1
|
Lopez-Pajares V, Bhaduri A, Zhao Y, Gowrishankar G, Donohue LKH, Guo MG, Siprashvili Z, Miao W, Nguyen DT, Yang X, Li AM, Tung ASH, Shanderson RL, Winge MCG, Meservey LM, Srinivasan S, Meyers RM, Guerrero A, Ji AL, Garcia OS, Tao S, Gambhir SS, Long JZ, Ye J, Khavari PA. Glucose modulates IRF6 transcription factor dimerization to enable epidermal differentiation. Cell Stem Cell 2025:S1934-5909(25)00088-8. [PMID: 40120584 DOI: 10.1016/j.stem.2025.02.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 12/18/2024] [Accepted: 02/28/2025] [Indexed: 03/25/2025]
Abstract
Non-energetic roles for glucose are largely unclear, as is the interplay between transcription factors (TFs) and ubiquitous biomolecules. Metabolomic analyses uncovered elevation of intracellular glucose during differentiation of diverse cell types. Human and mouse tissue engineered with glucose sensors detected a glucose gradient that peaked in the outermost differentiated layers of the epidermis. Free glucose accumulation was essential for epidermal differentiation and required the SGLT1 glucose transporter. Glucose affinity chromatography uncovered glucose binding to diverse regulatory proteins, including the IRF6 TF. Direct glucose binding enabled IRF6 dimerization, DNA binding, genomic localization, and induction of IRF6 target genes, including essential pro-differentiation TFs GRHL1, GRHL3, HOPX, and PRDM1. These data identify a role for glucose as a gradient morphogen that modulates protein multimerization in cellular differentiation.
Collapse
Affiliation(s)
- Vanessa Lopez-Pajares
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA 94305, USA.
| | - Aparna Bhaduri
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA 94305, USA; Program in Cancer Biology, Stanford University, Stanford, CA 94305, USA
| | - Yang Zhao
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Gayatri Gowrishankar
- Departments of Bioengineering and Radiology, Stanford University, Stanford, CA 94305, USA; Molecular Imaging Program at Stanford, Stanford University, Stanford, CA 94305, USA
| | - Laura K H Donohue
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Genetics, Stanford University, Stanford, CA 94305, USA
| | - Margaret G Guo
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA 94305, USA; Program in Biomedical Informatics, Stanford University, Stanford, CA 94305, USA
| | - Zurab Siprashvili
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Weili Miao
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Duy T Nguyen
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Xue Yang
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA 94305, USA; Program in Cancer Biology, Stanford University, Stanford, CA 94305, USA
| | - Albert M Li
- Department of Radiation Oncology, Stanford University, Stanford, CA 94305, USA
| | - Alan Sheng-Hwa Tung
- Department of Pathology, Stanford University, Stanford, CA 94350, USA; Sarafan ChEM-H, Stanford University, Stanford, CA 94305, USA
| | - Ronald L Shanderson
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA 94305, USA; Program in Cancer Biology, Stanford University, Stanford, CA 94305, USA
| | - Marten C G Winge
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Lindsey M Meservey
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Biology, Stanford University, Stanford, CA 94305, USA
| | - Suhas Srinivasan
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Robin M Meyers
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Genetics, Stanford University, Stanford, CA 94305, USA
| | - Angela Guerrero
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Andrew L Ji
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Omar S Garcia
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Shiying Tao
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Sanjiv S Gambhir
- Departments of Bioengineering and Radiology, Stanford University, Stanford, CA 94305, USA; Molecular Imaging Program at Stanford, Stanford University, Stanford, CA 94305, USA
| | - Jonathan Z Long
- Department of Pathology, Stanford University, Stanford, CA 94350, USA; Sarafan ChEM-H, Stanford University, Stanford, CA 94305, USA
| | - Jiangbin Ye
- Department of Radiation Oncology, Stanford University, Stanford, CA 94305, USA
| | - Paul A Khavari
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA 94305, USA; Program in Cancer Biology, Stanford University, Stanford, CA 94305, USA; Veterans Affairs Palo Alto Healthcare System, Palo Alto, CA 94304, USA.
| |
Collapse
|
2
|
Song J, Wang C, Zhao T, Zhang Y, Xing J, Zhao X, Zhang Y, Zhang Z. Multi-omics approaches for biomarker discovery and precision diagnosis of prediabetes. Front Endocrinol (Lausanne) 2025; 16:1520436. [PMID: 40162315 PMCID: PMC11949806 DOI: 10.3389/fendo.2025.1520436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 02/24/2025] [Indexed: 04/02/2025] Open
Abstract
Recent advancements in multi-omics technologies have provided unprecedented opportunities to identify biomarkers associated with prediabetes, offering novel insights into its diagnosis and management. This review synthesizes the latest findings on prediabetes from multiple omics domains, including genomics, epigenomics, transcriptomics, proteomics, metabolomics, microbiomics, and radiomics. We explore how these technologies elucidate the molecular and cellular mechanisms underlying prediabetes and analyze potential biomarkers with predictive value in disease progression. Integrating multi-omics data helps address the limitations of traditional diagnostic methods, enabling early detection, personalized interventions, and improved patient outcomes. However, challenges such as data integration, standardization, and clinical validation and translation remain to be resolved. Future research leveraging artificial intelligence and machine learning is expected to further enhance the predictive power of multi-omics technologies, contributing to the precision diagnosis and tailored management of prediabetes.
Collapse
Affiliation(s)
- Jielin Song
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- TCM Institute of Sore and Ulcer, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Tianjin Institute of Traditional Chinese Medicine Surgery, Tianjin, China
| | - Chuanfu Wang
- Department of Encephalopathy, Liangping District Hospital of Traditional Chinese Medicine, Chongqing, China
| | - Tong Zhao
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- TCM Institute of Sore and Ulcer, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Tianjin Institute of Traditional Chinese Medicine Surgery, Tianjin, China
| | - Yu Zhang
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- TCM Institute of Sore and Ulcer, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Tianjin Institute of Traditional Chinese Medicine Surgery, Tianjin, China
| | - Jixiang Xing
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- TCM Institute of Sore and Ulcer, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Tianjin Institute of Traditional Chinese Medicine Surgery, Tianjin, China
| | - Xuelian Zhao
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- TCM Institute of Sore and Ulcer, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Tianjin Institute of Traditional Chinese Medicine Surgery, Tianjin, China
| | - Yunsha Zhang
- TCM Institute of Sore and Ulcer, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Tianjin Institute of Traditional Chinese Medicine Surgery, Tianjin, China
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Zhaohui Zhang
- TCM Institute of Sore and Ulcer, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Tianjin Institute of Traditional Chinese Medicine Surgery, Tianjin, China
- Department of Traditional Chinese Medicine Surgery, The Second Affiliated Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|
3
|
Sandforth L, Kullmann S, Sandforth A, Fritsche A, Jumpertz-von Schwartzenberg R, Stefan N, Birkenfeld AL. Prediabetes remission to reduce the global burden of type 2 diabetes. Trends Endocrinol Metab 2025:S1043-2760(25)00004-9. [PMID: 39955249 DOI: 10.1016/j.tem.2025.01.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 12/12/2024] [Accepted: 01/15/2025] [Indexed: 02/17/2025]
Abstract
Prediabetes is a highly prevalent and increasingly common condition affecting a significant proportion of the global population. The heterogeneous nature of prediabetes presents a challenge in identifying individuals who particularly benefit from lifestyle or other therapeutic interventions aiming at preventing type 2 diabetes (T2D) and associated comorbidities. The phenotypic characteristics of individuals at risk for diabetes are associated with both specific risk profiles for progression and a differential potential to facilitate prediabetes remission and reduce the risk of future T2D. This review examines the current definition and global prevalence of prediabetes and evaluates the potential of prediabetes remission to reduce the alarming increase in the global burden of T2D.
Collapse
Affiliation(s)
- Leontine Sandforth
- Institute for Diabetes Research and Metabolic Diseases of Helmholtz Munich at the University of Tübingen, Tübingen, Germany; Internal Medicine IV, Endocrinology, Diabetology, and Nephrology, University Hospital Tübingen, Tübingen, Germany; German Center for Diabetes Research, Tübingen, Germany
| | - Stephanie Kullmann
- Institute for Diabetes Research and Metabolic Diseases of Helmholtz Munich at the University of Tübingen, Tübingen, Germany; Internal Medicine IV, Endocrinology, Diabetology, and Nephrology, University Hospital Tübingen, Tübingen, Germany; German Center for Diabetes Research, Tübingen, Germany
| | - Arvid Sandforth
- Institute for Diabetes Research and Metabolic Diseases of Helmholtz Munich at the University of Tübingen, Tübingen, Germany; Internal Medicine IV, Endocrinology, Diabetology, and Nephrology, University Hospital Tübingen, Tübingen, Germany; German Center for Diabetes Research, Tübingen, Germany
| | - Andreas Fritsche
- Institute for Diabetes Research and Metabolic Diseases of Helmholtz Munich at the University of Tübingen, Tübingen, Germany; Internal Medicine IV, Endocrinology, Diabetology, and Nephrology, University Hospital Tübingen, Tübingen, Germany; German Center for Diabetes Research, Tübingen, Germany
| | - Reiner Jumpertz-von Schwartzenberg
- Institute for Diabetes Research and Metabolic Diseases of Helmholtz Munich at the University of Tübingen, Tübingen, Germany; Internal Medicine IV, Endocrinology, Diabetology, and Nephrology, University Hospital Tübingen, Tübingen, Germany; German Center for Diabetes Research, Tübingen, Germany; M3 Research Center, Malignom, Metabolome, Microbiome, 72076 Tübingen, Germany; Cluster of Excellence EXC 2124 'Controlling Microbes to Fight Infections' (CMFI), University of Tübingen, Tübingen, Germany
| | - Norbert Stefan
- Institute for Diabetes Research and Metabolic Diseases of Helmholtz Munich at the University of Tübingen, Tübingen, Germany; Internal Medicine IV, Endocrinology, Diabetology, and Nephrology, University Hospital Tübingen, Tübingen, Germany; German Center for Diabetes Research, Tübingen, Germany
| | - Andreas L Birkenfeld
- Institute for Diabetes Research and Metabolic Diseases of Helmholtz Munich at the University of Tübingen, Tübingen, Germany; Internal Medicine IV, Endocrinology, Diabetology, and Nephrology, University Hospital Tübingen, Tübingen, Germany; German Center for Diabetes Research, Tübingen, Germany; Department of Diabetes, Life Sciences, and Medicine, Cardiovascular Medicine and Life Sciences, King's College London, London, UK.
| |
Collapse
|
4
|
Kaddi C, Tao M, Bergeler S, George K, Geerts H, van der Graaf PH, Batista JL, Foster M, Ortemann-Renon C, Zaher A, An Haack K, Zaph S. Quantitative Systems Pharmacology-Based Digital Twins Approach Supplements Clinical Trial Data for Enzyme Replacement Therapies in Pompe Disease. Clin Pharmacol Ther 2025; 117:579-588. [PMID: 39632463 DOI: 10.1002/cpt.3498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 10/28/2024] [Indexed: 12/07/2024]
Abstract
Pompe disease is a rare, progressive neuromuscular disease caused by deficient lysosomal glycogen degradation, and includes both late-onset (LOPD) and severe infantile-onset (IOPD) phenotypes. Due to very small patient numbers in IOPD and the high phenotypic heterogeneity observed in this population, a quantitative systems pharmacology (QSP)-based "digital twin" approach was developed to perform an in silico comparison of the efficacy of avalglucosidase alfa vs. the standard of care, in a virtual population of IOPD patients. A QSP model was developed that represents key elements of Pompe disease pathophysiology, including tissue glycogen accumulation and the elevation of the biomarker urine Hex4 in both LOPD and IOPD patients. In this approach, the QSP model was used to generate digital twins of each IOPD patient enrolled in the avalglucosidase alfa clinical program, considering their respective disease burden, demographics, and treatment history. This virtual cohort supplemented clinical observations by simulating and comparing tissue glycogen and urine Hex4 following avalglucosidase alfa treatment vs. the standard of care. The digital twin analysis supports the interpretation that the enhanced reduction in urine Hex4 observed following avalglucosidase alfa treatment is attributable to greater tissue glycogen clearance. Overall, this study provides mechanism-based insight into avalglucosidase alfa efficacy across the phenotypic spectrum of Pompe disease and demonstrates the value of applying a QSP-based digital twin analysis to support rare disease drug development.
Collapse
Affiliation(s)
- Chanchala Kaddi
- Translational Disease Modeling, Translational Medicine and Early Development, Sanofi, Cambridge, Massachusetts, USA
| | - Mengdi Tao
- Translational Disease Modeling, Translational Medicine and Early Development, Sanofi, Bridgewater, New Jersey, USA
| | - Silke Bergeler
- Certara Applied Biosimulation, Lawrenceville, New Jersey, USA
| | - Kelly George
- Global Medical Affairs, Sanofi, Cambridge, Massachusetts, USA
| | - Hugo Geerts
- Certara Applied Biosimulation, Princeton, New Jersey, USA
| | | | - Julie L Batista
- Rare Disease Registries Global Medical Affairs, Sanofi, Cambridge, Massachusetts, USA
| | - Meredith Foster
- Rare Disease Registries Global Medical Affairs, Sanofi, Cambridge, Massachusetts, USA
| | | | - Atef Zaher
- Rare Diseases and Rare Blood Disorders, Clinical Development, Sanofi, Quebec City, Quebec, Canada
| | - Kristina An Haack
- Rare Diseases and Rare Blood Disorders, Clinical Development, Sanofi, Gentilly, France
| | - Susana Zaph
- Translational Disease Modeling, Translational Medicine and Early Development, Sanofi, Bridgewater, New Jersey, USA
| |
Collapse
|
5
|
Minamimoto R, Kato K, Naganawa S. Clinical scenarios of unusual FDG uptake in muscle. Jpn J Radiol 2025; 43:177-185. [PMID: 39412643 PMCID: PMC11790741 DOI: 10.1007/s11604-024-01672-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 09/26/2024] [Indexed: 02/04/2025]
Abstract
Glucose is essential for muscle function and its uptake is influenced by aerobic conditions, hormonal regulations, and exercise. 18F-Fluorodeoxyglucose (FDG), a glucose analog used in PET/CT scans, can show incidental uptake in muscles, and thus careful interpretation is required to avoid misdiagnosis. Proper patient preparation and understanding of the clinical scenarios affecting FDG uptake are crucial for accurate PET/CT interpretation, thus ensuring precise diagnoses and avoiding unnecessary interventions. This review emphasizes the need to consider patient-specific factors in evaluating incidental FDG uptake in muscle.
Collapse
Affiliation(s)
- Ryogo Minamimoto
- Department of Integrated Image Information Analysis, Nagoya University Graduate School of Medicine, 65, Tsurumaicho, Shouwa-ku, Nagoya, Aichi, 466-8550, Japan.
