1
|
Stratmann B, Eggers B, Mattern Y, de Carvalho TS, Marcus-Alic K, Tschoepe D. Maladaptive response following glucose overload in GLUT4-overexpressing H9C2 cardiomyoblasts. Diabetes Obes Metab 2024; 26:2379-2389. [PMID: 38528822 DOI: 10.1111/dom.15553] [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: 09/25/2023] [Revised: 02/29/2024] [Accepted: 02/29/2024] [Indexed: 03/27/2024]
Abstract
BACKGROUND Glucose overload drives diabetic cardiomyopathy by affecting the tricarboxylic acid pathway. However, it is still unknown how cells could overcome massive chronic glucose influx on cellular and structural level. METHODS/MATERIALS Expression profiles of hyperglycemic, glucose transporter-4 (GLUT4) overexpressing H9C2 (KE2) cardiomyoblasts loaded with 30 mM glucose (KE230L) and wild type (WT) cardiomyoblasts loaded with 30 mM glucose (WT30L) were compared using proteomics, real-time polymerase quantitative chain reaction analysis, or Western blotting, and immunocytochemistry. RESULTS The findings suggest that hyperglycemic insulin-sensitive cells at the onset of diabetic cardiomyopathy present complex changes in levels of structural cell-related proteins like tissue inhibitor of metalloproteases-1 (1.3 fold), intercellular adhesion molecule 1 (1.8 fold), type-IV-collagen (3.2 fold), chaperones (Glucose-Regulated Protein 78: 1.8 fold), autophagy (Autophagosome Proteins LC3A, LC3B: 1.3 fold), and in unfolded protein response (UPR; activating transcription factor 6α expression: 2.3 fold and processing: 2.4 fold). Increased f-actin levels were detectable with glucose overload by immnocytochemistry. Effects on energy balance (1.6 fold), sirtuin expression profile (Sirtuin 1: 0.7 fold, sirtuin 3: 1.9 fold, and sirtuin 6: 4.2 fold), and antioxidant enzymes (Catalase: 0.8 fold and Superoxide dismutase 2: 1.5 fold) were detected. CONCLUSION In conclusion, these findings implicate induction of chronic cell distress by sustained glucose accumulation with a non-compensatory repair reaction not preventing final cell death. This might explain the chronic long lasting pathogenesis observed in developing heart failure in diabetes mellitus.
Collapse
Affiliation(s)
- Bernd Stratmann
- Herz- and Diabeteszentrum NRW, Diabeteszentrum, Ruhr-Universität Bochum, Bad Oeynhausen, Germany
| | - Britta Eggers
- Medizinisches Proteom-Center, Medical Faculty, Ruhr-University Bochum, Bochum, Germany
- Medical Proteome Analysis, Centre for Protein Diagnostics (PRODI), Ruhr-University Bochum, Bochum, Germany
| | - Yvonne Mattern
- Herz- and Diabeteszentrum NRW, Diabeteszentrum, Ruhr-Universität Bochum, Bad Oeynhausen, Germany
| | - Tayana Silva de Carvalho
- Herz- and Diabeteszentrum NRW, Diabeteszentrum, Ruhr-Universität Bochum, Bad Oeynhausen, Germany
| | - Katrin Marcus-Alic
- Medizinisches Proteom-Center, Medical Faculty, Ruhr-University Bochum, Bochum, Germany
- Medical Proteome Analysis, Centre for Protein Diagnostics (PRODI), Ruhr-University Bochum, Bochum, Germany
| | - Diethelm Tschoepe
- Herz- and Diabeteszentrum NRW, Diabeteszentrum, Ruhr-Universität Bochum, Bad Oeynhausen, Germany
- Stiftung DHD (Der herzkranke Diabetiker) Stiftung in der Deutschen Diabetes-Stiftung, Bad Oeynhausen, Germany
| |
Collapse
|
2
|
Dreher SI, Grubba P, von Toerne C, Moruzzi A, Maurer J, Goj T, Birkenfeld AL, Peter A, Loskill P, Hauck SM, Weigert C. IGF1 promotes human myotube differentiation toward a mature metabolic and contractile phenotype. Am J Physiol Cell Physiol 2024; 326:C1462-C1481. [PMID: 38690930 PMCID: PMC11371365 DOI: 10.1152/ajpcell.00654.2023] [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: 11/29/2023] [Revised: 02/26/2024] [Accepted: 02/26/2024] [Indexed: 05/03/2024]
Abstract
Skeletal muscle mediates the beneficial effects of exercise, thereby improving insulin sensitivity and reducing the risk for type 2 diabetes. Current human skeletal muscle models in vitro are incapable of fully recapitulating its physiological functions especially muscle contractility. By supplementation of insulin-like growth factor 1 (IGF1), a growth factor secreted by myofibers in vivo, we aimed to overcome these limitations. We monitored the differentiation process starting from primary human CD56-positive myoblasts in the presence/absence of IGF1 in serum-free medium in daily collected samples for 10 days. IGF1-supported differentiation formed thicker multinucleated myotubes showing physiological contraction upon electrical pulse stimulation (EPS) following day 6. Myotubes without IGF1 were almost incapable of contraction. IGF1 treatment shifted the proteome toward skeletal muscle-specific proteins that contribute to myofibril and sarcomere assembly, striated muscle contraction, and ATP production. Elevated PPARGC1A, MYH7, and reduced MYH1/2 suggest a more oxidative phenotype further demonstrated by higher abundance of proteins of the respiratory chain and elevated mitochondrial respiration. IGF1-treatment also upregulated glucose transporter (GLUT)4 and increased insulin-dependent glucose uptake compared with myotubes differentiated without IGF1. To conclude, addition of IGF1 to serum-free medium significantly improves the differentiation of human myotubes that showed enhanced myofibril formation, response to electrical pulse stimulation, oxidative respiratory capacity, and glucose metabolism overcoming limitations of previous standards. This novel protocol enables investigation of muscular exercise on a molecular level.NEW & NOTEWORTHY Human skeletal muscle models are highly valuable to study how exercise prevents type 2 diabetes without invasive biopsies. Current models did not fully recapitulate the function of skeletal muscle especially during exercise. By supplementing insulin-like growth factor 1 (IGF1), the authors developed a functional human skeletal muscle model characterized by inducible contractility and increased oxidative and insulin-sensitive metabolism. The novel protocol overcomes the limitations of previous standards and enables investigation of exercise on a molecular level.
Collapse
Affiliation(s)
- Simon I Dreher
- Department for Diagnostic Laboratory Medicine, Institute for Clinical Chemistry and Pathobiochemistry, University Hospital Tübingen, Tübingen, Germany
| | - Paul Grubba
- Department for Diagnostic Laboratory Medicine, Institute for Clinical Chemistry and Pathobiochemistry, University Hospital Tübingen, Tübingen, Germany
| | - Christine von Toerne
- Metabolomics and Proteomics Core Helmholtz Center Munich, German Research Center for Environmental Health, Neuherberg, Germany
| | - Alessia Moruzzi
- NMI Natural and Medical Sciences Institute at the University of Tübingen, Reutlingen, Germany
- Department for Microphysiological Systems, Institute of Biomedical Engineering, Faculty of Medicine, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Jennifer Maurer
- Department for Diagnostic Laboratory Medicine, Institute for Clinical Chemistry and Pathobiochemistry, University Hospital Tübingen, Tübingen, Germany
| | - Thomas Goj
- Department for Diagnostic Laboratory Medicine, Institute for Clinical Chemistry and Pathobiochemistry, University Hospital Tübingen, Tübingen, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Andreas L Birkenfeld
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Zentrum München, University of Tübingen, Tübingen, Germany
- Department of Internal Medicine IV, University Hospital Tübingen, Tübingen, Germany
| | - Andreas Peter
- Department for Diagnostic Laboratory Medicine, Institute for Clinical Chemistry and Pathobiochemistry, University Hospital Tübingen, Tübingen, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Zentrum München, University of Tübingen, Tübingen, Germany
| | - Peter Loskill
- NMI Natural and Medical Sciences Institute at the University of Tübingen, Reutlingen, Germany
- Department for Microphysiological Systems, Institute of Biomedical Engineering, Faculty of Medicine, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Stefanie M Hauck
- Metabolomics and Proteomics Core Helmholtz Center Munich, German Research Center for Environmental Health, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Cora Weigert
- Department for Diagnostic Laboratory Medicine, Institute for Clinical Chemistry and Pathobiochemistry, University Hospital Tübingen, Tübingen, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Zentrum München, University of Tübingen, Tübingen, Germany
| |
Collapse
|
3
|
Zanni-Ruiz E, Mayorga L, Pavarotti M. Flow cytometry protocol for GLUT4-myc detection on cell surfaces. Biosci Rep 2024; 44:BSR20231987. [PMID: 38533799 PMCID: PMC11016532 DOI: 10.1042/bsr20231987] [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: 11/22/2023] [Revised: 03/05/2024] [Accepted: 03/26/2024] [Indexed: 03/28/2024] Open
Abstract
Insulin and muscle contraction trigger GLUT4 translocation to the plasma membrane, which increases glucose uptake by muscle cells. Insulin resistance and Type 2 diabetes are the result of impaired GLUT4 translocation. Quantifying GLUT4 translocation is essential for comprehending the intricacies of both physiological and pathophysiological processes involved in glucose metabolism. The most commonly used methods for measuring GLUT4 translocation are the ELISA-type assay and the immunofluorescence assay. While some reports suggest that flow cytometry could be useful in quantifying GLUT4 translocation, this technique is not frequently used. Much of our current understanding of the regulation of GLUT4 has been based on experiments using the rat myoblast cell line (L6 cell) which expresses GLUT4 with a myc epitope on the exofacial loop. In the present study, we use the L6-GLUT4myc cell line to develop a flow cytometry-based approach to detect GLUT4 translocation. Flow cytometry offers the advantages of both immunofluorescence and ELISA-based assays. It allows easy identification of separate cell populations in the sample, similar to immunofluorescence, while providing results based on a population-level analysis of multiple individual cells, like an ELISA-based assay. Our results demonstrate a 0.6-fold increase with insulin stimulation compared with basal conditions. Finally, flow cytometry consistently yielded results across different experiments and exhibited sensitivity under the tested conditions.
Collapse
Affiliation(s)
- Emilia Zanni-Ruiz
- Laboratorio de Transporte Intracelular, Instituto de Histología y Embriología de Mendoza Dr. Mario H Burgos, Mendoza, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
- Universidad Nacional de Cuyo, Mendoza, Argentina
| | - Luis Segundo Mayorga
- Laboratorio de Transporte Intracelular, Instituto de Histología y Embriología de Mendoza Dr. Mario H Burgos, Mendoza, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
- Facultad de Ciencias Exactas y Naturales, Universidad Nacional de Cuyo, Mendoza, Argentina
- Universidad Nacional de Cuyo, Mendoza, Argentina
| | - Martin Alejandro Pavarotti
- Laboratorio de Transporte Intracelular, Instituto de Histología y Embriología de Mendoza Dr. Mario H Burgos, Mendoza, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
- Universidad Nacional de Cuyo, Mendoza, Argentina
| |
Collapse
|
4
|
Suaifan GARY, Alkhawaja B, Shehadeh MB, Sharmaa M, Hor Kuan C, Okechukwu PN. Glucosamine substituted sulfonylureas: IRS-PI3K-PKC-AKT-GLUT4 insulin signalling pathway intriguing agent. RSC Med Chem 2024; 15:695-703. [PMID: 38389876 PMCID: PMC10880904 DOI: 10.1039/d3md00647f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 12/30/2023] [Indexed: 02/24/2024] Open
Abstract
Normally, skeletal muscle accounts for 70-80% of insulin-stimulated glucose uptake in the postprandial hyperglycemia state. Consequently, abnormalities in glucose uptake by skeletal muscle or insulin resistance (IR) are deemed as initial metabolic defects in the pathogenesis of type 2 diabetes mellitus (T2DM). Globally, T2DM is growing in exponential proportion. The majority of T2DM patients are treated with sulfonylureas in combination with other drugs to improve insulin sensitivity. Glycosylated sulfonylureas (sulfonylurea-glucosamine analogues) are modified analogues of sulfonylurea that have been previously reported to possess antidiabetic activity. The aim of this study was to evaluate the impact of glycosylated sulfonylureas on the insulin signalling pathway at the molecular level using L6 skeletal muscle cell (in vitro) and extracted soleus muscle (ex vivo) models. To create an in vitro model, insulin resistance was established utilizing a high insulin-glucose approach in differentiated L6 muscle cells from Rattus norvegicus. Additionally, for the ex vivo model, extracted soleus muscles, adult Sprague-Dawley rats were subjected to a solution containing 25 mmol L-1 glucose and 100 mmol L-1 insulin for 24 hours to induce insulin resistance. After insulin resistance, compounds under investigation and standard medicines (metformin and glimepiride) were tested. The differential expression of PI3K, IRS-1, PKC, AKT2, and GLUT4 genes involved in the insulin signaling pathway was evaluated using qPCR. The evaluated glycosylated sulfonylurea analogues exhibited a significant increase in the gene expression of insulin-dependent pathways both in vitro and ex vivo, confirming the rejuvenation of the impaired insulin signaling pathway genes. Altogether, glycosylated sulfonylurea analogues described in this study represent potential therapeutic anti-diabetic drugs.
Collapse
Affiliation(s)
- Ghadeer A R Y Suaifan
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, The University of Jordan Amman 11942 Jordan
| | - Bayan Alkhawaja
- Faculty of Pharmacy and Medical Sciences, University of Petra Amman 11196 Jordan
| | - Mayadah B Shehadeh
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, The University of Jordan Amman 11942 Jordan
| | - Mridula Sharmaa
- Department of Food and Nutrition, Faculty of Applied Sciences, UCSI University Kuala Lumpur 56000 Malaysia
| | - Chan Hor Kuan
- Department of Biotechnology, Faculty of Applied Sciences, UCSI University Kuala Lumpur 56000 Malaysia
- Faculty of Pharmacy and Medical Sciences, University of Petra Amman 11196 Jordan
| | - Patrick Nwabueze Okechukwu
- Department of Food and Nutrition, Faculty of Applied Sciences, UCSI University Kuala Lumpur 56000 Malaysia
| |
Collapse
|
5
|
Zuhri UM, Yuliana ND, Fadilah F, Erlina L, Purwaningsih EH, Khatib A. Exploration of the main active metabolites from Tinospora crispa (L.) Hook. f. & Thomson stem as insulin sensitizer in L6.C11 skeletal muscle cell by integrating in vitro, metabolomics, and molecular docking. JOURNAL OF ETHNOPHARMACOLOGY 2024; 319:117296. [PMID: 37820996 DOI: 10.1016/j.jep.2023.117296] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 10/05/2023] [Accepted: 10/06/2023] [Indexed: 10/13/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Tinospora crispa (L.) Hook. f. & Thomson stem (TCS) has long been used as folk medicine for the treatment of diabetes mellitus. Previous study revealed that TCS possesses multi-ingredients and multi-targets characteristic potential as insulin sensitizer activity. However, its mechanisms of action and molecular targets are still obscure. AIM OF THE STUDY In the present study, we investigated the effects of TCS against insulin resistance in muscle cells through integrating in vitro experiment and identifying its active biomarker using metabolomics and in molecular docking validation. MATERIALS AND METHODS We used centrifugal partition chromatography (CPC) to isolate 33 fractions from methanolic extract of TCS, and then used UHPLC-Orbitrap-HRMS to identify the detectable metabolites in each fraction. We assessed the insulin sensitization activity of each fraction using enzyme-linked immunosorbent assay (ELISA), and then used confocal immunocytochemistry microscopy to measure the translocation of glucose transporter 4 (GLUT4) to the cell membrane. The identified active metabolites were further simulated for its molecular docking interaction using Autodock Tools. RESULTS The polar fractions of TCS significantly increased insulin sensitivity, as measured by the inhibition of phosphorylated insulin receptor substrate-1 (pIRS1) at serine-312 residue (ser312) also the increasing number of translocated GLUT4 and glycogen content. We identified 58 metabolites of TCS, including glycosides, flavonoids, alkaloids, coumarins, and nucleotides groups. The metabolomics and molecular docking simulations showed the presence of minor metabolites consisting of tinoscorside D, higenamine, and tinoscorside A as the active compounds. CONCLUSIONS Our findings suggest that TCS is a promising new treatment for insulin resistance and the identification of the active metabolites in TCS could lead to the development of new drugs therapies for diabetes that target these pathways.
