51
|
Edick AM, Auclair O, Burgos SA. Role of Grb10 in mTORC1-dependent regulation of insulin signaling and action in human skeletal muscle cells. Am J Physiol Endocrinol Metab 2020; 318:E173-E183. [PMID: 31794259 DOI: 10.1152/ajpendo.00025.2019] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Growth factor receptor-bound protein 10 (Grb10) is an adaptor protein that binds to the insulin receptor, upon which insulin signaling and action are thought to be inhibited. Grb10 is also a substrate for the mechanistic target of rapamycin complex 1 (mTORC1) that mediates its feedback inhibition on phosphatidylinositide 3-kinase (PI3K)/Akt signaling. To characterize the function of Grb10 and its regulation by mTORC1 in human muscle, primary skeletal muscle cells were isolated from healthy lean young men and then induced to differentiate into myotubes. Knockdown of Grb10 enhanced insulin-induced PI3K/Akt signaling and glucose uptake in myotubes, reinforcing the notion underlying its function as a negative regulator of insulin action in human muscle. The increased insulin responsiveness in Grb10-silenced myotubes was associated with a higher abundance of the insulin receptor. Furthermore, insulin and amino acids independently and additively stimulated phosphorylation of Grb10 at Ser476. However, acute inhibition of mTORC1 with rapamycin blocked Grb10 Ser476 phosphorylation and repressed a negative-feedback loop on PI3K/Akt signaling that increased myotube responsiveness to insulin. Chronic rapamycin treatment reduced Grb10 protein abundance in conjunction with increased insulin receptor protein levels. Based on these findings, we propose that mTORC1 controls PI3K/Akt signaling through modulation of insulin receptor abundance by Grb10. These findings have potential implications for obesity-linked insulin resistance, as well as clinical use of mTORC1 inhibitors.
Collapse
Affiliation(s)
- Ashlin M Edick
- Department of Animal Science, McGill University, Sainte-Anne-de-Bellevue, Quebec, Canada
| | - Olivia Auclair
- Department of Animal Science, McGill University, Sainte-Anne-de-Bellevue, Quebec, Canada
| | - Sergio A Burgos
- Department of Animal Science, McGill University, Sainte-Anne-de-Bellevue, Quebec, Canada
- Department of Medicine, McGill University, Montreal, Quebec, Canada
- Metabolic Disorders and Complications Program, Research Institute of McGill University Health Centre, Montreal, Quebec, Canada
| |
Collapse
|
52
|
Sayed S, Ahmed M, El-Shehawi A, Alkafafy M, Al-Otaibi S, El-Sawy H, Farouk S, El-Shazly S. Ginger Water Reduces Body Weight Gain and Improves Energy Expenditure in Rats. Foods 2020; 9:E38. [PMID: 31906567 PMCID: PMC7023345 DOI: 10.3390/foods9010038] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 12/26/2019] [Accepted: 12/30/2019] [Indexed: 12/22/2022] Open
Abstract
Obesity is a serious global problem that causes predisposition to numerous serious diseases. The current study aims to investigate the effect of ginger water on body weight and energy expenditure through modulation of mRNA expression of carbohydrate and lipid metabolism. A white colored liquid obtained during freeze-drying of fresh rhizomes of Zingiber officinal was collected and named ginger water. It was used to treat rats, then blood and tissue samples were collected from the liver and white adipose at the end of the experiment. The serum was prepared and used for biochemical assays, while tissue samples were used for RNA isolation and gene expression analysis via Reverse transcription polymerase chain reaction (RT-PCR). Results of High Performance Liquid Chromatography (HPLC) analysis of ginger water revealed the presence of chrysin and galangin at concentrations of 0.24 µg/mL and 0.53 µg/mL, respectively. Average body weight gain decreased significantly in groups that received ginger water. In addition, both total cholesterol and serum triacylglycerol were reduced in the groups that received ginger water. Furthermore, mRNA expression of Sterol regulatory element-binding protein 1 (SREBP-1c) in the liver and leptin in adipose tissues were downregulated, while those of adiponectin, hepatic carnitine palmitoyltransferase1 (CPT-1), acyl-coA oxidase (ACO), Glucose transporter 2 (GLUT-2), and pyruvate kinase (PK) were upregulated in ginger water-treated groups. These results clearly revealed the lowering body weight gain effect of ginger water, which most likely occurs at the transcriptional level of energy metabolizing proteins.