- Department of Radiology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan.
| | - Katsuhiko Kato
- Division of Advanced Information Health Sciences, Department of Integrated Health Sciences, Functional Medical Imaging, Biomedical Imaging Sciences, Nagoya, Aichi, Japan
| | - Shinji Naganawa
- Department of Radiology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| |
Collapse
|
6
|
Gross‐Valle C, Jacobs TC, Dijck‐Brouwer JDA, Lubberts J, Bakker BM, Bakker SJL, van der Veen Y, Schreuder AB, Derks TGJ, van der Krogt J, Groen J, Heiner‐Fokkema MR. The relation between dietary polysaccharide intake and urinary excretion of tetraglucoside. J Inherit Metab Dis 2025; 48:e12801. [PMID: 39460557 PMCID: PMC11668232 DOI: 10.1002/jimd.12801] [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: 05/05/2024] [Revised: 09/12/2024] [Accepted: 09/16/2024] [Indexed: 10/28/2024]
Abstract
The urinary metabolite tetraglucoside (Glc4) is a potential biomarker for hepatic glycogen storage diseases (GSDs). Glc4 is believed to reflect body glycogen content and/or turnover. However, dietary polysaccharide intake may influence Glc4 excretion, potentially limiting the utility of Glc4 as a monitoring biomarker in hepatic GSDs. We aimed to investigate the association of dietary polysaccharide intake with Glc4 excretion. Urinary Glc4 excretion (mmol/mmol creatinine and mmol/24 h) was analyzed using a validated LC-MS/MS method. Data was analyzed from 65 kidney transplant recipients and 58 healthy kidney donors in the TransplantLines cohort study. Spearman's correlation and multivariable linear regression analyses were performed. In the multivariable analysis, dry lean body mass (DLBM), dietary polysaccharide intake, transplantation status, age, sex, and glycated hemoglobin (HbA1c) served as independent variables. Daily variation was examined in 21 healthy individuals of urinary Glc4 excretion in 2-h collections over a 24-h period. Mixed generalized additive models were built to study the association of prior polysaccharide intake with Glc4 excretion. No (univariate) associations were found between polysaccharide intake and Glc4 excretion. However, a significant interaction between DLBM and polysaccharide on 24 h Glc4 excretion was observed in the multivariate analysis. Glc4 excretion throughout the day exhibited no relationship to prior polysaccharide intake. Our findings suggest an indirect effect of polysaccharide intake on Glc4 excretion, potentially due to changes in muscle glycogen content and/or turnover. We have found no evidence that dietary polysaccharides under normal intakes increase urinary Glc4 directly.
Collapse
Affiliation(s)
- Candelas Gross‐Valle
- Department of Laboratory Medicine, Laboratory of Metabolic DiseasesUniversity of Groningen, University Medical Center GroningenGroningenThe Netherlands
| | - Tessa C. Jacobs
- Department of Laboratory Medicine, Laboratory of Metabolic DiseasesUniversity of Groningen, University Medical Center GroningenGroningenThe Netherlands
| | - Janneke D. A. Dijck‐Brouwer
- Department of Laboratory MedicineUniversity of Groningen, University Medical Center GroningenGroningenThe Netherlands
| | - Janniek Lubberts
- Department of Laboratory MedicineUniversity of Groningen, University Medical Center GroningenGroningenThe Netherlands
| | - Barbara M. Bakker
- Laboratory of PediatricsUniversity of Groningen, University Medical Center GroningenGroningenThe Netherlands
| | - Stephan J. L. Bakker
- Department of Internal MedicineUniversity of Groningen, University Medical Center GroningenGroningenThe Netherlands
| | - Yvonne van der Veen
- Department of Internal MedicineUniversity of Groningen, University Medical Center GroningenGroningenThe Netherlands
| | - Andrea B. Schreuder
- Section of Metabolic Diseases, Beatrix Children's HospitalUniversity of Groningen, University Medical Center GroningenGroningenThe Netherlands
| | - Terry G. J. Derks
- Section of Metabolic Diseases, Beatrix Children's HospitalUniversity of Groningen, University Medical Center GroningenGroningenThe Netherlands
| | - Jennifer van der Krogt
- Department of Laboratory Medicine, Laboratory of Metabolic DiseasesUniversity of Groningen, University Medical Center GroningenGroningenThe Netherlands
| | - Joost Groen
- Department of Laboratory Medicine, Laboratory of Metabolic DiseasesUniversity of Groningen, University Medical Center GroningenGroningenThe Netherlands
| | - M. Rebecca Heiner‐Fokkema
- Department of Laboratory Medicine, Laboratory of Metabolic DiseasesUniversity of Groningen, University Medical Center GroningenGroningenThe Netherlands
| |
Collapse
|
7
|
Zhang L, Liu X, Hu J, Quan H, Lee SK, Korivi M, Wang L, Li T, Li W. Aerobic exercise attenuates high-fat diet-induced glycometabolism impairments in skeletal muscle of rat: role of EGR-1/PTP1B signaling pathway. Nutr Metab (Lond) 2024; 21:113. [PMID: 39741281 DOI: 10.1186/s12986-024-00888-8] [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: 08/31/2024] [Accepted: 12/15/2024] [Indexed: 01/02/2025] Open
Abstract
OBJECTIVE Impaired skeletal muscle glycogen synthesis contributes to insulin resistance (IR). Aerobic exercise reported to ameliorate IR by augmenting insulin signaling, however the detailed mechanism behind this improvement remains unclear. This study investigated whether aerobic exercise enhances glycogen anabolism and insulin sensitivity via EGR-1/PTP1B signaling pathway in skeletal muscle of rats. METHODS Sprague-Dawley rats fed a high-fat diet (HFD), and performed treadmill exercise training for 6-week. Oral glucose tolerance test was conducted to confirm the IR. Periodic Acid-Schiff (PAS) staining and anthrone colorimetry were used to assess the skeletal muscle glycogen. RT-qPCR, western blot, and immunofluorescence were used to detect the EGR-1/PTP1B pathway and associated signaling molecules. RESULTS We found that exercise training significantly decreased blood glucose, insulin, and homeostasis model assessment for IR (HOMA-IR) against HFD-induced elevation. Decreased muscle glycogen content due to HFD was significantly restored after exercise training. Exercise training promoted mRNA expressions of Irs1, Akt, and Glut4, while inhibited Gsk-3β expression against HFD. Next, the decreased IRS1 (phosphorylated/total), AKT (phosphorylated/total), and GLUT4, and increased GSK-3β proteins with HFD were significantly reversed by exercise. Furthermore, HFD-induced overexpression of EGR-1 and PTP1B evidenced by mRNA, protein, and immunofluorescence intensity, were substantially inhibited by exercise, which may contribute to promote insulin sensitivity and glycogen anabolism. CONCLUSIONS Aerobic exercise training promotes insulin sensitivity and skeletal muscle glycogen synthesis in HFD-fed rats. The beneficial effects of exercise might be mediated by EGR-1/PTP1B signaling pathway in skeletal muscle, however further studies are necessary to confirm this mechanism.
Collapse
Affiliation(s)
- Liangzhi Zhang
- College of Physical Education and Health Sciences, Zhejiang Normal University, 688 Yingbin Road, Jinhua, 321004, Zhejiang Province, China
| | - Xiaojie Liu
- College of Physical Education and Health Sciences, Zhejiang Normal University, 688 Yingbin Road, Jinhua, 321004, Zhejiang Province, China
| | - Jing Hu
- Department of Clinical Medicine, Medical College, Jinhua University of Vocational Technology, Jinhua, Zhejiang, China
| | - Helong Quan
- Exercise Capacity Assessment and Promotion Research Center, School of Physical Education, Northeast Normal University, Changchun, Jilin, China
| | - Sang Ki Lee
- Department of Sport Science, College of Natural Science, Chungnam National University, Deajeon, Korea
| | - Mallikarjuna Korivi
- College of Physical Education and Health Sciences, Zhejiang Normal University, 688 Yingbin Road, Jinhua, 321004, Zhejiang Province, China
| | - Lifeng Wang
- College of Physical Education and Health Sciences, Zhejiang Normal University, 688 Yingbin Road, Jinhua, 321004, Zhejiang Province, China
| | - Ting Li
- College of Physical Education and Health Sciences, Zhejiang Normal University, 688 Yingbin Road, Jinhua, 321004, Zhejiang Province, China.
| | - Wei Li
- College of Physical Education and Health Sciences, Zhejiang Normal University, 688 Yingbin Road, Jinhua, 321004, Zhejiang Province, China.
| |
Collapse
|
8
|
Januario E, Barakat A, Rajsundar A, Fatima Z, Nanda Palienkar V, Bullapur AV, Singh Brar S, Kharel P, Koyappathodi Machingal MM, Backosh A. A Comprehensive Review of Pathophysiological Link Between Non-alcoholic Fatty Liver Disease, Insulin Resistance, and Metabolic Syndrome. Cureus 2024; 16:e75677. [PMID: 39807459 PMCID: PMC11725408 DOI: 10.7759/cureus.75677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/13/2024] [Indexed: 01/16/2025] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a chronic condition characterized by hepatic steatosis in the absence of significant alcohol consumption and is increasingly recognized as the hepatic manifestation of metabolic syndrome (MetS). This review aims to explore the molecular mechanisms underlying the interaction between NAFLD, insulin resistance (IR), and MetS, with a focus on identifying therapeutic targets. A comprehensive review of existing literature on NAFLD, IR, and MetS was conducted. The review indicates that IR contributes to hepatic lipid accumulation through increased lipolysis, elevated free fatty acid flux, and impaired fatty acid oxidation, while MetS exacerbates the condition by promoting visceral adiposity, chronic inflammation, and impaired lipid metabolism. Additionally, dysbiosis and increased intestinal permeability in the gut-liver axis worsen IR, leading to a vicious cycle of metabolic dysfunction. In conclusion, addressing these interconnected pathways could enhance therapeutic strategies and reduce the burden of NAFLD-related complications.
Collapse
Affiliation(s)
| | - Aly Barakat
- Internal Medicine, Medway NHS Foundation Trust, Gillingham, GBR
| | | | - Zahra Fatima
- Medicine, Dr. VRK Women's Medical College, Aziznagar, IND
| | | | | | | | - Punam Kharel
- Medicine, Sir Salimullah Medical College, Dhaka, BGD
| | | | - Amena Backosh
- Orthopedics, Medway Maritime Hospital, Gillingham, GBR
| |
Collapse
|
9
|
Yadav M, Verma S, Tiwari P, Mugale MN. Unraveling the mechanisms of hepatogenous diabetes and its therapeutic perspectives. Life Sci 2024; 353:122934. [PMID: 39089644 DOI: 10.1016/j.lfs.2024.122934] [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: 05/09/2024] [Revised: 06/26/2024] [Accepted: 07/25/2024] [Indexed: 08/04/2024]
Abstract
The review focused mainly on the pathogenesis of hepatogenous diabetes (HD) in liver cirrhosis (LC). This review reveals parallels between the mechanisms of metabolic dysfunction observed in LC and type II diabetes (T2DM), suggesting a shared pathway leading to HD. It underscores the role of insulin in HD pathogenesis, highlighting key factors such as insulin signaling, glucose metabolism, insulin resistance (IR), and the influence of adipocytes. Furthermore, the impact of adipose tissue accumulation, fatty acid metabolism, and pro-inflammatory cytokines like Tumor necrosis factor-α (TNF-α) on IR are discussed in the context of HD. Altered signaling pathways, disruptions in the endocrine system, liver inflammation, changes in muscle mass and composition, and modifications to the gut microbiota collectively contribute to the complex interplay linking cirrhosis and HD. This study highlights how important it is to identify and treat this complex condition in cirrhotic patients by thoroughly analyzing the link between cirrhosis, IR, and HD. It also emphasizes the vitality of targeted interventions. Cellular and molecular investigations into IR have revealed potential therapeutic targets for managing and preventing HD.
Collapse
Affiliation(s)
- Manisha Yadav
- Division of Toxicology and Experimental Medicine, CSIR-Central Drug Research Institute (CSIR-CDRI), Lucknow 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Smriti Verma
- Division of Toxicology and Experimental Medicine, CSIR-Central Drug Research Institute (CSIR-CDRI), Lucknow 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Purnima Tiwari
- Division of Toxicology and Experimental Medicine, CSIR-Central Drug Research Institute (CSIR-CDRI), Lucknow 226031, India
| | - Madhav Nilakanth Mugale
- Division of Toxicology and Experimental Medicine, CSIR-Central Drug Research Institute (CSIR-CDRI), Lucknow 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| |
Collapse
|
10
|
Mäkinen S, Sree S, Ala-Nisula T, Kultalahti H, Koivunen P, Koistinen HA. Activation of the hypoxia-inducible factor pathway by roxadustat improves glucose metabolism in human primary myotubes from men. Diabetologia 2024; 67:1943-1954. [PMID: 38814443 PMCID: PMC11410918 DOI: 10.1007/s00125-024-06185-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Accepted: 04/19/2024] [Indexed: 05/31/2024]
Abstract
AIMS/HYPOTHESIS Hypoxia-inducible factor prolyl 4-hydroxylase (HIF-P4H) enzymes regulate adaptive cellular responses to low oxygen concentrations. Inhibition of HIF-P4Hs leads to stabilisation of hypoxia-inducible factors (HIFs) and activation of the HIF pathway affecting multiple biological processes to rescue cells from hypoxia. As evidence from animal models suggests that HIF-P4H inhibitors could be used to treat metabolic disorders associated with insulin resistance, we examined whether roxadustat, an HIF-P4H inhibitor approved for the treatment of renal anaemia, would have an effect on glucose metabolism in primary human myotubes. METHODS Primary skeletal muscle cell cultures, established from biopsies of vastus lateralis muscle from men with normal glucose tolerance (NGT) (n=5) or type 2 diabetes (n=8), were treated with roxadustat. Induction of HIF target gene expression was detected with quantitative real-time PCR. Glucose uptake and glycogen synthesis were investigated with radioactive tracers. Glycolysis and mitochondrial respiration rates were measured with a Seahorse analyser. RESULTS Exposure to roxadustat stabilised nuclear HIF1α protein expression in human myotubes. Treatment with roxadustat led to induction of HIF target gene mRNAs for GLUT1 (also known as SLC2A1), HK2, MCT4 (also known as SLC16A4) and HIF-P4H-2 (also known as PHD2 or EGLN1) in myotubes from donors with NGT, with a blunted response in myotubes from donors with type 2 diabetes. mRNAs for LDHA, PDK1 and GBE1 were induced to a similar degree in myotubes from donors with NGT or type 2 diabetes. Exposure of myotubes to roxadustat led to a 1.4-fold increase in glycolytic rate in myotubes from men with NGT (p=0.0370) and a 1.7-fold increase in myotubes from donors with type 2 diabetes (p=0.0044), with no difference between the groups (p=0.1391). Exposure to roxadustat led to a reduction in basal mitochondrial respiration in both groups (p<0.01). Basal glucose uptake rates were similar in myotubes from donors with NGT (20.2 ± 2.7 pmol mg-1 min-1) and type 2 diabetes (25.3 ± 4.4 pmol mg-1 min-1, p=0.4205). Treatment with roxadustat enhanced insulin-stimulated glucose uptake in myotubes from donors with NGT (1.4-fold vs insulin-only condition, p=0.0023). The basal rate of glucose incorporation into glycogen was lower in myotubes from donors with NGT (233 ± 12.4 nmol g-1 h-1) than in myotubes from donors with type 2 diabetes (360 ± 40.3 nmol g-1 h-1, p=0.0344). Insulin increased glycogen synthesis by 1.9-fold (p=0.0025) in myotubes from donors with NGT, whereas roxadustat did not affect their basal or insulin-stimulated glycogen synthesis. Insulin increased glycogen synthesis by 1.7-fold (p=0.0031) in myotubes from donors with type 2 diabetes. While basal glycogen synthesis was unaffected by roxadustat, pretreatment with roxadustat enhanced insulin-stimulated glycogen synthesis in myotubes from donors with type 2 diabetes (p=0.0345). CONCLUSIONS/INTERPRETATION Roxadustat increases glycolysis and inhibits mitochondrial respiration in primary human myotubes regardless of diabetes status. Roxadustat may also improve insulin action on glycogen synthesis in myotubes from donors with type 2 diabetes.