Collapse
Affiliation(s)
- Ummu Mastna Zuhri
- Doctoral Program in Biomedical Science, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - Nancy Dewi Yuliana
- Department of Food Science and Technology, Bogor Agricultural University, Bogor, Indonesia
| | - Fadilah Fadilah
- Doctoral Program in Biomedical Science, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia; Department of Medical Chemistry, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia.
| | - Linda Erlina
- Department of Medical Chemistry, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - Erni Hernawati Purwaningsih
- Doctoral Program in Biomedical Science, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia; Department of Medical Pharmacy, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - Alfi Khatib
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, International Islamic University Malaysia, Kuantan, Malaysia
| |
Collapse
|
6
|
Yang AJT, Mohammad A, Finch MS, Tsiani E, Spencer G, Necakov A, MacPherson REK. Influence of metabolic stress and metformin on synaptic protein profile in SH-SY5Y-derived neurons. Physiol Rep 2023; 11:10.14814/phy2.15852. [PMID: 38010200 PMCID: PMC10680579 DOI: 10.14814/phy2.15852] [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: 08/28/2023] [Revised: 10/02/2023] [Accepted: 10/02/2023] [Indexed: 11/29/2023] Open
Abstract
Insulin resistance (IR) is associated with reductions in neuronal proteins often observed with Alzheimer's disease (AD), however, the mechanisms through which IR promotes neurodegeneration/AD pathogenesis are poorly understood. Metformin (MET), a potent activator of the metabolic regulator AMPK is used to treat IR but its effectiveness for AD is unclear. We have previously shown that chronic AMPK activation impairs neurite growth and protein synthesis in SH-SY5Y neurons, however, AMPK activation in IR was not explored. Therefore, we examined the effects of MET-driven AMPK activation with and without IR. Retinoic acid-differentiated SH-SY5Y neurons were treated with: (1) Ctl: 24 h vehicle followed by 24 h Vehicle; (2) HI: 100 nM insulin (24 h HI followed by 24 h HI); or (3) MET: 24 h vehicle followed by 24 h 2 mM metformin; (4) HI/MET: 24 h 100 nM insulin followed by 24 h 100 nM INS+2 mM MET. INS and INS/MET groups saw impairments in markers of insulin signaling (Akt S473, mTOR S2448, p70s6k T389, and IRS-1S636) demonstrating IR was not recovered with MET treatment. All treatment groups showed reductions in neuronal markers (post-synaptic marker HOMER1 mRNA content and synapse marker synaptophysin protein content). INS and MET treatments showed a reduction in the content of the mature neuronal marker NeuN that was prevented by INS/MET. Similarly, increases in cell size/area, neurite length/area observed with INS and MET, were prevented with INS/MET. These findings indicate that IR and MET impair neuronal markers through distinct pathways and suggest that MET is ineffective in treating IR-driven impairments in neurons.
Collapse
Affiliation(s)
- Alex J. T. Yang
- Department of Health Sciences, Faculty of Applied Health SciencesBrock UniversitySt CatharinesOntarioCanada
| | - Ahmad Mohammad
- Department of Health Sciences, Faculty of Applied Health SciencesBrock UniversitySt CatharinesOntarioCanada
| | - Michael S. Finch
- Department of Health Sciences, Faculty of Applied Health SciencesBrock UniversitySt CatharinesOntarioCanada
| | - Evangelia Tsiani
- Department of Health Sciences, Faculty of Applied Health SciencesBrock UniversitySt CatharinesOntarioCanada
| | - Gaynor Spencer
- Department of Biological SciencesBrock UniversitySt CatharinesOntarioCanada
- Centre for NeuroscienceBrock UniversitySt. CatharinesOntarioCanada
| | - Aleksandar Necakov
- Department of Biological SciencesBrock UniversitySt CatharinesOntarioCanada
- Centre for NeuroscienceBrock UniversitySt. CatharinesOntarioCanada
| | - Rebecca E. K. MacPherson
- Department of Health Sciences, Faculty of Applied Health SciencesBrock UniversitySt CatharinesOntarioCanada
- Centre for NeuroscienceBrock UniversitySt. CatharinesOntarioCanada
| |
Collapse
|
7
|
Antonescu CN, Ishikura S, Bilan PJ, Klip A. Measurement of GLUT4 Traffic to and from the Cell Surface in Muscle Cells. Curr Protoc 2023; 3:e803. [PMID: 37367531 DOI: 10.1002/cpz1.803] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/28/2023]
Abstract
Elevated blood glucose following a meal is cleared by insulin-stimulated glucose entry into muscle and fat cells. The hormone increases the amount of the glucose transporter GLUT4 at the plasma membrane in these tissues at the expense of preformed intracellular pools. In addition, muscle contraction also increases glucose uptake via a gain in GLUT4 at the plasma membrane. Regulation of GLUT4 levels at the cell surface could arise from alterations in the rate of its exocytosis, endocytosis, or both. Hence, methods that can independently measure these traffic parameters for GLUT4 are essential to understanding the mechanism of regulation of membrane traffic of the transporter. Here, we describe cell population-based assays to measure the steady-state levels of GLUT4 at the cell surface, as well as to separately measure the rates of GLUT4 endocytosis and endocytosis. © 2023 Wiley Periodicals LLC. Basic Protocol 1: Measuring steady-state cell surface GLUT4myc Basic Protocol 2: Measuring steady-state cell surface GLUT4-HA Basic Protocol 3: Measuring GLUT4myc endocytosis Basic Protocol 4: Measuring GLUT4myc exocytosis.
Collapse
Affiliation(s)
- Costin N Antonescu
- Department of Chemistry and Biology, Toronto Metropolitan University, Toronto, Canada
| | | | - Philip J Bilan
- Program in Cell Biology, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Amira Klip
- Program in Cell Biology, The Hospital for Sick Children, Toronto, Ontario, Canada
| |
Collapse
|
8
|
van Laar A, Grootaert C, Rajkovic A, Desmet T, Beerens K, Van Camp J. Rare Sugar Metabolism and Impact on Insulin Sensitivity along the Gut-Liver-Muscle Axis In Vitro. Nutrients 2023; 15:1593. [PMID: 37049441 PMCID: PMC10096767 DOI: 10.3390/nu15071593] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/21/2023] [Accepted: 03/22/2023] [Indexed: 03/29/2023] Open
Abstract
Rare sugars have recently attracted attention as potential sugar replacers. Understanding the biochemical and biological behavior of these sugars is of importance in (novel) food formulations and prevention of type 2 diabetes. In this study, we investigated whether rare sugars may positively affect intestinal and liver metabolism, as well as muscle insulin sensitivity, compared to conventional sugars. Rare disaccharide digestibility, hepatic metabolism of monosaccharides (respirometry) and the effects of sugars on skeletal muscle insulin sensitivity (impaired glucose uptake) were investigated in, respectively, Caco-2, HepG2 and L6 cells or a triple coculture model with these cells. Glucose and fructose, but not l-arabinose, acutely increased extracellular acidification rate (ECAR) responses in HepG2 cells and impaired glucose uptake in L6 cells following a 24 h exposure at 28 mM. Cellular bioenergetics and digestion experiments with Caco-2 cells indicate that especially trehalose (α1-1α), D-Glc-α1,2-D-Gal, D-Glc-α1,2-D-Rib and D-Glc-α1,3-L-Ara experience delayed digestion and reduced cellular impact compared to maltose (α1-4), without differences on insulin-stimulated glucose uptake in a short-term setup with a Caco-2/HepG2/L6 triple coculture. These results suggest a potential for l-arabinose and specific rare disaccharides to improve metabolic health; however, additional in vivo research with longer sugar exposures should confirm their beneficial impact on insulin sensitivity in humans.
Collapse
Affiliation(s)
- Amar van Laar
- NutriFOODChem, Department of Food Technology, Safety and Health, Faculty of Bioscience Engineering, Ghent University, 9000 Ghent, Belgium
| | - Charlotte Grootaert
- NutriFOODChem, Department of Food Technology, Safety and Health, Faculty of Bioscience Engineering, Ghent University, 9000 Ghent, Belgium
| | - Andreja Rajkovic
- NutriFOODChem, Department of Food Technology, Safety and Health, Faculty of Bioscience Engineering, Ghent University, 9000 Ghent, Belgium
- Food Microbiology and Food Preservation, Department of Food Technology, Safety and Health, Faculty of Bioscience Engineering, Ghent University, 9000 Ghent, Belgium
| | - Tom Desmet
- Centre for Synthetic Biology, Department of Biotechnology, Faculty of Bioscience Engineering, Ghent University, 9000 Ghent, Belgium
| | - Koen Beerens
- Centre for Synthetic Biology, Department of Biotechnology, Faculty of Bioscience Engineering, Ghent University, 9000 Ghent, Belgium
| | - John Van Camp
- NutriFOODChem, Department of Food Technology, Safety and Health, Faculty of Bioscience Engineering, Ghent University, 9000 Ghent, Belgium
| |
Collapse
|
9
|
Sheng CY, Son YH, Jang J, Park SJ. In vitro skeletal muscle models for type 2 diabetes. BIOPHYSICS REVIEWS 2022; 3:031306. [PMID: 36124295 PMCID: PMC9478902 DOI: 10.1063/5.0096420] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Accepted: 08/18/2022] [Indexed: 06/15/2023]
Abstract
Type 2 diabetes mellitus, a metabolic disorder characterized by abnormally elevated blood sugar, poses a growing social, economic, and medical burden worldwide. The skeletal muscle is the largest metabolic organ responsible for glucose homeostasis in the body, and its inability to properly uptake sugar often precedes type 2 diabetes. Although exercise is known to have preventative and therapeutic effects on type 2 diabetes, the underlying mechanism of these beneficial effects is largely unknown. Animal studies have been conducted to better understand the pathophysiology of type 2 diabetes and the positive effects of exercise on type 2 diabetes. However, the complexity of in vivo systems and the inability of animal models to fully capture human type 2 diabetes genetics and pathophysiology are two major limitations in these animal studies. Fortunately, in vitro models capable of recapitulating human genetics and physiology provide promising avenues to overcome these obstacles. This review summarizes current in vitro type 2 diabetes models with focuses on the skeletal muscle, interorgan crosstalk, and exercise. We discuss diabetes, its pathophysiology, common in vitro type 2 diabetes skeletal muscle models, interorgan crosstalk type 2 diabetes models, exercise benefits on type 2 diabetes, and in vitro type 2 diabetes models with exercise.
Collapse
Affiliation(s)
- Christina Y. Sheng
- Biohybrid Systems Group, Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University School of Medicine, Atlanta, Georgia 30322, USA
| | - Young Hoon Son
- Biohybrid Systems Group, Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University School of Medicine, Atlanta, Georgia 30322, USA
| | | | - Sung-Jin Park
- Biohybrid Systems Group, Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University School of Medicine, Atlanta, Georgia 30322, USA
| |
Collapse
|
10
|
Pavlovic K, Krako Jakovljevic N, Isakovic AM, Ivanovic T, Markovic I, Lalic NM. Therapeutic vs. Suprapharmacological Metformin Concentrations: Different Effects on Energy Metabolism and Mitochondrial Function in Skeletal Muscle Cells in vitro. Front Pharmacol 2022; 13:930308. [PMID: 35873556 PMCID: PMC9299382 DOI: 10.3389/fphar.2022.930308] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 06/16/2022] [Indexed: 11/30/2022] Open
Abstract
Metformin is an oral antidiabetic agent that has been widely used in clinical practice for over 60 years, and is currently the most prescribed antidiabetic drug worldwide. However, the molecular mechanisms of metformin action in different tissues are still not completely understood. Although metformin-induced inhibition of mitochondrial respiratory chain Complex I and activation of AMP-activated protein kinase have been observed in many studies, published data is inconsistent. Furthermore, metformin concentrations used for in vitro studies and their pharmacological relevance are a common point of debate. The aim of this study was to explore the effects of different metformin concentrations on energy metabolism and activity of relevant signaling pathways in C2C12 muscle cells in vitro. In order to determine if therapeutic metformin concentrations have an effect on skeletal muscle cells, we used micromolar metformin concentrations (50 µM), and compared the effects with those of higher, millimolar concentrations (5 mM), that have already been established to affect mitochondrial function and AMPK activity. We conducted all experiments in conditions of high (25 mM) and low glucose (5.5 mM) concentration, in order to discern the role of glucose availability on metformin action. According to our results, micromolar metformin treatment did not cause Complex I inhibition nor AMPK activation. Also, cells cultured in low glucose medium were more sensitive to Complex I inhibition, mitochondrial membrane depolarization and AMPK activation by millimolar metformin, but cells cultured in high glucose medium were more prone to induction of ROS production. In conclusion, even though suprapharmacological metformin concentrations cause Complex I inhibition and AMPK activation in skeletal muscle cells in vitro, therapeutic concentrations cause no such effect. This raises the question if these mechanisms are relevant for therapeutic effects of metformin in skeletal muscle.
