylation of insulin receptor substrate acts as a bona fide regulator of insulin signaling and its implications for cancer cell migration. Cancer Gene Ther 2024; 31:599-611. [PMID: 38272982 PMCID: PMC11016467 DOI: 10.1038/s41417-024-00729-z] [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: 09/21/2023] [Revised: 01/03/2024] [Accepted: 01/09/2024] [Indexed: 01/27/2024]
Abstract
Irregularities in insulin signaling have significantly increased the risk of various cancers, yet the precise underlying mechanisms remain unclear. Within our study, we observed that inhibiting neddylation enhances cancer cell migration across different cancer types by activating both insulin receptor substrates 1 and 2 (IRS1 and IRS2), along with the PI3K/AKT signaling pathway. Notably, in the context of high-grade serous carcinoma (HGSC) patients, whether they had type 2 diabetes mellitus or not, IRS1 and IRS2 displayed a parallel relationship with each other while exhibiting an inverse relationship with NEDD8. We also identified C-CBL as an E3 ligase responsible for neddylating IRS1 and IRS2, with clinical evidence further confirming a reciprocal relationship between C-CBL and pAKT, thereby reinforcing the tumor suppressive role of C-CBL. Altogether, these findings suggest that neddylation genuinely participates in IRS1 and IRS2-dependent insulin signaling, effectively suppressing cancer cell migration. Thus, caution is advised when considering neddylation inhibitors as a treatment option for cancer patients, particularly those presenting with insulin signaling dysregulations linked to conditions like obesity-related type 2 diabetes or hyperinsulinemia.
Collapse
Affiliation(s)
- Jun Bum Park
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
- Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
| | - Geon Ho Moon
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
- Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
| | - Ara Cho
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, and Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
| | - Minji Kwon
- Department of Pathology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
| | - Jong-Wan Park
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
- Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
| | - Eugene C Yi
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, and Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
| | - Haeryoung Kim
- Department of Pathology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
| | - Junji Fukuda
- Faculty of Engineering, Yokohama National University, Yokohama, 240-8501, Japan
| | - Cheol Kwak
- Department of Urology, Seoul National University Hospital, Seoul, 03080, Republic of Korea
| | - Young-Gyu Ko
- Division of Life Sciences, Korea University, Seoul, 02841, Republic of Korea
| | - Yang-Sook Chun
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea.
- Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea.
- Department of Physiology, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea.
| |
Collapse
|
19
|
Ivovic A, Yung JHM, Oprescu AI, Vlavcheski F, Mori Y, Rahman SMN, Ye W, Eversley JA, Wheeler MB, Woo M, Tsiani E, Giacca A. β-Cell Insulin Resistance Plays a Causal Role in Fat-Induced β-Cell Dysfunction In Vitro and In Vivo. Endocrinology 2024; 165:bqae044. [PMID: 38578954 PMCID: PMC11033845 DOI: 10.1210/endocr/bqae044] [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: 03/28/2023] [Revised: 03/27/2024] [Accepted: 04/04/2024] [Indexed: 04/07/2024]
Abstract
In the classical insulin target tissues of liver, muscle, and adipose tissue, chronically elevated levels of free fatty acids (FFA) impair insulin signaling. Insulin signaling molecules are also present in β-cells where they play a role in β-cell function. Therefore, inhibition of the insulin/insulin-like growth factor 1 pathway may be involved in fat-induced β-cell dysfunction. To address the role of β-cell insulin resistance in FFA-induced β-cell dysfunction we co-infused bisperoxovanadate (BPV) with oleate or olive oil for 48 hours in rats. BPV, a tyrosine phosphatase inhibitor, acts as an insulin mimetic and is devoid of any antioxidant effect that could prevent β-cell dysfunction, unlike most insulin sensitizers. Following fat infusion, rats either underwent hyperglycemic clamps for assessment of β-cell function in vivo or islets were isolated for ex vivo assessment of glucose-stimulated insulin secretion (GSIS). We also incubated islets with oleate or palmitate and BPV for in vitro assessment of GSIS and Akt (protein kinase B) phosphorylation. Next, mice with β-cell specific deletion of PTEN (phosphatase and tensin homolog; negative regulator of insulin signaling) and littermate controls were infused with oleate for 48 hours, followed by hyperglycemic clamps or ex vivo evaluation of GSIS. In rat experiments, BPV protected against fat-induced impairment of β-cell function in vivo, ex vivo, and in vitro. In mice, β-cell specific deletion of PTEN protected against oleate-induced β-cell dysfunction in vivo and ex vivo. These data support the hypothesis that β-cell insulin resistance plays a causal role in FFA-induced β-cell dysfunction.
Collapse
Affiliation(s)
- Aleksandar Ivovic
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Justin Hou Ming Yung
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Andrei I Oprescu
- Institute of Medical Science, Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Filip Vlavcheski
- Department of Health Sciences, Brock University, St. Catharines, ON L2S 3A1, Canada
| | - Yusaku Mori
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
- Division of Diabetes, Metabolism, and Endocrinology, Anti-Glycation Research Section, Department of Medicine, Showa University School of Medicine, Shinagawa, Tokyo 142-8555, Japan
| | - S M Niazur Rahman
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Wenyue Ye
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Judith A Eversley
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Michael B Wheeler
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Minna Woo
- Institute of Medical Science, Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
- Toronto General Hospital Research Institute, University Health Network, University of Toronto, Toronto, ON M5G 2C4, Canada
- Division of Endocrinology, Department of Medicine, University Health Network, University of Toronto, Toronto, ON M5G 2C4, Canada
- Department of Medicine, Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
- Banting and Best Diabetes Centre, University of Toronto, Toronto, ON M5G 2C4, Canada
| | - Evangelia Tsiani
- Department of Health Sciences, Brock University, St. Catharines, ON L2S 3A1, Canada
| | - Adria Giacca
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
- Institute of Medical Science, Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
- Department of Medicine, Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
- Banting and Best Diabetes Centre, University of Toronto, Toronto, ON M5G 2C4, Canada
| |
Collapse
|
20
|
Song H, Hao Z, Feng H, Li R, Zhang R, Limesand SW, Zhao Y, Chen X. Insulin resistance and dyslipidemia in low-birth-weight goat kids. Front Vet Sci 2024; 11:1370640. [PMID: 38596462 PMCID: PMC11002208 DOI: 10.3389/fvets.2024.1370640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 03/06/2024] [Indexed: 04/11/2024] Open
Abstract
Low birth weight (LBW) impairs the development and health of livestock by affecting postnatal growth performance and metabolic health in adulthood. Previous studies on indigenous goats in southwest China showed that LBW goat kids had higher mortality and morbidity rates, including hepatic dyslipidemia and liver damage. However, the mechanism of insulin resistance affecting lipid metabolism under LBW conditions remains unclear. In this study, we conducted in vivo glucose-insulin metabolic studies, measured biochemical parameters, and analyzed related regulatory pathways. Both glucose tolerance tests and insulin tolerance tests indicated insulin resistance in LBW goat kids compared to controls (p < 0.05). The marker of insulin resistance, homeostasis model assessment (HOMA), was 2.85-fold higher in LBW than in control goats (p < 0.01). Additionally, elevated levels of free fatty acids in both plasma and skeletal muscle were observed in LBW goats compared to normal birth weight (NBW) goats (p < 0.05). Transcriptome analysis revealed impairments in lipid metabolism and insulin signaling in LBW goats. The observed lipid accumulation was associated with the upregulation of genes linked to fatty acid uptake and transport (FABP3), fatty acid oxidation (PPARA), triacylglycerol synthesis (LPIN1 and DGAT1), oxidative stress (ANKRD2), and insulin resistance (PGC1α). Furthermore, the insulin receptor substrate 2 (IRS2) was lower in the liver of LBW goat kids (p < 0.05). While there was no change in insulin function in skeletal muscle, LBW may lead to lipid accumulation in skeletal muscle by interfering with insulin function in the liver. These findings collectively impact the health and growth performance of livestock.
Collapse
Affiliation(s)
- Huihui Song
- College of Animal Science and Technology, Southwest University, Chongqing Key Laboratory of Herbivore Science, Chongqing, China
| | - Zhuohang Hao
- College of Animal Science and Technology, Southwest University, Chongqing Key Laboratory of Herbivore Science, Chongqing, China
| | - Hehan Feng
- College of Animal Science and Technology, Southwest University, Chongqing Key Laboratory of Herbivore Science, Chongqing, China
| | - Rui Li
- College of Animal Science and Technology, Southwest University, Chongqing Key Laboratory of Herbivore Science, Chongqing, China
| | - Ran Zhang
- Yunnan Center for Animal Disease Control and Prevention, Kunming, Yunnan, China
| | - Sean W. Limesand
- School of Animal and Comparative Biomedical Sciences, The University of Arizona, Tucson, AZ, United States
| | - Yongju Zhao
- College of Animal Science and Technology, Southwest University, Chongqing Key Laboratory of Herbivore Science, Chongqing, China
| | - Xiaochuan Chen
- College of Animal Science and Technology, Southwest University, Chongqing Key Laboratory of Herbivore Science, Chongqing, China
| |
Collapse
|
21
|
Yu Y, Yang J, Zheng L, Su H, Cao S, Jiang X, Liu X, Liu W, Wang Z, Meng F, Xu H, Wen D, Sun C, Song X, Vidal-Puig A, Cao L. Dysfunction of Akt/FoxO3a/Atg7 regulatory loop magnifies obesity-regulated muscular mass decline. Mol Metab 2024; 81:101892. [PMID: 38331318 PMCID: PMC10876605 DOI: 10.1016/j.molmet.2024.101892] [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: 01/09/2024] [Accepted: 01/30/2024] [Indexed: 02/10/2024] Open
Abstract
BACKGROUND Myoprotein degradation accelerates in obese individuals, resulting in a decline in muscular mass. Atg7 plays a crucial role in regulating protein stability and function through both autophagy-dependent and independent pathways. As obesity progresses, the expression of Atg7 gradually rises in muscle tissue. Nonetheless, the precise impact and mechanism of Atg7 in promoting muscle mass decline in obesity remain uncertain. The study aimed to elucidate the role and underly mechanism of Atg7 action in the context of obesity-induced muscle mass decline. METHODS In this study, we established a murine model of high-fat diet-induced obesity (DIO) and introduced adeno-associated virus delivery of short hairpin RNA to knock down Atg7 (shAtg7) into the gastrocnemius muscle. We then examined the expressions of Atg7 and myoprotein degradation markers in the gastrocnemius tissues of obese patients and mice using immunofluorescence and western blotting techniques. To further investigate the effects of Atg7, we assessed skeletal muscle cell diameter and the myoprotein degradation pathway in C2C12 and HSkMC cells in the presence or absence of Atg7. Immunofluorescence staining for MyHC and western blotting were utilized for this purpose. To understand the transcriptional regulation of Atg7 in response to myoprotein degradation, we conducted luciferase reporter assays and chromatin immunoprecipitation experiments to examine whether FoxO3a enhances the transcription of Atg7. Moreover, we explored the role of Akt in Atg7-mediated regulation and its relevance to obesity-induced muscle mass decline. This was accomplished by Akt knockdown, treatment with MK2206, and GST pulldown assays to assess the interaction between Atg7 and Akt. RESULTS After 20 weeks of being on a high-fat diet, obesity was induced, leading to a significant decrease in the gastrocnemius muscle area and a decline in muscle performance. This was accompanied by a notable increase in Atg7 protein expression (p < 0.01). Similarly, in gastrocnemius tissues of obese patients when compared to nonobese individuals, there was a significant increase in both Atg7 (p < 0.01) and TRIM63 (p < 0.01) levels. When palmitic acid was administered to C2C12 cells, it resulted in increased Atg7 (p < 0.01), LC3Ⅱ/Ⅰ (p < 0.01), and p62 levels (p < 0.01). Additionally, it promoted FoxO3a-mediated transcription of Atg7. The knockdown of Atg7 in the gastrocnemius partially reversed DIO-induced muscle mass decline. Furthermore, when Atg7 was knocked down in C2C12 and HSkMC cells, it mitigated palmitic acid-induced insulin resistance, increased the p-Akt/Akt ratio (p < 0.01), and reduced TRIM63 (p < 0.01). Muscular atrophy mediated by Atg7 was reversed by genetic knockdown of Akt and treatment with the p-Akt inhibitor MK2206. Palmitic acid administration increased the binding between Atg7 and Akt (p < 0.01) while weakening the binding of PDK1 (p < 0.01) and PDK2 (p < 0.01) to Akt. GST pulldown assays demonstrated that Atg7 directly interacted with the C-terminal domain of Akt. CONCLUSION The consumption of a high-fat diet, along with lipid-induced effects, led to the inhibition of Akt signaling, which, in turn, promoted FoxO3a-mediated transcription, increasing Atg7 levels in muscle cells. The excess Atg7 inhibited the phosphorylation of Akt, leading to a cyclic activation of FoxO3a and exacerbating the decline in muscle mass regulated by obesity. Consequently, Atg7 serves as a regulatory point in determining the decline in muscle mass induced by obesity.