Collapse
Affiliation(s)
- Selina Mäkinen
- Minerva Foundation Institute for Medical Research, Helsinki, Finland
- Department of Medicine, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Sreesha Sree
- Minerva Foundation Institute for Medical Research, Helsinki, Finland
- Department of Medicine, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Tuulia Ala-Nisula
- Biocenter Oulu, Faculty of Biochemistry and Molecular Medicine, Oulu Center for Cell-Matrix Research, University of Oulu, Oulu, Finland
| | - Henric Kultalahti
- Minerva Foundation Institute for Medical Research, Helsinki, Finland
- Department of Medicine, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Peppi Koivunen
- Biocenter Oulu, Faculty of Biochemistry and Molecular Medicine, Oulu Center for Cell-Matrix Research, University of Oulu, Oulu, Finland
| | - Heikki A Koistinen
- Minerva Foundation Institute for Medical Research, Helsinki, Finland.
- Department of Medicine, University of Helsinki and Helsinki University Hospital, Helsinki, Finland.
| |
Collapse
|
11
|
Chiang WY, Yu HW, Wu MC, Huang YM, Chen YQ, Lin JW, Liu YW, You LR, Chiou A, Kuo JC. Matrix mechanics regulates muscle regeneration by modulating kinesin-1 activity. Biomaterials 2024; 308:122551. [PMID: 38593710 DOI: 10.1016/j.biomaterials.2024.122551] [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: 07/24/2023] [Revised: 03/22/2024] [Accepted: 03/25/2024] [Indexed: 04/11/2024]
Abstract
Sarcopenia, a prevalent muscle disease characterized by muscle mass and strength reduction, is associated with impaired skeletal muscle regeneration. However, the influence of the biomechanical properties of sarcopenic skeletal muscle on the efficiency of the myogenic program remains unclear. Herein, we established a mouse model of sarcopenia and observed a reduction in stiffness within the sarcopenic skeletal muscle in vivo. To investigate whether the biomechanical properties of skeletal muscle directly impact the myogenic program, we established an in vitro system to explore the intrinsic mechanism involving matrix stiffness control of myogenic differentiation. Our findings identify the microtubule motor protein, kinesin-1, as a mechano-transduction hub that senses and responds to matrix stiffness, crucial for myogenic differentiation and muscle regeneration. Specifically, kinesin-1 activity is positively regulated by stiff matrices, facilitating its role in transporting mitochondria and enhancing translocation of the glucose transporter GLUT4 to the cell surface for glucose uptake. Conversely, the softer matrices significantly suppress kinesin-1 activity, leading to the accumulation of mitochondria around nuclei and hindering glucose uptake by inhibiting GLUT4 membrane translocation, consequently impairing myogenic differentiation. The insights gained from the in-vitro system highlight the mechano-transduction significance of kinesin-1 motor proteins in myogenic differentiation. Furthermore, our study confirms that enhancing kinesin-1 activity in the sarcopenic mouse model restores satellite cell expansion, myogenic differentiation, and muscle regeneration. Taken together, our findings provide a potential target for improving muscle regeneration in sarcopenia.
Collapse
Affiliation(s)
- Wan-Yu Chiang
- Institute of Biochemistry and Molecular Biology, National Yang Ming Chiao Tung University, Taipei, 11221, Taiwan
| | - Helen Wenshin Yu
- Institute of Biochemistry and Molecular Biology, National Yang Ming Chiao Tung University, Taipei, 11221, Taiwan
| | - Ming-Chung Wu
- Institute of Biochemistry and Molecular Biology, National Yang Ming Chiao Tung University, Taipei, 11221, Taiwan
| | - Yi-Man Huang
- Institute of Biochemistry and Molecular Biology, National Yang Ming Chiao Tung University, Taipei, 11221, Taiwan
| | - Yin-Quan Chen
- Cancer and Immunology Research Center, National Yang Ming Chiao Tung University, Taipei, 11221, Taiwan
| | - Jong-Wei Lin
- Institute of Biochemistry and Molecular Biology, National Yang Ming Chiao Tung University, Taipei, 11221, Taiwan
| | - Yen-Wenn Liu
- Institute of Biochemistry and Molecular Biology, National Yang Ming Chiao Tung University, Taipei, 11221, Taiwan
| | - Li-Ru You
- Institute of Biochemistry and Molecular Biology, National Yang Ming Chiao Tung University, Taipei, 11221, Taiwan
| | - Arthur Chiou
- Institute of Biophotonics, National Yang Ming Chiao Tung University, Taipei, 11221, Taiwan
| | - Jean-Cheng Kuo
- Institute of Biochemistry and Molecular Biology, National Yang Ming Chiao Tung University, Taipei, 11221, Taiwan; Cancer and Immunology Research Center, National Yang Ming Chiao Tung University, Taipei, 11221, Taiwan.
| |
Collapse
|
12
|
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; 16:1059-1079. [PMID: 38916734 PMCID: PMC11964417 DOI: 10.14336/ad.2024.0162] [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: 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&V 811, Boston, MA 02111, USA
| |
Collapse
|
13
|
Logesh R, Hari B, Chidambaram K, Das N. Molecular effects of Vitamin-D and PUFAs metabolism in skeletal muscle combating Type-II diabetes mellitus. Gene 2024; 904:148216. [PMID: 38307219 DOI: 10.1016/j.gene.2024.148216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Revised: 01/10/2024] [Accepted: 01/25/2024] [Indexed: 02/04/2024]
Abstract
Multiple post-receptor intracellular alterations such as impaired glucose transfer, glucose phosphorylation, decreased glucose oxidation, and glycogen production contribute to insulin resistance (IR) in skeletal muscle, manifested by diminished insulin-stimulated glucose uptake. Type-2 diabetes mellites (T2DM) has caused by IR, which is also seen in obese patients and those with metabolic syndrome. The Vitamin-D receptor (VDR) and poly unsaturated fatty acids (PUFAs) roles in skeletal muscle growth, shapes, and function for combating type-2 diabetes have been clarified throughout this research. VDR and PUFAs appears to show a variety of effects on skeletal muscle, in addition it shows a promising role on bone and mineral homeostasis. Individuals having T2DM are reported to suffer from severe muscular weakness and alterations in shape of the muscle. Several studies have investigated the effect on VDR on muscular strength and mass, which leads to Vitamin-D deficiency (VDD) in individuals, in which most commonly seen in elderly. VDR has been shown to affect skeletal cellular proliferation, intracellular calcium handling, as well as genomic activity in a variety of different ways such as muscle metabolism, insulin sensitivity, which is the major characteristic pathogenesis for IR in combating T2DM. The identified VDR gene polymorphisms are ApaI, TaqI, FokI, and BsmI that are associated with T2DM. This review collates informations on the mechanisms by which VDR activation takes place in skeletal muscles. Despite the significant breakthroughs made in recent decades, various studies show that IR affects VDR and PUFAs metabolism in skeletal muscle. Therefore, this review collates the data to show the role of VDR and PUFAs in the skeletal muscles to combat T2DM.
Collapse
Affiliation(s)
- Rajan Logesh
- Department of Pharmacognosy, JSS College of Pharmacy, Mysuru, JSS Academy of Higher Education & Research, Karnataka, India.
| | - Balaji Hari
- TIFAC CORE in Herbal Drugs, Department of Pharmacognosy, JSS Academy of Higher Education & Research, JSS College of Pharmacy, The Nilgiris, Ooty 643001, Tamil Nadu, India
| | - Kumarappan Chidambaram
- Department of Pharmacology, College of Pharmacy, King Khalid University, Al-Qara, Asir Province, Saudi Arabia
| | - Niranjan Das
- Department of Chemistry, Iswar Chandra Vidyasagar College, Belonia 799155, Tripura, India
| |
Collapse
|
14
|
Pearce B, Pearce K. Mitochondrial dysfunction and diabetes in South Africa: A review. ENDOCRINE AND METABOLIC SCIENCE 2024; 14:100157. [DOI: 10.1016/j.endmts.2024.100157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2025] Open
|
15
|
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
|
16
|
Szablewski L. Changes in Cells Associated with Insulin Resistance. Int J Mol Sci 2024; 25:2397. [PMID: 38397072 PMCID: PMC10889819 DOI: 10.3390/ijms25042397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Revised: 02/10/2024] [Accepted: 02/14/2024] [Indexed: 02/25/2024] Open
Abstract
Insulin is a polypeptide hormone synthesized and secreted by pancreatic β-cells. It plays an important role as a metabolic hormone. Insulin influences the metabolism of glucose, regulating plasma glucose levels and stimulating glucose storage in organs such as the liver, muscles and adipose tissue. It is involved in fat metabolism, increasing the storage of triglycerides and decreasing lipolysis. Ketone body metabolism also depends on insulin action, as insulin reduces ketone body concentrations and influences protein metabolism. It increases nitrogen retention, facilitates the transport of amino acids into cells and increases the synthesis of proteins. Insulin also inhibits protein breakdown and is involved in cellular growth and proliferation. On the other hand, defects in the intracellular signaling pathways of insulin may cause several disturbances in human metabolism, resulting in several chronic diseases. Insulin resistance, also known as impaired insulin sensitivity, is due to the decreased reaction of insulin signaling for glucose levels, seen when glucose use in response to an adequate concentration of insulin is impaired. Insulin resistance may cause, for example, increased plasma insulin levels. That state, called hyperinsulinemia, impairs metabolic processes and is observed in patients with type 2 diabetes mellitus and obesity. Hyperinsulinemia may increase the risk of initiation, progression and metastasis of several cancers and may cause poor cancer outcomes. Insulin resistance is a health problem worldwide; therefore, mechanisms of insulin resistance, causes and types of insulin resistance and strategies against insulin resistance are described in this review. Attention is also paid to factors that are associated with the development of insulin resistance, the main and characteristic symptoms of particular syndromes, plus other aspects of severe insulin resistance. This review mainly focuses on the description and analysis of changes in cells due to insulin resistance.
Collapse
Affiliation(s)
- Leszek Szablewski
- Chair and Department of General Biology and Parasitology, Medical University of Warsaw, Chałubińskiego Str. 5, 02-004 Warsaw, Poland
| |
Collapse
|
17
|
Kal S, Mahata S, Jati S, Mahata SK. Mitochondrial-derived peptides: Antidiabetic functions and evolutionary perspectives. Peptides 2024; 172:171147. [PMID: 38160808 PMCID: PMC10838678 DOI: 10.1016/j.peptides.2023.171147] [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: 11/28/2023] [Revised: 12/27/2023] [Accepted: 12/27/2023] [Indexed: 01/03/2024]
Abstract
Mitochondrial-derived peptides (MDPs) are a novel class of bioactive microproteins encoded by short open-reading frames (sORF) in mitochondrial DNA (mtDNA). Currently, three types of MDPs have been identified: Humanin (HN), MOTS-c (Mitochondrial ORF within Twelve S rRNA type-c), and SHLP1-6 (small Humanin-like peptide, 1 to 6). The 12 S ribosomal RNA (MT-RNR1) gene harbors the sequence for MOTS-c, whereas HN and SHLP1-6 are encoded by the 16 S ribosomal RNA (MT-RNR2) gene. Special genetic codes are used in mtDNA as compared to nuclear DNA: (i) ATA and ATT are used as start codons in addition to the standard start codon ATG; (ii) AGA and AGG are used as stop codons instead of coding for arginine; (iii) the standard stop codon UGA is used to code for tryptophan. While HN, SHLP6, and MOTS-c are encoded by the H (heavy owing to high guanine + thymine base composition)-strand of the mtDNA, SHLP1-5 are encoded by the L (light owing to less guanine + thymine base composition)-strand. MDPs attenuate disease pathology including Type 1 diabetes (T1D), Type 2 diabetes (T2D), gestational diabetes, Alzheimer's disease (AD), cardiovascular diseases, prostate cancer, and macular degeneration. The current review will focus on the MDP regulation of T2D, T1D, and gestational diabetes along with an emphasis on the evolutionary pressures for conservation of the amino acid sequences of MDPs.
Collapse
Affiliation(s)
- Satadeepa Kal
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Sumana Mahata
- Department of Anesthesiology, Riverside University Health System, Moreno Valley, CA, USA
| | - Suborno Jati
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, CA, USA
| | - Sushil K Mahata
- Department of Medicine, University of California San Diego, La Jolla, CA, USA; VA San Diego Healthcare System, San Diego, CA, USA.
| |
Collapse
|
18
|
Yazıcı D, Demir SÇ, Sezer H. Insulin Resistance, Obesity, and Lipotoxicity. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1460:391-430. [PMID: 39287860 DOI: 10.1007/978-3-031-63657-8_14] [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
Lipotoxicity, originally used to describe the destructive effects of excess fat accumulation on glucose metabolism, causes functional impairments in several metabolic pathways, both in adipose tissue and peripheral organs, like liver, heart, pancreas, and muscle. Ectopic lipid accumulation in the kidneys, liver, and heart has important clinical counterparts like diabetic nephropathy in type 2 diabetes mellitus, obesity-related glomerulopathy, nonalcoholic fatty liver disease, and cardiomyopathy. Insulin resistance due to lipotoxicity indirectly lead to reproductive system disorders, like polycystic ovary syndrome. Lipotoxicity has roles in insulin resistance and pancreatic beta-cell dysfunction. Increased circulating levels of lipids and the metabolic alterations in fatty acid utilization and intracellular signaling have been related to insulin resistance in muscle and liver. Different pathways, like novel protein kinase c pathways and the JNK-1 pathway, are involved as the mechanisms of how lipotoxicity leads to insulin resistance in nonadipose tissue organs, such as liver and muscle. Mitochondrial dysfunction plays a role in the pathogenesis of insulin resistance. Endoplasmic reticulum stress, through mainly increased oxidative stress, also plays an important role in the etiology of insulin resistance, especially seen in non-alcoholic fatty liver disease. Visceral adiposity and insulin resistance both increase the cardiometabolic risk, and lipotoxicity seems to play a crucial role in the pathophysiology of these associations.