Collapse
Affiliation(s)
- Kasja Pavlovic
- Clinic for Endocrinology, Diabetes and Metabolic Diseases, University Clinical Center of Serbia, Belgrade, Serbia
| | - Nina Krako Jakovljevic
- Clinic for Endocrinology, Diabetes and Metabolic Diseases, University Clinical Center of Serbia, Belgrade, Serbia
| | - Andjelka M Isakovic
- Faculty of Medicine, Institute of Medical and Clinical Biochemistry, University of Belgrade, Belgrade, Serbia
| | - Tijana Ivanovic
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Ivanka Markovic
- Faculty of Medicine, Institute of Medical and Clinical Biochemistry, University of Belgrade, Belgrade, Serbia
| | - Nebojsa M Lalic
- Clinic for Endocrinology, Diabetes and Metabolic Diseases, University Clinical Center of Serbia, Belgrade, Serbia
| |
Collapse
|
11
|
Stratmann B, Eggers B, Mattern Y, Silva de Carvalho T, Marcus K, Tschoepe D. Chronic Hyperglycaemia Inhibits Tricarboxylic Acid Cycle in Rat Cardiomyoblasts Overexpressing Glucose Transporter Type 4. Int J Mol Sci 2022; 23:ijms23137255. [PMID: 35806260 PMCID: PMC9266806 DOI: 10.3390/ijms23137255] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 06/23/2022] [Accepted: 06/27/2022] [Indexed: 11/26/2022] Open
Abstract
An oversupply of nutrients with a loss of metabolic flexibility and subsequent cardiac dysfunction are hallmarks of diabetic cardiomyopathy. Even if excess substrate is offered, the heart suffers energy depletion as metabolic fluxes are diminished. To study the effects of a high glucose supply, a stably glucose transporter type 4 (GLUT4)-overexpressing cell line presenting an onset of diabetic cardiomyopathy-like phenotype was established. Long-term hyperglycaemia effects were analysed. Rat cardiomyoblasts overexpressing GLUT4 (H9C2KE2) were cultured under normo- and hyperglycaemic conditions for long-term. Expression profiles of several proteins were compared to non-transfected H9C2 cells (H9C2) using RT-qPCR, proteomics-based analysis, or Western blotting. GLUT4 surface analysis, glucose uptake, and cell morphology changes as well as apoptosis/necrosis measurements were performed using flow cytometry. Additionally, brain natriuretic peptide (BNP) levels, reactive oxygen species (ROS) formation, glucose consumption, and lactate production were quantified. Long-term hyperglycaemia in H9C2KE2 cells induced increased GLUT4 presence on the cell surface and was associated with exaggerated glucose influx and lactate production. On the metabolic level, hyperglycaemia affected the tricarboxylic acid (TCA) cycle with accumulation of fumarate. This was associated with increased BNP-levels, oxidative stress, and lower antioxidant response, resulting in pronounced apoptosis and necrosis. Chronic glucose overload in cardiomyoblasts induced by GLUT4 overexpression and hyperglycaemia resulted in metabolically stimulated proteome profile changes and metabolic alterations on the TCA level.
Collapse
Affiliation(s)
- Bernd Stratmann
- Herz- and Diabeteszentrum NRW, Diabeteszentrum, Ruhr Universität Bochum, 32545 Bad Oeynhausen, Germany; (Y.M.); (T.S.d.C.); (D.T.)
- Correspondence: ; Tel.: +49-(0)-5731/973768
| | - Britta Eggers
- Medizinisches Proteom-Center, Centre for Translational and Behavioural Neurosciences, Medical Faculty, Ruhr-University Bochum, 44801 Bochum, Germany; (B.E.); (K.M.)
- Medical Proteome Analysis, Centre for Protein Diagnostics (PRODI), Ruhr-University Bochum, 44801 Bochum, Germany
| | - Yvonne Mattern
- Herz- and Diabeteszentrum NRW, Diabeteszentrum, Ruhr Universität Bochum, 32545 Bad Oeynhausen, Germany; (Y.M.); (T.S.d.C.); (D.T.)
| | - Tayana Silva de Carvalho
- Herz- and Diabeteszentrum NRW, Diabeteszentrum, Ruhr Universität Bochum, 32545 Bad Oeynhausen, Germany; (Y.M.); (T.S.d.C.); (D.T.)
| | - Katrin Marcus
- Medizinisches Proteom-Center, Centre for Translational and Behavioural Neurosciences, Medical Faculty, Ruhr-University Bochum, 44801 Bochum, Germany; (B.E.); (K.M.)
- Medical Proteome Analysis, Centre for Protein Diagnostics (PRODI), Ruhr-University Bochum, 44801 Bochum, Germany
| | - Diethelm Tschoepe
- Herz- and Diabeteszentrum NRW, Diabeteszentrum, Ruhr Universität Bochum, 32545 Bad Oeynhausen, Germany; (Y.M.); (T.S.d.C.); (D.T.)
- Stiftung DHD (Der herzkranke Diabetiker) Stiftung in der Deutschen Diabetes-Stiftung, 32545 Bad Oeynhausen, Germany
| |
Collapse
|
12
|
Komati A, Anand A, Nagendla NK, Madhusudana K, Mudiam MKR, Babu KS, Tiwari AK. Bombax ceiba
calyx displays antihyperglycemic activity via improving insulin secretion and sensitivity: Identification of bioactive phytometabolomes by UPLC‐QTof‐MS/MS. J Food Sci 2022; 87:1865-1881. [DOI: 10.1111/1750-3841.16093] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 01/04/2022] [Accepted: 02/01/2022] [Indexed: 12/28/2022]
Affiliation(s)
- Anusha Komati
- Centre for Natural Products & Traditional Knowledge CSIR‐Indian Institute of Chemical Technology Hyderabad India
- Academy of Scientific & Innovative Research (AcSIR) Ghaziabad India
| | - Ajay Anand
- Centre for Natural Products & Traditional Knowledge CSIR‐Indian Institute of Chemical Technology Hyderabad India
- Academy of Scientific & Innovative Research (AcSIR) Ghaziabad India
- Carver College of Medicine, Department of Pathology, University Of Iowa Iowa City USA
| | - Narendra Kumar Nagendla
- Analytical & Structural Chemistry Department CSIR‐Indian Institute of Chemical Technology Hyderabad India
- Academy of Scientific & Innovative Research (AcSIR) Ghaziabad India
| | - Kuncha Madhusudana
- Applied Biology Division CSIR‐Indian Institute of Chemical Technology Hyderabad India
| | - Mohana Krishna Reddy Mudiam
- Analytical & Structural Chemistry Department CSIR‐Indian Institute of Chemical Technology Hyderabad India
- Academy of Scientific & Innovative Research (AcSIR) Ghaziabad India
| | - Katragadda Suresh Babu
- Centre for Natural Products & Traditional Knowledge CSIR‐Indian Institute of Chemical Technology Hyderabad India
- Academy of Scientific & Innovative Research (AcSIR) Ghaziabad India
| | - Ashok Kumar Tiwari
- Centre for Natural Products & Traditional Knowledge CSIR‐Indian Institute of Chemical Technology Hyderabad India
- Academy of Scientific & Innovative Research (AcSIR) Ghaziabad India
| |
Collapse
|
13
|
Sung HK, Mitchell PL, Gross S, Marette A, Sweeney G. ALY688 elicits adiponectin-mimetic signaling and improves insulin action in skeletal muscle cells. Am J Physiol Cell Physiol 2021; 322:C151-C163. [PMID: 34910600 DOI: 10.1152/ajpcell.00603.2020] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Adiponectin is well established to mediate many beneficial metabolic effects, and this has stimulated great interest in development and validation of adiponectin receptor agonists as pharmaceutical tools. This study investigated the effects of ALY688, a peptide-based adiponectin receptor agonist, in rat L6 skeletal muscle cells. ALY688 significantly increased phosphorylation of several adiponectin downstream effectors, including AMPK, ACC and p38MAPK, assessed by immunoblotting and immunofluorescence microscopy. Temporal analysis using cells expressing an Akt biosensor demonstrated that ALY688 enhanced insulin sensitivity. This effect was associated with increased insulin-stimulated Akt and IRS-1 phosphorylation. The functional metabolic significance of these signaling effects was examined by measuring glucose uptake in myoblasts stably overexpressing the glucose transporter GLUT4. ALY688 treatment both increased glucose uptake itself and enhanced insulin-stimulated glucose uptake. In the model of high glucose/high insulin (HGHI)-induced insulin resistant cells, both temporal studies using the Akt biosensor as well as immunoblotting assessing Akt and IRS-1 phosphorylation indicated that ALY688 significantly reduced insulin resistance. Importantly, we observed that ALY688 administration to high-fat high sucrose fed mice also improve glucose handling, validating its efficacy in vivo. In summary, these data indicate that ALY688 activates adiponectin signaling pathways in skeletal muscle, leading to improved insulin sensitivity and beneficial metabolic effects.
Collapse
Affiliation(s)
| | - Patricia L Mitchell
- Quebec Heart and Lung Institute (IUCPQ), and Institute of Nutrition and Functional Foods (INAF), Laval University, Quebec, Canada
| | - Sean Gross
- Department of Biomedical Engineering, OHSU Center for Spatial Systems Biomedicine, Knight Cancer Institute, Oregon Health and Sciences University, Portland, OR, United States
| | - Andre Marette
- Quebec Heart and Lung Institute (IUCPQ), and Institute of Nutrition and Functional Foods (INAF), Laval University, Quebec, Canada
| | - Gary Sweeney
- Department of Biology, York University, Toronto, ON, Canada
| |
Collapse
|
14
|
Beckerman M, Harel C, Michael I, Klip A, Bilan PJ, Gallagher EJ, LeRoith D, Lewis EC, Karnieli E, Levenberg S. GLUT4-overexpressing engineered muscle constructs as a therapeutic platform to normalize glycemia in diabetic mice. SCIENCE ADVANCES 2021; 7:eabg3947. [PMID: 34644106 PMCID: PMC8514095 DOI: 10.1126/sciadv.abg3947] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Accepted: 08/23/2021] [Indexed: 05/29/2023]
Abstract
Skeletal muscle insulin resistance is a main defect in type 2 diabetes (T2D), which is associated with impaired function and content of glucose transporter type 4 (GLUT4). GLUT4 overexpression in skeletal muscle tissue can improve glucose homeostasis. Therefore, we created an engineered muscle construct (EMC) composed of GLUT4-overexpressing (OEG4) cells. The ability of the engineered implants to reduce fasting glucose levels was tested in diet-induced obesity mice. Decrease and stabilization of basal glucose levels were apparent up to 4 months after implantation. Analysis of the retrieved constructs showed elevated expression of myokines and proteins related to metabolic processes. In addition, we validated the efficiency of OEG4-EMCs in insulin-resistant mice. Following high glucose load administration, mice showed improved glucose tolerance. Our data indicate that OEG4-EMC implant is an efficient mode for restoring insulin sensitivity and improving glucose homeostasis in diabetic mice. Such procedure is a potential innovative modality for T2D therapy.
Collapse
Affiliation(s)
- Margarita Beckerman
- Faculty of Biomedical Engineering, Technion—Israel Institute of Technology, Haifa, Israel
- Rina and Avner Schneur Center of Diabetes Research, Technion—Israel Institute of Technology, Haifa, Israel
| | - Chava Harel
- Rina and Avner Schneur Center of Diabetes Research, Technion—Israel Institute of Technology, Haifa, Israel
- Rappaport Faculty of Medicine, Technion—Israel Institute of Technology, Haifa, Israel
| | - Inbal Michael
- Faculty of Biomedical Engineering, Technion—Israel Institute of Technology, Haifa, Israel
| | - Amira Klip
- Program in Cell Biology, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Philip J. Bilan
- Program in Cell Biology, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Emily J. Gallagher
- Division of Endocrinology, Diabetes and Bone Diseases, Samuel Bronfman Department of Medicine, Ichan School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Derek LeRoith
- Division of Endocrinology, Diabetes and Bone Diseases, Samuel Bronfman Department of Medicine, Ichan School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Eli C. Lewis
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Eddy Karnieli
- Rina and Avner Schneur Center of Diabetes Research, Technion—Israel Institute of Technology, Haifa, Israel
- Rappaport Faculty of Medicine, Technion—Israel Institute of Technology, Haifa, Israel
| | - Shulamit Levenberg
- Faculty of Biomedical Engineering, Technion—Israel Institute of Technology, Haifa, Israel
- Rina and Avner Schneur Center of Diabetes Research, Technion—Israel Institute of Technology, Haifa, Israel
| |
Collapse
|
15
|
Feraco A, Gorini S, Armani A, Camajani E, Rizzo M, Caprio M. Exploring the Role of Skeletal Muscle in Insulin Resistance: Lessons from Cultured Cells to Animal Models. Int J Mol Sci 2021; 22:ijms22179327. [PMID: 34502235 PMCID: PMC8430804 DOI: 10.3390/ijms22179327] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 08/22/2021] [Accepted: 08/25/2021] [Indexed: 12/12/2022] Open
Abstract
Skeletal muscle is essential to maintain vital functions such as movement, breathing, and thermogenesis, and it is now recognized as an endocrine organ. Muscles release factors named myokines, which can regulate several physiological processes. Moreover, skeletal muscle is particularly important in maintaining body homeostasis, since it is responsible for more than 75% of all insulin-mediated glucose disposal. Alterations of skeletal muscle differentiation and function, with subsequent dysfunctional expression and secretion of myokines, play a key role in the pathogenesis of obesity, type 2 diabetes, and other metabolic diseases, finally leading to cardiometabolic complications. Hence, a deeper understanding of the molecular mechanisms regulating skeletal muscle function related to energy metabolism is critical for novel strategies to treat and prevent insulin resistance and its cardiometabolic complications. This review will be focused on both cellular and animal models currently available for exploring skeletal muscle metabolism and endocrine function.
Collapse
Affiliation(s)
- Alessandra Feraco
- Laboratory of Cardiovascular Endocrinology, IRCCS San Raffaele Roma, 00166 Rome, Italy; (A.F.); (S.G.); (A.A.)
- Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Roma Open University, 00166 Rome, Italy;
| | - Stefania Gorini
- Laboratory of Cardiovascular Endocrinology, IRCCS San Raffaele Roma, 00166 Rome, Italy; (A.F.); (S.G.); (A.A.)
| | - Andrea Armani
- Laboratory of Cardiovascular Endocrinology, IRCCS San Raffaele Roma, 00166 Rome, Italy; (A.F.); (S.G.); (A.A.)
- Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Roma Open University, 00166 Rome, Italy;
| | - Elisabetta Camajani
- Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Roma Open University, 00166 Rome, Italy;
- PhD Programme in Endocrinological Sciences, Department of Experimental Medicine, University of Rome “La Sapienza”, 00161 Rome, Italy
| | - Manfredi Rizzo
- Promise Department, School of Medicine, University of Palermo, 90127 Palermo, Italy;
| | - Massimiliano Caprio
- Laboratory of Cardiovascular Endocrinology, IRCCS San Raffaele Roma, 00166 Rome, Italy; (A.F.); (S.G.); (A.A.)
- Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Roma Open University, 00166 Rome, Italy;
- Correspondence: ; Tel.: +39-065-225-3419
| |
Collapse
|
16
|
Shamshoum H, Vlavcheski F, MacPherson REK, Tsiani E. Rosemary extract activates AMPK, inhibits mTOR and attenuates the high glucose and high insulin-induced muscle cell insulin resistance. Appl Physiol Nutr Metab 2021; 46:819-827. [PMID: 33471600 DOI: 10.1139/apnm-2020-0592] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Impaired action of insulin in skeletal muscle, termed insulin resistance, leads to increased blood glucose levels resulting in compensatory increase in insulin levels. The elevated blood glucose and insulin levels exacerbate insulin resistance and contribute to the pathogenesis of type 2 diabetes mellitus. In previous studies we found attenuation of free fatty acid-induced muscle cell insulin resistance by rosemary extract (RE). In the present study we investigated the effects of RE on high glucose (HG) and high insulin (HI)-induced muscle cell insulin resistance. Exposure of L6 myotubes to 25 mmol/L glucose and 100 nmol/L insulin for 24 h, to mimic hyperglycemia and hyperinsulinemia, abolished the acute insulin-stimulated glucose uptake, increased the serine phosphorylation of IRS-1 and the phosphorylation/activation of mTOR and p70S6K. Treatment with RE significantly improved the insulin-stimulated glucose uptake and increased the acute insulin-stimulated tyrosine phosphorylation while reducing the HG+HI-induced serine phosphorylation of IRS-1 and phosphorylation of mTOR and p70S6K. Additionally, treatment with RE significantly increased the phosphorylation of AMPK, its downstream effector ACC and the plasma membrane GLUT4 levels. Our data indicate a potential of RE to counteract muscle cell insulin resistance and more studies are required to investigate its effectiveness in vivo. Novelty: RE phosphorylated muscle cell AMPK and ACC under both normal and HG+HI conditions. The HG+HI-induced serine phosphorylation of IRS-1 and activation of mTOR and p70S6K were attenuated by RE. RE restored the insulin-stimulated glucose uptake by enhancing GLUT4 glucose transporter translocation to plasma membrane.
Collapse
Affiliation(s)
- Hesham Shamshoum
- Department of Health Sciences, Brock University, St. Catharines, ON, Canada.,Centre for Bone and Muscle Health, Brock University, St. Catharines, ON, Canada
| | - Filip Vlavcheski
- Department of Health Sciences, Brock University, St. Catharines, ON, Canada.,Centre for Bone and Muscle Health, Brock University, St. Catharines, ON, Canada
| | - Rebecca E K MacPherson
- Department of Health Sciences, Brock University, St. Catharines, ON, Canada.,Centre for Bone and Muscle Health, Brock University, St. Catharines, ON, Canada
| | - Evangelia Tsiani
- Department of Health Sciences, Brock University, St. Catharines, ON, Canada.,Centre for Bone and Muscle Health, Brock University, St. Catharines, ON, Canada
| |
Collapse
|
17
|
Pavlović K, Lalić N. Cell models for studying muscle insulin resistance. MEDICINSKI PODMLADAK 2021. [DOI: 10.5937/mp72-31381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
Abstract
Type 2 diabetes is one of the most prevalent chronic diseases in the world today. Insulin resistance - a reduced responsiveness of tissues to insulin - is a hallmark of type 2 diabetes pathology. Skeletal muscle plays a pivotal role in glucose homeostasis - it is responsible for the majority of insulin-mediated glucose disposal and thus is one of the tissues most affected by insulin resistance. To study the molecular mechanisms of a disease, researchers often turn to cell models - they are inexpensive, easy to use, and exist in a controlled environment with few unknown variables. Cell models for exploring muscle insulin resistance are constructed using primary cell cultures or immortalised cell lines and treating them with fatty acids, high insulin or high glucose concentrations. The choice of cell culture, concentration and duration of the treatment and the methods for measuring insulin sensitivity, in order to confirm the model, are rarely discussed. Choosing an appropriate and physiologically relevant model for a particular topic of interest is required in order for the results to be reproducible, relevant, comparable and translatable to more complex biological systems. Cell models enable research that would otherwise be inaccessible but, especially when studying human disease, they do not serve a purpose if they are not in line with the biological reality. This review aims to summarise and critically evaluate the most commonly used cell models of muscle insulin resistance: the rationale for choosing these exact treatments and conditions, the protocols for constructing the models and the measurable outcomes used for confirming insulin resistance in the cells.
Collapse
|
18
|
Goff M, Chen G. Long-term treatment with insulin and retinoic acid increased glucose utilization in L6 muscle cells via glycogenesis. Biochem Cell Biol 2020; 98:683-697. [PMID: 33215509 DOI: 10.1139/bcb-2020-0131] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
The skeletal muscle regulates glucose homeostasis. Here, the effects of vitamin A metabolites including retinoic acid (RA) alone, and in combination with insulin, on glucose utilization were investigated in rat L6 muscle cells during the differentiation process. L6 cells were treated with differentiation medium containing retinol, retinal, RA, and (or) insulin. The glucose levels and pH values in the medium were measured every 2 days. The expression levels of insulin signaling and glycogen synthesis proteins, as well as glycogen content were determined. Retinal and RA reduced the glucose content and pH levels in the medium of the L6 cells. RA acted synergistically with insulin to reduce glucose and pH levels in the medium. The RA- and insulin-mediated reduction of glucose in the medium only occurred when glucose levels were at or above 15 mmol/L. Insulin-induced phosphorylation of Akt Thr308 was further enhanced by RA treatment through the activation of retinoic acid receptor. RA acted synergistically with insulin to phosphorylate glycogen synthase kinase 3β, and dephosphorylate glycogen synthase (GS), which was associated with increases in the protein and mRNA levels of GS. Increases in glycogen content were induced by insulin, and was further enhanced in the presence of RA. We conclude that activation of the RA signaling pathway enhanced insulin-induced glucose utilization in differentiating L6 cells through increases in glycogenesis.
Collapse
Affiliation(s)
- Matthew Goff
- Department of Nutrition, University of Tennessee at Knoxville, Knoxville, Tennessee, USA
- Department of Nutrition, University of Tennessee at Knoxville, Knoxville, Tennessee, USA
| | - Guoxun Chen
- Department of Nutrition, University of Tennessee at Knoxville, Knoxville, Tennessee, USA
- Department of Nutrition, University of Tennessee at Knoxville, Knoxville, Tennessee, USA
| |
Collapse
|
19
|
Heydarpour F, Sajadimajd S, Mirzarazi E, Haratipour P, Joshi T, Farzaei MH, Khan H, Echeverría J. Involvement of TGF-β and Autophagy Pathways in Pathogenesis of Diabetes: A Comprehensive Review on Biological and Pharmacological Insights. Front Pharmacol 2020; 11:498758. [PMID: 33041786 PMCID: PMC7522371 DOI: 10.3389/fphar.2020.498758] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Accepted: 08/27/2020] [Indexed: 12/21/2022] Open
Abstract
Despite recent advancements in clinical drugs, diabetes treatment still needs further progress. As such, ongoing research has attempted to determine the precise molecular mechanisms of the disorder. Specifically, evidence supports that several signaling pathways play pivotal roles in the development of diabetes. However, the exact molecular mechanisms of diabetes still need to be explored. This study examines exciting new hallmarks for the strict involvement of autophagy and TGF-β signaling pathways in the pathogenesis of diabetes and the design of novel therapeutic strategies. Dysregulated autophagy in pancreatic β cells due to hyperglycemia, oxidative stress, and inflammation is associated with diabetes and accompanied by dysregulated autophagy in insulin target tissues and the progression of diabetic complications. Consequently, several therapeutic agents such as adiponectin, ezetimibe, GABA tea, geniposide, liraglutide, guava extract, and vitamin D were shown to inhibit diabetes and its complications through modulation of the autophagy pathway. Another pathway, TGF-β signaling pathway, appears to play a part in the progression of diabetes, insulin resistance, and autoimmunity in both type 1 and 2 diabetes and complications in diabetes. Subsequently, drugs that target TGF-β signaling, especially naturally derived ones such as resveratrol, puerarin, curcumin, hesperidin, and silymarin, as well as Propolis, Lycopus lucidus, and Momordica charantia extracts, may become promising alternatives to current drugs in diabetes treatment. This review provides keen insights into novel therapeutic strategies for the medical care of diabetes.
Collapse
Affiliation(s)
- Fatemeh Heydarpour
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Soraya Sajadimajd
- Departament of Biology, Faculty of Sciences, Razi University, Kermanshah, Iran
| | - Elahe Mirzarazi
- Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran
| | - Pouya Haratipour
- Department of Chemistry, Sharif University of Technology, Tehran, Iran.,PhytoPharmacology Interest Group (PPIG), Universal Scientific Education and Research Network (USERN), Los Angeles, CA, United States
| | - Tanuj Joshi
- Department of Pharmaceutical Sciences, Faculty of Technology, Kumaun University, Nainital, India
| | - Mohammad Hosein Farzaei
- Pharmaceutical Sciences Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Haroon Khan
- Department of Pharmacy, Abdul Wali Khan University, Mardan, Pakistan
| | - Javier Echeverría
- Departamento de Ciencias del Ambiente, Facultad de Química y Biología, Universidad de Santiago de Chile, Santiago, Chile
| |
Collapse
|
20
|
Govindaraju K, Suganya KSU. In vitro anti-diabetic assessment of guavanoic acid functionalized gold nanoparticles in regulating glucose transport using L6 rat skeletal muscle cells. RSC Med Chem 2020; 11:814-822. [PMID: 33479677 DOI: 10.1039/d0md00125b] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 04/27/2020] [Indexed: 12/22/2022] Open
Abstract
Guavanoic acid functionalized gold nanoparticles exhibit anti-diabetic potential by improving insulin dependent glucose uptake in L6 rat skeletal muscle cells. The mode of action of the gold nanoparticles was established from the glucose uptake assay in the presence and absence of genistein and wortmannin. The anti-diabetic efficacy of guavanoic acid functionalized gold nanoparticles was put forth by in vitro assays like for PTP 1B, α-amylase and α-glucosidase enzyme activities. Studies on cytotoxicity revealed 50% inhibition of cells at 265 ± 0.01 μg mL-1. In the LDH enzyme release assay on differentiated L6 myoblasts treated with different concentrations (1-100 μg mL-1) of guavanoic acid functionalized gold nanoparticles, a viability of 75% at 100 μg mL-1 was observed.
Collapse
Affiliation(s)
- K Govindaraju
- Centre for Ocean Research (DST-FIST Sponsored Centre) , Sathyabama Institute of Science and Technology , Chennai-600 119 , India . ;
| | - K S Uma Suganya
- Centre for Ocean Research (DST-FIST Sponsored Centre) , Sathyabama Institute of Science and Technology , Chennai-600 119 , India . ; .,Department of Biotechnology and Biochemical Engineering , Sree Chitra Thirunal College of Engineering , Pappanamcode , Thiruvananthapuram , Kerala-695018 , India
| |
Collapse
|
21
|
Vlavcheski F, Den Hartogh DJ, Giacca A, Tsiani E. Amelioration of High-Insulin-Induced Skeletal Muscle Cell Insulin Resistance by Resveratrol Is Linked to Activation of AMPK and Restoration of GLUT4 Translocation. Nutrients 2020; 12:E914. [PMID: 32230718 PMCID: PMC7230755 DOI: 10.3390/nu12040914] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 03/17/2020] [Accepted: 03/23/2020] [Indexed: 12/14/2022] Open
Abstract
Insulin resistance, the hallmark of type 2 diabetes mellitus (T2DM), is linked to hyperinsulinemia, which develops to counterbalance initial peripheral hormone resistance. Studies indicate that chronically elevated levels of insulin lead to skeletal muscle insulin resistance by deregulating steps within the insulin signaling cascade. The polyphenol resveratrol (RSV) has been shown to have antidiabetic properties in vitro and in vivo. In the present study, we examined the effect of RSV on high insulin (HI)-induced insulin resistance in skeletal muscle cells in vitro and investigated the mechanisms involved. Parental and GLUT4myc-overexpressing L6 rat skeletal muscle cells were used. [3H]2-deoxyglucose (2DG) uptake was measured, and total and phosphorylated levels of specific proteins were examined by immunoblotting. Exposure of L6 cells to HI levels (100 nM) for 24 h decreased the acute-insulin-stimulated 2DG uptake, indicating insulin resistance. HI increased ser307 and ser636/639 phosphorylation of IRS-1 (to 184% ± 12% and 225% ± 28.9% of control, with p < 0.001 and p < 0.01, respectively) and increased the phosphorylation levels of mTOR (174% ± 6.7% of control, p < 0.01) and p70 S6K (228% ± 33.5% of control, p < 0.01). Treatment with RSV abolished these HI-induced responses. Furthermore, RSV increased the activation of AMPK and restored the insulin-mediated increase in plasma membrane GLUT4 glucose transporter levels. These data suggest that RSV has a potential to counteract the HI-induced muscle insulin resistance.
Collapse
Affiliation(s)
- Filip Vlavcheski
- Department of Health Sciences, Brock University, St. Catharines, ON L2S 3A1, Canada; (F.V.); (D.J.D.H.)
- Centre for Bone and Muscle Health, Brock University, St. Catharines, ON L2S 3A1, Canada
| | - Danja J. Den Hartogh
- Department of Health Sciences, Brock University, St. Catharines, ON L2S 3A1, Canada; (F.V.); (D.J.D.H.)
- Centre for Bone and Muscle Health, Brock University, St. Catharines, ON L2S 3A1, Canada
| | - Adria Giacca
- Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada;
- Department of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
- Institute of Medical Sciences, University of Toronto, Toronto, ON M5S 1A8T, Canada
- Banting and Best Diabetes Centre, University of Toronto, Toronto, ON M5G 2C4, Canada
| | - Evangelia Tsiani
- Department of Health Sciences, Brock University, St. Catharines, ON L2S 3A1, Canada; (F.V.); (D.J.D.H.)
- Centre for Bone and Muscle Health, Brock University, St. Catharines, ON L2S 3A1, Canada
| |
Collapse
|
22
|
Ren G, Kim T, Papizan JB, Okerberg CK, Kothari VM, Zaid H, Bilan PJ, Araya-Ramirez F, Littlefield LA, Bowers RL, Mahurin AJ, Nickles MM, Ludvigsen R, He X, Grandjean PW, Mathews ST. Phosphorylation status of fetuin-A is critical for inhibition of insulin action and is correlated with obesity and insulin resistance. Am J Physiol Endocrinol Metab 2019; 317:E250-E260. [PMID: 31084489 DOI: 10.1152/ajpendo.00089.2018] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Fetuin-A (Fet-A), a hepatokine associated with insulin resistance, obesity, and incident type 2 diabetes, is shown to exist in both phosphorylated and dephosphorylated forms in circulation. However, studies on fetuin-A phosphorylation status in insulin-resistant conditions and its functional significance are limited. We demonstrate that serum phosphofetuin-A (Ser312) levels were significantly elevated in high-fat diet-induced obese mice, insulin-resistant Zucker diabetic fatty rats, and in individuals with obesity who are insulin resistant. Unlike serum total fetuin-A, serum phosphofetuin-A was associated with body weight, insulin, and markers of insulin resistance. To characterize potential mechanisms, fetuin-A was purified from Hep3B human hepatoma cells. Hep3B Fet-A was phosphorylated (Ser312) and inhibited insulin-stimulated glucose uptake and glycogen synthesis in L6GLUT4 myoblasts. Furthermore, single (Ser312Ala) and double (Ser312Ala + Ser120Ala) phosphorylation-defective Fet-A mutants were without effect on glucose uptake and glycogen synthesis in L6GLUT4 myoblasts. Together, our studies demonstrate that phosphorylation status of Fet-A (Ser312) is associated with obesity and insulin resistance and raise the possibility that Fet-A phosphorylation may play a role in regulation of insulin action.