Collapse
Affiliation(s)
- Yang Yu
- Institute of Health Sciences, China Medical University, Shenyang 110122, Liaoning, China; College of Basic Medical Science, Key Laboratory of Medical Cell Biology, Ministry of Education, Key Laboratory of Liaoning Province, China Medical University, Shenyang 110122, Liaoning, China
| | - Jing Yang
- College of Basic Medical Science, Key Laboratory of Medical Cell Biology, Ministry of Education, Key Laboratory of Liaoning Province, China Medical University, Shenyang 110122, Liaoning, China
| | - Lixia Zheng
- Institute of Health Sciences, China Medical University, Shenyang 110122, Liaoning, China; College of Basic Medical Science, Key Laboratory of Medical Cell Biology, Ministry of Education, Key Laboratory of Liaoning Province, China Medical University, Shenyang 110122, Liaoning, China
| | - Han Su
- Institute of Health Sciences, China Medical University, Shenyang 110122, Liaoning, China; College of Basic Medical Science, Key Laboratory of Medical Cell Biology, Ministry of Education, Key Laboratory of Liaoning Province, China Medical University, Shenyang 110122, Liaoning, China
| | - Sunrun Cao
- Institute of Health Sciences, China Medical University, Shenyang 110122, Liaoning, China; College of Basic Medical Science, Key Laboratory of Medical Cell Biology, Ministry of Education, Key Laboratory of Liaoning Province, China Medical University, Shenyang 110122, Liaoning, China
| | - Xuehan Jiang
- Institute of Health Sciences, China Medical University, Shenyang 110122, Liaoning, China; College of Basic Medical Science, Key Laboratory of Medical Cell Biology, Ministry of Education, Key Laboratory of Liaoning Province, China Medical University, Shenyang 110122, Liaoning, China
| | - Xiyan Liu
- Institute of Health Sciences, China Medical University, Shenyang 110122, Liaoning, China; College of Basic Medical Science, Key Laboratory of Medical Cell Biology, Ministry of Education, Key Laboratory of Liaoning Province, China Medical University, Shenyang 110122, Liaoning, China
| | - Weiwei Liu
- Institute of Health Sciences, China Medical University, Shenyang 110122, Liaoning, China
| | - Zhuo Wang
- Institute of Health Sciences, China Medical University, Shenyang 110122, Liaoning, China; College of Basic Medical Science, Key Laboratory of Medical Cell Biology, Ministry of Education, Key Laboratory of Liaoning Province, China Medical University, Shenyang 110122, Liaoning, China
| | - Fang Meng
- Institute of Health Sciences, China Medical University, Shenyang 110122, Liaoning, China; College of Basic Medical Science, Key Laboratory of Medical Cell Biology, Ministry of Education, Key Laboratory of Liaoning Province, China Medical University, Shenyang 110122, Liaoning, China
| | - Hongde Xu
- Institute of Health Sciences, China Medical University, Shenyang 110122, Liaoning, China; College of Basic Medical Science, Key Laboratory of Medical Cell Biology, Ministry of Education, Key Laboratory of Liaoning Province, China Medical University, Shenyang 110122, Liaoning, China
| | - Deliang Wen
- Institute of Health Sciences, China Medical University, Shenyang 110122, Liaoning, China
| | - Chen Sun
- Institute of Health Sciences, China Medical University, Shenyang 110122, Liaoning, China; Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning, China.
| | - Xiaoyu Song
- Institute of Health Sciences, China Medical University, Shenyang 110122, Liaoning, China; College of Basic Medical Science, Key Laboratory of Medical Cell Biology, Ministry of Education, Key Laboratory of Liaoning Province, China Medical University, Shenyang 110122, Liaoning, China.
| | - Antonio Vidal-Puig
- MRC Metabolic Diseases Unit, Wellcome Trust-Medical Research Council Institute of Metabolic Science, University of Cambridge, CB2 1TN, Cambridge, UK; Centro de Investigacion Principe Felipe, Valencia, Spain; Cambridge University Nanjing Centre of Technology and Innovation, Nanjing, China.
| | - Liu Cao
- Institute of Health Sciences, China Medical University, Shenyang 110122, Liaoning, China; College of Basic Medical Science, Key Laboratory of Medical Cell Biology, Ministry of Education, Key Laboratory of Liaoning Province, China Medical University, Shenyang 110122, Liaoning, China.
| |
Collapse
|
22
|
Basheer M, Bolus M, Basheer A, Loai A, Nimer A. Olive Oil's Attenuating Effects on Lipotoxicity. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1460:869-882. [PMID: 39287875 DOI: 10.1007/978-3-031-63657-8_29] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
Dietary fatty acids play a role in the pathogenesis of obesity-associated nonalcoholic fatty liver disease. Lipotoxicity in obesity mediates insulin resistance, endothelial dysfunction, atherosclerosis, and gut microbiota dysbiosis. Cardiovascular complications are the main cause of morbidity and mortality in obese, insulin-resistant, and type 2 diabetes mellitus patients.Interventions targeting lipotoxicity are the main issue in preventing its multiple insults. Lifestyle modifications including healthy eating and regular exercise are the primary recommendations. Treatments also include drugs targeting energy intake, energy disposal, lipotoxic liver injury, and the resulting inflammation, fibrogenesis, and cirrhosis.Diet and nutrition have been linked to insulin resistance, an increased risk of developing type 2 diabetes, and impaired postprandial lipid metabolism. Low-fat diets are associated with higher survival. The Mediterranean diet includes an abundance of olive oil. Extra-virgin olive oil is the main source of monounsaturated fatty acids in Mediterranean diets. An olive oil-rich diet decreases triglyceride accumulation in the liver, improves postprandial triglyceride levels, improves glucose and insulin secretions, and upregulates GLUT-2 expression in the liver. The exact molecular mechanisms of olive oil's effects are unknown, but decreasing NF-kB activation, decreasing LDL oxidation, and improving insulin resistance by reducing the production of inflammatory cytokines (TNF-α and IL-6) and upregulating kinases and JNK-mediated phosphorylation of IRS-1 are possible principal mechanisms. Olive oil phenolic compounds also modulate gut microbiota diversity, which also affects lipotoxicity.In this review, we document lipotoxicity in obesity manifestations and the beneficial health effects of the Mediterranean diet derived from monounsaturated fatty acids, mainly from olive oil.
Collapse
Affiliation(s)
- Maamoun Basheer
- Department of Internal Medicine A, Galilee Medical Center, Nahariya, Israel
| | - Mariana Bolus
- Department of Internal Medicine A, Galilee Medical Center, Nahariya, Israel
| | - Areej Basheer
- Department of Internal Medicine A, Galilee Medical Center, Nahariya, Israel
- Nutrition and Diet Services, Hillel Yaffe, Hadera, Israel
| | - Arraf Loai
- Department of Internal Medicine A, Galilee Medical Center, Nahariya, Israel
| | - Assy Nimer
- Department of Internal Medicine A, Galilee Medical Center, Nahariya, Israel.
- Faculty of Medicine at Galilee, Bar-Ilan University, Safed, Israel.
| |
Collapse
|
23
|
Swain J, Jadhao P, Sravya SL, Teli B, Lavanya K, Singh J, Sahoo A, Das S. Mitochondrial Dysfunction and Imeglimin: A New Ray of Hope for the Treatment of Type-2 Diabetes Mellitus. Mini Rev Med Chem 2024; 24:1575-1589. [PMID: 37861052 DOI: 10.2174/0113895575260225230921062013] [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: 05/01/2023] [Revised: 07/03/2023] [Accepted: 07/26/2023] [Indexed: 10/21/2023]
Abstract
Diabetes is a rapidly growing health challenge and epidemic in many developing countries, including India. India, being the diabetes capital of the world, has the dubious dual distinction of being the leading nations for both undernutrition and overnutrition. Diabetes prevalence has increased in both rural and urban areas, affected the younger population and increased the risk of complications and economic burden. These alarming statistics ring an alarm bell to achieve glycemic targets in the affected population in order to decrease diabetes-related morbidity and mortality. In the recent years, diabetes pathophysiology has been extended from an ominous triad through octet and dirty dozen etc. There is a new scope to target multiple pathways at the molecular level to achieve a better glycemic target and further prevent micro- and macrovascular complications. Mitochondrial dysfunction has a pivotal role in both β-cell failure and insulin resistance. Hence, targeting this molecular pathway may help with both insulin secretion and peripheral tissue sensitization to insulin. Imeglimin is the latest addition to our anti-diabetic armamentarium. As imeglimin targets, this root cause of defective energy metabolism and insulin resistance makes it a new add-on therapy in different diabetic regimes to achieve the proper glycemic targets. Its good tolerability and efficacy profiles in recent studies shows a new ray of hope in the journey to curtail diabetes-related morbidity.
Collapse
Affiliation(s)
- Jayshree Swain
- Department of Endocrinology, Institute of Medical Sciences and SUM Hospital, Siksha 'O' Anusandhan Deemed to be University, Bhubaneswar, India
| | - Pooja Jadhao
- Department of Endocrinology, Institute of Medical Sciences and SUM Hospital, Siksha 'O' Anusandhan Deemed to be University, Bhubaneswar, India
| | - S L Sravya
- Department of Endocrinology, Institute of Medical Sciences and SUM Hospital, Siksha 'O' Anusandhan Deemed to be University, Bhubaneswar, India
| | - Brij Teli
- Department of Endocrinology, Institute of Medical Sciences and SUM Hospital, Siksha 'O' Anusandhan Deemed to be University, Bhubaneswar, India
| | - Kasukurti Lavanya
- Department of Endocrinology, Institute of Medical Sciences and SUM Hospital, Siksha 'O' Anusandhan Deemed to be University, Bhubaneswar, India
| | - Jaspreet Singh
- Department of Endocrinology, Institute of Medical Sciences and SUM Hospital, Siksha 'O' Anusandhan Deemed to be University, Bhubaneswar, India
| | - Abhay Sahoo
- Department of Endocrinology, Institute of Medical Sciences and SUM Hospital, Siksha 'O' Anusandhan Deemed to be University, Bhubaneswar, India
| | - Srijit Das
- Department of Human & Clinical Anatomy, College of Medicine & Health Sciences, Sultan Qaboos University, Muscat 123, Sultanate of Oman
| |
Collapse
|
24
|
Bagherzadeh-Rahmani B, Marzetti E, Karami E, Campbell BI, Fakourian A, Haghighi AH, Mousavi SH, Heinrich KM, Brazzi L, Jung F, Baker JS, Patel DI. Tirzepatide and exercise training in obesity. Clin Hemorheol Microcirc 2024; 87:465-480. [PMID: 38640145 DOI: 10.3233/ch-242134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/21/2024]
Abstract
OBJECTIVES The purpose of this study was to investigate the effects of 6 weeks of resistance training (RT) combined with aerobic training (AT) and Tirzepatide supplementation on lipid profiles, insulin resistance, anthropometric characteristics and physical fitness in prediabetic obese soldiers. METHODS 61 obese men were randomly divided into six groups: Placebo; Tirzepatide 5 mg (T5); Tirzepatide 2.5 mg (T2.5); Hypertrophy, Strength, Power-Circuit Training+Placebo (Ex+P); Hypertrophy, Strength, Power-Circuit Training+Tirzepatide 5 mg (Ex+T5); Hypertrophy, Strength, Power-Circuit Training+Tirzepatide 2.5 mg (Ex+T2.5). All training groups performed aerobic training (AT) after resistance training. Subjects trained for six weeks, three sessions per week. Before and after the intervention period, the participants were evaluated for anthropometric measures, body composition [body weight, body mass index (BMI), waist circumference (WC), waist to hip ratio (WHR) and fat mass (FM)], cardiorespiratory fitness (VO2max), and muscle strength (chest press 1RM and leg press 1RM). Blood biochemistry evaluations included triglyceride (TG), total cholesterol (TC), low density lipoprotein cholesterol (LDL-C), high-density lipoprotein cholesterol (HDL-C), fasting blood glucose (FBG), insulin level and insulin resistance (HOMA-IR). To evaluate the differences between the groups, ANCOVA statistical method was used along with Bonferroni's post hoc test, and the significance level was P < 0.05. RESULTS Body weight, BMI, WC, FM, FBG, LDL-C, TC, TG and HOMA-IR were significantly decreased in Ex+P, Ex+T5 and Ex+T2.5 groups compared to Placebo, T5 and T2.5 groups. WHR significantly decreased in Ex+P, Ex+T5 and Ex+T2.5 groups compared to Placebo group. HDL-C, chest press and leg press significantly increased in Ex+P, Ex+T5 and Ex+T2.5 groups compared to Placebo, T5 and T2.5 groups. VO2max significantly increased and insulin significantly decreased in Ex+P group compared to Placebo, T5 and T2.5 groups. FM, FBG and TG were significantly decreased in both the T2.5 and T5 groups compared to Placebo group. HOMA-IR, LDL-C and TC significantly decreased in the T5 group compared to Placebo group. Also, leg press significantly increased in Ex+P group compared to all other groups. CONCLUSIONS Performing six weeks of combined resistance and aerobic training in the form of RT+AT alone is more effective than the simultaneous use of Tirzepatide on cardiorespiratory fitness, strength, and modulating insulin levels. Taking Tirzepatide in doses of 5 mg and 2.5 mg in combination with exercise training did not have a significant advantage over exercise training alone. Finally, taking Tirzepatide in doses of 5 mg or 2.5 mg in combination with exercise training is not significantly superior to each other.