Collapse
Affiliation(s)
- Dilek Yazıcı
- Koç University Medical School, Section of Endocrinology and Metabolism, Koç University Hospital, Topkapi, Istanbul, Turkey.
| | - Selin Çakmak Demir
- Koç University Medical School, Section of Endocrinology and Metabolism, Koç University Hospital, Topkapi, Istanbul, Turkey
| | - Havva Sezer
- Koç University Medical School, Section of Endocrinology and Metabolism, Koç University Hospital, Topkapi, Istanbul, Turkey
| |
Collapse
|
19
|
Li R, He M, Yang Q, Liang Z, Li Y, Huang L, Wu R, Huang J. Association between serum creatinine and type 2 diabetes in the Chinese population: a retrospective cohort study. Sci Rep 2023; 13:6806. [PMID: 37100791 PMCID: PMC10133309 DOI: 10.1038/s41598-023-33878-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Accepted: 04/20/2023] [Indexed: 04/28/2023] Open
Abstract
The relationship between serum creatinine and type 2 diabetes is limited. We aimed to investigate the association of baseline serum creatinine and new-onset type 2 diabetes in Chinese population. This retrospective cohort study was conducted using data from the health screening program in China. The population were divided into four groups based on serum creatinine levels, and the outcome of interest was the occurrence of a diabetic event. Cox proportional risk model was used to assess the independent effect of baseline serum creatinine level on future diabetes risk. Sensitivity and subgroup analysis were used to verify the reliability of the results. After an average follow-up of 3.12 years, among 201,298 individuals aged ≥ 20 years, 3389 patients developed diabetes. Compared with participants in quartile 2-4 (> 51.6umol/L for female, > 71.8umol/L for male,), a significantly higher risk of new-onset Type 2 Diabetes (OR, 1.15; 95%CI: 1.07-1.23) was found in those in quartile 1 (< 51.6umol/L for female, < 71.8umol/L for male). Moreover, Similar results were found in various subgroups stratified by age, BMI, TG, TC, FPG and family history group. Low serum creatinine is independently associated with increased risk of type 2 diabetes in Chinese adults. It was also stable in various subgroups stratified.
Collapse
Affiliation(s)
- Rugang Li
- Department of Nephrology, Yuebei People's Hospital, No. 133 South Huimin Road, Shaoguan, 512026, Guangdong, China
| | - Min He
- Department of Nephrology, Yuebei People's Hospital, No. 133 South Huimin Road, Shaoguan, 512026, Guangdong, China
| | - Qilin Yang
- Department of Critical Care, the Second Affiliated Hospital of Guangzhou Medical University, No. 250 Changgang East Road, Haizhu District, Guangzhou, Guangdong, China
| | - Zezhi Liang
- Department of Nephrology, Yuebei People's Hospital, No. 133 South Huimin Road, Shaoguan, 512026, Guangdong, China
| | - Ying Li
- Department of Nephrology, Yuebei People's Hospital, No. 133 South Huimin Road, Shaoguan, 512026, Guangdong, China
| | - Ling Huang
- Department of Nephrology, Yuebei People's Hospital, No. 133 South Huimin Road, Shaoguan, 512026, Guangdong, China
| | - Rong Wu
- Department of Cardiology, Chenzhou Third People's Hospital, Chenzhou, 423000, Hunan, China
| | - Jieping Huang
- Department of Nephrology, Yuebei People's Hospital, No. 133 South Huimin Road, Shaoguan, 512026, Guangdong, China.
| |
Collapse
|
20
|
Cea LA, Vásquez W, Hernández-Salinas R, Vielma AZ, Castillo-Ruiz M, Velarde V, Salgado M, Sáez JC. Skeletal Muscle Atrophy Induced by Diabetes Is Mediated by Non-Selective Channels and Prevented by Boldine. Biomolecules 2023; 13:708. [PMID: 37189454 PMCID: PMC10136059 DOI: 10.3390/biom13040708] [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: 01/31/2023] [Revised: 03/23/2023] [Accepted: 04/04/2023] [Indexed: 05/17/2023] Open
Abstract
Individuals with diabetes mellitus present a skeletal muscle myopathy characterized by atrophy. However, the mechanism underlying this muscular alteration remains elusive, which makes it difficult to design a rational treatment that could avoid the negative consequences in muscles due to diabetes. In the present work, the atrophy of skeletal myofibers from streptozotocin-induced diabetic rats was prevented with boldine, suggesting that non-selective channels inhibited by this alkaloid are involved in this process, as has previously shown for other muscular pathologies. Accordingly, we found a relevant increase in sarcolemma permeability of skeletal myofibers of diabetic animals in vivo and in vitro due to de novo expression of functional connexin hemichannels (Cx HCs) containing connexins (Cxs) 39, 43, and 45. These cells also expressed P2X7 receptors, and their inhibition in vitro drastically reduced sarcolemma permeability, suggesting their participation in the activation of Cx HCs. Notably, sarcolemma permeability of skeletal myofibers was prevented by boldine treatment that blocks Cx43 and Cx45 HCs, and now we demonstrated that it also blocks P2X7 receptors. In addition, the skeletal muscle alterations described above were not observed in diabetic mice with myofibers deficient in Cx43/Cx45 expression. Moreover, murine myofibers cultured for 24 h in high glucose presented a drastic increase in sarcolemma permeability and levels of NLRP3, a molecular member of the inflammasome, a response that was also prevented by boldine, suggesting that, in addition to the systemic inflammatory response found in diabetes, high glucose can promote the expression of functional Cx HCs and activation of the inflammasome in skeletal myofibers. Therefore, Cx43 and Cx45 HCs play a critical role in myofiber degeneration, and boldine could be considered a potential therapeutic agent to treat muscular complications due to diabetes.
Collapse
Affiliation(s)
- Luis A. Cea
- Instituto de Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Av. Llano Subercaseaux 2801, San Miguel, Santiago 8910060, Chile
| | - Walter Vásquez
- Departamento de Fisiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile
| | - Romina Hernández-Salinas
- Centro de Fisiología Celular e Integrativa, Facultad de Medicina, Universidad del Desarrollo, Santiago 7610000, Chile
| | - Alejandra Z. Vielma
- Escuela de Odontología, Facultad de Medicina Clínica Alemana, Universidad del Desarrollo, Santiago 7610000, Chile
| | - Mario Castillo-Ruiz
- Escuela de Química y Farmacia, Facultad de Medicina, Universidad Andres Bello, Santiago 8370149, Chile
- Departamento de Ciencias Químicas y Biológicas, Facultad de Ciencias de la Salud, Universidad Bernardo O’Higgins, Santiago 8370854, Chile
| | - Victoria Velarde
- Instituto de Fisiología, Universidad de Valparaíso, Valparaíso 2340000, Chile
| | - Magdiel Salgado
- Instituto de Neurociencias, Centro Interdisciplinario de Neurociencias de Valparaíso, Universidad de Valparaíso, Pasaje Harrington 287, Playa Ancha, Valparaíso 2340000, Chile
| | - Juan C. Sáez
- Instituto de Neurociencias, Centro Interdisciplinario de Neurociencias de Valparaíso, Universidad de Valparaíso, Pasaje Harrington 287, Playa Ancha, Valparaíso 2340000, Chile
| |
Collapse
|
21
|
Youxiang C, Lin Z, Zekai C, Weijun X. Resting and exercise metabolic characteristics in obese children with insulin resistance. Front Physiol 2022; 13:1049560. [DOI: 10.3389/fphys.2022.1049560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 11/22/2022] [Indexed: 12/05/2022] Open
Abstract
Purpose: This study aimed to explore the characteristics of resting energy expenditure (REE) and lipid metabolism during incremental load exercise in obese children and adolescents with insulin resistance (IR) to provide evidence for exercise intervention in obese children and adolescents with IR.Method: From July 2019 to August 2021, 195 obese children and adolescents aged 13–17 were recruited through a summer camp. The participants were divided into IR (n = 67) and no-IR (without insulin resistance, n = 128) groups and underwent morphology, blood indicators, body composition, and resting energy consumption gas metabolism tests. Thirty participants each were randomly selected from the IR and no-IR groups to carry out the incremental treadmill test.Results: Significant metabolic differences in resting and exercise duration were found between the IR and no-IR groups. In the resting state, the resting metabolic equivalents (4.33 ± 0.94 ml/min/kg vs. 3.91 ± 0.73 ml/min/kg, p = 0.001) and REE (2464.03 ± 462.29 kcal/d vs. 2143.88 ± 380.07 kcal/d, p < 0.001) in the IR group were significantly higher than in the no-IR group. During exercise, the absolute maximal fat oxidation (0.33 ± 0.07 g/min vs. 0.36 ± 0.09 g/min, p = 0.002) in the IR group was significantly lower than in the no-IR group; maximal fat oxidation intensity (130.9 ± 8.9 bpm vs. 139.9 ± 7.4 bpm, p = 0.040) was significantly lower in the IR group.Conclusion: Significant resting and exercise metabolic differences were found between obese IR and no-IR children and adolescents. Obese IR children and adolescents have higher REE and lower maximal fat oxidation intensity than obese no-IR children and adolescents.
Collapse
|
22
|
Bielczyk-Maczynska E, Zhao M, Zushin PJH, Schnurr TM, Kim HJ, Li J, Nallagatla P, Sangwung P, Park CY, Cornn C, Stahl A, Svensson KJ, Knowles JW. G protein-coupled receptor 151 regulates glucose metabolism and hepatic gluconeogenesis. Nat Commun 2022; 13:7408. [PMID: 36456565 PMCID: PMC9715671 DOI: 10.1038/s41467-022-35069-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 11/16/2022] [Indexed: 12/03/2022] Open
Abstract
Human genetics has been instrumental in identification of genetic variants linked to type 2 diabetes. Recently a rare, putative loss-of-function mutation in the orphan G-protein coupled receptor 151 (GPR151) was found to be associated with lower odds ratio for type 2 diabetes, but the mechanism behind this association has remained elusive. Here we show that Gpr151 is a fasting- and glucagon-responsive hepatic gene which regulates hepatic gluconeogenesis. Gpr151 ablation in mice leads to suppression of hepatic gluconeogenesis genes and reduced hepatic glucose production in response to pyruvate. Importantly, the restoration of hepatic Gpr151 levels in the Gpr151 knockout mice reverses the reduced hepatic glucose production. In this work, we establish a previously unknown role of Gpr151 in the liver that provides an explanation to the lowered type 2 diabetes risk in individuals with nonsynonymous mutations in GPR151.
Collapse
Affiliation(s)
- Ewa Bielczyk-Maczynska
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA.
- Stanford Diabetes Research Center, Stanford University School of Medicine, Stanford, CA, USA.
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA.
| | - Meng Zhao
- Stanford Diabetes Research Center, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Peter-James H Zushin
- Department of Nutritional Sciences and Toxicology, University of California at Berkeley, Berkeley, CA, USA
| | - Theresia M Schnurr
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Diabetes Research Center, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Hyun-Jung Kim
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Jiehan Li
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Diabetes Research Center, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Pratima Nallagatla
- Genetics Bioinformatics Service Center, Stanford University School of Medicine, Stanford, CA, USA
| | - Panjamaporn Sangwung
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Diabetes Research Center, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Chong Y Park
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Diabetes Research Center, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Cameron Cornn
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Andreas Stahl
- Department of Nutritional Sciences and Toxicology, University of California at Berkeley, Berkeley, CA, USA
| | - Katrin J Svensson
- Stanford Diabetes Research Center, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Joshua W Knowles
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA.
- Stanford Diabetes Research Center, Stanford University School of Medicine, Stanford, CA, USA.
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA.
- Stanford Prevention Research Center, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
23
|
Inflammaging: Implications in Sarcopenia. Int J Mol Sci 2022; 23:ijms232315039. [PMID: 36499366 PMCID: PMC9740553 DOI: 10.3390/ijms232315039] [Citation(s) in RCA: 80] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 11/25/2022] [Accepted: 11/28/2022] [Indexed: 12/02/2022] Open
Abstract
In a world in which life expectancy is increasing, understanding and promoting healthy aging becomes a contemporary demand. In the elderly, a sterile, chronic and low-grade systemic inflammation known as "inflammaging" is linked with many age-associated diseases. Considering sarcopenia as a loss of strength and mass of skeletal muscle related to aging, correlations between these two terms have been proposed. Better knowledge of the immune system players in skeletal muscle would help to elucidate their implications in sarcopenia. Characterizing the activators of damage sensors and the downstream effectors explains the inference with skeletal muscle performance. Sarcopenia has also been linked to chronic diseases such as diabetes, metabolic syndrome and obesity. Implications of inflammatory signals from these diseases negatively affect skeletal muscle. Autophagic mechanisms are closely related with the inflammasome, as autophagy eliminates stress signaling sent by damage organelles, but also acts with an immunomodulatory function affecting immune cells and cytokine release. The use of melatonin, an antioxidant, ROS scavenger and immune and autophagy modulator, or senotherapeutic compounds targeting senescent cells could represent strategies to counteract inflammation. This review aims to present the many factors regulating skeletal muscle inflammaging and their major implications in order to understand the molecular mechanisms involved in sarcopenia.
Collapse
|
24
|
Nieblas B, Pérez-Treviño P, García N. Role of mitochondria-associated endoplasmic reticulum membranes in insulin sensitivity, energy metabolism, and contraction of skeletal muscle. Front Mol Biosci 2022; 9:959844. [PMID: 36275635 PMCID: PMC9585326 DOI: 10.3389/fmolb.2022.959844] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 08/04/2022] [Indexed: 11/29/2022] Open
Abstract
Skeletal muscle has a critical role in the regulation of the energy balance of the organism, particularly as the principal tissue responsible for insulin-stimulated glucose disposal and as the major site of peripheral insulin resistance (IR), which has been related to accumulation of lipid intermediates, reduced oxidative capacity of mitochondria and endoplasmic reticulum (ER) stress. These organelles form contact sites, known as mitochondria-associated ER membranes (MAMs). This interconnection seems to be involved in various cellular processes, including Ca2+ transport and energy metabolism; therefore, MAMs could play an important role in maintaining cellular homeostasis. Evidence suggests that alterations in MAMs may contribute to IR. However, the evidence does not refer to a specific subcellular location, which is of interest due to the fact that skeletal muscle is constituted by oxidative and glycolytic fibers as well as different mitochondrial populations that appear to respond differently to stimuli and pathological conditions. In this review, we show the available evidence of possible differential responses in the formation of MAMs in skeletal muscle as well as its role in insulin signaling and the beneficial effect it could have in the regulation of energetic metabolism and muscular contraction.