Collapse
Affiliation(s)
- Guang Ren
- Department of Nutrition and Dietetics, Auburn University , Auburn, Alabama
| | - Teayoun Kim
- Department of Nutrition and Dietetics, Auburn University , Auburn, Alabama
| | - James B Papizan
- Department of Nutrition and Dietetics, Auburn University , Auburn, Alabama
| | - Carl K Okerberg
- Department of Nutrition and Dietetics, Auburn University , Auburn, Alabama
| | - Vishal M Kothari
- Department of Nutrition and Dietetics, Auburn University , Auburn, Alabama
| | - Hilal Zaid
- Cell Biology Program, The Hospital for Sick Children , Toronto , Canada
| | - Phillip J Bilan
- Cell Biology Program, The Hospital for Sick Children , Toronto , Canada
| | | | | | | | - A Jack Mahurin
- School of Kinesiology, Auburn University , Auburn, Alabama
| | - Mary M Nickles
- Department of Nutrition and Dietetics, Samford University , Birmingham, Alabama
| | - Rebecca Ludvigsen
- Department of Nutrition and Dietetics, Auburn University , Auburn, Alabama
| | - Xiaoming He
- Department of Nutrition and Dietetics, Auburn University , Auburn, Alabama
| | | | - Suresh T Mathews
- Department of Nutrition and Dietetics, Auburn University , Auburn, Alabama
- Department of Nutrition and Dietetics, Samford University , Birmingham, Alabama
| |
Collapse
|
23
|
Gohil AR, Deshmukh SK, Bhattacharya V, Lavhale R, Verekar S, Kate AS. Exophiarin, an isocoumarin from the fungus Exophiala sp. with antihyperglycemic activity. Nat Prod Res 2019; 35:1573-1581. [PMID: 31170816 DOI: 10.1080/14786419.2019.1624957] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Chemical characterization of ethyl acetate extract of Exophiala sp. has afforded the isolation of three compounds including a new isocoumarin named exophiarin (1). Exophiala sp. was obtained from the soil containing dumped organic waste (litter). Initially, LC-UV-MS analysis of the extract of Exophiala sp. revealed the presence of a new compound having molecular weight 438 (1) and previously reported TPI-2 and TPI-5. The novelty was established using advanced database search comprising of biological source, molecular weight and UV profile. 1D, 2D NMR and HRMS data have been used for structure elucidation. Exophiarin with TPI-2 and TPI-5 have displayed moderate improvement in glucose uptake activity when tested in rat skeletal muscle cell line L6.
Collapse
Affiliation(s)
- Akash R Gohil
- Natural Products Microbiology, Piramal Enterprises Limited, Mumbai, India.,Natural Products Bioactivity Lab, Piramal Enterprises Limited, Mumbai, India
| | - Sunil K Deshmukh
- Natural Products Microbiology, Piramal Enterprises Limited, Mumbai, India
| | | | - Rahul Lavhale
- Natural Products Bioactivity Lab, Piramal Enterprises Limited, Mumbai, India
| | - Shilpa Verekar
- Natural Products Microbiology, Piramal Enterprises Limited, Mumbai, India
| | - Abhijeet S Kate
- Natural Products Bioactivity Lab, Piramal Enterprises Limited, Mumbai, India.,National Institute of Pharmaceutical Education and Research - Ahmedabad, Gandhinagar, Gujarat, India
| |
Collapse
|
24
|
Bone DBJ, Meister J, Knudsen JR, Dattaroy D, Cohen A, Lee R, Lu H, Metzger D, Jensen TE, Wess J. Skeletal Muscle-Specific Activation of G q Signaling Maintains Glucose Homeostasis. Diabetes 2019; 68:1341-1352. [PMID: 30936140 PMCID: PMC6610017 DOI: 10.2337/db18-0796] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Accepted: 03/22/2019] [Indexed: 12/19/2022]
Abstract
Skeletal muscle (SKM) insulin resistance plays a central role in the pathogenesis of type 2 diabetes. Because G-protein-coupled receptors (GPCRs) represent excellent drug targets, we hypothesized that activation of specific functional classes of SKM GPCRs might lead to improved glucose homeostasis in type 2 diabetes. At present, little is known about the in vivo metabolic roles of the various distinct GPCR signaling pathways operative in SKM. In this study, we tested the hypothesis that selective activation of SKM Gq signaling can improve SKM glucose uptake and whole-body glucose homeostasis under physiological and pathophysiological conditions. Studies with transgenic mice expressing a Gq-linked designer GPCR selectively in SKM cells demonstrated that receptor-mediated activation of SKM Gq signaling greatly promoted glucose uptake into SKM and significantly improved glucose homeostasis in obese, glucose-intolerant mice. These beneficial metabolic effects required the activity of SKM AMPK. In contrast, obese mutant mice that lacked both Gαq and Gα11 selectively in SKM showed severe deficits in glucose homeostasis. Moreover, GPCR-mediated activation of Gq signaling also stimulated glucose uptake in primary human SKM cells. Taken together, these findings strongly suggest that agents capable of enhancing SKM Gq signaling may prove useful as novel antidiabetic drugs.
Collapse
Affiliation(s)
- Derek B J Bone
- Molecular Signaling Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD
| | - Jaroslawna Meister
- Molecular Signaling Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD
| | - Jonas R Knudsen
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Diptadip Dattaroy
- Molecular Signaling Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD
| | - Amanda Cohen
- Molecular Signaling Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD
| | - Regina Lee
- Molecular Signaling Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD
| | - Huiyan Lu
- Mouse Transgenic Core Facility, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD
| | - Daniel Metzger
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS UMR 7104, INSERM U1258, Université de Strasbourg, Illkirch, France
| | - Thomas E Jensen
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Jürgen Wess
- Molecular Signaling Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD
| |
Collapse
|
25
|
Sasikumar P, Lekshmy K, Sini S, Prabha B, Kumar NA, Sivan VV, Jithin MM, Jayamurthy P, Shibi IG, Radhakrishnan KV. Isolation and characterization of resveratrol oligomers from the stem bark of Hopea ponga (Dennst.) Mabb. And their antidiabetic effect by modulation of digestive enzymes, protein glycation and glucose uptake in L6 myocytes. JOURNAL OF ETHNOPHARMACOLOGY 2019; 236:196-204. [PMID: 30844488 DOI: 10.1016/j.jep.2019.01.046] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 11/24/2018] [Accepted: 01/22/2019] [Indexed: 06/09/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Hopea ponga (Dennst.) Mabb. Is used in traditional herbal formulations for diabetes complications. The aim of this study is to evaluate the antidiabetic effect of extracts and compounds from H. ponga. MATERIALS AND METHODS Silica gel column chromatography was performed to identify various chemical components of the plant extract. Different extracts of H. ponga and isolated compounds were screened for their antidiabetic effect by modulation of digestive enzymes and protein glycation. The effect of glucose uptake by the compounds and the pathways through which the compounds mediate the glucose uptake potential were confirmed by fluorescent microscopy, flow cytometry and western blot analysis. RESULTS Acetone and ethanol extracts of the stem bark of Hopea ponga (Dennst.) Mabb. Afforded six resveratrol oligomers namely, E-resveratrol (1), (-)-ε-viniferin (2), (-)-α-viniferin (3), trihydroxyphenanthrene glucoside (THPG) (4), vaticaphenol A (5), (-)-hopeaphenol (6), along with four phytosterols. The structures were determined on the basis of spectroscopic analyses including nuclear magnetic resonance (NMR) spectroscopy and high resolution mass spectrometry (HRMS) data. Compounds 1-5 and 7-10 were tested for their α-glucosidase, α-amylase and glycation inhibitiory activities. All the resveratrol oligomers (1-5) showed prominent α-glucosidase inhibition with IC50 values, 12.56 ± 1.00, 23.98 ± 1.11, 7.17 ± 1.10, 31.74 ± 0.42 and 16.95 ± 0.39 μM, respectively. Molecular docking studies also supported the observed α-glucosidase inhibition. Compound 3 displayed IC50 values of 4.85 ± 0.06 and 27.10 ± 0.04 μM in α-amylase and glycation inhibitory assays activity. The 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) assay revealed that the compounds 3 and 4 were found to be less toxic at a concentration of 100 μM (<10%) and 25 μM (<20%), respectively. The effect of glucose uptake performed by 2-(N-(7-Nitrobenz-2-oxa-1,3-diazol-4-yl)amino)-2-deoxyglucose (2-NBDG) in L6 myoblast were measured by fluorescent microscopy and flow cytometry. The compounds 3 and 4 showed 2-NBDG uptake of 49.6% and 38.8% respectively. By examining the molecular pathway through which the compounds elicit their glucose uptake potential, it was observed that both the compounds mainly act via AMPK pathway. CONCLUSION This is the first report on the isolation of compounds from H. ponga. Altogether, the results of this study reveal the antidiabetic effects of H. ponga extracts and isolated compounds promoting traditional use of this plant in the treatment of diabetes.
Collapse
Affiliation(s)
- P Sasikumar
- Chemical Science and Technology Division, CSIR-National Institute for Interdisciplinary Science and Technology, Thiruvananthapuram, 695019, India
| | - K Lekshmy
- Agroprocessing and Technology Division, CSIR-National Institute for Interdisciplinary Science and Technology, Thiruvananthapuram, 695019, India
| | - S Sini
- Agroprocessing and Technology Division, CSIR-National Institute for Interdisciplinary Science and Technology, Thiruvananthapuram, 695019, India
| | - B Prabha
- Chemical Science and Technology Division, CSIR-National Institute for Interdisciplinary Science and Technology, Thiruvananthapuram, 695019, India
| | - N Anil Kumar
- M. S. Swaminathan Research Foundation-Community Agrobiodiversity Centre (MSSRF-CAbC), Puthurvayal, Wayanad, Kerala, India
| | - V V Sivan
- M. S. Swaminathan Research Foundation-Community Agrobiodiversity Centre (MSSRF-CAbC), Puthurvayal, Wayanad, Kerala, India
| | - M M Jithin
- M. S. Swaminathan Research Foundation-Community Agrobiodiversity Centre (MSSRF-CAbC), Puthurvayal, Wayanad, Kerala, India
| | - P Jayamurthy
- Agroprocessing and Technology Division, CSIR-National Institute for Interdisciplinary Science and Technology, Thiruvananthapuram, 695019, India; Academy of Scientific and Innovative Research (AcSIR), Thiruvananthapuram, 695019, India
| | - I G Shibi
- Department of Chemistry, Sree Narayana College, Chempazhanthy, Thiruvananthapuram, Kerala, India
| | - K V Radhakrishnan
- Chemical Science and Technology Division, CSIR-National Institute for Interdisciplinary Science and Technology, Thiruvananthapuram, 695019, India; Academy of Scientific and Innovative Research (AcSIR), Thiruvananthapuram, 695019, India.
| |
Collapse
|
26
|
Uma Suganya KS, Govindaraju K, Veena Vani C, Premanathan M, Ganesh Kumar VK. In vitro biological evaluation of anti-diabetic activity of organic-inorganic hybrid gold nanoparticles. IET Nanobiotechnol 2019; 13:226-229. [PMID: 31051455 DOI: 10.1049/iet-nbt.2018.5139] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Diabetes mellitus has been considered as a heterogeneous metabolic disorder characterised by complete or relative impairment in the production of insulin by pancreatic β-cells or insulin resistance. In the present study, propanoic acid, an active biocomponent isolated from Cassia auriculata is employed for the synthesis of propanoic acid functionalised gold nanoparticles (Pa@AuNPs) and its anti-diabetic activity has been demonstrated in vitro. In vitro cytotoxicity of synthesised Pa@AuNPs was performed in L6 myotubes. The mode of action of Pa@AuNPs exhibiting anti-diabetic potential was validated by glucose uptake assay in the presence of Genistein (insulin receptor tyrosine kinase inhibitor) and Wortmannin (Phosphatidyl inositide kinase inhibitor). Pa@AuNPs exhibited significant glucose uptake in L6 myotubes with maximum uptake at 50 ng/ml. Assays were performed to study the potential of Pa@AuNPs in the inhibition of protein-tyrosine phosphatase 1B, α-glucosidases, and α-amylase activity.
Collapse
Affiliation(s)
| | - Kasivelu Govindaraju
- Centre for Ocean Research, Sathyabama Institute of Science and Technology, Chennai-600 119, India.
| | - Chitoor Veena Vani
- Centre for Ocean Research, Sathyabama Institute of Science and Technology, Chennai-600 119, India
| | - Mariappan Premanathan
- Central Bioscience Research Laboratories (CBRL), Department of Biology, College of Science, Al-Zulfi, Majmaah University, Kingdom of Saudi Arabia
| | | |
Collapse
|
27
|
In vitro experimental models for examining the skeletal muscle cell biology of exercise: the possibilities, challenges and future developments. Pflugers Arch 2018; 471:413-429. [PMID: 30291430 DOI: 10.1007/s00424-018-2210-4] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Revised: 09/18/2018] [Accepted: 09/25/2018] [Indexed: 12/11/2022]
Abstract
Exercise provides a cornerstone in the prevention and treatment of several chronic diseases. The use of in vivo exercise models alone cannot fully establish the skeletal muscle-specific mechanisms involved in such health-promoting effects. As such, models that replicate exercise-like effects in vitro provide useful tools to allow investigations that are not otherwise possible in vivo. In this review, we provide an overview of experimental models currently used to induce exercise-like effects in skeletal muscle in vitro. In particular, the appropriateness of electrical pulse stimulation and several pharmacological compounds to resemble exercise, as well as important technical considerations, are addressed. Each model covered herein provides a useful tool to investigate different aspects of exercise with a level of abstraction not possible in vivo. That said, none of these models are perfect under all circumstances, and the choice of model (and terminology) used should be informed by the specific research question whilst accounting for the several inherent limitations of each model. Further work is required to develop and optimise the current experimental models used, such as combination with complementary techniques during treatment, and thereby improve their overall utility and impact within muscle biology research.