Collapse
Affiliation(s)
- Behnam Bagherzadeh-Rahmani
- Department of Exercise Physiology, Faculty of Sport Sciences, Hakim Sabzevari University, Sabzevar, Iran
| | - Emanuele Marzetti
- Department of Geriatrics and Orthopedics, UniversitÀ Cattolica del Sacro Cuore, Rome, Italy
- Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Rome, Italy
| | - Esmail Karami
- Department of Physiology, Faculty of Medicine, AJA University of Medical Sciences, Tehran, Iran
| | - Bill I Campbell
- Performance & Physique Enhancement Laboratory, University of South Florida, Tampa, FL, USA
| | - Ali Fakourian
- Department of Exercise Physiology, Central Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Amir Hossein Haghighi
- Department of Exercise Physiology, Faculty of Sport Sciences, Hakim Sabzevari University, Sabzevar, Iran
| | - Seyyed Hossein Mousavi
- Department of Cardiology, School of Medicine, AJA University of Medical Sciences, Tehran, Iran
| | - Katie M Heinrich
- Department of Kinesiology, Kansas State University, Manhattan, KS, USA
| | - Luca Brazzi
- Department of Surgical Sciences, University of Turin, Turin, Italy
| | - Friedrich Jung
- Institute of Biotechnology, Molecular Cell Biology, Brandenburg University of Technology Cottbus-Senftenberg, Senftenberg, Germany
| | - Julien S Baker
- Department of Sport, Physical Education and Health, Hong Kong Baptist University, Kowloon, Hong Kong
| | - Darpan I Patel
- School of Nursing, University of Texas Medical Branch, Galveston, TX, USA
| |
Collapse
|
25
|
Liu J, Liu T, Ren S(V, Zhu C, Bouso E, Mamlouk S, Rau CD, Wang Y, Gao C. Metabolic status differentiates Trp53inp2 function in pressure-overload induced heart failure. Front Cardiovasc Med 2023; 10:1226586. [PMID: 38188257 PMCID: PMC10766701 DOI: 10.3389/fcvm.2023.1226586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 11/30/2023] [Indexed: 01/09/2024] Open
Abstract
Cardiometabolic disorders encompass a broad range of cardiovascular complications associated with metabolic dysfunction. These conditions have an increasing share in the health burden worldwide due to worsening endemic of hypertension, obesity, and diabetes. Previous studies have identified Tumor Protein p53-inducible Nuclear Protein 2 (Trp53inp2) as a molecular link between hyperglycemia and cardiac hypertrophy. However, its role in cardiac pathology has never been determined in vivo. In this study, we generated a cardiac specific knockout model of Trp53inp2 (Trp53inp2-cKO) and investigated the impact of Trp53inp2 inactivation on the pathogenesis of heart failure under mechanic or/and metabolic stresses. Based on echocardiography assessment, inactivation of Trp53inp2 in heart led to accelerated onset of HFrEF in response to pressure-overload, with significantly reduced ejection fraction and elevated heart failure marker genes comparing to the control mice. In contrast, inactivation of Trp53inp2 ameliorated cardiac dysfunction induced by combined stresses of high fat diet and moderate pressure overload (Cardiometabolic Disorder Model). Moreover, Trp53inp2 inactivation led to reduced expression of glucose metabolism genes in lean, pressure-overloaded hearts. However, the same set of genes were significantly induced in the Trp53inp2-cKO hearts under both mechanical and metabolic stresses. In summary, we have demonstrated for the first time that cardiomyocyte Trp53inp2 has diametrically differential roles in the pathogenesis of heart failure and glucose regulation under mechanical vs. mechanical plus metabolic stresses. This insight suggests that Trp53inp2 may exacerbate the cardiac dysfunction during pressure overload injury but have a protective effect in cardiac diastolic function in cardiometabolic disease.
Collapse
Affiliation(s)
- Jianfang Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Tian Liu
- Department of Pharmacology and Systems Physiology, University of Cincinnati, Cincinnati, OH, United States
| | - Shuxun (Vincent) Ren
- Signature Research Program in Cardiovascular and Metabolic Diseases, DukeNUS Medical School, Singapore, Singapore
| | - Cansheng Zhu
- Department of Pharmacology and Systems Physiology, University of Cincinnati, Cincinnati, OH, United States
| | - Eyad Bouso
- Department of Pharmacology and Systems Physiology, University of Cincinnati, Cincinnati, OH, United States
| | - Samir Mamlouk
- Department of Pharmacology and Systems Physiology, University of Cincinnati, Cincinnati, OH, United States
| | - Christoph D. Rau
- Department of Genetics, School of Medicine, University of North Carolina, Chapel Hill, NC, United States
| | - Yibin Wang
- Signature Research Program in Cardiovascular and Metabolic Diseases, DukeNUS Medical School, Singapore, Singapore
| | - Chen Gao
- Department of Pharmacology and Systems Physiology, University of Cincinnati, Cincinnati, OH, United States
| |
Collapse
|
26
|
Martemucci G, Fracchiolla G, Muraglia M, Tardugno R, Dibenedetto RS, D’Alessandro AG. Metabolic Syndrome: A Narrative Review from the Oxidative Stress to the Management of Related Diseases. Antioxidants (Basel) 2023; 12:2091. [PMID: 38136211 PMCID: PMC10740837 DOI: 10.3390/antiox12122091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 11/15/2023] [Accepted: 11/27/2023] [Indexed: 12/24/2023] Open
Abstract
Metabolic syndrome (MS) is a growing disorder affecting thousands of people worldwide, especially in industrialised countries, increasing mortality. Oxidative stress, hyperglycaemia, insulin resistance, inflammation, dysbiosis, abdominal obesity, atherogenic dyslipidaemia and hypertension are important factors linked to MS clusters of different pathologies, such as diabesity, cardiovascular diseases and neurological disorders. All biochemical changes observed in MS, such as dysregulation in the glucose and lipid metabolism, immune response, endothelial cell function and intestinal microbiota, promote pathological bridges between metabolic syndrome, diabesity and cardiovascular and neurodegenerative disorders. This review aims to summarise metabolic syndrome's involvement in diabesity and highlight the link between MS and cardiovascular and neurological diseases. A better understanding of MS could promote a novel strategic approach to reduce MS comorbidities.
Collapse
Affiliation(s)
- Giovanni Martemucci
- Department of Agricultural and Environmental Sciences, University of Bari Aldo Moro, 70126 Bari, Italy;
| | - Giuseppe Fracchiolla
- Department of Pharmacy–Drug Sciences, University of Bari Aldo Moro, 70126 Bari, Italy; (M.M.); (R.T.); (R.S.D.)
| | - Marilena Muraglia
- Department of Pharmacy–Drug Sciences, University of Bari Aldo Moro, 70126 Bari, Italy; (M.M.); (R.T.); (R.S.D.)
| | - Roberta Tardugno
- Department of Pharmacy–Drug Sciences, University of Bari Aldo Moro, 70126 Bari, Italy; (M.M.); (R.T.); (R.S.D.)
| | - Roberta Savina Dibenedetto
- Department of Pharmacy–Drug Sciences, University of Bari Aldo Moro, 70126 Bari, Italy; (M.M.); (R.T.); (R.S.D.)
| | | |
Collapse
|
27
|
Busebee B, Ghusn W, Cifuentes L, Acosta A. Obesity: A Review of Pathophysiology and Classification. Mayo Clin Proc 2023; 98:1842-1857. [PMID: 37831039 PMCID: PMC10843116 DOI: 10.1016/j.mayocp.2023.05.026] [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: 12/15/2022] [Revised: 05/18/2023] [Accepted: 05/30/2023] [Indexed: 10/14/2023]
Abstract
Obesity is a chronic, multifactorial, and morbid disease. In the United States, 69% of adults are overweight or have obesity, and the global prevalence of obesity is increasing. Obesity is influenced by genetic, neurologic, metabolic, enteric, and behavioral processes. It remains a key modifiable risk factor for many comorbid diseases, including cardiovascular disease, diabetes mellitus, and cancer. Whereas there are recent and significant advances in obesity therapy, including diets, lifestyle modifications, pharmacotherapies, endoscopic procedures, and bariatric surgeries, there is an immense need for a better understanding of the heterogeneity in the pathophysiologic process of obesity and outcomes. Here we review salient pathophysiologic mechanisms underlying the development and morbidity of obesity as well as pathophysiologically based classification systems that inform current obesity management and may inform improved and individualized management in the future.
Collapse
Affiliation(s)
| | - Wissam Ghusn
- Precision Medicine for Obesity Program, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN
| | - Lizeth Cifuentes
- Precision Medicine for Obesity Program, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN
| | - Andres Acosta
- Precision Medicine for Obesity Program, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN.
| |
Collapse
|
28
|
Sahoo OS, Mitra R, Bhattacharjee A, Kar S, Mukherjee O. Is Diabetes Mellitus a Predisposing Factor for Helicobacter pylori Infections? Curr Diab Rep 2023; 23:195-205. [PMID: 37213058 DOI: 10.1007/s11892-023-01511-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/23/2023] [Indexed: 05/23/2023]
Abstract
PURPOSE OF REVIEW This review aims to analyse the consistency of reports suggesting the role of Diabetes Mellitus in the pathogenesis of Helicobacter pylori (H. pylori). RECENT FINDINGS There have been numerous controversies citing the prevalence of H. pylori infections in patients suffering from type 2 diabetes mellitus (T2DM). This review investigates the possible crosstalk between H. pylori infections and T2DM and also designs a meta-analysis to quantify the association. Subgroup analyses have also been conducted to deduce factors like geography and testing techniques, in playing a role in stratification analysis. Based on a scientific literature survey and meta-analysis of databases from 1996 to 2022, a trend towards more frequent H. pylori infections in patients with diabetes mellitus was observed. The highly diversified nature of H. pylori infections across age, gender, and geographical regions requires large interventional studies to evaluate its long-term association with diabetes mellitus. Further possible linkage of the prevalence of diabetes mellitus concomitant with that of H. pylori infected patients has also been delineated in the review.
Collapse
Affiliation(s)
- Om Saswat Sahoo
- Department of Biotechnology, National Institute of Technology Durgapur, Durgapur, West Bengal, India, 713209
| | - Rhiti Mitra
- Department of Biotechnology, National Institute of Technology Durgapur, Durgapur, West Bengal, India, 713209
| | - Arghyadeep Bhattacharjee
- Department of Biotechnology, National Institute of Technology Durgapur, Durgapur, West Bengal, India, 713209
- Department of Microbiology, Kingston College of Science, Beruanpukuria, Barasat, West Bengal, India, 700129
| | - Samarjit Kar
- Department of Mathematics, National Institute of Technology Durgapur, Durgapur, West Bengal, India, 713209
| | - Oindrilla Mukherjee
- Department of Biotechnology, National Institute of Technology Durgapur, Durgapur, West Bengal, India, 713209.
| |
Collapse
|
29
|
Zarini S, Zemski Berry KA, Kahn DE, Garfield A, Perreault L, Kerege A, Bergman BC. Deoxysphingolipids: Atypical Skeletal Muscle Lipids Related to Insulin Resistance in Humans That Decrease Insulin Sensitivity In Vitro. Diabetes 2023; 72:884-897. [PMID: 37186949 PMCID: PMC10281238 DOI: 10.2337/db22-1018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 04/19/2023] [Indexed: 05/17/2023]
Abstract
Sphingolipids are thought to promote skeletal muscle insulin resistance. Deoxysphingolipids (dSLs) are atypical sphingolipids that are increased in the plasma of individuals with type 2 diabetes and cause β-cell dysfunction in vitro. However, their role in human skeletal muscle is unknown. We found that dSL species are significantly elevated in muscle of individuals with obesity and type 2 diabetes compared with athletes and lean individuals and are inversely related to insulin sensitivity. Furthermore, we observed a significant reduction in muscle dSL content in individuals with obesity who completed a combined weight loss and exercise intervention. Increased dSL content in primary human myotubes caused a decrease in insulin sensitivity associated with increased inflammation, decreased AMPK phosphorylation, and altered insulin signaling. Our findings reveal a central role for dSL in human muscle insulin resistance and suggest dSLs as therapeutic targets for the treatment and prevention of type 2 diabetes. ARTICLE HIGHLIGHTS Deoxysphingolipids (dSLs) are atypical sphingolipids elevated in the plasma of individuals with type 2 diabetes, and their role in muscle insulin resistance has not been investigated. We evaluated dSL in vivo in skeletal muscle from cross-sectional and longitudinal insulin-sensitizing intervention studies and in vitro in myotubes manipulated to synthesize higher dSLs. dSLs were increased in the muscle of people with insulin resistance, inversely correlated to insulin sensitivity, and significantly decreased after an insulin-sensitizing intervention; increased intracellular dSL concentrations cause myotubes to become more insulin resistant. Reduction of muscle dSL levels is a potential novel therapeutic target to prevent/treat skeletal muscle insulin resistance.