Collapse
Affiliation(s)
- Bianca Nieblas
- Escuela de Medicina y Ciencias de la Salud, Tecnologico de Monterrey, Monterrey, Nuevo León, México
- Experimental Medicine and Advanced Therapies, The Institute for Obesity Research, Tecnologico de Monterrey, Monterrey, Nuevo León, México
| | - Perla Pérez-Treviño
- Experimental Medicine and Advanced Therapies, The Institute for Obesity Research, Tecnologico de Monterrey, Monterrey, Nuevo León, México
| | - Noemí García
- Escuela de Medicina y Ciencias de la Salud, Tecnologico de Monterrey, Monterrey, Nuevo León, México
- Experimental Medicine and Advanced Therapies, The Institute for Obesity Research, Tecnologico de Monterrey, Monterrey, Nuevo León, México
- *Correspondence: Noemí García,
| |
Collapse
|
25
|
Verlande A, Chun SK, Song WA, Oettler D, Knot HJ, Masri S. Exogenous detection of 13C-glucose metabolism in tumor and diet-induced obesity models. Front Physiol 2022; 13:1023614. [PMID: 36277179 PMCID: PMC9581140 DOI: 10.3389/fphys.2022.1023614] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Accepted: 09/20/2022] [Indexed: 11/13/2022] Open
Abstract
Metabolic rewiring is a hallmark feature prevalent in cancer cells as well as insulin resistance (IR) associated with diet-induced obesity (DIO). For instance, tumor metabolism shifts towards an enhanced glycolytic state even under aerobic conditions. In contrast, DIO triggers lipid-induced IR by impairing insulin signaling and reducing insulin-stimulated glucose uptake. Based on physiological differences in systemic metabolism, we used a breath analysis approach to discriminate between different pathological states using glucose oxidation as a readout. We assessed glucose utilization in lung cancer-induced cachexia and DIO mouse models using a U-13C glucose tracer and stable isotope sensors integrated into an indirect calorimetry system. Our data showed increased 13CO2 expired by tumor-bearing (TB) mice and a reduction in exhaled 13CO2 in the DIO model. Taken together, our findings illustrate high glucose uptake and consumption in TB animals and decreased glucose uptake and oxidation in obese mice with an IR phenotype. Our work has important translational implications for the utility of stable isotopes in breath-based detection of glucose homeostasis in models of lung cancer progression and DIO.
Collapse
Affiliation(s)
- Amandine Verlande
- Department of Biological Chemistry, University of California, Irvine, Irvine, CA, United States
| | - Sung Kook Chun
- Department of Biological Chemistry, University of California, Irvine, Irvine, CA, United States
| | - Wei A. Song
- Department of Biological Chemistry, University of California, Irvine, Irvine, CA, United States
| | | | - Harm J. Knot
- TSE Systems Inc., Chesterfield, MO, United States
| | - Selma Masri
- Department of Biological Chemistry, University of California, Irvine, Irvine, CA, United States
| |
Collapse
|
26
|
Oxidative Stress in Type 2 Diabetes: The Case for Future Pediatric Redoxomics Studies. Antioxidants (Basel) 2022; 11:antiox11071336. [PMID: 35883827 PMCID: PMC9312244 DOI: 10.3390/antiox11071336] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 06/28/2022] [Accepted: 06/30/2022] [Indexed: 01/27/2023] Open
Abstract
Considerable evidence supports the role of oxidative stress in adult type 2 diabetes (T2D). Due to increasing rates of pediatric obesity, lack of physical activity, and consumption of excess food calories, it is projected that the number of children living with insulin resistance, prediabetes, and T2D will markedly increase with enormous worldwide economic costs. Understanding the factors contributing to oxidative stress and T2D risk may help develop optimal early intervention strategies. Evidence suggests that oxidative stress, triggered by excess dietary fat consumption, causes excess mitochondrial hydrogen peroxide emission in skeletal muscle, alters redox status, and promotes insulin resistance leading to T2D. The pathophysiological events arising from excess calorie-induced mitochondrial reactive oxygen species production are complex and not yet investigated in children. Systems medicine is an integrative approach leveraging conventional medical information and environmental factors with data obtained from “omics” technologies such as genomics, proteomics, and metabolomics. In adults with T2D, systems medicine shows promise in risk assessment and predicting drug response. Redoxomics is a branch of systems medicine focusing on “omics” data related to redox status. Systems medicine with a complementary emphasis on redoxomics can potentially optimize future healthcare strategies for adults and children with T2D.
Collapse
|
27
|
Li M, Chi X, Wang Y, Setrerrahmane S, Xie W, Xu H. Trends in insulin resistance: insights into mechanisms and therapeutic strategy. Signal Transduct Target Ther 2022; 7:216. [PMID: 35794109 PMCID: PMC9259665 DOI: 10.1038/s41392-022-01073-0] [Citation(s) in RCA: 294] [Impact Index Per Article: 98.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 06/15/2022] [Accepted: 06/20/2022] [Indexed: 02/06/2023] Open
Abstract
The centenary of insulin discovery represents an important opportunity to transform diabetes from a fatal diagnosis into a medically manageable chronic condition. Insulin is a key peptide hormone and mediates the systemic glucose metabolism in different tissues. Insulin resistance (IR) is a disordered biological response for insulin stimulation through the disruption of different molecular pathways in target tissues. Acquired conditions and genetic factors have been implicated in IR. Recent genetic and biochemical studies suggest that the dysregulated metabolic mediators released by adipose tissue including adipokines, cytokines, chemokines, excess lipids and toxic lipid metabolites promote IR in other tissues. IR is associated with several groups of abnormal syndromes that include obesity, diabetes, metabolic dysfunction-associated fatty liver disease (MAFLD), cardiovascular disease, polycystic ovary syndrome (PCOS), and other abnormalities. Although no medication is specifically approved to treat IR, we summarized the lifestyle changes and pharmacological medications that have been used as efficient intervention to improve insulin sensitivity. Ultimately, the systematic discussion of complex mechanism will help to identify potential new targets and treat the closely associated metabolic syndrome of IR.
Collapse
Affiliation(s)
- Mengwei Li
- The Engineering Research Center of Synthetic Peptide Drug Discovery and Evaluation of Jiangsu Province, China Pharmaceutical University, Nanjing, 210009, China
- State Key Laboratory of Natural Medicines, Ministry of Education, China Pharmaceutical University, Nanjing, 210009, China
| | - Xiaowei Chi
- Development Center for Medical Science & Technology National Health Commission of the People's Republic of China, 100044, Beijing, China
| | - Ying Wang
- The Engineering Research Center of Synthetic Peptide Drug Discovery and Evaluation of Jiangsu Province, China Pharmaceutical University, Nanjing, 210009, China
- State Key Laboratory of Natural Medicines, Ministry of Education, China Pharmaceutical University, Nanjing, 210009, China
| | | | - Wenwei Xie
- The Engineering Research Center of Synthetic Peptide Drug Discovery and Evaluation of Jiangsu Province, China Pharmaceutical University, Nanjing, 210009, China
- State Key Laboratory of Natural Medicines, Ministry of Education, China Pharmaceutical University, Nanjing, 210009, China
| | - Hanmei Xu
- The Engineering Research Center of Synthetic Peptide Drug Discovery and Evaluation of Jiangsu Province, China Pharmaceutical University, Nanjing, 210009, China.
- State Key Laboratory of Natural Medicines, Ministry of Education, China Pharmaceutical University, Nanjing, 210009, China.
| |
Collapse
|
28
|
Zhang T, Wang S, Li L, Zhu A, Wang Q. Associating diethylhexyl phthalate to gestational diabetes mellitus via adverse outcome pathways using a network-based approach. THE SCIENCE OF THE TOTAL ENVIRONMENT 2022; 824:153932. [PMID: 35182638 DOI: 10.1016/j.scitotenv.2022.153932] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 02/11/2022] [Accepted: 02/12/2022] [Indexed: 06/14/2023]
Abstract
Gestational diabetes mellitus (GDM) is a common pregnancy complication that is harmful to both the woman and fetus. Several epidemiological studies have found that exposure to diethylhexyl phthalate (DEHP), an endocrine disruptor ubiquitous in the environment, may be associated with GDM. This study aims to investigate the mechanism between DEHP and GDM using the adverse outcome pathway (AOP) framework, which can integrate information from different sources to elucidate the causal pathways between chemicals and adverse outcomes. We applied a network-based workflow to integrate diverse information to generate computational AOPs and accelerate the AOP development. The interactions among DEHP, genes, phenotypes, and GDM were retrieved from several publicly available databases, including the Comparative Toxicogenomics Database (CTD), Computational Toxicology (CompTox) Chemicals Dashboard, DisGeNET, MalaCards, Gene Ontology (GO), and Kyoto Encyclopedia of Genes and Genomes (KEGG). Based on the above interactions, a DEHP-Gene-Phenotype-GDM network consisting of 52 nodes and 227 edges was formed to support AOP construction. The filtered genes and phenotypes were assembled as molecular initiating events (MIEs) and key events (KEs) according to the upstream and downstream relationships, generating a computational AOP (cAOP) network. Based on the Organization for Economic Co-operation and Development handbook of AOPs, a cAOP was assessed and applied to determine the effects of DEHP on GDM. DEHP could increase TNF-α, downregulate the glucose uptake process, and lead to GDM. Overall, this study revealed the utility of computational methods in integrating a variety of datasets, supporting AOP development, and facilitating a better understanding of the underlying mechanism of exposure to chemicals on human health.
Collapse
Affiliation(s)
- Tao Zhang
- Department of Toxicology, School of Public Health, Peking University, Beijing 100191, China
| | - Shuo Wang
- Department of Toxicology, School of Public Health, Peking University, Beijing 100191, China
| | - Ludi Li
- Department of Toxicology, School of Public Health, Peking University, Beijing 100191, China
| | - An Zhu
- Department of Toxicology, School of Public Health, Peking University, Beijing 100191, China; Key laboratory of Ministry of Education for Gastrointestinal Cancer, School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350108, China
| | - Qi Wang
- Department of Toxicology, School of Public Health, Peking University, Beijing 100191, China.
| |
Collapse
|
29
|
Glucose deprivation-induced glycogen degradation and viability are altered in peripheral blood mononuclear cells of type 2 diabetes patients. UKRAINIAN BIOCHEMICAL JOURNAL 2022. [DOI: 10.15407/ubj94.01.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
30
|
Wasserman DH. Insulin, Muscle Glucose Uptake, and Hexokinase: Revisiting the Road Not Taken. Physiology (Bethesda) 2022; 37:115-127. [PMID: 34779282 PMCID: PMC8977147 DOI: 10.1152/physiol.00034.2021] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 11/05/2021] [Accepted: 11/07/2021] [Indexed: 12/25/2022] Open
Abstract
Research conducted over the last 50 yr has provided insight into the mechanisms by which insulin stimulates glucose transport across the skeletal muscle cell membrane Transport alone, however, does not result in net glucose uptake as free glucose equilibrates across the cell membrane and is not metabolized. Glucose uptake requires that glucose is phosphorylated by hexokinases. Phosphorylated glucose cannot leave the cell and is the substrate for metabolism. It is indisputable that glucose phosphorylation is essential for glucose uptake. Major advances have been made in defining the regulation of the insulin-stimulated glucose transporter (GLUT4) in skeletal muscle. By contrast, the insulin-regulated hexokinase (hexokinase II) parallels Robert Frost's "The Road Not Taken." Here the case is made that an understanding of glucose phosphorylation by hexokinase II is necessary to define the regulation of skeletal muscle glucose uptake in health and insulin resistance. Results of studies from different physiological disciplines that have elegantly described how hexokinase II can be regulated are summarized to provide a framework for potential application to skeletal muscle. Mechanisms by which hexokinase II is regulated in skeletal muscle await rigorous examination.
Collapse
Affiliation(s)
- David H Wasserman
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee
| |
Collapse
|
31
|
Leitner BP, Siebel S, Akingbesote ND, Zhang X, Perry RJ. Insulin and cancer: a tangled web. Biochem J 2022; 479:583-607. [PMID: 35244142 PMCID: PMC9022985 DOI: 10.1042/bcj20210134] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 02/13/2022] [Accepted: 02/15/2022] [Indexed: 12/13/2022]
Abstract
For a century, since the pioneering work of Otto Warburg, the interwoven relationship between metabolism and cancer has been appreciated. More recently, with obesity rates rising in the U.S. and worldwide, epidemiologic evidence has supported a link between obesity and cancer. A substantial body of work seeks to mechanistically unpack the association between obesity, altered metabolism, and cancer. Without question, these relationships are multifactorial and cannot be distilled to a single obesity- and metabolism-altering hormone, substrate, or factor. However, it is important to understand the hormone-specific associations between metabolism and cancer. Here, we review the links between obesity, metabolic dysregulation, insulin, and cancer, with an emphasis on current investigational metabolic adjuncts to standard-of-care cancer treatment.
Collapse
Affiliation(s)
- Brooks P. Leitner
- Departments of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, CT, U.S.A
- Departments of Internal Medicine, Yale School of Medicine, New Haven, CT, U.S.A
| | - Stephan Siebel
- Departments of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, CT, U.S.A
- Departments of Internal Medicine, Yale School of Medicine, New Haven, CT, U.S.A
- Departments of Pediatrics, Yale School of Medicine, New Haven, CT, U.S.A
| | - Ngozi D. Akingbesote
- Departments of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, CT, U.S.A
- Departments of Internal Medicine, Yale School of Medicine, New Haven, CT, U.S.A
| | - Xinyi Zhang
- Departments of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, CT, U.S.A
- Departments of Internal Medicine, Yale School of Medicine, New Haven, CT, U.S.A
| | - Rachel J. Perry
- Departments of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, CT, U.S.A
- Departments of Internal Medicine, Yale School of Medicine, New Haven, CT, U.S.A
| |
Collapse
|
32
|
Young BE, Padilla J, Finsen SH, Fadel PJ, Mortensen SP. Role of Endothelin-1 Receptors in Limiting Leg Blood Flow and Glucose Uptake During Hyperinsulinemia in Type 2 Diabetes. Endocrinology 2022; 163:6515918. [PMID: 35084435 PMCID: PMC8852254 DOI: 10.1210/endocr/bqac008] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Indexed: 01/29/2023]
Abstract
Skeletal muscle insulin resistance is a hallmark of individuals with type 2 diabetes mellitus (T2D). In healthy individuals insulin stimulates vasodilation, which is markedly blunted in T2D; however, the mechanism(s) remain incompletely understood. Investigations in rodents indicate augmented endothelin-1 (ET-1) action as a major contributor. Human studies have been limited to young obese participants and focused exclusively on the ET-1 A (ETA) receptor. Herein, we have hypothesized that ETA receptor antagonism would improve insulin-stimulated vasodilation and glucose uptake in T2D, with further improvements observed during concurrent ETA + ET-1 B (ETB) antagonism. Arterial pressure (arterial line), leg blood flow (LBF; Doppler), and leg glucose uptake (LGU) were measured at rest, during hyperinsulinemia alone, and hyperinsulinemia with (1) femoral artery infusion of BQ-123, the selective ETA receptor antagonist (n = 10 control, n = 9 T2D) and then (2) addition of BQ-788 (selective ETB antagonist) for blockade of ETA and ETB receptors (n = 7 each). The LBF responses to hyperinsulinemia alone tended to be lower in T2D (controls: ∆161 ± 160 mL/minute; T2D: ∆58 ± 43 mL/minute, P = .08). BQ-123 during hyperinsulinemia augmented LBF to a greater extent in T2D (% change: controls: 14 ± 23%; T2D: 38 ± 21%, P = .029). LGU following BQ-123 increased similarly between groups (P = .85). Concurrent ETA + ETB antagonism did not further increase LBF or LGU in either group. Collectively, these findings suggest that during hyperinsulinemia ETA receptor activation restrains vasodilation more in T2D than controls while limiting glucose uptake similarly in both groups, with no further effect of ETB receptors (NCT04907838).