Collapse
|
28
|
Anti-diabetic effect and mechanism of Kursi Wufarikun Ziyabit in L6 rat skeletal muscle cells. J Pharmacol Sci 2018; 137:212-219. [DOI: 10.1016/j.jphs.2018.06.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2018] [Revised: 05/23/2018] [Accepted: 06/11/2018] [Indexed: 02/03/2023] Open
|
29
|
Turner MC, Player DJ, Martin NR, Akam EC, Lewis MP. The effect of chronic high insulin exposure upon metabolic and myogenic markers in C2C12 skeletal muscle cells and myotubes. J Cell Biochem 2018; 119:5686-5695. [DOI: 10.1002/jcb.26748] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Accepted: 01/25/2018] [Indexed: 11/08/2022]
Affiliation(s)
- Mark C. Turner
- School of Sport, Exercise and Health Sciences, National Centre for Sport and Exercise MedicineLoughborough UniversityLeicestershireUnited Kingdom
| | - Darren J. Player
- School of Sport, Exercise and Health Sciences, National Centre for Sport and Exercise MedicineLoughborough UniversityLeicestershireUnited Kingdom
- Institute of Orthopaedics and Musculoskeletal ScienceDivision of Surgery and Interventional ScienceUniversity College LondonStanmoreMiddlesexUnited Kingdom
| | - Neil R.W. Martin
- School of Sport, Exercise and Health Sciences, National Centre for Sport and Exercise MedicineLoughborough UniversityLeicestershireUnited Kingdom
| | - Elizabeth C. Akam
- School of Sport, Exercise and Health Sciences, National Centre for Sport and Exercise MedicineLoughborough UniversityLeicestershireUnited Kingdom
| | - Mark P. Lewis
- School of Sport, Exercise and Health Sciences, National Centre for Sport and Exercise MedicineLoughborough UniversityLeicestershireUnited Kingdom
| |
Collapse
|
30
|
Jaldin-Fincati JR, Bilan PJ, Klip A. GLUT4 Translocation in Single Muscle Cells in Culture: Epitope Detection by Immunofluorescence. Methods Mol Biol 2018; 1713:175-192. [PMID: 29218526 DOI: 10.1007/978-1-4939-7507-5_14] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
GLUT4 is the major glucose transporter in skeletal muscle. GLUT4 cycles to and from the plasma membrane and its exocytic rate is accelerated by insulin and muscle contraction to achieve a new steady state with more GLUT4 proteins at the muscle cell surface. To gain a better understanding of the molecular and cellular mechanisms that govern GLUT4 protein recycling, we developed an in vitro model in which myc-epitope-tagged GLUT4 or GLUT4-GFP is expressed in L6 skeletal muscle cells. The myc-epitope is inserted into an exofacial domain that is accessible to anti-myc antibodies from the outside of non-permeabilized cells, allowing one to count the number of transporters at the cell surface. This enables one to perform single-cell analysis using confocal fluorescence microscopy to quantify cell surface GLUT4myc or GLUT4myc-GFP in cells co-transfected with diverse cDNA constructs, treated with siRNAs, or co-stained with antibodies for other proteins of interest. Herein, we describe the methodology to perform these experimental approaches in insulin-stimulated L6 muscle cells.
Collapse
Affiliation(s)
- Javier R Jaldin-Fincati
- Cell Biology Program, Research Institute, The Hospital for Sick Children, 686 Bay Street, PGCRL, Room 19.9709, Toronto, ON, Canada, M5G 0A4
| | - Philip J Bilan
- Cell Biology Program, Research Institute, The Hospital for Sick Children, 686 Bay Street, PGCRL, Room 19.9709, Toronto, ON, Canada, M5G 0A4
| | - Amira Klip
- Cell Biology Program, Research Institute, The Hospital for Sick Children, 686 Bay Street, PGCRL, Room 19.9709, Toronto, ON, Canada, M5G 0A4.
| |
Collapse
|
31
|
Ahlstrom P, Rai E, Chakma S, Cho HH, Rengasamy P, Sweeney G. Adiponectin improves insulin sensitivity via activation of autophagic flux. J Mol Endocrinol 2017; 59:339-350. [PMID: 28954814 DOI: 10.1530/jme-17-0096] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2017] [Accepted: 09/04/2017] [Indexed: 12/26/2022]
Abstract
Skeletal muscle insulin resistance is known to play an important role in the pathogenesis of diabetes, and one potential causative cellular mechanism is endoplasmic reticulum (ER) stress. Adiponectin mediates anti-diabetic effects via direct metabolic actions and by improving insulin sensitivity, and we recently demonstrated an important role in stimulation of autophagy by adiponectin. However, there is limited knowledge on crosstalk between autophagy and ER stress in skeletal muscle and in particular how they are regulated by adiponectin. Here, we utilized the model of high insulin/glucose (HIHG)-induced insulin resistance, determined by measuring Akt phosphorylation (T308 and S473) and glucose uptake in L6 skeletal muscle cells. HIHG reduced autophagic flux measured by LC3 and p62 Western blotting and tandem fluorescent RFP/GFP-LC3 immunofluorescence (IF). HIHG also induced ER stress assessed by thioflavin T/KDEL IF, pIRE1, pPERK, peIF2α and ATF6 Western blotting and induction of a GRP78-mCherry reporter. Induction of autophagy by adiponectin or rapamycin attenuated HIHG-induced ER stress and improved insulin sensitivity. The functional significance of enhanced autophagy was validated by demonstrating a lack of improved insulin sensitivity in response to adiponectin in autophagy-deficient cells generated by overexpression of dominant negative mutant of Atg5. In summary, adiponectin-induced autophagy in skeletal muscle cells alleviated HIHG-induced ER stress and insulin resistance.
Collapse
Affiliation(s)
| | - Esther Rai
- Department of BiologyYork University, Toronto, Canada
| | | | - Hee Ho Cho
- Department of BiologyYork University, Toronto, Canada
| | | | - Gary Sweeney
- Department of BiologyYork University, Toronto, Canada
| |
Collapse
|
32
|
Nikolić N, Görgens SW, Thoresen GH, Aas V, Eckel J, Eckardt K. Electrical pulse stimulation of cultured skeletal muscle cells as a model for in vitro exercise - possibilities and limitations. Acta Physiol (Oxf) 2017; 220:310-331. [PMID: 27863008 DOI: 10.1111/apha.12830] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Revised: 06/28/2016] [Accepted: 11/06/2016] [Indexed: 12/19/2022]
Abstract
The beneficial health-related effects of exercise are well recognized, and numerous studies have investigated underlying mechanism using various in vivo and in vitro models. Although electrical pulse stimulation (EPS) for the induction of muscle contraction has been used for quite some time, its application on cultured skeletal muscle cells of animal or human origin as a model of in vitro exercise is a more recent development. In this review, we compare in vivo exercise and in vitro EPS with regard to effects on signalling, expression level and metabolism. We provide a comprehensive overview of different EPS protocols and their applications, discuss technical aspects of this model including critical controls and the importance of a proper maintenance procedure and finally discuss the limitations of the EPS model.
Collapse
Affiliation(s)
- N. Nikolić
- Department of Pharmaceutical Biosciences; School of Pharmacy; University of Oslo; Oslo Norway
| | - S. W. Görgens
- Paul-Langerhans-Group for Integrative Physiology; German Diabetes Center; Düsseldorf Germany
| | - G. H. Thoresen
- Department of Pharmaceutical Biosciences; School of Pharmacy; University of Oslo; Oslo Norway
- Department of Pharmacology; Institute of Clinical Medicine; Faculty of Medicine; University of Oslo; Oslo Norway
| | - V. Aas
- Department of Life Sciences and Health; Oslo and Akershus University College of Applied Sciences; Oslo Norway
| | - J. Eckel
- Paul-Langerhans-Group for Integrative Physiology; German Diabetes Center; Düsseldorf Germany
- German Center for Diabetes Research (DZD e.V.); Düsseldorf Germany
| | - K. Eckardt
- Department of Nutrition; Institute for Basic Medical Sciences; Faculty of Medicine; University of Oslo; Oslo Norway
| |
Collapse
|
33
|
Choudhary AK, Dey CS. Nuclear co-repressor (NCoR) is required to maintain insulin sensitivity in C 2 C 12 myotubes. Cell Biol Int 2016; 41:204-212. [PMID: 27935220 DOI: 10.1002/cbin.10711] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2016] [Accepted: 11/26/2016] [Indexed: 12/30/2022]
Abstract
Nuclear co-repressor (NCoR) regulates peripheral insulin sensitivity; however, its role in modulating insulin sensitivity in skeletal muscle remains elusive. Present study investigated protein expression and effect of NCoR on insulin sensitivity in murine skeletal muscle cell line C2 C12 . Myotubes as compared to myoblasts of C2 C12 cells were found to be more sensitive in response to insulin as increase in insulin-stimulated phosphorylation of AKT at serine 473 residue (pAKTS473 ) was significantly higher in myotubes. Incidentally, reduced protein level of NCoR coincided with differentiation of myoblasts into myotubes of C2 C12 cells. However, insulin stimulation per se failed to affect protein level of NCoR either in myoblasts or myotubes of C2 C12 cells. To assess the role of NCoR on insulin sensitivity, NCoR was transiently knocked down using siRNA in myotubes of C2 C12 . In fact, transient silencing of NCoR led to significant reduction in insulin-stimulated pAKTS473 and impaired glucose uptake. This observation is in contrast to published studies where NCoR has been reported to negatively regulate insulin signaling cascade. Furthermore, transient silencing of NCoR failed to improve insulin sensitivity in chronic hyperinsulinemia-induced insulin-resistant model of C2 C12 cells. Importantly, inhibition of lysosomal protein degradation pathway using ammonium chloride restored protein level of NCoR but failed to increase glucose uptake in serum-starved C2 C12 myotubes. Collectively, data from present study show differential protein level of NCoR under different cell state (myoblast and myotubes) of C2 C12 cells and NCoR proves to be vital for maintaining insulin sensitivity in C2 C12 myotubes.
Collapse
Affiliation(s)
- Abhijeet K Choudhary
- Kusuma School of Biological Sciences (KSBS), Indian Institute of Technology (IIT)-Delhi, Hauz Khas, New Delhi, 110016, India
| | - Chinmoy S Dey
- Kusuma School of Biological Sciences (KSBS), Indian Institute of Technology (IIT)-Delhi, Hauz Khas, New Delhi, 110016, India
| |
Collapse
|
34
|
Kachko I, Traitel T, Goldbart R, Silbert L, Katz M, Bashan N, Jelinek R, Rudich A, Kost J. Polymeric carrier-mediated intracellular delivery of phosphatidylinositol-3,4,5-trisphosphate to overcome insulin resistance. J Drug Target 2016; 23:698-709. [PMID: 26453165 DOI: 10.3109/1061186x.2015.1052076] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
BACKGROUND Phosphatidylinositol-3,4,5-trisphosphate (PIP3) is a major lipid second messenger in insulin-mediated signalling towards the metabolic actions of this hormone in muscle and fat. PURPOSE Assessing the intracellular transport of exogenous PIP3 attached to a polymeric carrier in an attempt to overcome cellular insulin resistance. METHODS Artificial chromatic bio-mimetic membrane vesicles composed of dimyristoylphosphatidylcholine and polydiacetylene were applied to screen the polymeric carriers. PIP3 cellular localization and bio-activity was assessed by fluorescent and live-cell microscopy in L6 muscle cells and in 3T3-L1 adipocytes. RESULTS AND DISCUSSION We demonstrate that a specific-branched polyethylenimine (PEI-25, 25 kDa) carrier forms complexes with PIP3 that interact with the bio-mimetic membrane vesicles in a manner predictive of their interaction with cells: In L6 muscle cells, PEI-25/fluorescent-PIP3 complexes are retarded at the cell perimeter. PEI-25/PIP3 complexes retain their bio-activity, engaging signalling steps downstream of PIP3, even in muscle cells rendered insulin resistant by exposure to high glucose/high insulin. CONCLUSIONS Inducing insulin actions by intracellular PIP3 delivery (PEI-25/PIP3 complexes) in some forms of insulin-resistant cells provides the first proof-of-principle for the potential therapeutic use of PIP3 in a "second-messenger agonist" approach. In addition, utilization of an artificial bio-mimetic membrane platform to screen for highly efficient PIP3 delivery predicts biological function in cells.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Assaf Rudich
- c Department of Clinical Biochemistry , and.,d The National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev , Beer-Sheva , Israel
| | | |
Collapse
|
35
|
Proteomics of Skeletal Muscle: Focus on Insulin Resistance and Exercise Biology. Proteomes 2016; 4:proteomes4010006. [PMID: 28248217 PMCID: PMC5217365 DOI: 10.3390/proteomes4010006] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Revised: 01/25/2016] [Accepted: 01/28/2016] [Indexed: 12/21/2022] Open
Abstract
Skeletal muscle is the largest tissue in the human body and plays an important role in locomotion and whole body metabolism. It accounts for ~80% of insulin stimulated glucose disposal. Skeletal muscle insulin resistance, a primary feature of Type 2 diabetes, is caused by a decreased ability of muscle to respond to circulating insulin. Physical exercise improves insulin sensitivity and whole body metabolism and remains one of the most promising interventions for the prevention of Type 2 diabetes. Insulin resistance and exercise adaptations in skeletal muscle might be a cause, or consequence, of altered protein expressions profiles and/or their posttranslational modifications (PTMs). Mass spectrometry (MS)-based proteomics offer enormous promise for investigating the molecular mechanisms underlying skeletal muscle insulin resistance and exercise-induced adaptation; however, skeletal muscle proteomics are challenging. This review describes the technical limitations of skeletal muscle proteomics as well as emerging developments in proteomics workflow with respect to samples preparation, liquid chromatography (LC), MS and computational analysis. These technologies have not yet been fully exploited in the field of skeletal muscle proteomics. Future studies that involve state-of-the-art proteomics technology will broaden our understanding of exercise-induced adaptations as well as molecular pathogenesis of insulin resistance. This could lead to the identification of new therapeutic targets.
Collapse
|
36
|
Stringer DM, Zahradka P, Taylor CG. Glucose transporters: cellular links to hyperglycemia in insulin resistance and diabetes. Nutr Rev 2016; 73:140-54. [PMID: 26024537 DOI: 10.1093/nutrit/nuu012] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Abnormal expression and/or function of mammalian hexose transporters contribute to the hallmark hyperglycemia of diabetes. Due to different roles in glucose handling, various organ systems possess specific transporters that may be affected during the diabetic state. Diabetes has been associated with higher rates of intestinal glucose transport, paralleled by increased expression of both active and facilitative transporters and a shift in the location of transporters within the enterocyte, events that occur independent of intestinal hyperplasia and hyperglycemia. Peripheral tissues also exhibit deregulated glucose transport in the diabetic state, most notably defective translocation of transporters to the plasma membrane and reduced capacity to clear glucose from the bloodstream. Expression of renal active and facilitative glucose transporters increases as a result of diabetes, leading to elevated rates of glucose reabsorption. However, this may be a natural response designed to combat elevated blood glucose concentrations and not necessarily a direct effect of insulin deficiency. Functional foods and nutraceuticals, by modulation of glucose transporter activity, represent a potential dietary tool to aid in the management of hyperglycemia and diabetes.