Collapse
Affiliation(s)
- Simona Zarini
- Division of Endocrinology, Metabolism, and Diabetes, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Karin A. Zemski Berry
- Division of Endocrinology, Metabolism, and Diabetes, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Darcy E. Kahn
- Division of Endocrinology, Metabolism, and Diabetes, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Amanda Garfield
- Division of Endocrinology, Metabolism, and Diabetes, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Leigh Perreault
- Division of Endocrinology, Metabolism, and Diabetes, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Anna Kerege
- Division of Endocrinology, Metabolism, and Diabetes, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Bryan C. Bergman
- Division of Endocrinology, Metabolism, and Diabetes, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO
| |
Collapse
|
30
|
Cavaliere G, Cimmino F, Trinchese G, Catapano A, Petrella L, D'Angelo M, Lucchin L, Mollica MP. From Obesity-Induced Low-Grade Inflammation to Lipotoxicity and Mitochondrial Dysfunction: Altered Multi-Crosstalk between Adipose Tissue and Metabolically Active Organs. Antioxidants (Basel) 2023; 12:1172. [PMID: 37371902 DOI: 10.3390/antiox12061172] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 05/23/2023] [Accepted: 05/27/2023] [Indexed: 06/29/2023] Open
Abstract
Obesity is a major risk factor for several metabolic diseases, including type 2 diabetes, hyperlipidemia, cardiovascular diseases, and brain disorders. Growing evidence suggests the importance of inter-organ metabolic communication for the progression of obesity and the subsequent onset of related disorders. This review provides a broad overview of the pathophysiological processes that from adipose tissue dysfunction leading to altered multi-tissue crosstalk relevant to regulating energy homeostasis and the etiology of obesity. First, a comprehensive description of the role of adipose tissue was reported. Then, attention was turned toward the unhealthy expansion of adipose tissue, low-grade inflammatory state, metabolic inflexibility, and mitochondrial dysfunction as root causes of systemic metabolic alterations. In addition, a short spot was devoted to iron deficiency in obese conditions and the role of the hepcidin-ferroportin relationship in the management of this issue. Finally, different classes of bioactive food components were described with a perspective to enhance their potential preventive and therapeutic use against obesity-related diseases.
Collapse
Affiliation(s)
- Gina Cavaliere
- Department of Pharmaceutical Sciences, University of Perugia, 06126 Perugia, Italy
- Centro Servizi Metrologici e Tecnologici Avanzati (CeSMA), Complesso Universitario di Monte Sant'Angelo, 80126 Naples, Italy
| | - Fabiano Cimmino
- Centro Servizi Metrologici e Tecnologici Avanzati (CeSMA), Complesso Universitario di Monte Sant'Angelo, 80126 Naples, Italy
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy
| | - Giovanna Trinchese
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy
| | - Angela Catapano
- Centro Servizi Metrologici e Tecnologici Avanzati (CeSMA), Complesso Universitario di Monte Sant'Angelo, 80126 Naples, Italy
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy
| | - Lidia Petrella
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy
| | - Margherita D'Angelo
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy
| | - Lucio Lucchin
- Dietetics and Clinical Nutrition, Bolzano Health District, 39100 Bolzano, Italy
| | - Maria Pina Mollica
- Centro Servizi Metrologici e Tecnologici Avanzati (CeSMA), Complesso Universitario di Monte Sant'Angelo, 80126 Naples, Italy
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy
- Task Force on Microbiome Studies, University of Naples Federico II, 80138 Naples, Italy
| |
Collapse
|
31
|
Moellering DR, Smith-Johnston K, Kelley C, Sammy MJ, Benedict J, Brock G, Johnson J, Baskin KK, Jarjour WN, Belury MA, Reiser PJ, Nagareddy PR, Hanaoka BY. Association between skeletal muscle mitochondrial dysfunction and insulin resistance in patients with rheumatoid arthritis: a case-control study. Arthritis Res Ther 2023; 25:85. [PMID: 37210569 PMCID: PMC10199606 DOI: 10.1186/s13075-023-03065-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 05/09/2023] [Indexed: 05/22/2023] Open
Abstract
BACKGROUND Insulin resistance affects a substantial proportion of patients with rheumatoid arthritis (RA). Skeletal muscle mitochondrial dysfunction results in the accumulation of lipid intermediates that interfere with insulin signaling. We therefore sought to determine if lower oxidative phosphorylation and muscle mitochondrial content are associated with insulin resistance in patients with RA. METHODS This was a cross-sectional prospective study of RA patients. Matsuda index from the glucose tolerance test was used to estimate insulin sensitivity. Mitochondrial content was measured by citrate synthase (CS) activity in snap-frozen muscle samples. Mitochondrial function was measured by using high-resolution respirometry of permeabilized muscle fibers and electron transport chain complex IV enzyme kinetics in isolated mitochondrial subpopulations. RESULTS RA participants demonstrated lower insulin sensitivity as measured by the Matsuda index compared to controls [median 3.95 IQR (2.33, 5.64) vs. 7.17 (5.83, 7.75), p = 0.02]. There was lower muscle mitochondrial content among RA vs. controls [median 60 mU/mg IQR (45, 80) vs. 79 mU/mg (65, 97), p = 0.03]. Notably, OxPhos normalized to mitochondrial content was higher among RA vs. controls [mean difference (95% CI) = 0.14 (0.02, 0.26), p = 0.03], indicating a possible compensatory mechanism for lower mitochondrial content or lipid overload. Among RA participants, the activity of muscle CS activity was not correlated with the Matsuda index (ρ = - 0.05, p = 0.84), but it was positively correlated with self-reported (IPAQ) total MET-minutes/week (ρ = 0.44, p = 0.03) and Actigraph-measured time on physical activity (MET rate) (ρ = 0.47, p = 0.03). CONCLUSIONS Mitochondrial content and function were not associated with insulin sensitivity among participants with RA. However, our study demonstrates a significant association between muscle mitochondrial content and physical activity level, highlighting the potential for future exercise interventions that enhance mitochondrial efficiency in RA patients.
Collapse
Affiliation(s)
- Douglas R Moellering
- Department of Nutrition Sciences, School of Health Professions, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Kelley Smith-Johnston
- Department of Nutrition Sciences, School of Health Professions, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Christian Kelley
- Center for Exercise Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Melissa J Sammy
- Department of Nutrition Sciences, School of Health Professions, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Jason Benedict
- Department of Biomedical Bioinformatics, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Guy Brock
- Department of Biomedical Bioinformatics, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Jillian Johnson
- Department of Surgery, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Kedryn K Baskin
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Wael N Jarjour
- Division of Rheumatology and Immunology, Department of Internal Medicine, The Ohio State University, Columbus, OH, USA
| | - Martha A Belury
- Department of Human Sciences, College of Education and Human Ecology, The Ohio State University, Columbus, OH, USA
| | - Peter J Reiser
- Division of Biosciences, College of Dentistry, The Ohio State University, Columbus, OH, USA
| | - Prabhakara R Nagareddy
- Department of Surgery, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Beatriz Y Hanaoka
- Division of Rheumatology and Immunology, Department of Internal Medicine, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
32
|
Schleh MW, Ryan BJ, Ahn C, Ludzki AC, Varshney P, Gillen JB, Van Pelt DW, Pitchford LM, Howton SM, Rode T, Chenevert TL, Hummel SL, Burant CF, Horowitz JF. Metabolic dysfunction in obesity is related to impaired suppression of fatty acid release from adipose tissue by insulin. Obesity (Silver Spring) 2023; 31:1347-1361. [PMID: 36988872 PMCID: PMC10192005 DOI: 10.1002/oby.23734] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Revised: 12/02/2022] [Accepted: 01/01/2023] [Indexed: 03/30/2023]
Abstract
OBJECTIVE The aims of this study were: 1) to assess relationships among insulin-mediated glucose uptake with standard clinical outcomes and deep-phenotyping measures (including fatty acid [FA] rate of appearance [FA Ra] into the systemic circulation); and 2) to examine the contribution of adipocyte size, fibrosis, and proteomic profile to FA Ra regulation. METHODS A total of 66 adults with obesity (BMI = 34 [SD 3] kg/m2 ) were assessed for insulin sensitivity (hyperinsulinemic-euglycemic clamp), and stable isotope dilution methods quantified glucose, FA, and glycerol kinetics in vivo. Abdominal subcutaneous adipose tissue (aSAT) and skeletal muscle biopsies were collected, and magnetic resonance imaging quantified liver and visceral fat content. RESULTS Insulin-mediated FA Ra suppression associated with insulin-mediated glucose uptake (r = 0.51; p < 0.01) and negatively correlated with liver (r = -0.36; p < 0.01) and visceral fat (r = -0.42; p < 0.01). aSAT proteomics from subcohorts of participants with low FA Ra suppression (n = 8) versus high FA Ra suppression (n = 8) demonstrated greater extracellular matrix collagen protein in low versus high FA Ra suppression. Skeletal muscle lipidomics (n = 18) revealed inverse correlations of FA Ra suppression with acyl-chain length of acylcarnitine (r = -0.42; p = 0.02) and triacylglycerol (r = -0.51; p < 0.01), in addition to insulin-mediated glucose uptake (acylcarnitine: r = -0.49; p < 0.01, triacylglycerol: r = -0.40; p < 0.01). CONCLUSIONS Insulin's ability to suppress FA release from aSAT in obesity is related to enhanced insulin-mediated glucose uptake and metabolic health in peripheral tissues.
Collapse
Affiliation(s)
- Michael W Schleh
- Substrate Metabolism Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Benjamin J Ryan
- Substrate Metabolism Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Cheehoon Ahn
- Substrate Metabolism Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Alison C Ludzki
- Substrate Metabolism Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Pallavi Varshney
- Substrate Metabolism Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Jenna B Gillen
- Substrate Metabolism Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, Michigan, USA
- Faculty of Kinesiology and Physical Education, University of Toronto, Toronto, Ontario, Canada
| | - Douglas W Van Pelt
- Substrate Metabolism Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Lisa M Pitchford
- Substrate Metabolism Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Suzette M Howton
- Substrate Metabolism Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Thomas Rode
- Substrate Metabolism Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Thomas L Chenevert
- Department of Radiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Scott L Hummel
- Division of Cardiology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
- Veterans Affairs Health System, Ann Arbor, Michigan, USA
| | - Charles F Burant
- Division of Metabolism, Endocrinology, and Diabetes, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Jeffrey F Horowitz
- Substrate Metabolism Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
33
|
Grapentine S, Singh RK, Bakovic M. Skeletal Muscle Consequences of Phosphatidylethanolamine Synthesis Deficiency. FUNCTION 2023; 4:zqad020. [PMID: 37342414 PMCID: PMC10278983 DOI: 10.1093/function/zqad020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 04/24/2023] [Accepted: 04/25/2023] [Indexed: 06/22/2023] Open
Abstract
The maintenance of phospholipid homeostasis is increasingly being implicated in metabolic health. Phosphatidylethanolamine (PE) is the most abundant phospholipid on the inner leaflet of cellular membranes, and we have previously shown that mice with a heterozygous ablation of the PE synthesizing enzyme, Pcyt2 (Pcyt2+/-), develop obesity, insulin resistance, and NASH. Skeletal muscle is a major determinant of systemic energy metabolism, making it a key player in metabolic disease development. Both the total PE levels and the ratio of PE to other membrane lipids in skeletal muscle are implicated in insulin resistance; however, the underlying mechanisms and the role of Pcyt2 regulation in this association remain unclear. Here, we show how reduced phospholipid synthesis due to Pcyt2 deficiency causes Pcyt2+/- skeletal muscle dysfunction and metabolic abnormalities. Pcyt2+/- skeletal muscle exhibits damage and degeneration, with skeletal muscle cell vacuolization, disordered sarcomeres, mitochondria ultrastructure irregularities and paucity, inflammation, and fibrosis. There is intramuscular adipose tissue accumulation, and major disturbances in lipid metabolism with impaired FA mobilization and oxidation, elevated lipogenesis, and long-chain fatty acyl-CoA, diacylglycerol, and triacylglycerol accumulation. Pcyt2+/- skeletal muscle exhibits perturbed glucose metabolism with elevated glycogen content, impaired insulin signaling, and reduced glucose uptake. Together, this study lends insight into the critical role of PE homeostasis in skeletal muscle metabolism and health with broad implications on metabolic disease development.