Collapse
Affiliation(s)
- Benjamin E Young
- Department of Kinesiology, University of Texas at Arlington, Arlington, TX 76019, USA
- Correspondence: Benjamin E. Young, PhD, Department of Kinesiology, College of Nursing and Health Innovation, University of Texas at Arlington, 411 S. Nedderman Dr., Pickard Hall, room 504, Arlington, TX 76019, USA.
| | - Jaume Padilla
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO 65211, USA
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO 65211, USA
| | - Stine H Finsen
- Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense, Denmark
| | - Paul J Fadel
- Department of Kinesiology, University of Texas at Arlington, Arlington, TX 76019, USA
| | - Stefan P Mortensen
- Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
33
|
DeVito LM, Dennis EA, Kahn BB, Shulman GI, Witztum JL, Sadhu S, Nickels J, Spite M, Smyth S, Spiegel S. Bioactive lipids and metabolic syndrome-a symposium report. Ann N Y Acad Sci 2022; 1511:87-106. [PMID: 35218041 DOI: 10.1111/nyas.14752] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 01/10/2022] [Indexed: 11/27/2022]
Abstract
Recent research has shed light on the cellular and molecular functions of bioactive lipids that go far beyond what was known about their role as dietary lipids. Bioactive lipids regulate inflammation and its resolution as signaling molecules. Genetic studies have identified key factors that can increase the risk of cardiovascular diseases and metabolic syndrome through their effects on lipogenesis. Lipid scientists have explored how these signaling pathways affect lipid metabolism in the liver, adipose tissue, and macrophages by utilizing a variety of techniques in both humans and animal models, including novel lipidomics approaches and molecular dynamics models. Dissecting out these lipid pathways can help identify mechanisms that can be targeted to prevent or treat cardiometabolic conditions. Continued investigation of the multitude of functions mediated by bioactive lipids may reveal additional components of these pathways that can provide a greater understanding of metabolic homeostasis.
Collapse
Affiliation(s)
| | | | - Barbara B Kahn
- Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts
| | | | | | | | - Joseph Nickels
- Genesis Biotechnology Group, Hamilton Township, New Jersey
| | - Matthew Spite
- Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts
| | - Susan Smyth
- University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Sarah Spiegel
- Virginia Commonwealth University School of Medicine, Richmond, Virginia
| |
Collapse
|
34
|
Lee-Ødegård S, Olsen T, Norheim F, Drevon CA, Birkeland KI. Potential Mechanisms for How Long-Term Physical Activity May Reduce Insulin Resistance. Metabolites 2022; 12:metabo12030208. [PMID: 35323652 PMCID: PMC8950317 DOI: 10.3390/metabo12030208] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 02/20/2022] [Accepted: 02/22/2022] [Indexed: 02/06/2023] Open
Abstract
Insulin became available for the treatment of patients with diabetes 100 years ago, and soon thereafter it became evident that the biological response to its actions differed markedly between individuals. This prompted extensive research into insulin action and resistance (IR), resulting in the universally agreed fact that IR is a core finding in patients with type 2 diabetes mellitus (T2DM). T2DM is the most prevalent form of diabetes, reaching epidemic proportions worldwide. Physical activity (PA) has the potential of improving IR and is, therefore, a cornerstone in the prevention and treatment of T2DM. Whereas most research has focused on the acute effects of PA, less is known about the effects of long-term PA on IR. Here, we describe a model of potential mechanisms behind reduced IR after long-term PA to guide further mechanistic investigations and to tailor PA interventions in the therapy of T2DM. The development of such interventions requires knowledge of normal glucose metabolism, and we briefly summarize an integrated physiological perspective on IR. We then describe the effects of long-term PA on signaling molecules involved in cellular responses to insulin, tissue-specific functions, and whole-body IR.
Collapse
Affiliation(s)
- Sindre Lee-Ødegård
- Department of Clinical Medicine, Faculty of Medicine, University of Oslo, 0372 Oslo, Norway;
| | - Thomas Olsen
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, 0372 Oslo, Norway; (T.O.); (F.N.); (C.A.D.)
| | - Frode Norheim
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, 0372 Oslo, Norway; (T.O.); (F.N.); (C.A.D.)
| | - Christian Andre Drevon
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, 0372 Oslo, Norway; (T.O.); (F.N.); (C.A.D.)
- Vitas Ltd. Analytical Services, Oslo Science Park, 0349 Oslo, Norway
| | - Kåre Inge Birkeland
- Department of Clinical Medicine, Faculty of Medicine, University of Oslo, 0372 Oslo, Norway;
- Correspondence:
| |
Collapse
|
35
|
Hulett NA, Scalzo RL, Reusch JEB. Glucose Uptake by Skeletal Muscle within the Contexts of Type 2 Diabetes and Exercise: An Integrated Approach. Nutrients 2022; 14:647. [PMID: 35277006 PMCID: PMC8839578 DOI: 10.3390/nu14030647] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2021] [Revised: 01/25/2022] [Accepted: 01/28/2022] [Indexed: 02/01/2023] Open
Abstract
Type 2 diabetes continues to negatively impact the health of millions. The inability to respond to insulin to clear blood glucose (insulin resistance) is a key pathogenic driver of the disease. Skeletal muscle is the primary tissue for maintaining glucose homeostasis through glucose uptake via insulin-dependent and -independent mechanisms. Skeletal muscle is also responsive to exercise-meditated glucose transport, and as such, exercise is a cornerstone for glucose management in people with type 2 diabetes. Skeletal muscle glucose uptake requires a concert of events. First, the glucose-rich blood must be transported to the skeletal muscle. Next, the glucose must traverse the endothelium, extracellular matrix, and skeletal muscle membrane. Lastly, intracellular metabolic processes must be activated to maintain the diffusion gradient to facilitate glucose transport into the cell. This review aims to examine the physiology at each of these steps in healthy individuals, analyze the dysregulation affecting these pathways associated with type 2 diabetes, and describe the mechanisms by which exercise acts to increase glucose uptake.
Collapse
Affiliation(s)
- Nicholas A. Hulett
- Department of Medicine, Anschutz Medical Campus, University of Colorado, Aurora, CO 80045, USA; (N.A.H.); (R.L.S.)
| | - Rebecca L. Scalzo
- Department of Medicine, Anschutz Medical Campus, University of Colorado, Aurora, CO 80045, USA; (N.A.H.); (R.L.S.)
- Rocky Mountain Regional Veterans Affairs Medical Center, Aurora, CO 80045, USA
- Center for Women’s Health Research, Anschutz Medical Campus, University of Colorado, Aurora, CO 80045, USA
| | - Jane E. B. Reusch
- Department of Medicine, Anschutz Medical Campus, University of Colorado, Aurora, CO 80045, USA; (N.A.H.); (R.L.S.)
- Rocky Mountain Regional Veterans Affairs Medical Center, Aurora, CO 80045, USA
- Center for Women’s Health Research, Anschutz Medical Campus, University of Colorado, Aurora, CO 80045, USA
| |
Collapse
|
36
|
Camastra S, Ferrannini E. Role of anatomical location, cellular phenotype and perfusion of adipose tissue in intermediary metabolism: A narrative review. Rev Endocr Metab Disord 2022; 23:43-50. [PMID: 35031911 PMCID: PMC8873050 DOI: 10.1007/s11154-021-09708-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/22/2021] [Indexed: 02/07/2023]
Abstract
It is well-established that adipose tissue accumulation is associated with insulin resistance through multiple mechanisms. One major metabolic link is the classical Randle cycle: enhanced release of free fatty acids (FFA) from hydrolysis of adipose tissue triglycerides impedes insulin-mediated glucose uptake in muscle tissues. Less well studied are the different routes of this communication. First, white adipose tissue depots may be regionally distant from muscle (i.e., gluteal fat and diaphragm muscle) or contiguous to muscle but separated by a fascia (Scarpa's fascia in the abdomen, fascia lata in the thigh). In this case, released FFA outflow through the venous drainage and merge into arterial plasma to be transported to muscle tissues. Next, cytosolic triglycerides can directly, i.e., within the cell, provide FFA to myocytes (but also pancreatic ß-cells, renal tubular cells, etc.). Finally, adipocyte layers or lumps may be adjacent to, but not anatomically segregated, from muscle, as is typically the case for epicardial fat and cardiomyocytes. As regulation of these three main delivery paths is different, their separate contribution to substrate competition at the whole-body level is uncertain. Another important link between fat and muscle is vascular. In the resting state, blood flow is generally higher in adipose tissue than in muscle. In the insulinized state, fat blood flow is directly related to whole-body insulin resistance whereas muscle blood flow is not; consequently, fractional (i.e., flow-adjusted) glucose uptake is stimulated in muscle but not fat. Thus, reduced blood supply is a major factor for the impairment of in vivo insulin-mediated glucose uptake in both subcutaneous and visceral fat. In contrast, the insulin resistance of glucose uptake in resting skeletal muscle is predominantly a cellular defect.
Collapse
Affiliation(s)
- Stefania Camastra
- Department of Clinical & Experimental Medicine, University of Pisa, Pisa, Italy.
| | | |
Collapse
|
37
|
Metformin and Insulin Resistance: A Review of the Underlying Mechanisms behind Changes in GLUT4-Mediated Glucose Transport. Int J Mol Sci 2022; 23:ijms23031264. [PMID: 35163187 PMCID: PMC8836112 DOI: 10.3390/ijms23031264] [Citation(s) in RCA: 101] [Impact Index Per Article: 33.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 01/21/2022] [Accepted: 01/21/2022] [Indexed: 02/06/2023] Open
Abstract
Metformin is the most commonly used treatment to increase insulin sensitivity in insulin-resistant (IR) conditions such as diabetes, prediabetes, polycystic ovary syndrome, and obesity. There is a well-documented correlation between glucose transporter 4 (GLUT4) expression and the level of IR. Therefore, the observed increase in peripheral glucose utilization after metformin treatment most likely comes from the induction of GLUT4 expression and its increased translocation to the plasma membrane. However, the mechanisms behind this effect and the critical metformin targets are still largely undefined. The present review explores the evidence for the crucial role of changes in the expression and activation of insulin signaling pathway mediators, AMPK, several GLUT4 translocation mediators, and the effect of posttranscriptional modifications based on previously published preclinical and clinical models of metformin’s mode of action in animal and human studies. Our aim is to provide a comprehensive review of the studies in this field in order to shed some light on the complex interactions between metformin action, GLUT4 expression, GLUT4 translocation, and the observed increase in peripheral insulin sensitivity.
Collapse
|
38
|
Lee SH, Park SY, Choi CS. Insulin Resistance: From Mechanisms to Therapeutic Strategies. Diabetes Metab J 2022; 46:15-37. [PMID: 34965646 PMCID: PMC8831809 DOI: 10.4093/dmj.2021.0280] [Citation(s) in RCA: 365] [Impact Index Per Article: 121.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 12/27/2021] [Indexed: 11/12/2022] Open
Abstract
Insulin resistance is the pivotal pathogenic component of many metabolic diseases, including type 2 diabetes mellitus, and is defined as a state of reduced responsiveness of insulin-targeting tissues to physiological levels of insulin. Although the underlying mechanism of insulin resistance is not fully understood, several credible theories have been proposed. In this review, we summarize the functions of insulin in glucose metabolism in typical metabolic tissues and describe the mechanisms proposed to underlie insulin resistance, that is, ectopic lipid accumulation in liver and skeletal muscle, endoplasmic reticulum stress, and inflammation. In addition, we suggest potential therapeutic strategies for addressing insulin resistance.
Collapse
Affiliation(s)
- Shin-Hae Lee
- Korea Mouse Metabolic Phenotyping Center (KMMPC), Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, Korea
| | - Shi-Young Park
- Korea Mouse Metabolic Phenotyping Center (KMMPC), Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, Korea
| | - Cheol Soo Choi
- Korea Mouse Metabolic Phenotyping Center (KMMPC), Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, Korea
- Department of Internal Medicine, Gachon University Gil Medical Center, Incheon, Korea
- Division of Molecular Medicine, Gachon University College of Medicine, Incheon, Korea
- Corresponding author: Cheol Soo Choi https://orcid.org/0000-0001-9627-058X Division of Molecular Medicine, Gachon University College of Medicine, 21 Namdongdaero 774beon-gil, Namdong-gu, Incheon 21565, Korea E-mail:
| |
Collapse
|
39
|
Sedivy P, Dusilova T, Hajek M, Burian M, Krššák M, Dezortova M. In Vitro 31P MR Chemical Shifts of In Vivo-Detectable Metabolites at 3T as a Basis Set for a Pilot Evaluation of Skeletal Muscle and Liver 31P Spectra with LCModel Software. Molecules 2021; 26:molecules26247571. [PMID: 34946652 PMCID: PMC8703310 DOI: 10.3390/molecules26247571] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 12/06/2021] [Accepted: 12/10/2021] [Indexed: 11/24/2022] Open
Abstract
Most in vivo 31P MR studies are realized on 3T MR systems that provide sufficient signal intensity for prominent phosphorus metabolites. The identification of these metabolites in the in vivo spectra is performed by comparing their chemical shifts with the chemical shifts measured in vitro on high-field NMR spectrometers. To approach in vivo conditions at 3T, a set of phantoms with defined metabolite solutions were measured in a 3T whole-body MR system at 7.0 and 7.5 pH, at 37 °C. A free induction decay (FID) sequence with and without 1H decoupling was used. Chemical shifts were obtained of phosphoenolpyruvate (PEP), phosphatidylcholine (PtdC), phosphocholine (PC), phosphoethanolamine (PE), glycerophosphocholine (GPC), glycerophosphoetanolamine (GPE), uridine diphosphoglucose (UDPG), glucose-6-phosphate (G6P), glucose-1-phosphate (G1P), 2,3-diphosphoglycerate (2,3-DPG), nicotinamide adenine dinucleotide (NADH and NAD+), phosphocreatine (PCr), adenosine triphosphate (ATP), adenosine diphosphate (ADP), and inorganic phosphate (Pi). The measured chemical shifts were used to construct a basis set of 31P MR spectra for the evaluation of 31P in vivo spectra of muscle and the liver using LCModel software (linear combination model). Prior knowledge was successfully employed in the analysis of previously acquired in vivo data.