Collapse
Affiliation(s)
- Danielle M Stringer
- D.M. Stringer was with the Department of Human Nutritional Sciences, University of Manitoba, and the Canadian Centre for Agri-Food Research in Health and Medicine, St. Boniface Hospital Research Centre, Winnipeg, MB, Canada at the time of manuscript preparation. C.G. Taylor is with the Department of Human Nutritional Sciences, University of Manitoba; the Department of Physiology, University of Manitoba; and the Canadian Centre for Agri-Food Research in Health and Medicine, St. Boniface Hospital Research Centre, Winnipeg, Manitoba, Canada. P. Zahradka is with the Department of Human Nutritional Sciences, University of Manitoba; the Department of Physiology, University of Manitoba; and the Canadian Centre for Agri-Food Research in Health and Medicine, St. Boniface Hospital Research Centre, Winnipeg, Manitoba, Canada.
| | - Peter Zahradka
- D.M. Stringer was with the Department of Human Nutritional Sciences, University of Manitoba, and the Canadian Centre for Agri-Food Research in Health and Medicine, St. Boniface Hospital Research Centre, Winnipeg, MB, Canada at the time of manuscript preparation. C.G. Taylor is with the Department of Human Nutritional Sciences, University of Manitoba; the Department of Physiology, University of Manitoba; and the Canadian Centre for Agri-Food Research in Health and Medicine, St. Boniface Hospital Research Centre, Winnipeg, Manitoba, Canada. P. Zahradka is with the Department of Human Nutritional Sciences, University of Manitoba; the Department of Physiology, University of Manitoba; and the Canadian Centre for Agri-Food Research in Health and Medicine, St. Boniface Hospital Research Centre, Winnipeg, Manitoba, Canada
| | - Carla G Taylor
- D.M. Stringer was with the Department of Human Nutritional Sciences, University of Manitoba, and the Canadian Centre for Agri-Food Research in Health and Medicine, St. Boniface Hospital Research Centre, Winnipeg, MB, Canada at the time of manuscript preparation. C.G. Taylor is with the Department of Human Nutritional Sciences, University of Manitoba; the Department of Physiology, University of Manitoba; and the Canadian Centre for Agri-Food Research in Health and Medicine, St. Boniface Hospital Research Centre, Winnipeg, Manitoba, Canada. P. Zahradka is with the Department of Human Nutritional Sciences, University of Manitoba; the Department of Physiology, University of Manitoba; and the Canadian Centre for Agri-Food Research in Health and Medicine, St. Boniface Hospital Research Centre, Winnipeg, Manitoba, Canada
| |
Collapse
|
37
|
Han S, Park JS, Lee S, Jeong AL, Oh KS, Ka HI, Choi HJ, Son WC, Lee WY, Oh SJ, Lim JS, Lee MS, Yang Y. CTRP1 protects against diet-induced hyperglycemia by enhancing glycolysis and fatty acid oxidation. J Nutr Biochem 2016; 27:43-52. [DOI: 10.1016/j.jnutbio.2015.08.018] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Revised: 08/10/2015] [Accepted: 08/11/2015] [Indexed: 12/25/2022]
|
38
|
Enhancement of glucose uptake by Ficus hispida methanolic extract through phosphatidylinositol 3 kinase and inhibiting aldose reductase – An in vitro analysis. Eur J Integr Med 2015. [DOI: 10.1016/j.eujim.2014.12.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
39
|
Souto Padron de Figueiredo A, Salmon AB, Bruno F, Jimenez F, Martinez HG, Halade GV, Ahuja SS, Clark RA, DeFronzo RA, Abboud HE, El Jamali A. Nox2 mediates skeletal muscle insulin resistance induced by a high fat diet. J Biol Chem 2015; 290:13427-39. [PMID: 25825489 DOI: 10.1074/jbc.m114.626077] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Indexed: 01/16/2023] Open
Abstract
Inflammation and oxidative stress through the production of reactive oxygen species (ROS) are consistently associated with metabolic syndrome/type 2 diabetes. Although the role of Nox2, a major ROS-generating enzyme, is well described in host defense and inflammation, little is known about its potential role in insulin resistance in skeletal muscle. Insulin resistance induced by a high fat diet was mitigated in Nox2-null mice compared with wild-type mice after 3 or 9 months on the diet. High fat feeding increased Nox2 expression, superoxide production, and impaired insulin signaling in skeletal muscle tissue of wild-type mice but not in Nox2-null mice. Exposure of C2C12 cultured myotubes to either high glucose concentration, palmitate, or H2O2 decreases insulin-induced Akt phosphorylation and glucose uptake. Pretreatment with catalase abrogated these effects, indicating a key role for H2O2 in mediating insulin resistance. Down-regulation of Nox2 in C2C12 cells by shRNA prevented insulin resistance induced by high glucose or palmitate but not H2O2. These data indicate that increased production of ROS in insulin resistance induced by high glucose in skeletal muscle cells is a consequence of Nox2 activation. This is the first report to show that Nox2 is a key mediator of insulin resistance in skeletal muscle.
Collapse
Affiliation(s)
| | - Adam B Salmon
- The Sam and Ann Barshop Institute for Longevity and Aging Studies 15355 Lambda Drive, San Antonio, Texas 78245, and Audie L. Murphy Hospital, South Texas Veterans Health Care System, San Antonio, Texas 78229-3900
| | - Francesca Bruno
- From the Department of Medicine, University of Texas Health Science Center, San Antonio, Texas 78229-3900
| | - Fabio Jimenez
- From the Department of Medicine, University of Texas Health Science Center, San Antonio, Texas 78229-3900
| | - Herman G Martinez
- From the Department of Medicine, University of Texas Health Science Center, San Antonio, Texas 78229-3900
| | - Ganesh V Halade
- From the Department of Medicine, University of Texas Health Science Center, San Antonio, Texas 78229-3900
| | - Seema S Ahuja
- From the Department of Medicine, University of Texas Health Science Center, San Antonio, Texas 78229-3900, Audie L. Murphy Hospital, South Texas Veterans Health Care System, San Antonio, Texas 78229-3900
| | - Robert A Clark
- From the Department of Medicine, University of Texas Health Science Center, San Antonio, Texas 78229-3900, Audie L. Murphy Hospital, South Texas Veterans Health Care System, San Antonio, Texas 78229-3900
| | - Ralph A DeFronzo
- From the Department of Medicine, University of Texas Health Science Center, San Antonio, Texas 78229-3900, Audie L. Murphy Hospital, South Texas Veterans Health Care System, San Antonio, Texas 78229-3900
| | - Hanna E Abboud
- From the Department of Medicine, University of Texas Health Science Center, San Antonio, Texas 78229-3900, Audie L. Murphy Hospital, South Texas Veterans Health Care System, San Antonio, Texas 78229-3900
| | - Amina El Jamali
- From the Department of Medicine, University of Texas Health Science Center, San Antonio, Texas 78229-3900,
| |
Collapse
|
40
|
Sylow L, Møller LLV, Kleinert M, Richter EA, Jensen TE. Stretch-stimulated glucose transport in skeletal muscle is regulated by Rac1. J Physiol 2015; 593:645-56. [PMID: 25416624 DOI: 10.1113/jphysiol.2014.284281] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Accepted: 11/13/2014] [Indexed: 11/08/2022] Open
Abstract
KEY POINTS Rac1 regulates stretch-stimulated (i.e. mechanical stress) glucose transport in muscle. Actin depolymerization decreases stretch-induced glucose transport in skeletal muscle. Rac1 is a required part of the mechanical stress-component of the contraction-stimulus to glucose transport in skeletal muscle. ABSTRACT An alternative to the canonical insulin signalling pathway for glucose transport is muscle contraction/exercise. Mechanical stress is an integrated part of the muscle contraction/relaxation cycle, and passive stretch stimulates muscle glucose transport. However, the signalling mechanism regulating stretch-stimulated glucose transport is not well understood. We recently reported that the actin cytoskeleton regulating GTPase, Rac1, was activated in mouse muscle in response to stretching. Rac1 is a regulator of contraction- and insulin-stimulated glucose transport, however, its role in stretch-stimulated glucose transport and signalling is unknown. We therefore investigated whether stretch-induced glucose transport in skeletal muscle required Rac1 and the actin cytoskeleton. We used muscle-specific inducible Rac1 knockout mice as well as pharmacological inhibitors of Rac1 and the actin cytoskeleton in isolated soleus and extensor digitorum longus muscles. In addition, the role of Rac1 in contraction-stimulated glucose transport during conditions without mechanical load on the muscles was evaluated in loosely hanging muscles and muscles in which cross-bridge formation was blocked by the myosin ATPase inhibitors BTS and Blebbistatin. Knockout as well as pharmacological inhibition of Rac1 reduced stretch-stimulated glucose transport by 30-50% in soleus and extensor digitorum longus muscle. The actin depolymerizing agent latrunculin B similarly decreased glucose transport in response to stretching by 40-50%. Rac1 inhibition reduced contraction-stimulated glucose transport by 30-40% in tension developing muscle but did not affect contraction-stimulated glucose transport in muscles in which force development was prevented. Our findings suggest that Rac1 and the actin cytoskeleton regulate stretch-stimulated glucose transport and that Rac1 is a required part of the mechanical stress-component of the contraction-stimulus to glucose transport in skeletal muscle.
Collapse
Affiliation(s)
- Lykke Sylow
- Molecular Physiology Group, Department of Nutrition, Exercise and Sports, University of Copenhagen, Denmark
| | | | | | | | | |
Collapse
|
41
|
Ha BG, Yonezawa T, Son MJ, Woo JT, Ohba S, Chung UI, Yagasaki K. Antidiabetic effect of nepodin, a component of Rumex roots, and its modes of action in vitro and in vivo. Biofactors 2014; 40:436-47. [PMID: 24756979 DOI: 10.1002/biof.1165] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2014] [Revised: 03/18/2014] [Accepted: 04/04/2014] [Indexed: 11/08/2022]
Abstract
Many active components derived from edible natural resources such as plant extracts have recently attracted attention for their potential use as functional foods or drugs for preventing and treating metabolic diseases such as diabetes. To obtain a novel modulator of glucose metabolism, we conducted screening of a small compound library in cultured L6 myotubes. We identified nepodin that stimulated glucose uptake dose-dependently in differentiated L6 myotubes. The stimulatory effect of nepodin on glucose uptake was abrogated by a 5'-adenosine monophosphate-activated protein kinase (AMPK) inhibitor. In addition, nepodin stimulated the phosphorylation of AMPK. Nepodin also stimulated the translocation of GLUT4 to the plasma membrane in L6 myoblasts transfected with a Glut4 cDNA-coding vector and in differentiated L6 myotubes. In in vivo study, nepodin suppressed the increases in fasting blood glucose levels and improved the glucose intolerance of C57BL/KsJ-db/db mice, a type 2 diabetic animal model. Nepodin rescued the impaired phosphorylation of AMPK in the skeletal muscle of db/db mice. These results suggest that nepodin has an antidiabetic effect, which is at least partly mediated by stimulation of GLUT4 translocation via AMPK activation by nepodin.
Collapse
Affiliation(s)
- Byung Geun Ha
- Department of Nutriproteomics, Graduate School of Medicine, The University of Tokyo, Bunkyo-Ku, Tokyo, Japan
| | | | | | | | | | | | | |
Collapse
|
42
|
Son MJ, Miura Y, Yagasaki K. Mechanisms for antidiabetic effect of gingerol in cultured cells and obese diabetic model mice. Cytotechnology 2014; 67:641-52. [PMID: 24794903 DOI: 10.1007/s10616-014-9730-3] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2014] [Accepted: 04/15/2014] [Indexed: 12/16/2022] Open
Abstract
There have been studies on health beneficial effects of ginger and its components. However, there still remain certain aspects that are not well defined in their anti-hyperglycemic effects. Our aims were to find evidence of possible mechanisms for antidiabetic action of [6]-gingerol, a pungent component of ginger, employing a rat skeletal muscle-derived cell line, a rat-derived pancreatic β-cell line, and type 2 diabetic model animals. The antidiabetic effect of [6]-gingerol was investigated through studies on glucose uptake in L6 myocytes and on pancreatic β-cell protective ability from reactive oxygen species (ROS) in RIN-5F cells. Its in vivo effect was also examined using obese diabetic db/db mice. [6]-Gingerol increased glucose uptake under insulin absent condition and induced 5' adenosine monophosphate-activated protein kinase phosphorylation in L6 myotubes. Promotion by [6]-gingerol of glucose transporter 4 (GLUT4) translocation to plasma membrane was visually demonstrated by immunocytochemistry in L6 myoblasts transfected with glut4 cDNA-coding vector. [6]-Gingerol suppressed advanced glycation end product-induced rise of ROS levels in RIN-5F pancreatic β-cells. [6]-Gingerol feeding suppressed the increases in fasting blood glucose levels and improved glucose intolerance in db/db mice. [6]-Gingerol regulated hepatic gene expression of enzymes related to glucose metabolism toward decreases in gluconeogenesis and glycogenolysis as well as an increase in glycogenesis, thereby contributing to reductions in hepatic glucose production and hence blood glucose concentrations. These in vitro and in vivo results strongly suggest that [6]-gingerol has antidiabetic potential through multiple mechanisms.
Collapse
Affiliation(s)
- Myoung Jin Son
- Department of Applied Life Science, Graduate School of Agricultural Science, Tokyo University of Agriculture and Technology, Fuchu, Tokyo, 183-8509, Japan
| | | | | |
Collapse
|
43
|
Hu D, Peng J, Zhang X, Zheng H, Yan S, Zhang Y, Guan Q, Ding Q. Thyroid hormone exacerbates vasoconstriction in insulin resistance: The role of ONOO−. Eur J Pharmacol 2014; 730:41-50. [DOI: 10.1016/j.ejphar.2014.02.018] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2013] [Revised: 02/14/2014] [Accepted: 02/17/2014] [Indexed: 10/25/2022]
|
44
|
Lam YY, Janovská A, McAinch AJ, Belobrajdic DP, Hatzinikolas G, Game P, Wittert GA. The use of adipose tissue-conditioned media to demonstrate the differential effects of fat depots on insulin-stimulated glucose uptake in a skeletal muscle cell line. Obes Res Clin Pract 2013; 5:e1-e78. [PMID: 24331010 DOI: 10.1016/j.orcp.2010.12.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2010] [Revised: 12/05/2010] [Accepted: 12/15/2010] [Indexed: 01/04/2023]
Abstract
SUMMARY We aimed to study the depot-specific effect of adipose tissue on insulin sensitivity of skeletal muscle in vitro. Adipose tissue-conditioned medium (CM) was generated from visceral and subcutaneous fat from obese subjects. CM from visceral as compared to subcutaneous fat had higher concentrations of interleukin (IL)-6 (15-fold; P < 0.05) and IL-8 (8-fold; P < 0.05). CM from visceral fat (1:128 dilution) reduced insulin-stimulated glucose uptake in L6 myotubes by 19% (P < 0.05), an effect mediated by a nuclear factor kappa B (NFκB)/mammalian target of rapamycin complex 1 (mTORC1)-dependent pathway and partially reversed by neutralizing IL-6. IL-6 at a concentration comparable to that in CM from visceral fat reduced insulin-stimulated glucose uptake by 53% (P < 0.05), an effect abolished by inhibiting NFκB or mTORC1. We demonstrated the utility of the CM-myotube system and identified IL-6 as a major cytokine mediating visceral fat-induced muscle insulin resistance.:
Collapse
Affiliation(s)
- Yan Y Lam
- Discipline of Medicine, University of Adelaide, Adelaide, SA 5005, Australia.