Collapse
Affiliation(s)
- Sophie Grapentine
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph N1G 2W1, Canada
| | - Rathnesh K Singh
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph N1G 2W1, Canada
| | - Marica Bakovic
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph N1G 2W1, Canada
| |
Collapse
|
34
|
Adipose tissue macrophages and their role in obesity-associated insulin resistance: an overview of the complex dynamics at play. Biosci Rep 2023; 43:232519. [PMID: 36718668 PMCID: PMC10011338 DOI: 10.1042/bsr20220200] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 01/18/2023] [Accepted: 01/26/2023] [Indexed: 02/01/2023] Open
Abstract
Obesity, a major global health concern, is characterized by serious imbalance between energy intake and expenditure leading to excess accumulation of fat in adipose tissue (AT). A state of chronic low-grade AT inflammation is prevalent during obesity. The adipose tissue macrophages (ATM) with astounding heterogeneity and complex regulation play a decisive role in mediating obesity-induced insulin resistance. Adipose-derived macrophages were broadly classified as proinflammatory M1 and anti-inflammatory M2 subtypes but recent reports have proclaimed several novel and intermediate profiles, which are crucial in understanding the dynamics of macrophage phenotypes during development of obesity. Lipid-laden hypertrophic adipocytes release various chemotactic signals that aggravate macrophage infiltration into AT skewing toward mostly proinflammatory status. The ratio of M1-like to M2-like macrophages is increased substantially resulting in copious secretion of proinflammatory mediators such as TNFα, IL-6, IL-1β, MCP-1, fetuin-A (FetA), etc. further worsening insulin resistance. Several AT-derived factors could influence ATM content and activation. Apart from being detrimental, ATM exerts beneficial effects during obesity. Recent studies have highlighted the prime role of AT-resident macrophage subpopulations in not only effective clearance of excess fat and dying adipocytes but also in controlling vascular integrity, adipocyte secretions, and fibrosis within obese AT. The role of ATM subpopulations as friend or foe is determined by an intricate interplay of such factors arising within hyperlipidemic microenvironment of obese AT. The present review article highlights some of the key research advances in ATM function and regulation, and appreciates the complex dynamics of ATM in the pathophysiologic scenario of obesity-associated insulin resistance.
Collapse
|
35
|
Kyrgiafini MA, Sarafidou T, Giannoulis T, Chatziparasidou A, Christoforidis N, Mamuris Z. Gene-by-Sex Interactions: Genome-Wide Association Study Reveals Five SNPs Associated with Obesity and Overweight in a Male Population. Genes (Basel) 2023; 14:genes14040799. [PMID: 37107557 PMCID: PMC10137758 DOI: 10.3390/genes14040799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Revised: 03/23/2023] [Accepted: 03/24/2023] [Indexed: 03/29/2023] Open
Abstract
Obesity is a chronic health problem associated with severe complications and with an increasing prevalence in the Western world. Body-fat composition and distribution are closely associated with obesity, but the human body’s composition is a sexually dimorphic trait, as differences between the two sexes are evident even from fetal life. The effect of sex hormones contributes to this phenomenon. However, studies investigating gene-by-sex interactions for obesity are limited. Therefore, the aim of the present study was to identify single-nucleotide polymorphisms (SNPs) associated with obesity and overweight in a male population. A genome-wide association study (GWAS) that included 104 control, 125 overweight, and 61 obese subjects revealed four SNPs associated with overweight (rs7818910, rs7863750, rs1554116, and rs7500401) and one SNP (rs114252547) associated with obesity in males. An in silico functional annotation was subsequently used to further investigate their role. Most of the SNPs were found in genes regulating energy metabolism and homeostasis, and some of them were expression quantitative trait loci (eQTL). These findings contribute to the understanding of the molecular mechanisms underlying obesity-related traits, especially in males, and pave the road for future research toward the improvement of the diagnosis and therapy of obese individuals.
Collapse
Affiliation(s)
- Maria-Anna Kyrgiafini
- Laboratory of Genetics, Comparative and Evolutionary Biology, Department of Biochemistry and Biotechnology, University of Thessaly, Viopolis, Mezourlo, 41500 Larissa, Greece
| | - Theologia Sarafidou
- Laboratory of Genetics, Comparative and Evolutionary Biology, Department of Biochemistry and Biotechnology, University of Thessaly, Viopolis, Mezourlo, 41500 Larissa, Greece
| | - Themistoklis Giannoulis
- Laboratory of Biology, Genetics and Bioinformatics, Department of Animal Sciences, University of Thessaly, Gaiopolis, 41336 Larissa, Greece
| | - Alexia Chatziparasidou
- Embryolab IVF Unit, St. 173-175 Ethnikis Antistaseos, Kalamaria, 55134 Thessaloniki, Greece
| | | | - Zissis Mamuris
- Laboratory of Genetics, Comparative and Evolutionary Biology, Department of Biochemistry and Biotechnology, University of Thessaly, Viopolis, Mezourlo, 41500 Larissa, Greece
- Correspondence:
| |
Collapse
|
36
|
Bielawiec P, Dziemitko S, Konstantynowicz-Nowicka K, Chabowski A, Dzięcioł J, Harasim-Symbor E. Cannabidiol improves muscular lipid profile by affecting the expression of fatty acid transporters and inhibiting de novo lipogenesis. Sci Rep 2023; 13:3694. [PMID: 36879113 PMCID: PMC9988888 DOI: 10.1038/s41598-023-30872-w] [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: 12/09/2022] [Accepted: 03/02/2023] [Indexed: 03/08/2023] Open
Abstract
Obesity is one of the principal public health concerns leading to disturbances in glucose and lipid metabolism, which is a risk factor for several chronic diseases, including insulin resistance, type 2 diabetes mellitus, and cardiovascular diseases. In recent years, it turned out that cannabidiol (CBD) is a potential therapeutic agent in the treatment of obesity and its complications. Therefore, in the present study, we used CBD therapy (intraperitoneal injections in a dose of 10 mg/kg of body mass for 14 days) in a rat model of obesity induced by a high-fat diet (HFD). Gas-liquid chromatography and Western blotting were applied in order to determine the intramuscular lipid content and total expression of selected proteins in the white and red gastrocnemius muscle, respectively. Based on fatty acid composition, we calculated de novo lipogenesis ratio (16:0/18:2n-6), desaturation ratio (18:1n-9/18:0), and elongation ratios (18:0/16:0, 20:0/18:0, 22:0/20:0 and 24:0/22:0), in the selected lipid fractions. Two-week CBD administration significantly reduced the intramuscular fatty acids (FAs) accumulation and inhibited de novo lipogenesis in different lipid pools (in the free fatty acid, diacylglycerol, and triacylglycerol fractions) in both muscle types, which coincided with a decrease in the expression of membrane fatty acid transporters (fatty acid translocase, membrane-associated fatty acid binding protein, and fatty acid transport proteins 1 and 4). Moreover, CBD application profoundly improved the elongation and desaturation ratios, which was in line with downregulated expression of enzymes from the family of elongases and desaturases regardless of the metabolism presented by the muscle type. To our knowledge, this study is the first that outlines the novel effects of CBD action on skeletal muscle with different types of metabolism (oxidative vs. glycolytic).
Collapse
Affiliation(s)
- Patrycja Bielawiec
- Department of Physiology, Medical University of Bialystok, Bialystok, Poland.
| | - Sylwia Dziemitko
- Department of Physiology, Medical University of Bialystok, Bialystok, Poland
| | | | - Adrian Chabowski
- Department of Physiology, Medical University of Bialystok, Bialystok, Poland
| | - Janusz Dzięcioł
- Department of Human Anatomy, Medical University of Bialystok, Bialystok, Poland
| | - Ewa Harasim-Symbor
- Department of Physiology, Medical University of Bialystok, Bialystok, Poland
| |
Collapse
|
37
|
Kirwan JP, Heintz EC, Rebello CJ, Axelrod CL. Exercise in the Prevention and Treatment of Type 2 Diabetes. Compr Physiol 2023; 13:4559-4585. [PMID: 36815623 DOI: 10.1002/cphy.c220009] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/24/2023]
Abstract
Type 2 diabetes is a systemic, multifactorial disease that is a leading cause of morbidity and mortality globally. Despite a rise in the number of available medications and treatments available for management, exercise remains a first-line prevention and intervention strategy due to established safety, efficacy, and tolerability in the general population. Herein we review the predisposing risk factors for, prevention, pathophysiology, and treatment of type 2 diabetes. We emphasize key cellular and molecular adaptive processes that provide insight into our evolving understanding of how, when, and what types of exercise may improve glycemic control. © 2023 American Physiological Society. Compr Physiol 13:1-27, 2023.
Collapse
Affiliation(s)
- John P Kirwan
- Integrative Physiology and Molecular Medicine Laboratory, Pennington Biomedical Research Center, Baton Rouge, Louisiana, USA
| | - Elizabeth C Heintz
- Integrative Physiology and Molecular Medicine Laboratory, Pennington Biomedical Research Center, Baton Rouge, Louisiana, USA
| | - Candida J Rebello
- Integrative Physiology and Molecular Medicine Laboratory, Pennington Biomedical Research Center, Baton Rouge, Louisiana, USA
| | - Christopher L Axelrod
- Integrative Physiology and Molecular Medicine Laboratory, Pennington Biomedical Research Center, Baton Rouge, Louisiana, USA
| |
Collapse
|
38
|
Sanches JM, Zhao LN, Salehi A, Wollheim CB, Kaldis P. Pathophysiology of type 2 diabetes and the impact of altered metabolic interorgan crosstalk. FEBS J 2023; 290:620-648. [PMID: 34847289 DOI: 10.1111/febs.16306] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 10/14/2021] [Accepted: 11/29/2021] [Indexed: 02/06/2023]
Abstract
Diabetes is a complex and multifactorial disease that affects millions of people worldwide, reducing the quality of life significantly, and results in grave consequences for our health care system. In type 2 diabetes (T2D), the lack of β-cell compensatory mechanisms overcoming peripherally developed insulin resistance is a paramount factor leading to disturbed blood glucose levels and lipid metabolism. Impaired β-cell functions and insulin resistance have been studied extensively resulting in a good understanding of these pathways but much less is known about interorgan crosstalk, which we define as signaling between tissues by secreted factors. Besides hormones and organokines, dysregulated blood glucose and long-lasting hyperglycemia in T2D is associated with changes in metabolism with metabolites from different tissues contributing to the development of this disease. Recent data suggest that metabolites, such as lipids including free fatty acids and amino acids, play important roles in the interorgan crosstalk during the development of T2D. In general, metabolic remodeling affects physiological homeostasis and impacts the development of T2D. Hence, we highlight the importance of metabolic interorgan crosstalk in this review to gain enhanced knowledge of the pathophysiology of T2D, which may lead to new therapeutic approaches to treat this disease.