Collapse
Affiliation(s)
- Petr Sedivy
- MR-Unit, Department of Diagnostic and Interventional Radiology, Institute for Clinical and Experimental Medicine, Videnska 1958/9, 140 21 Prague, Czech Republic; (P.S.); (T.D.); (M.H.); (M.B.)
| | - Tereza Dusilova
- MR-Unit, Department of Diagnostic and Interventional Radiology, Institute for Clinical and Experimental Medicine, Videnska 1958/9, 140 21 Prague, Czech Republic; (P.S.); (T.D.); (M.H.); (M.B.)
| | - Milan Hajek
- MR-Unit, Department of Diagnostic and Interventional Radiology, Institute for Clinical and Experimental Medicine, Videnska 1958/9, 140 21 Prague, Czech Republic; (P.S.); (T.D.); (M.H.); (M.B.)
| | - Martin Burian
- MR-Unit, Department of Diagnostic and Interventional Radiology, Institute for Clinical and Experimental Medicine, Videnska 1958/9, 140 21 Prague, Czech Republic; (P.S.); (T.D.); (M.H.); (M.B.)
| | - Martin Krššák
- Division of Endocrinology and Metabolism, Department of Internal Medicine III, Medical University of Vienna, 1090 Vienna, Austria;
- High-Field MR Center, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, 1090 Vienna, Austria
| | - Monika Dezortova
- MR-Unit, Department of Diagnostic and Interventional Radiology, Institute for Clinical and Experimental Medicine, Videnska 1958/9, 140 21 Prague, Czech Republic; (P.S.); (T.D.); (M.H.); (M.B.)
- Correspondence: ; Tel.: +420-23605-5245
| |
Collapse
|
40
|
Zhang Y, Zhao H, Liu B, Shu H, Zhang L, Bao M, Yi W, Tan Y, Ji X, Zhang C, Zhao N, Pang G, He D, Wang Y, Li L, Yi J, Lu C. Human serum metabolomic analysis reveals progression for high blood pressure in type 2 diabetes mellitus. BMJ Open Diabetes Res Care 2021; 9:9/1/e002337. [PMID: 34711543 PMCID: PMC8557281 DOI: 10.1136/bmjdrc-2021-002337] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 10/05/2021] [Indexed: 11/04/2022] Open
Abstract
INTRODUCTION Type 2 diabetes mellitus (T2DM) is one of the most ordinary metabolic disorders and manifests as a high blood sugar level; 80%-90% of patients with T2DM will develop high blood pressure (HBP), which exacerbates irreversible organ damage. Understanding the metabolic basis of HBP is essential to facilitating early diagnosis and prompt treatments of diabetic complications. RESEARCH DESIGN AND METHODS 34 patients who originally had T2DM and then developed HBP within 1 year were selected from physical examination participants. Using ultrahigh-performance liquid chromatography quadrupole time-of-flight metabolomic analysis, we compared the metabolomic profile of patients with 30 healthy controls. The results showed a clear discrimination in metabolomic profiles between T2DM and T2DM+HBP when employing orthogonal projection to latent structure with discriminant analysis with electrospray ionization modes. RESULTS Eight differential metabolites changed significantly during disease progression, among which L-isoleucine, L-glutamic acid, pyroglutamic acid and linoleic acid decreased, while sphinganine, Cer(d18:0/16:0), Cer(d18:0/18:0), and citric acid increased. These metabolites are associated with the γ-glutamyl cycle, tricarboxylic acid cycle, and ceramide metabolism. CONCLUSIONS These novel serum biomarkers may improve the management of T2DM and HBP complications, thus reducing the use of incorrect medical care.
Collapse
Affiliation(s)
- Yin Zhang
- China Academy of Chinese Medical Sciences, Beijing, China
- Yichun University, Yichun, Jiangxi, China
| | - Heru Zhao
- China Academy of Chinese Medical Sciences, Beijing, China
- Yichun University, Yichun, Jiangxi, China
| | - Bin Liu
- China Academy of Chinese Medical Sciences, Beijing, China
| | - Haiyang Shu
- Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Lulu Zhang
- China Academy of Chinese Medical Sciences, Beijing, China
- Yichun University, Yichun, Jiangxi, China
| | - Mei Bao
- China Academy of Chinese Medical Sciences, Beijing, China
- Yichun University, Yichun, Jiangxi, China
| | - Wenjun Yi
- China Academy of Chinese Medical Sciences, Beijing, China
| | - Yong Tan
- China Academy of Chinese Medical Sciences, Beijing, China
| | - Xinyu Ji
- China Academy of Chinese Medical Sciences, Beijing, China
| | - Chi Zhang
- China Academy of Chinese Medical Sciences, Beijing, China
| | - Ning Zhao
- China Academy of Chinese Medical Sciences, Beijing, China
| | - Guoming Pang
- Traditional Chinese Medicine Hospital of Kaifeng, Kaifeng, China
| | - Dan He
- Jiangxi University of Traditional Chinese Medicine, Nanchang, Jiangxi, China
| | - Yuexi Wang
- China Academy of Chinese Medical Sciences, Beijing, China
| | - Li Li
- China Academy of Chinese Medical Sciences, Beijing, China
| | | | - Cheng Lu
- China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
41
|
Öhman T, Teppo J, Datta N, Mäkinen S, Varjosalo M, Koistinen HA. Skeletal muscle proteomes reveal downregulation of mitochondrial proteins in transition from prediabetes into type 2 diabetes. iScience 2021; 24:102712. [PMID: 34235411 PMCID: PMC8246593 DOI: 10.1016/j.isci.2021.102712] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 02/17/2021] [Accepted: 06/08/2021] [Indexed: 12/22/2022] Open
Abstract
Skeletal muscle insulin resistance is a central defect in the pathogenesis of type 2 diabetes (T2D). Here, we analyzed skeletal muscle proteome in 148 vastus lateralis muscle biopsies obtained from men covering all glucose tolerance phenotypes: normal, impaired fasting glucose (IFG), impaired glucose tolerance (IGT) and T2D. Skeletal muscle proteome was analyzed by a sequential window acquisition of all theoretical mass spectra (SWATH-MS) proteomics technique. Our data indicate a downregulation in several proteins involved in mitochondrial electron transport or respiratory chain complex assembly already in IFG and IGT muscles, with most profound decreases observed in T2D. Additional phosphoproteomic analysis reveals altered phosphorylation in several signaling pathways in IFG, IGT, and T2D muscles, including those regulating glucose metabolic processes, and the structure of muscle cells. These data reveal several alterations present in skeletal muscle already in prediabetes and highlight impaired mitochondrial energy metabolism in the trajectory from prediabetes into T2D. Skeletal muscle proteome from men with all stages of glucose tolerance was analyzed Phosphoproteomics reveal altered phosphorylation in IFG, IGT, and T2D muscles OXPHOS proteins are decreased in prediabetic muscles, with most decrease in T2D
Collapse
Affiliation(s)
- Tiina Öhman
- University of Helsinki, Molecular Systems Biology Research Group and Proteomics Unit, Institute of Biotechnology, 00014 Helsinki, Finland
| | - Jaakko Teppo
- University of Helsinki, Molecular Systems Biology Research Group and Proteomics Unit, Institute of Biotechnology, 00014 Helsinki, Finland.,University of Helsinki, Drug Research Program, Faculty of Pharmacy, 00014 Helsinki, Finland
| | - Neeta Datta
- University of Helsinki, Department of Medicine, Helsinki University Hospital, Haartmaninkatu 4, PO BOX 340, 00029 HUS, Helsinki, Finland.,Minerva Foundation Institute for Medical Research, Tukholmankatu 8, 00290 Helsinki, Finland
| | - Selina Mäkinen
- University of Helsinki, Department of Medicine, Helsinki University Hospital, Haartmaninkatu 4, PO BOX 340, 00029 HUS, Helsinki, Finland.,Minerva Foundation Institute for Medical Research, Tukholmankatu 8, 00290 Helsinki, Finland
| | - Markku Varjosalo
- University of Helsinki, Molecular Systems Biology Research Group and Proteomics Unit, Institute of Biotechnology, 00014 Helsinki, Finland
| | - Heikki A Koistinen
- University of Helsinki, Department of Medicine, Helsinki University Hospital, Haartmaninkatu 4, PO BOX 340, 00029 HUS, Helsinki, Finland.,Minerva Foundation Institute for Medical Research, Tukholmankatu 8, 00290 Helsinki, Finland
| |
Collapse
|
42
|
Batista TM, Haider N, Kahn CR. Defining the underlying defect in insulin action in type 2 diabetes. Diabetologia 2021; 64:994-1006. [PMID: 33730188 PMCID: PMC8916220 DOI: 10.1007/s00125-021-05415-5] [Citation(s) in RCA: 92] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Accepted: 01/29/2021] [Indexed: 01/08/2023]
Abstract
Insulin resistance is one of the earliest defects in the pathogenesis of type 2 diabetes. Over the past 50 years, elucidation of the insulin signalling network has provided important mechanistic insights into the abnormalities of glucose, lipid and protein metabolism that underlie insulin resistance. In classical target tissues (liver, muscle and adipose tissue), insulin binding to its receptor initiates a broad signalling cascade mediated by changes in phosphorylation, gene expression and vesicular trafficking that result in increased nutrient utilisation and storage, and suppression of catabolic processes. Insulin receptors are also expressed in non-classical targets, such as the brain and endothelial cells, where it helps regulate appetite, energy expenditure, reproductive hormones, mood/behaviour and vascular function. Recent progress in cell biology and unbiased molecular profiling by mass spectrometry and DNA/RNA-sequencing has provided a unique opportunity to dissect the determinants of insulin resistance in type 2 diabetes and the metabolic syndrome; best studied are extrinsic factors, such as circulating lipids, amino acids and other metabolites and exosomal microRNAs. More challenging has been defining the cell-intrinsic factors programmed by genetics and epigenetics that underlie insulin resistance. In this regard, studies using human induced pluripotent stem cells and tissues point to cell-autonomous alterations in signalling super-networks, involving changes in phosphorylation and gene expression both inside and outside the canonical insulin signalling pathway. Understanding how these multi-layered molecular networks modulate insulin action and metabolism in different tissues will open new avenues for therapy and prevention of type 2 diabetes and its associated pathologies.
Collapse
Affiliation(s)
- Thiago M Batista
- Section of Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Nida Haider
- Section of Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - C Ronald Kahn
- Section of Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
43
|
Nguyen Huu T, Park J, Zhang Y, Park I, Yoon HJ, Woo HA, Lee SR. Redox Regulation of PTEN by Peroxiredoxins. Antioxidants (Basel) 2021; 10:antiox10020302. [PMID: 33669370 PMCID: PMC7920247 DOI: 10.3390/antiox10020302] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 02/08/2021] [Accepted: 02/10/2021] [Indexed: 12/20/2022] Open
Abstract
Phosphatase and tensin homolog deleted on chromosome 10 (PTEN) is known as a tumor suppressor gene that is frequently mutated in numerous human cancers and inherited syndromes. PTEN functions as a negative regulator of PI3K/Akt signaling pathway by dephosphorylating phosphatidylinositol (3, 4, 5)-trisphosphate (PIP3) to phosphatidylinositol (4, 5)-bisphosphate (PIP2), which leads to the inhibition of cell growth, proliferation, cell survival, and protein synthesis. PTEN contains a cysteine residue in the active site that can be oxidized by peroxides, forming an intramolecular disulfide bond between Cys124 and Cys71. Redox regulation of PTEN by reactive oxygen species (ROS) plays a crucial role in cellular signaling. Peroxiredoxins (Prxs) are a superfamily of peroxidase that catalyzes reduction of peroxides and maintains redox homeostasis. Mammalian Prxs have 6 isoforms (I-VI) and can scavenge cellular peroxides. It has been demonstrated that Prx I can preserve and promote the tumor-suppressive function of PTEN by preventing oxidation of PTEN under benign oxidative stress via direct interaction. Also, Prx II-deficient cells increased PTEN oxidation and insulin sensitivity. Furthermore, Prx III has been shown to protect PTEN from oxidation induced by 15s-HpETE and 12s-HpETE, these are potent inflammatory and pro-oxidant mediators. Understanding the tight connection between PTEN and Prxs is important for providing novel therapies. Herein, we summarized recent studies focusing on the relationship of Prxs and the redox regulation of PTEN.
Collapse
Affiliation(s)
- Thang Nguyen Huu
- Department of Biochemistry, Research Center for Aging and Geriatrics, Research Institute of Medical Sciences, Chonnam National University Medical School, Gwangju 501-190, Korea; (T.N.H.); (I.P.); (H.J.Y.)
- Department of Biomedical Sciences, Research Center for Aging and Geriatrics, Research Institute of Medical Sciences, Chonnam National University Medical School, Gwangju 501-190, Korea
| | - Jiyoung Park
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 120-750, Korea;
| | - Ying Zhang
- Department of Cell Biology, School of Medicine, Jiangsu University, Zhenjiang 212013, China;
| | - Iha Park
- Department of Biochemistry, Research Center for Aging and Geriatrics, Research Institute of Medical Sciences, Chonnam National University Medical School, Gwangju 501-190, Korea; (T.N.H.); (I.P.); (H.J.Y.)
- Department of Biomedical Sciences, Research Center for Aging and Geriatrics, Research Institute of Medical Sciences, Chonnam National University Medical School, Gwangju 501-190, Korea
| | - Hyun Joong Yoon
- Department of Biochemistry, Research Center for Aging and Geriatrics, Research Institute of Medical Sciences, Chonnam National University Medical School, Gwangju 501-190, Korea; (T.N.H.); (I.P.); (H.J.Y.)
| | - Hyun Ae Woo
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 120-750, Korea;
- Correspondence: (H.A.W.); (S.-R.L.); Tel.: +82-2-3277-4654 (H.A.W.); +82-61-379-2775 (S.-R.L.); Fax: +82-2-3277-3760 (H.A.W.); +82-61-379-2782 (S.-R.L.)
| | - Seung-Rock Lee
- Department of Biochemistry, Research Center for Aging and Geriatrics, Research Institute of Medical Sciences, Chonnam National University Medical School, Gwangju 501-190, Korea; (T.N.H.); (I.P.); (H.J.Y.)