| | - Alena Janovská
- Discipline of Medicine, University of Adelaide, Adelaide, SA 5005, Australia
| | - Andrew J McAinch
- School of Biomedical and Health Sciences, Victoria University, Melbourne, VIC 3000, Australia
| | | | - George Hatzinikolas
- Discipline of Medicine, University of Adelaide, Adelaide, SA 5005, Australia
| | - Philip Game
- Discipline of Surgery, University of Adelaide, Adelaide, SA 5005, Australia
| | - Gary A Wittert
- Discipline of Medicine, University of Adelaide, Adelaide, SA 5005, Australia
| |
Collapse
|
45
|
Smadja-Lamère N, Shum M, Déléris P, Roux PP, Abe JI, Marette A. Insulin activates RSK (p90 ribosomal S6 kinase) to trigger a new negative feedback loop that regulates insulin signaling for glucose metabolism. J Biol Chem 2013; 288:31165-76. [PMID: 24036112 DOI: 10.1074/jbc.m113.474148] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
We previously demonstrated that the mTORC1/S6K1 pathway is activated by insulin and nutrient overload (e.g. amino acids (AA)), which leads to the inhibition of the PI3K/Akt pathway via the inhibitory serine phosphorylation of IRS-1, notably on serine 1101 (Ser-1101). However, even in the absence of AA, insulin can still promote IRS-1 Ser-1101 phosphorylation by other kinases that remain to be fully characterized. Here, we describe a new negative regulator of IRS-1, the p90 ribosomal S6 kinase (RSK). Computational analyses revealed that Ser-1101 within IRS-1 falls into the consensus motif of RSK. Moreover, recombinant RSK phosphorylated IRS-1 C-terminal fragment on Ser-1101, which was prevented by mutations of this site or when a kinase-inactive mutant of RSK was used. Using antibodies directed toward the phosphorylation sites located in the activation segment of RSK (Ser-221 or Ser-380), we found that insulin activates RSK in L6 myocytes in the absence of AA overload. Inhibition of RSK using either the pharmacological inhibitor BI-D1870 or after adenoviral expression of a dominant negative RSK1 mutant (RSK1-DN) showed that RSK selectively phosphorylates IRS-1 on Ser-1101. Accordingly, expression of the RSK1-DN mutant in L6 myocytes and FAO hepatic cells improved insulin action on glucose uptake and glucose production, respectively. Furthermore, RSK1 inhibition prevented insulin resistance in L6 myocytes chronically exposed to high glucose and high insulin. These results show that RSK is a novel regulator of insulin signaling and glucose metabolism and a potential mediator of insulin resistance, notably through the negative phosphorylation of IRS-1 on Ser-1101.
Collapse
Affiliation(s)
- Nicolas Smadja-Lamère
- From the Quebec Heart and Lung Institute, Laval University, 2705 Chemin Ste-Foy, Ste-Foy (Quebec) G1V4G5, Canada
| | | | | | | | | | | |
Collapse
|
46
|
Resistin disrupts glycogen synthesis under high insulin and high glucose levels by down-regulating the hepatic levels of GSK3β. Gene 2013; 529:50-6. [PMID: 23860320 DOI: 10.1016/j.gene.2013.06.085] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2013] [Revised: 06/25/2013] [Accepted: 06/27/2013] [Indexed: 11/21/2022]
Abstract
The effect of mouse resistin on hepatic insulin resistance in vivo and in vitro, and its possible molecular mechanism were examined. Focusing on liver glycogen metabolism and gluconeogenesis, which are important parts of glucose metabolism, in primary cultures of rat hepatocytes we found that glycogen content was significantly lower (P<0.05) after treatment with recombinant murine resistin only in the presence of insulin plus glucose stimulation. Protein levels of factors in the insulin signaling pathway involved in glycogen synthesis were examined by Western blot analysis, with the only significant change observed being the level of phosphorylated (at Ser 9) glycogen synthase kinase-3β (GSK-3β) (P<0.001). No differences in the protein levels for the insulin receptor β (IRβ), insulin receptor substrates (IRS1 and IRS2), phosphatidylinositol 3-kinase (PI3K), protein kinase B (Akt) or their phosphorylated forms were observed between control and resistin treated primary rat hepatocytes. In a mouse model with high liver-specific expression of resistin, fasting blood glucose levels and liver glycogen content changed. Fasting blood glucose levels were significantly higher (P<0.001) in the model mice, compared to the control mice, while the glycogen content of the liver tissue was about 60% of that of the control mice (P<0.05). The gluconeogenic response was not altered between the experimental and control mice. The level of phosphorylated GSK-3β in the liver tissue was also decreased (P<0.05) in the model mice, consistent with the results from the primary rat hepatocytes. Our results suggest that resistin reduces the levels of GSK-3β phosphorylated at Ser 9 leading to impaired hepatic insulin action in primary rat hepatocytes and in a mouse model with high liver-specific expression of resistin.
Collapse
|
47
|
Xue R, Hao DD, Sun JP, Li WW, Zhao MM, Li XH, Chen Y, Zhu JH, Ding YJ, Liu J, Zhu YC. Hydrogen sulfide treatment promotes glucose uptake by increasing insulin receptor sensitivity and ameliorates kidney lesions in type 2 diabetes. Antioxid Redox Signal 2013; 19:5-23. [PMID: 23293908 PMCID: PMC3676664 DOI: 10.1089/ars.2012.5024] [Citation(s) in RCA: 104] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
AIMS To examine if hydrogen sulfide (H2S) can promote glucose uptake and provide amelioration in type 2 diabetes. RESULTS Treatment with sodium hydrosulfide (NaHS, an H2S donor) increased glucose uptake in both myotubes and adipocytes. The H2S gas solution showed similar effects. The NaHS effects were blocked by an siRNA-mediated knockdown of the insulin receptor (IR). NaHS also increased phosphorylation of the IR, PI3K, and Akt. In Goto-Kakizaki (GK) diabetic rats, chronic NaHS treatment (30 μmol·kg(-1)·day(-1)) decreased fasting blood glucose, increased insulin sensitivity, and increased glucose tolerance with increased phosphorylation of PI3K and Akt in muscles. Similar insulin-sensitizing effects of NaHS treatment were also observed in Wistar rats. Moreover, glucose uptake was reduced in the cells with siRNA-mediated knockdown of the H2S-generating enzyme cystathionine γ-lyase in the presence or absence of exogenous H2S. Moreover, chronic NaHS treatment reduced oxygen species and the number of crescentic glomeruli in the kidney of GK rats. INNOVATION AND CONCLUSION This study provides the first piece of evidence for the insulin-sensitizing effect of NaHS/H2S in the both in vitro and in vivo models of insulin resistance. REBOUND TRACK: This work was rejected during a standard peer review and rescued by the Rebound Peer Review (Antoxid Redox Signal 16: 293-296, 2012) with the following serving as open reviewers: Jin-Song Bian, Samuel Dudley, Hideo Kimura, and Xian Wang.
Collapse
Affiliation(s)
- Rong Xue
- Department of Physiology and Pathophysiology, Fudan University Shanghai Medical College, Shanghai, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Chen YH, Heneidi S, Lee JM, Layman LC, Stepp DW, Gamboa GM, Chen BS, Chazenbalk G, Azziz R. miRNA-93 inhibits GLUT4 and is overexpressed in adipose tissue of polycystic ovary syndrome patients and women with insulin resistance. Diabetes 2013; 62:2278-86. [PMID: 23493574 PMCID: PMC3712080 DOI: 10.2337/db12-0963] [Citation(s) in RCA: 199] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Approximately 70% of women with polycystic ovary syndrome (PCOS) have intrinsic insulin resistance (IR) above and beyond that associated with body mass, including dysfunctional glucose metabolism in adipose tissue (AT). In AT, analysis of the IRS/PI3-K/AKT pathway signaling components identified only GLUT4 expression to be significantly lower in PCOS patients and in control subjects with IR. We examined the role of miRNAs, particularly in the regulation of GLUT4, the insulin-sensitive glucose transporter, in the AT of PCOS and matched control subjects. PCOS AT was determined to have a differentially expressed miRNA profile, including upregulated miR-93, -133, and -223. GLUT4 is a highly predicted target for miR-93, while miR-133 and miR-223 have been demonstrated to regulate GLUT4 expression in cardiomyocytes. Expression of miR-93 revealed a strong correlation between the homeostasis model assessment of IR in vivo values and GLUT4 and miR-93 but not miR-133 and -223 expression in human AT. Overexpression of miR-93 resulted in downregulation of GLUT4 gene expression in adipocytes through direct targeting of the GLUT4 3'UTR, while inhibition of miR-93 activity led to increased GLUT4 expression. These results point to a novel mechanism for regulating insulin-stimulated glucose uptake via miR-93 and demonstrate upregulated miR-93 expression in all PCOS, and in non-PCOS women with IR, possibly accounting for the IR of the syndrome. In contrast, miR-133 and miR-223 may have a different, although yet to be defined, role in the IR of PCOS.
Collapse
Affiliation(s)
- Yen-Hao Chen
- Department of Obstetrics/Gynecology, Georgia Regents University, Augusta, Georgia
| | - Saleh Heneidi
- Department of Obstetrics/Gynecology, Georgia Regents University, Augusta, Georgia
| | - Jung-Min Lee
- Department of Internal Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Lawrence C. Layman
- Section of Reproductive Endocrinology, Infertility, & Genetics, Georgia Regents University, Augusta, Georgia
- Institute of Molecular Medicine and Genetics, Georgia Regents University, Augusta, Georgia
- Neuroscience Program, Georgia Regents University, Augusta, Georgia
| | - David W. Stepp
- Department of Physiology, Georgia Regents University, Augusta, Georgia
- Vascular Biology Center, Georgia Regents University, Augusta, Georgia
| | | | - Bo-Shiun Chen
- Institute of Molecular Medicine and Genetics, Georgia Regents University, Augusta, Georgia
- Neuroscience Program, Georgia Regents University, Augusta, Georgia
| | - Gregorio Chazenbalk
- Department of Obstetrics/Gynecology, Center for Health Sciences, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, California
| | - Ricardo Azziz
- Department of Obstetrics/Gynecology, Georgia Regents University, Augusta, Georgia
- Department of Medicine, Georgia Regents University, Augusta, Georgia
- Corresponding author: Ricardo Azziz,
| |
Collapse
|
49
|
Vaughan RA, Garcia-Smith R, Bisoffi M, Conn CA, Trujillo KA. Ubiquinol rescues simvastatin-suppression of mitochondrial content, function and metabolism: implications for statin-induced rhabdomyolysis. Eur J Pharmacol 2013; 711:1-9. [PMID: 23624330 DOI: 10.1016/j.ejphar.2013.04.009] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2012] [Revised: 04/03/2013] [Accepted: 04/04/2013] [Indexed: 01/03/2023]
Abstract
Statin medications diminish cholesterol biosynthesis and are commonly prescribed to reduce cardiovascular disease. Statins also reduce production of ubiquinol, a vital component of mitochondrial energy production; ubiquinol reduction may contribute to rhabdomyolysis. Human rhabdomyosarcoma cells were treated with either ethanol and dimethyl sulfoxide (DMSO) control, or simvastatin at 5 µM or 10 µM, or simvastatin at 5 µM with ubiquinol at 0.5 µM or 1.0 µM for 24 h or 48 h. PGC-1α RNA levels were determined using quantitative reverse transcriptase polymerase chain reaction (qRT-PCR). Mitochondrial content was determined using flow cytometry and immunocytochemistry. Metabolism was determined by quantification of extracellular acidification rate and oxygen consumption rate. Treatment of human rhabdomyosarcoma cells with simvastatin significantly reduced oxidative, total metabolism, and cellular ATP content in a time- and dose-dependent manner which was rescued by concurrent treatment with ubiquinol. Treatment with simvastatin significantly reduced mitochondrial content as well as cell viability which were both rescued by simultaneous treatment with ubiquinol. This work demonstrates that the addition of ubiquinol to current statin treatment regimens may protect muscle cells from myopathies.
Collapse
Affiliation(s)
- Roger A Vaughan
- Department of Health, Exercise and Sports Science, University of New Mexico, Albuquerque, NM 87131, USA.
| | | | | | | | | |
Collapse
|
50
|
Solomon TPJ, Knudsen SH, Karstoft K, Winding K, Holst JJ, Pedersen BK. Examining the effects of hyperglycemia on pancreatic endocrine function in humans: evidence for in vivo glucotoxicity. J Clin Endocrinol Metab 2012; 97:4682-91. [PMID: 23043193 DOI: 10.1210/jc.2012-2097] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
CONTEXT Investigating the impact of hyperglycemia on pancreatic endocrine function promotes our understanding of the pathophysiology of hyperglycemia-related disease. OBJECTIVE The objective of the study was to test the hypothesis that experimental hyperglycemia impairs insulin and glucagon secretion. DESIGN A randomized, crossover in healthy controls, compared with type 2 diabetic patients. SETTING The study was conducted at a university hospital. PARTICIPANTS Normal glucose-tolerant subjects (n = 10) and patients with type 2 diabetes (n = 10), individually matched by age, sex, and body mass index. INTERVENTIONS Normal glucose-tolerant subjects underwent 24 h of experimental hyperglycemia (+5.4 mm above basal). Subjects with type 2 diabetes did not undergo an intervention. MAIN OUTCOME MEASURES Insulin secretion, glucagon secretion, insulin sensitivity, disposition index, and endogenous glucose production (via [6,6-(2)H(2)]glucose infusion) were measured during hyperglycemic clamps combined with infusion of glucagon-like peptide (GLP)-1(7-36) (0.5 pmol/kg · min) and injection of arginine (5 g). RESULTS Insulin secretion was correlated with glucagon suppression in subjects with normal glucose tolerance only. Individuals with type 2 diabetes had lower insulin sensitivity (-33 ± 11%) and insulin secretory responses to glucose, GLP-1, and arginine (-40 ± 11, -58 ± 7, and -36 ± 13%, respectively) and higher plasma glucagon and endogenous glucose production compared with normal glucose-tolerant subjects (all P < 0.05). After 24 h of experimental hyperglycemia, insulin sensitivity (-29 ± 10%), disposition index (-24 ± 16%), and GLP-1- (-19 ± 7%) and arginine-stimulated (-15 ± 10%) insulin secretion were decreased in normal glucose-tolerant subjects (all P < 0.05). However, plasma glucagon responses were not affected. Furthermore, experimental hyperglycemia abolished the correlation between insulin secretion and glucagon suppression. CONCLUSIONS Experimental hyperglycemia impaired pancreatic β-cell function but did not acutely impair α-cell glucagon secretion in normal glucose-tolerant subjects.
Collapse
Affiliation(s)
- Thomas P J Solomon
- The Centre of Inflammation and Metabolism, Department of Infectious Diseases, Rigshospitalet, Copenhagen 2100, Denmark.
| | | | | | | | | | | |
Collapse
|