Collapse
Affiliation(s)
| | - Li Na Zhao
- Department of Clinical Sciences, Lund University, Malmö, Sweden
| | - Albert Salehi
- Department of Clinical Sciences, Lund University, Malmö, Sweden
| | - Claes B Wollheim
- Department of Clinical Sciences, Lund University, Malmö, Sweden.,Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland
| | - Philipp Kaldis
- Department of Clinical Sciences, Lund University, Malmö, Sweden
| |
Collapse
|
39
|
Saeidi A, Nouri-Habashi A, Razi O, Ataeinosrat A, Rahmani H, Mollabashi SS, Bagherzadeh-Rahmani B, Aghdam SM, Khalajzadeh L, Al Kiyumi MH, Hackney AC, Laher I, Heinrich KM, Zouhal H. Astaxanthin Supplemented with High-Intensity Functional Training Decreases Adipokines Levels and Cardiovascular Risk Factors in Men with Obesity. Nutrients 2023; 15:nu15020286. [PMID: 36678157 PMCID: PMC9866205 DOI: 10.3390/nu15020286] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Revised: 12/29/2022] [Accepted: 01/03/2023] [Indexed: 01/09/2023] Open
Abstract
The aim of this study was to investigate the effects of 12 weeks of high-intensity training with astaxanthin supplementation on adipokine levels, insulin resistance and lipid profiles in males with obesity. Sixty-eight males with obesity were randomly stratified into four groups of seventeen subjects each: control group (CG), supplement group (SG), training group (TG), and training plus supplement group (TSG). Participants underwent 12 weeks of treatment with astaxanthin or placebo (20 mg/d capsule daily). The training protocol consisted of 36 sessions of high-intensity functional training (HIFT), 60 min/sessions, and three sessions/week. Metabolic profiles, body composition, anthropometrical measurements, cardio-respiratory indices and adipokine [Cq1/TNF-related protein 9 and 2 (CTRP9 and CTRP2) levels, and growth differentiation factors 8 and 15 (GDF8 and GDF15)] were measured. There were significant differences for all indicators between the groups (p < 0.05). Post-hoc analysis indicated that the levels of CTRP9, CTRP2, and GDF8 were different from CG (p < 0.05), although levels of GDF15 were similar to CG (p > 0.05). Levels of GDF8 were similar in the SG and TG groups (p > 0.05), with reductions of GDF15 levels in both training groups (p < 0.05). A total of 12 weeks of astaxanthin supplementation and exercise training decreased adipokines levels, body composition (weight, %fat), anthropometrical factors (BMI), and improved lipid and metabolic profiles. These benefits were greater for men with obesity in the TSG group.
Collapse
Affiliation(s)
- Ayoub Saeidi
- Department of Physical Education and Sport Sciences, Faculty of Humanities and Social Sciences, University of Kurdistan, Sanandaj 66177-15175, Iran
| | - Akbar Nouri-Habashi
- Department of Exercise Physiology and Corrective Movements, Faculty of Sport Sciences, Urmia University, Urmia 57561-51818, Iran
- Correspondence: (A.N.-H.); (M.H.A.K.)
| | - Omid Razi
- Department of Exercise Physiology, Faculty of Physical Education and Sports Science, Razi University, Kermanshah 94Q5+6G3, Iran
| | - Ali Ataeinosrat
- Department of Physical Education and Sport Science, Science and Research Branch, Islamic Azad University, Tehran 14778-93855, Iran
| | - Hiwa Rahmani
- Faculty of Physical Education and Sports Science, Alzahra University, Tehran 19938 93973, Iran
| | | | - Behnam Bagherzadeh-Rahmani
- Department of Exercise Physiology, Faculty of Sport Sciences, Hakim Sabzevari University, Sabzevar M3J+373, Iran
| | - Shahin Mahmoudi Aghdam
- Department of Exercise Physiology, Central Tehran Branch, Islamic Azad University, Tehran 14778-93855, Iran
| | - Leila Khalajzadeh
- Department of Exercise Physiology, Central Tehran Branch, Islamic Azad University, Tehran 14778-93855, Iran
| | - Maisa Hamed Al Kiyumi
- Department of Family Medicine and Public Health, Sultan Qaboos University Hospital, Muscat H5QC+36M, Oman
- Correspondence: (A.N.-H.); (M.H.A.K.)
| | - Anthony C. Hackney
- Department of Exercise & Sport Science, University of North Carolina, Chapel Hill, NC 27599, USA
- Department of Nutrition, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Ismail Laher
- Department of Anesthesiology, Pharmacology and Therapeutics, The University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Katie M. Heinrich
- Department of Kinesiology, College of Health and Human Sciences, Kansas State University, Manhattan, KS 66506, USA
| | - Hassane Zouhal
- Laboratoire Mouvement, Sport, Santé, University of Rennes, M2S—EA 1274, 35000 Rennes, France
- Institut International des Sciences du Sport (2I2S), 35850 Irodouer, France
| |
Collapse
|
40
|
Calderón-DuPont D, Torre-Villalvazo I, Díaz-Villaseñor A. Is insulin resistance tissue-dependent and substrate-specific? The role of white adipose tissue and skeletal muscle. Biochimie 2023; 204:48-68. [PMID: 36099940 DOI: 10.1016/j.biochi.2022.08.021] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 08/19/2022] [Accepted: 08/31/2022] [Indexed: 01/12/2023]
Abstract
Insulin resistance (IR) refers to a reduction in the ability of insulin to exert its metabolic effects in organs such as adipose tissue (AT) and skeletal muscle (SM), leading to chronic diseases such as type 2 diabetes, hepatic steatosis, and cardiovascular diseases. Obesity is the main cause of IR, however not all subjects with obesity develop clinical insulin resistance, and not all clinically insulin-resistant people have obesity. Recent evidence implies that IR onset is tissue-dependent (AT or SM) and/or substrate-specific (glucometabolic or lipometabolic). Therefore, the aims of the present review are 1) to describe the glucometabolic and lipometabolic activities of insulin in AT and SM in the maintenance of whole-body metabolic homeostasis, 2) to discuss the pathophysiology of substrate-specific IR in AT and SM, and 3) to highlight novel validated tests to assess tissue and substrate-specific IR that are easy to perform in clinical practice. In AT, glucometabolic IR reduces glucose availability for glycerol and fatty acid synthesis, thus decreasing the esterification and synthesis of signaling bioactive lipids. Lipometabolic IR in AT impairs the antilipolytic effect of insulin and lipogenesis, leading to an increase in circulating FFAs and generating lipotoxicity in peripheral tissues. In SM, glucometabolic IR reduces glucose uptake, whereas lipometabolic IR impairs mitochondrial lipid oxidation, increasing oxidative stress and inflammation, all of which lead to metabolic inflexibility. Understanding tissue-dependent and substrate-specific IR is of paramount importance for early detection before clinical manifestations and for the development of more specific treatments or direct interventions to prevent chronic life-threatening diseases.
Collapse
Affiliation(s)
- Diana Calderón-DuPont
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), Mexico City, 04510, Mexico; Doctorado en Ciencias Biomédicas, Universidad Nacional Autónoma de México (UNAM), Mexico City, 04510, Mexico
| | - Ivan Torre-Villalvazo
- Departamento de Fisiología de la Nutrición, Instituto Nacional en Ciencias Médicas y Nutricíon Salvador Zubirán, Mexico City, 14000, Mexico
| | - Andrea Díaz-Villaseñor
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), Mexico City, 04510, Mexico.
| |
Collapse
|
41
|
Handy RM, Holloway GP. Insights into the development of insulin resistance: Unraveling the interaction of physical inactivity, lipid metabolism and mitochondrial biology. Front Physiol 2023; 14:1151389. [PMID: 37153211 PMCID: PMC10157178 DOI: 10.3389/fphys.2023.1151389] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 04/07/2023] [Indexed: 05/09/2023] Open
Abstract
While impairments in peripheral tissue insulin signalling have a well-characterized role in the development of insulin resistance and type 2 diabetes (T2D), the specific mechanisms that contribute to these impairments remain debatable. Nonetheless, a prominent hypothesis implicates the presence of a high-lipid environment, resulting in both reactive lipid accumulation and increased mitochondrial reactive oxygen species (ROS) production in the induction of peripheral tissue insulin resistance. While the etiology of insulin resistance in a high lipid environment is rapid and well documented, physical inactivity promotes insulin resistance in the absence of redox stress/lipid-mediated mechanisms, suggesting alternative mechanisms-of-action. One possible mechanism is a reduction in protein synthesis and the resultant decrease in key metabolic proteins, including canonical insulin signaling and mitochondrial proteins. While reductions in mitochondrial content associated with physical inactivity are not required for the induction of insulin resistance, this could predispose individuals to the detrimental effects of a high-lipid environment. Conversely, exercise-training induced mitochondrial biogenesis has been implicated in the protective effects of exercise. Given mitochondrial biology may represent a point of convergence linking impaired insulin sensitivity in both scenarios of chronic overfeeding and physical inactivity, this review aims to describe the interaction between mitochondrial biology, physical (in)activity and lipid metabolism within the context of insulin signalling.
Collapse
|
42
|
Impact of Dysfunctional Adipose Tissue Depots on the Cardiovascular System. Int J Mol Sci 2022; 23:ijms232214296. [PMID: 36430774 PMCID: PMC9695168 DOI: 10.3390/ijms232214296] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 11/15/2022] [Accepted: 11/16/2022] [Indexed: 11/19/2022] Open
Abstract
Obesity with its associated complications represents a social, economic and health problem of utmost importance worldwide. Specifically, obese patients carry a significantly higher risk of developing cardiovascular disease compared to nonobese individuals. Multiple molecular mechanisms contribute to the impaired biological activity of the distinct adipose tissue depots in obesity, including secretion of proinflammatory mediators and reactive oxygen species, ultimately leading to an unfavorable impact on the cardiovascular system. This review summarizes data relating to the contribution of the main adipose tissue depots, including both remote (i.e., intra-abdominal, hepatic, skeletal, pancreatic, renal, and mesenteric adipose fat), and cardiac (i.e., the epicardial fat) adipose locations, on the cardiovascular system. Finally, we discuss both pharmacological and non-pharmacological strategies aimed at reducing cardiovascular risk through acting on adipose tissues, with particular attention to the epicardial fat.
Collapse
|
43
|
Al-Sabri MH, Behare N, Alsehli AM, Berkins S, Arora A, Antoniou E, Moysiadou EI, Anantha-Krishnan S, Cosmen PD, Vikner J, Moulin TC, Ammar N, Boukhatmi H, Clemensson LE, Rask-Andersen M, Mwinyi J, Williams MJ, Fredriksson R, Schiöth HB. Statins Induce Locomotion and Muscular Phenotypes in Drosophila melanogaster That Are Reminiscent of Human Myopathy: Evidence for the Role of the Chloride Channel Inhibition in the Muscular Phenotypes. Cells 2022; 11:3528. [PMID: 36428957 PMCID: PMC9688544 DOI: 10.3390/cells11223528] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 10/17/2022] [Accepted: 11/01/2022] [Indexed: 11/09/2022] Open
Abstract
The underlying mechanisms for statin-induced myopathy (SIM) are still equivocal. In this study, we employ Drosophila melanogaster to dissect possible underlying mechanisms for SIM. We observe that chronic fluvastatin treatment causes reduced general locomotion activity and climbing ability. In addition, transmission microscopy of dissected skeletal muscles of fluvastatin-treated flies reveals strong myofibrillar damage, including increased sarcomere lengths and Z-line streaming, which are reminiscent of myopathy, along with fragmented mitochondria of larger sizes, most of which are round-like shapes. Furthermore, chronic fluvastatin treatment is associated with impaired lipid metabolism and insulin signalling. Mechanistically, knockdown of the statin-target Hmgcr in the skeletal muscles recapitulates fluvastatin-induced mitochondrial phenotypes and lowered general locomotion activity; however, it was not sufficient to alter sarcomere length or elicit myofibrillar damage compared to controls or fluvastatin treatment. Moreover, we found that fluvastatin treatment was associated with reduced expression of the skeletal muscle chloride channel, ClC-a (Drosophila homolog of CLCN1), while selective knockdown of skeletal muscle ClC-a also recapitulated fluvastatin-induced myofibril damage and increased sarcomere lengths. Surprisingly, exercising fluvastatin-treated flies restored ClC-a expression and normalized sarcomere lengths, suggesting that fluvastatin-induced myofibrillar phenotypes could be linked to lowered ClC-a expression. Taken together, these results may indicate the potential role of ClC-a inhibition in statin-associated muscular phenotypes. This study underlines the importance of Drosophila melanogaster as a powerful model system for elucidating the locomotion and muscular phenotypes, promoting a better understanding of the molecular mechanisms underlying SIM.