- Department of Biomedical Sciences, Research Center for Aging and Geriatrics, Research Institute of Medical Sciences, Chonnam National University Medical School, Gwangju 501-190, Korea
- Correspondence: (H.A.W.); (S.-R.L.); Tel.: +82-2-3277-4654 (H.A.W.); +82-61-379-2775 (S.-R.L.); Fax: +82-2-3277-3760 (H.A.W.); +82-61-379-2782 (S.-R.L.)
| |
Collapse
|
44
|
Norris D, Yang P, Shin SY, Kearney AL, Kim HJ, Geddes T, Senior AM, Fazakerley DJ, Nguyen LK, James DE, Burchfield JG. Signaling Heterogeneity is Defined by Pathway Architecture and Intercellular Variability in Protein Expression. iScience 2021; 24:102118. [PMID: 33659881 PMCID: PMC7892930 DOI: 10.1016/j.isci.2021.102118] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 01/07/2021] [Accepted: 01/22/2021] [Indexed: 12/12/2022] Open
Abstract
Insulin's activation of PI3K/Akt signaling, stimulates glucose uptake by enhancing delivery of GLUT4 to the cell surface. Here we examined the origins of intercellular heterogeneity in insulin signaling. Akt activation alone accounted for ~25% of the variance in GLUT4, indicating that additional sources of variance exist. The Akt and GLUT4 responses were highly reproducible within the same cell, suggesting the variance is between cells (extrinsic) and not within cells (intrinsic). Generalized mechanistic models (supported by experimental observations) demonstrated that the correlation between the steady-state levels of two measured signaling processes decreases with increasing distance from each other and that intercellular variation in protein expression (as an example of extrinsic variance) is sufficient to account for the variance in and between Akt and GLUT4. Thus, the response of a population to insulin signaling is underpinned by considerable single-cell heterogeneity that is largely driven by variance in gene/protein expression between cells. Insulin signaling is heterogeneous between cells in the same population The temporal response of signaling components within a cell is highly reproducible Upstream responses (Akt) can only partially predict downstream response (GLUT4) Protein expression variance is a driver of intercellular signaling heterogeneity
Collapse
Affiliation(s)
- Dougall Norris
- Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia.,School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW 2006, Australia
| | - Pengyi Yang
- Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia.,School of Mathematics and Statistics, The University of Sydney, Sydney, NSW 2006, Australia.,Computational Systems Biology Group, Children's Medical Research Institute, Faculty of Medicine and Health, The University of Sydney, Westmead, NSW 2145, Australia
| | - Sung-Young Shin
- Department of Biochemistry and Molecular Biology, School of Biomedical Sciences, Monash University, Clayton, VIC 3800, Australia.,Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia
| | - Alison L Kearney
- Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia.,School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW 2006, Australia
| | - Hani Jieun Kim
- Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia.,School of Mathematics and Statistics, The University of Sydney, Sydney, NSW 2006, Australia.,Computational Systems Biology Group, Children's Medical Research Institute, Faculty of Medicine and Health, The University of Sydney, Westmead, NSW 2145, Australia
| | - Thomas Geddes
- Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia.,School of Mathematics and Statistics, The University of Sydney, Sydney, NSW 2006, Australia.,Computational Systems Biology Group, Children's Medical Research Institute, Faculty of Medicine and Health, The University of Sydney, Westmead, NSW 2145, Australia
| | - Alistair M Senior
- Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia.,School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW 2006, Australia
| | - Daniel J Fazakerley
- Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia
| | - Lan K Nguyen
- Department of Biochemistry and Molecular Biology, School of Biomedical Sciences, Monash University, Clayton, VIC 3800, Australia.,Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia
| | - David E James
- Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia.,School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW 2006, Australia.,Sydney Medical School, The University of Sydney, Sydney, NSW 2006, Australia
| | - James G Burchfield
- Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia.,School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW 2006, Australia
| |
Collapse
|
45
|
Gong L, Wang Z, Wang Z, Zhang Z. Sestrin2 as a Potential Target for Regulating Metabolic-Related Diseases. Front Endocrinol (Lausanne) 2021; 12:751020. [PMID: 34803916 PMCID: PMC8595836 DOI: 10.3389/fendo.2021.751020] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Accepted: 10/11/2021] [Indexed: 12/12/2022] Open
Abstract
Sestrin2 is a highly conserved protein that can be induced under a variety of stress conditions, including DNA damage, oxidative stress, endoplasmic reticulum (ER) stress, and metabolic stress. Numerous studies have shown that the AMP-activated protein kinase (AMPK)/mammalian target of rapamycin (mTOR) signaling pathway has a crucial role in the regulation of metabolism. Sestrin2 regulates metabolism via a number of pathways, including activation of AMPK, inhibition of the mTOR complex 1 (mTORC1), activation of mTOR complex 2 (mTORC2), inhibition of ER stress, and promotion of autophagy. Therefore, modulation of Sestrin2 activity may provide a potential therapeutic target for the prevention of metabolic diseases such as insulin resistance, diabetes, obesity, non-alcoholic fatty liver disease, and myocardial ischemia/reperfusion injury. In this review, we examined the regulatory relationship between Sestrin2 and the AMPK/mTOR signaling pathway and the effects of Sestrin2 on energy metabolism.
Collapse
|
46
|
Nie X, Chen Z, Pang L, Wang L, Jiang H, Chen Y, Zhang Z, Fu C, Ren B, Zhang J. Oral Nano Drug Delivery Systems for the Treatment of Type 2 Diabetes Mellitus: An Available Administration Strategy for Antidiabetic Phytocompounds. Int J Nanomedicine 2020; 15:10215-10240. [PMID: 33364755 PMCID: PMC7751584 DOI: 10.2147/ijn.s285134] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Accepted: 11/23/2020] [Indexed: 12/15/2022] Open
Abstract
In view of the worldwide serious health threat of type 2 diabetes mellitus (T2DM), natural sources of chemotherapies have been corroborated as the promising alternatives, with the excellent antidiabetic activities, bio-safety, and more cost-effective properties. However, their clinical application is somewhat limited, because of the poor solubility, instability in the gastrointestinal tract (GIT), low bioavailability, and so on. Nowadays, to develop nanoscaled systems has become a prominent strategy to improve the drug delivery of phytochemicals. In this review, we primarily summarized the intervention mechanisms of phytocompounds against T2DM and presented the recent advances in various nanosystems of antidiabetic phytocompounds. Selected nanosystems were grouped depending on their classification and structures, including polymeric NPs, lipid-based nanosystems, vesicular systems, inorganic nanocarriers, and so on. Based on this review, the state-of-the-art nanosystems for phytocompounds in T2DM treatment have been presented, suggesting the preponderance and potential of nanotechnologies.
Collapse
Affiliation(s)
- Xin Nie
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu611137, People’s Republic of China
| | - Zhejie Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, 999087, People’s Republic of China
| | - Lan Pang
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu611137, People’s Republic of China
| | - Lin Wang
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu611137, People’s Republic of China
| | - Huajuan Jiang
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu611137, People’s Republic of China
| | - Yi Chen
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu611137, People’s Republic of China
| | - Zhen Zhang
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu611137, People’s Republic of China
| | - Chaomei Fu
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu611137, People’s Republic of China
| | - Bo Ren
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu611137, People’s Republic of China
| | - Jinming Zhang
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu611137, People’s Republic of China
| |
Collapse
|
47
|
Mechanisms by which adiponectin reverses high fat diet-induced insulin resistance in mice. Proc Natl Acad Sci U S A 2020; 117:32584-32593. [PMID: 33293421 PMCID: PMC7768680 DOI: 10.1073/pnas.1922169117] [Citation(s) in RCA: 87] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Adiponectin has emerged as a potential therapy for type 2 diabetes mellitus, but the molecular mechanism by which adiponectin reverses insulin resistance remains unclear. Two weeks of globular adiponectin (gAcrp30) treatment reduced fasting plasma glucose, triglyceride (TAG), and insulin concentrations and reversed whole-body insulin resistance, which could be attributed to both improved insulin-mediated suppression of endogenous glucose production and increased insulin-stimulated glucose uptake in muscle and adipose tissues. These improvements in liver and muscle sensitivity were associated with ∼50% reductions in liver and muscle TAG and plasma membrane (PM)-associated diacylglycerol (DAG) content and occurred independent of reductions in total ceramide content. Reductions of PM DAG content in liver and skeletal muscle were associated with reduced PKCε translocation in liver and reduced PKCθ and PKCε translocation in skeletal muscle resulting in increased insulin-stimulated insulin receptor tyrosine1162 phosphorylation, IRS-1/IRS-2-associated PI3-kinase activity, and Akt-serine phosphorylation. Both gAcrp30 and full-length adiponectin (Acrp30) treatment increased eNOS/AMPK activation in muscle and muscle fatty acid oxidation. gAcrp30 and Acrp30 infusions also increased TAG uptake in epididymal white adipose tissue (eWAT), which could be attributed to increased lipoprotein lipase (LPL) activity. These data suggest that adiponectin and adiponectin-related molecules reverse lipid-induced liver and muscle insulin resistance by reducing ectopic lipid storage in these organs, resulting in decreased plasma membrane sn-1,2-DAG-induced nPKC activity and increased insulin signaling. Adiponectin mediates these effects by both promoting the storage of TAG in eWAT likely through stimulation of LPL as well as by stimulation of AMPK in muscle resulting in increased muscle fat oxidation.
Collapse
|
48
|
Watkins OC, Yong HEJ, Sharma N, Chan SY. A review of the role of inositols in conditions of insulin dysregulation and in uncomplicated and pathological pregnancy. Crit Rev Food Sci Nutr 2020; 62:1626-1673. [PMID: 33280430 DOI: 10.1080/10408398.2020.1845604] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Inositols, a group of 6-carbon polyols, are highly bioactive molecules derived from diet and endogenous synthesis. Inositols and their derivatives are involved in glucose and lipid metabolism and participate in insulin-signaling, with perturbations in inositol processing being associated with conditions involving insulin resistance, dysglycemia and dyslipidemia such as polycystic ovary syndrome and diabetes. Pregnancy is similarly characterized by substantial and complex changes in glycemic and lipidomic regulation as part of maternal adaptation and is also associated with physiological alterations in inositol processing. Disruptions in maternal adaptation are postulated to have a critical pathophysiological role in pregnancy complications such as gestational diabetes and pre-eclampsia. Inositol supplementation has shown promise as an intervention for the alleviation of symptoms in conditions of insulin resistance and for gestational diabetes prevention. However, the mechanisms behind these affects are not fully understood. In this review, we explore the role of inositols in conditions of insulin dysregulation and in pregnancy, and identify priority areas for research. We particularly examine the role and function of inositols within the maternal-placental-fetal axis in both uncomplicated and pathological pregnancies. We also discuss how inositols may mediate maternal-placental-fetal cross-talk, and regulate fetal growth and development, and suggest that inositols play a vital role in promoting healthy pregnancy.
Collapse
Affiliation(s)
- Oliver C Watkins
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Hannah E J Yong
- Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research, Singapore, Singapore
| | - Neha Sharma
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Shiao-Yng Chan
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research, Singapore, Singapore
| |
Collapse
|
49
|
Kim SM, Imm JY. The Effect of Chrysin-Loaded Phytosomes on Insulin Resistance and Blood Sugar Control in Type 2 Diabetic db/db Mice. Molecules 2020; 25:molecules25235503. [PMID: 33255372 PMCID: PMC7727825 DOI: 10.3390/molecules25235503] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 11/20/2020] [Accepted: 11/23/2020] [Indexed: 02/07/2023] Open
Abstract
Although a variety of beneficial health effects of natural flavonoids, including chrysin, has been suggested, poor solubility and bioavailability limit their practical use. As a promising delivery system, chrysin-loaded phytosomes (CPs) were prepared using egg phospholipid (EPL) at a 1:3 molar ratio and its antidiabetic effects were assessed in db/db diabetic mice. Male C57BLKS/J-db/db mice were fed a normal diet (control), chrysin diet (100 mg chrysin/kg), CP diet (100 mg chrysin equivalent/kg), metformin diet (200 mg/kg) or EPL diet (vehicle, the same amount of EPL used for CP preparation) for 9 weeks. Administration of CP significantly decreased fasting blood glucose and insulin levels in db/db mice compared with the control. An oral glucose tolerance test and homeostatic model assessment for insulin resistance were significantly improved in the CP group (p < 0.05). CP treatment suppressed gluconeogenesis via downregulation of phosphoenolpyruvate carboxykinase while it promoted glucose uptake in the skeletal muscle and liver of db/db mice (p < 0.05). The CP-mediated improved glucose utilization in the muscle was confirmed by upregulation of glucose transporter type 4, hexokinase2 and peroxisome proliferator-activated receptor γ during treatment (p < 0.05). The CP-induced promotion of GLUT4 plasma translocation was confirmed in the skeletal muscle of db/db mice (p < 0.05). Based on the results, CP showed greater antidiabetic performance compared to the control by ameliorating insulin resistance in db/db mice and phytosome can be used as an effective antidiabetic agent.
Collapse
|
50
|
Batista TM, Jayavelu AK, Wewer Albrechtsen NJ, Iovino S, Lebastchi J, Pan H, Dreyfuss JM, Krook A, Zierath JR, Mann M, Kahn CR. A Cell-Autonomous Signature of Dysregulated Protein Phosphorylation Underlies Muscle Insulin Resistance in Type 2 Diabetes. Cell Metab 2020; 32:844-859.e5. [PMID: 32888406 PMCID: PMC7875546 DOI: 10.1016/j.cmet.2020.08.007] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 05/26/2020] [Accepted: 08/11/2020] [Indexed: 02/06/2023]
Abstract
Skeletal muscle insulin resistance is the earliest defect in type 2 diabetes (T2D), preceding and predicting disease development. To what extent this reflects a primary defect or is secondary to tissue cross talk due to changes in hormones or circulating metabolites is unknown. To address this question, we have developed an in vitro disease-in-a-dish model using iPS cells from T2D patients differentiated into myoblasts (iMyos). We find that T2D iMyos in culture exhibit multiple defects mirroring human disease, including an altered insulin signaling, decreased insulin-stimulated glucose uptake, and reduced mitochondrial oxidation. More strikingly, global phosphoproteomic analysis reveals a multidimensional network of signaling defects in T2D iMyos going beyond the canonical insulin-signaling cascade, including proteins involved in regulation of Rho GTPases, mRNA splicing and/or processing, vesicular trafficking, gene transcription, and chromatin remodeling. These cell-autonomous defects and the dysregulated network of protein phosphorylation reveal a new dimension in the cellular mechanisms underlying the fundamental defects in T2D.
Collapse
Affiliation(s)
- Thiago M Batista
- Section of Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02215, USA
| | - Ashok Kumar Jayavelu
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, 82152 Martinsried, Germany
| | - Nicolai J Wewer Albrechtsen
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, 82152 Martinsried, Germany; Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark; Department of Clinical Biochemistry, Rigshospitalet, University of Copenhagen, Copenhagen 2100, Denmark
| | - Salvatore Iovino
- Section of Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02215, USA
| | - Jasmin Lebastchi
- Section of Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02215, USA
| | - Hui Pan
- Bioinformatics and Biostatistics Core, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02215, USA
| | - Jonathan M Dreyfuss
- Bioinformatics and Biostatistics Core, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02215, USA
| | - Anna Krook
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm 171 77, Sweden
| | - Juleen R Zierath
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark; Section of Integrative Physiology, Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm 171 76, Sweden
| | - Matthias Mann
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, 82152 Martinsried, Germany; Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
| | - C Ronald Kahn
- Section of Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02215, USA.
| |
Collapse
|