Collapse
Affiliation(s)
- Mohamed H. Al-Sabri
- Department of Surgical Sciences, Division of Functional Pharmacology and Neuroscience, Biomedical Center (BMC), Uppsala University, Husargatan 3, 751 24 Uppsala, Sweden
- Department of Pharmaceutical Biosciences, Uppsala University, 751 24 Uppsala, Sweden
| | - Neha Behare
- Department of Surgical Sciences, Division of Functional Pharmacology and Neuroscience, Biomedical Center (BMC), Uppsala University, Husargatan 3, 751 24 Uppsala, Sweden
| | - Ahmed M. Alsehli
- Department of Surgical Sciences, Division of Functional Pharmacology and Neuroscience, Biomedical Center (BMC), Uppsala University, Husargatan 3, 751 24 Uppsala, Sweden
- Faculty of Medicine, King Abdulaziz University and Hospital, Al Ehtifalat St., Jeddah 21589, Saudi Arabia
| | - Samuel Berkins
- Department of Surgical Sciences, Division of Functional Pharmacology and Neuroscience, Biomedical Center (BMC), Uppsala University, Husargatan 3, 751 24 Uppsala, Sweden
| | - Aadeya Arora
- Department of Surgical Sciences, Division of Functional Pharmacology and Neuroscience, Biomedical Center (BMC), Uppsala University, Husargatan 3, 751 24 Uppsala, Sweden
| | - Eirini Antoniou
- Department of Surgical Sciences, Division of Functional Pharmacology and Neuroscience, Biomedical Center (BMC), Uppsala University, Husargatan 3, 751 24 Uppsala, Sweden
| | - Eleni I. Moysiadou
- Department of Surgical Sciences, Division of Functional Pharmacology and Neuroscience, Biomedical Center (BMC), Uppsala University, Husargatan 3, 751 24 Uppsala, Sweden
| | - Sowmya Anantha-Krishnan
- Department of Surgical Sciences, Division of Functional Pharmacology and Neuroscience, Biomedical Center (BMC), Uppsala University, Husargatan 3, 751 24 Uppsala, Sweden
| | - Patricia D. Cosmen
- Department of Surgical Sciences, Division of Functional Pharmacology and Neuroscience, Biomedical Center (BMC), Uppsala University, Husargatan 3, 751 24 Uppsala, Sweden
| | - Johanna Vikner
- Department of Surgical Sciences, Division of Functional Pharmacology and Neuroscience, Biomedical Center (BMC), Uppsala University, Husargatan 3, 751 24 Uppsala, Sweden
| | - Thiago C. Moulin
- Department of Surgical Sciences, Division of Functional Pharmacology and Neuroscience, Biomedical Center (BMC), Uppsala University, Husargatan 3, 751 24 Uppsala, Sweden
- Faculty of Medicine, Department of Experimental Medical Science, Lund University, Sölvegatan 19, BMC F10, 221 84 Lund, Sweden
| | - Nourhene Ammar
- Institut de Génétique et Développement de Rennes (IGDR), Université de Rennes, CNRS, UMR6290, 35065 Rennes, France
| | - Hadi Boukhatmi
- Institut de Génétique et Développement de Rennes (IGDR), Université de Rennes, CNRS, UMR6290, 35065 Rennes, France
| | - Laura E. Clemensson
- Department of Surgical Sciences, Division of Functional Pharmacology and Neuroscience, Biomedical Center (BMC), Uppsala University, Husargatan 3, 751 24 Uppsala, Sweden
| | - Mathias Rask-Andersen
- Department of Immunology, Genetics and Pathology, Uppsala University, 752 37 Uppsala, Sweden
| | - Jessica Mwinyi
- Department of Surgical Sciences, Division of Functional Pharmacology and Neuroscience, Biomedical Center (BMC), Uppsala University, Husargatan 3, 751 24 Uppsala, Sweden
| | - Michael J. Williams
- Department of Surgical Sciences, Division of Functional Pharmacology and Neuroscience, Biomedical Center (BMC), Uppsala University, Husargatan 3, 751 24 Uppsala, Sweden
| | - Robert Fredriksson
- Department of Pharmaceutical Biosciences, Uppsala University, 751 24 Uppsala, Sweden
| | - Helgi B. Schiöth
- Department of Surgical Sciences, Division of Functional Pharmacology and Neuroscience, Biomedical Center (BMC), Uppsala University, Husargatan 3, 751 24 Uppsala, Sweden
| |
Collapse
|
44
|
Alsenousy AHA, El-Tahan RA, Ghazal NA, Piñol R, Millán A, Ali LMA, Kamel MA. The Anti-Obesity Potential of Superparamagnetic Iron Oxide Nanoparticles against High-Fat Diet-Induced Obesity in Rats: Possible Involvement of Mitochondrial Biogenesis in the Adipose Tissues. Pharmaceutics 2022; 14:pharmaceutics14102134. [PMID: 36297569 PMCID: PMC9607364 DOI: 10.3390/pharmaceutics14102134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 10/01/2022] [Accepted: 10/05/2022] [Indexed: 11/05/2022] Open
Abstract
BACKGROUND Obesity is a pandemic disease that is rapidly growing into a serious health problem and has economic impact on healthcare systems. This bleak image has elicited creative responses, and nanotechnology is a promising approach in obesity treatment. This study aimed to investigate the anti-obesity effect of superparamagnetic iron oxide nanoparticles (SPIONs) on a high-fat-diet rat model of obesity and compared their effect to a traditional anti-obesity drug (orlistat). METHODS The obese rats were treated daily with orlistat and/or SPIONs once per week for 8 weeks. At the end of the experiment, blood samples were collected for biochemical assays. Then, the animals were sacrificed to obtain white adipose tissues (WAT) and brown adipose tissues (BAT) for assessment of the expression of thermogenic genes and mitochondrial DNA copy number (mtDNA-CN). RESULTS For the first time, we reported promising ameliorating effects of SPIONs treatments against weight gain, hyperglycemia, adiponectin, leptin, and dyslipidemia in obese rats. At the molecular level, surprisingly, SPIONs treatments markedly corrected the disturbed expression and protein content of inflammatory markers and parameters controlling mitochondrial biogenesis and functions in BAT and WAT. CONCLUSIONS SPIONs have a powerful anti-obesity effect by acting as an inducer of WAT browning and activator of BAT functions.
Collapse
Affiliation(s)
- Aisha H. A. Alsenousy
- Department of Biochemistry, Medical Research Institute, Alexandria University, 165 El-Horeya Rd, Alexandria 21561, Egypt
- Correspondence: (A.H.A.A.); (M.A.K.)
| | - Rasha A. El-Tahan
- Department of Biochemistry, Medical Research Institute, Alexandria University, 165 El-Horeya Rd, Alexandria 21561, Egypt
| | - Nesma A. Ghazal
- Department of Biochemistry, Medical Research Institute, Alexandria University, 165 El-Horeya Rd, Alexandria 21561, Egypt
| | - Rafael Piñol
- Instituto de Nanociencia y Materiales de Aragón (INMA), CSIC-Universidad de Zaragoza, 50009 Zaragoza, Spain
| | - Angel Millán
- Instituto de Nanociencia y Materiales de Aragón (INMA), CSIC-Universidad de Zaragoza, 50009 Zaragoza, Spain
| | - Lamiaa M. A. Ali
- Department of Biochemistry, Medical Research Institute, Alexandria University, 165 El-Horeya Rd, Alexandria 21561, Egypt
- IBMM, University Montpellier, CNRS, ENSCM, 34093 Montpellier, France
| | - Maher A. Kamel
- Department of Biochemistry, Medical Research Institute, Alexandria University, 165 El-Horeya Rd, Alexandria 21561, Egypt
- Correspondence: (A.H.A.A.); (M.A.K.)
| |
Collapse
|
45
|
Kayampilly P, Roeser N, Rajendiran TM, Pennathur S, Afshinnia F. Acetyl Co-A Carboxylase Inhibition Halts Hyperglycemia Induced Upregulation of De Novo Lipogenesis in Podocytes and Proximal Tubular Cells. Metabolites 2022; 12:940. [PMID: 36295842 PMCID: PMC9610518 DOI: 10.3390/metabo12100940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 09/28/2022] [Accepted: 09/30/2022] [Indexed: 11/28/2022] Open
Abstract
The effect of glycemic stress on de novo lipogenesis (DNL) in podocytes and tubular epithelial cells is understudied. This study is aimed (A) to show the effect of glycemic stress on DNL, and (B) to assess the effect of acetyl-Co A (ACC) inhibition on halting upregulation of DNL, on the expression of other lipid regulatory genes in the DNL pathway, and on markers of fibrosis and apoptosis in podocytes and tubular epithelial cells. We used cultured mouse primary tubular epithelial cells, mouse proximal tubular (BUMPT) cells, and immortal mouse podocytes and measured their percentage of labeled 13C2-palmitate as a marker of DNL after incubation with 13C2 acetate in response to high glucose concentration (25 mM). We then tested the effect of ACC inhibition by complimentary strategies utilizing CRISPR/cas9 deletion or incubation with Acaca and Acacb GapmeRs or using a small molecule inhibitor on DNL under hyperglycemic concentration. Exposure to high glucose concentration (25 mM) compared to osmotic controlled low glucose concentration (5.5 mM) significantly increased labeled palmitate after 24 h up to 72 h in podocytes and primary tubular cells. Knocking out of the ACC coding Acaca and Acacb genes by CRISPR/cas9, downregulation of Acaca and Acacb by specific antisense LNA GapmeRs and inhibition of ACC by firsocostat similarly halted/mitigated upregulation of DNL and decreased markers of fibrosis and programmed cell death in podocytes and various tubular cells. ACC inhibition is a potential therapeutic target to mitigate or halt hyperglycemia-induced upregulation of DNL in podocytes and tubular cells.
Collapse
Affiliation(s)
- Pradeep Kayampilly
- Department of Internal Medicine-Nephrology, University of Michigan, Ann Arbor, MI 48105, USA
| | - Nancy Roeser
- Department of Internal Medicine-Nephrology, University of Michigan, Ann Arbor, MI 48105, USA
| | - Thekkelnaycke M Rajendiran
- Michigan Regional Comprehensive Metabolomics Resource Core, University of Michigan, Ann Arbor, MI 48105, USA
- Department of Pathology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Subramaniam Pennathur
- Department of Internal Medicine-Nephrology, University of Michigan, Ann Arbor, MI 48105, USA
- Michigan Regional Comprehensive Metabolomics Resource Core, University of Michigan, Ann Arbor, MI 48105, USA
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Farsad Afshinnia
- Department of Internal Medicine-Nephrology, University of Michigan, Ann Arbor, MI 48105, USA
| |
Collapse
|
46
|
Savikj M, Stocks B, Sato S, Caidahl K, Krook A, Deshmukh AS, Zierath JR, Wallberg-Henriksson H. Exercise timing influences multi-tissue metabolome and skeletal muscle proteome profiles in type 2 diabetic patients - A randomized crossover trial. Metabolism 2022; 135:155268. [PMID: 35908579 DOI: 10.1016/j.metabol.2022.155268] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 07/10/2022] [Accepted: 07/22/2022] [Indexed: 01/06/2023]
Abstract
AIMS/HYPOTHESIS Metabolic effects of exercise may partly depend on the time-of-day when exercise is performed. We tested the hypothesis that exercise timing affects the adaptations in multi-tissue metabolome and skeletal muscle proteome profiles in men with type 2 diabetes. METHODS Men fitting the inclusion (type 2 diabetes, age 45-68 years and body mass index 23-33 kg/m2) and exclusion criteria (insulin treatment, smoking, concurrent systemic disease, and regular exercise training) were included in a randomized crossover trial (n = 15). Participants included in this metabolomics and proteomics analysis fully completed all exercise sessions (n = 8). The trial consisted of two weeks of high-intensity interval training (HIT) (three sessions/week) either in the morning (08:00, n = 5) or afternoon (16:45, n = 3), a two-week wash-out period, and an additional two weeks of HIT at the opposing time. Participants and researchers were not blinded to group allocation. Blood, skeletal muscle and subcutaneous adipose tissue were obtained before the first, and after each training period. Broad-spectrum, untargeted proteomic analysis was performed on skeletal muscle, and metabolomic analysis was performed on all biosamples. Differential content was assessed by linear regression and pathway set enrichment analyses were performed. Coordinated metabolic changes across tissues were identified by Spearman correlation analysis. RESULTS Metabolic and proteomic profiles remained stable after two weeks of HIT, and individual metabolites and proteins were not altered, irrespective of the time of day at which the training was performed. However, coordinated changes in relevant metabolic pathways and protein categories were identified. Morning and afternoon HIT similarly increased plasma diacylglycerols, skeletal muscle acyl-carnitines, and subcutaneous adipose tissue sphingomyelins and lysophospholipids. Acyl-carnitines were central to training-induced metabolic cross-talk across tissues. Plasma carbohydrates, via the penthose phosphate pathway, were increased and skeletal muscle lipids were decreased after morning compared to afternoon HIT. Skeletal muscle lipoproteins were higher, and mitochondrial complex III abundance was lower after morning compared to afternoon HIT. CONCLUSIONS/INTERPRETATION We provide a comprehensive analysis of a multi-tissue metabolomic and skeletal muscle proteomic responses to training at different times of the day in men with type 2 diabetes. Increased circulating lipids and changes in adipose tissue lipid composition were common between morning and afternoon HIT. However, afternoon HIT increased skeletal muscle lipids and mitochondrial content to a greater degree than morning training. Thus, there is a diurnal component in the metabolomic and proteomic response to exercise in men with type 2 diabetes. The clinical relevance of this response warrants further investigation.
Collapse
Affiliation(s)
- Mladen Savikj
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Ben Stocks
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Shogo Sato
- Center for Epigenetics and Metabolism, INSERM U1233, Department of Biological Chemistry, School of Medicine, University of California, Irvine, Irvine, CA, USA
| | - Kenneth Caidahl
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden; Department of Clinical Physiology, Karolinska University Hospital, Stockholm, Sweden
| | - Anna Krook
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Atul S Deshmukh
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; The Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Juleen R Zierath
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden; Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | | |
Collapse
|
47
|
Sheng CY, Son YH, Jang J, Park SJ. In vitro skeletal muscle models for type 2 diabetes. BIOPHYSICS REVIEWS 2022; 3:031306. [PMID: 36124295 PMCID: PMC9478902 DOI: 10.1063/5.0096420] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Accepted: 08/18/2022] [Indexed: 06/15/2023]
Abstract
Type 2 diabetes mellitus, a metabolic disorder characterized by abnormally elevated blood sugar, poses a growing social, economic, and medical burden worldwide. The skeletal muscle is the largest metabolic organ responsible for glucose homeostasis in the body, and its inability to properly uptake sugar often precedes type 2 diabetes. Although exercise is known to have preventative and therapeutic effects on type 2 diabetes, the underlying mechanism of these beneficial effects is largely unknown. Animal studies have been conducted to better understand the pathophysiology of type 2 diabetes and the positive effects of exercise on type 2 diabetes. However, the complexity of in vivo systems and the inability of animal models to fully capture human type 2 diabetes genetics and pathophysiology are two major limitations in these animal studies. Fortunately, in vitro models capable of recapitulating human genetics and physiology provide promising avenues to overcome these obstacles. This review summarizes current in vitro type 2 diabetes models with focuses on the skeletal muscle, interorgan crosstalk, and exercise. We discuss diabetes, its pathophysiology, common in vitro type 2 diabetes skeletal muscle models, interorgan crosstalk type 2 diabetes models, exercise benefits on type 2 diabetes, and in vitro type 2 diabetes models with exercise.
Collapse
Affiliation(s)
- Christina Y. Sheng
- Biohybrid Systems Group, Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University School of Medicine, Atlanta, Georgia 30322, USA
| | - Young Hoon Son
- Biohybrid Systems Group, Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University School of Medicine, Atlanta, Georgia 30322, USA
| | | | - Sung-Jin Park
- Biohybrid Systems Group, Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University School of Medicine, Atlanta, Georgia 30322, USA
| |
Collapse
|
48
|
Blood and Urinary Biomarkers of Antipsychotic-Induced Metabolic Syndrome. Metabolites 2022; 12:metabo12080726. [PMID: 36005598 PMCID: PMC9416438 DOI: 10.3390/metabo12080726] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 07/29/2022] [Accepted: 08/03/2022] [Indexed: 12/15/2022] Open
Abstract
Metabolic syndrome (MetS) is a clustering of at least three of the following five medical conditions: abdominal obesity, high blood pressure, high blood sugar, high serum triglycerides, and low serum high-density lipoprotein (HDL). Antipsychotic (AP)-induced MetS (AIMetS) is the most common adverse drug reaction (ADR) of psychiatric pharmacotherapy. Herein, we review the results of studies of blood (serum and plasma) and urinary biomarkers as predictors of AIMetS in patients with schizophrenia (Sch). We reviewed 1440 studies examining 38 blood and 19 urinary metabolic biomarkers, including urinary indicators involved in the development of AIMetS. Among the results, only positive associations were revealed. However, at present, it should be recognized that there is no consensus on the role of any particular urinary biomarker of AIMetS. Evaluation of urinary biomarkers of the development of MetS and AIMetS, as one of the most common concomitant pathological conditions in the treatment of patients with psychiatric disorders, may provide a key to the development of strategies for personalized prevention and treatment of the condition, which is considered a complication of AP therapy for Sch in clinical practice.
Collapse
|
49
|
Huerta-Delgado AS, Roffe-Vazquez DN, Luna-Ceron E, Gonzalez-Gil AM, Casillas-Fikentscher A, Villarreal-Calderon JR, Enriquez C, de la Peña-Almaguer E, Castillo EC, Silva-Platas C, Garcia-Rivas G, Elizondo-Montemayor L. Association of irisin levels with cardiac magnetic resonance, inflammatory, and biochemical parameters in patients with chronic heart failure versus controls. Magn Reson Imaging 2022; 93:62-72. [PMID: 35842196 DOI: 10.1016/j.mri.2022.07.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 06/15/2022] [Accepted: 07/11/2022] [Indexed: 12/14/2022]
Abstract
BACKGROUND AND AIMS Chronic heart failure (CHF) represents a significant cause of morbidity and mortality globally. Metabolic maladaptation has proven to be critical in the progression of this condition. Preclinical studies have shown that irisin, an adipomyokine involved in metabolic regulations, can induce positive cardioprotective effects by improving cardiac remodeling, cardiomyocyte viability, calcium delivery, and reducing inflammatory mediators. However, data on clinical studies identifying the associations between irisin levels and functional imaging parameters are scarce in CHF patients. The objective of this study was to determine the association of irisin levels with cardiac imaging measurements through cardiac magnetic resonance, inflammatory markers, and biochemical parameters in patients with CHF compared with control subjects. METHODS AND RESULTS Thirty-two subjects diagnosed with CHF and thirty-two healthy controls were evaluated in a cross-sectional study. Serum irisin levels were significantly lower in patients with CHF than in controls. This is the first study to report a significant positive correlation between irisin levels and cardiac magnetic resonance parameters such as left ventricular ejection fraction, fraction shortening, and global radial strain. A negative correlation was demonstrated between irisin levels and brain natriuretic peptide, insulin levels, and Homeostatic model assessment for insulin resistance index. We did not observe significant correlations between irisin levels and inflammatory cytokines. CONCLUSIONS Given the importance of fraction shortening and global radial strain as accurate markers of ventricular wall motion, these results support the hypothesis that irisin may play an essential role in maintaining an adequate myocardial wall architecture, deformation, and thickness.
Collapse
Affiliation(s)
- Anna S Huerta-Delgado
- Tecnologico de Monterrey, Center for Research in Clinical Nutrition and Obesity, Escuela de Medicina, 64710 Monterrey, N.L., Mexico
| | - Daniel N Roffe-Vazquez
- Tecnologico de Monterrey, Center for Research in Clinical Nutrition and Obesity, Escuela de Medicina, 64710 Monterrey, N.L., Mexico
| | - Eder Luna-Ceron
- Tecnologico de Monterrey, Center for Research in Clinical Nutrition and Obesity, Escuela de Medicina, 64710 Monterrey, N.L., Mexico
| | - Adrian M Gonzalez-Gil
- Tecnologico de Monterrey, Center for Research in Clinical Nutrition and Obesity, Escuela de Medicina, 64710 Monterrey, N.L., Mexico
| | - Andrea Casillas-Fikentscher
- Tecnologico de Monterrey, Center for Research in Clinical Nutrition and Obesity, Escuela de Medicina, 64710 Monterrey, N.L., Mexico
| | - José R Villarreal-Calderon
- Tecnologico de Monterrey, Center for Research in Clinical Nutrition and Obesity, Escuela de Medicina, 64710 Monterrey, N.L., Mexico
| | - Cecilio Enriquez
- Tecnologico de Monterrey, Centro de Investigacion Biomedica, Hospital Zambrano Hellion, 66278 San Pedro Garza-Garcia, N.L., Mexico
| | - Erasmo de la Peña-Almaguer
- Tecnologico de Monterrey, Centro de Investigacion Biomedica, Hospital Zambrano Hellion, 66278 San Pedro Garza-Garcia, N.L., Mexico
| | - Elena C Castillo
- Tecnologico de Monterrey, Centro de Investigacion Biomedica, Hospital Zambrano Hellion, 66278 San Pedro Garza-Garcia, N.L., Mexico
| | - Christian Silva-Platas
- Tecnologico de Monterrey, Centro de Investigacion Biomedica, Hospital Zambrano Hellion, 66278 San Pedro Garza-Garcia, N.L., Mexico
| | - Gerardo Garcia-Rivas
- Tecnologico de Monterrey, Centro de Investigacion Biomedica, Hospital Zambrano Hellion, 66278 San Pedro Garza-Garcia, N.L., Mexico; Tecnologico de Monterrey, Cardiovascular Medicine and Metabolomics Research Group, Escuela de Medicina, 66278 San Pedro Garza-Garcia, N.L., Mexico
| | - Leticia Elizondo-Montemayor
- Tecnologico de Monterrey, Center for Research in Clinical Nutrition and Obesity, Escuela de Medicina, 64710 Monterrey, N.L., Mexico; Tecnologico de Monterrey, Cardiovascular Medicine and Metabolomics Research Group, Escuela de Medicina, 66278 San Pedro Garza-Garcia, N.L., Mexico.
| |
Collapse
|
50
|
Wu ZE, Kruger MC, Cooper GJS, Sequeira IR, McGill AT, Poppitt SD, Fraser K. Dissecting the relationship between plasma and tissue metabolome in a cohort of women with obesity: Analysis of subcutaneous and visceral adipose, muscle, and liver. FASEB J 2022; 36:e22371. [PMID: 35704337 DOI: 10.1096/fj.202101812r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 04/30/2022] [Accepted: 05/11/2022] [Indexed: 11/11/2022]
Abstract
Untargeted metabolomics of blood samples has become widely applied to study metabolic alterations underpinning disease and to identify biomarkers. However, understanding the relevance of a blood metabolite marker can be challenging if it is unknown whether it reflects the concentration in relevant tissues. To explore this field, metabolomic and lipidomic profiles of plasma, four sites of adipose tissues (ATs) from peripheral or central depot, two sites of muscle tissue, and liver tissue from a group of nondiabetic women with obesity who were scheduled to undergo bariatric surgery (n = 21) or other upper GI surgery (n = 5), were measured by liquid chromatography coupled with mass spectrometry. Relationships between plasma and tissue profiles were examined using Pearson correlation analysis subject to Benjamini-Hochberg correction. Plasma metabolites and lipids showed the highest number of significantly positive correlations with their corresponding concentrations in liver tissue, including lipid species of ceramide, mono- and di-hexosylceramide, sphingomyelin, phosphatidylcholine (PC), phosphatidylethanolamine (PE), lysophosphatidylethanolamine, dimethyl phosphatidylethanolamine, ether-linked PC, ether-linked PE, free fatty acid, cholesteryl ester, diacylglycerol and triacylglycerol, and polar metabolites linked to several metabolic functions and gut microbial metabolism. Plasma also showed significantly positive correlations with muscle for several phospholipid species and polar metabolites linked to metabolic functions and gut microbial metabolism, and with AT for several triacylglycerol species. In conclusion, plasma metabolomic and lipidomic profiles were reflective more of the liver profile than any of the muscle or AT sites examined in the present study. Our findings highlighted the importance of taking into consideration the metabolomic relationship of various tissues with plasma when postulating plasma metabolites marker to underlying mechanisms occurring in a specific tissue.
Collapse
Affiliation(s)
- Zhanxuan E Wu
- Food Chemistry and Structure, AgResearch Limited, Palmerston North, New Zealand.,School of Health Sciences, Massey University, Palmerston North, New Zealand.,High-Value Nutrition National Science Challenge, Auckland, New Zealand
| | - Marlena C Kruger
- School of Health Sciences, Massey University, Palmerston North, New Zealand.,Riddet Institute, Massey University, Palmerston North, New Zealand
| | - Garth J S Cooper
- School of Biological Sciences, University of Auckland, Auckland, New Zealand.,Department of Medicine, University of Auckland, Auckland, New Zealand.,Centre for Advanced Discovery and Experimental Therapeutics, School of Medical Sciences, University of Manchester, Manchester, UK
| | - Ivana R Sequeira
- High-Value Nutrition National Science Challenge, Auckland, New Zealand.,School of Biological Sciences, University of Auckland, Auckland, New Zealand.,Human Nutrition Unit, School of Biological Sciences, University of Auckland, Auckland, New Zealand
| | - Anne-Thea McGill
- Human Nutrition Unit, School of Biological Sciences, University of Auckland, Auckland, New Zealand
| | - Sally D Poppitt
- High-Value Nutrition National Science Challenge, Auckland, New Zealand.,Riddet Institute, Massey University, Palmerston North, New Zealand.,Department of Medicine, University of Auckland, Auckland, New Zealand.,Human Nutrition Unit, School of Biological Sciences, University of Auckland, Auckland, New Zealand
| | - Karl Fraser
- Food Chemistry and Structure, AgResearch Limited, Palmerston North, New Zealand.,High-Value Nutrition National Science Challenge, Auckland, New Zealand.,Riddet Institute, Massey University, Palmerston North, New Zealand
| |
Collapse
|