Collapse
Affiliation(s)
- Samy Sayed
- Department of Biotechnology, Faculty of Science, Taif University, Taif 21974, Saudi Arabia; (S.S.); (A.E.-S.); (M.A.); (S.A.-O.); (S.F.)
- Faculty of Agriculture, Cairo University, Giza 12613, Egypt
| | - Mohamed Ahmed
- Faculty of Veterinary Medicine, University of Sadat City, Sadat City 32958, Egypt;
| | - Ahmed El-Shehawi
- Department of Biotechnology, Faculty of Science, Taif University, Taif 21974, Saudi Arabia; (S.S.); (A.E.-S.); (M.A.); (S.A.-O.); (S.F.)
- Department of Genetics, Faculty of Agriculture, University of Alexandria, Alexandria 21526, Egypt
| | - Mohamed Alkafafy
- Department of Biotechnology, Faculty of Science, Taif University, Taif 21974, Saudi Arabia; (S.S.); (A.E.-S.); (M.A.); (S.A.-O.); (S.F.)
- Faculty of Veterinary Medicine, University of Sadat City, Sadat City 32958, Egypt;
| | - Saqer Al-Otaibi
- Department of Biotechnology, Faculty of Science, Taif University, Taif 21974, Saudi Arabia; (S.S.); (A.E.-S.); (M.A.); (S.A.-O.); (S.F.)
| | - Hanan El-Sawy
- Department of Nutrition and Clinical Nutrition, Faculty of Veterinary Medicine, Kafrelsheikh University, Kafrelsheikh 33516, Egypt;
| | - Samy Farouk
- Department of Biotechnology, Faculty of Science, Taif University, Taif 21974, Saudi Arabia; (S.S.); (A.E.-S.); (M.A.); (S.A.-O.); (S.F.)
| | - Samir El-Shazly
- Department of Biotechnology, Faculty of Science, Taif University, Taif 21974, Saudi Arabia; (S.S.); (A.E.-S.); (M.A.); (S.A.-O.); (S.F.)
- Department of Biochemistry, Faculty of Veterinary Medicine, Kafrelsheikh University, Kafr Elsheikh 33511, Egypt
| |
Collapse
|
53
|
Kusumastuti SA, Nugrahaningsih DAA, Wahyuningsih MSH. Centella asiatica (L.) extract attenuates inflammation and improve insulin sensitivity in a coculture of lipopolysaccharide (LPS)-induced 3T3-L1 adipocytes and RAW 264.7 macrophages. Drug Discov Ther 2019; 13:261-267. [DOI: 10.5582/ddt.2019.01052] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Affiliation(s)
- Siska Andrina Kusumastuti
- Center of Pharmaceutical and Medicine Technology, Agency for the Assessment and Application of Technology (BPPT), Jakarta, Indonesia
| | | | | |
Collapse
|
54
|
Zhou X, Fouda S, Zeng XY, Li D, Zhang K, Xu J, Ye JM. Characterization of the Therapeutic Profile of Albiflorin for the Metabolic Syndrome. Front Pharmacol 2019; 10:1151. [PMID: 31680948 PMCID: PMC6797612 DOI: 10.3389/fphar.2019.01151] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2019] [Accepted: 09/06/2019] [Indexed: 12/12/2022] Open
Abstract
Albiflorin (AF) is a small molecule (MW 481) isolated from Paeoniae radix, a plant used as a remedy for various conditions with pathogenesis shared by metabolic diseases. Reported here is our characterization of its therapeutic profiles in three mouse models with distinctive pathological features of metabolic syndrome (MetS). Our results firstly showed that AF alleviated high fat (HF) induced obesity and associated glucose intolerance, suggesting its therapeutic efficacy for MetS. In the type 2 diabetes (T2D) model induced by a combination of HF and low doses of streptozotocin, AF lowered hyperglycaemia and improved insulin-stimulated glucose disposal. In the non-alcoholic steatohepatitis-like model resulting from a HF and high cholesterol (HF-HC) diet, AF reversed the increased liver triglyceride and cholesterol, plasma aspartate aminotransferase, and liver TNFα mRNA levels. Consistent with its effect in promoting glucose disposal in HF-fed mice, AF stimulated glucose uptake and GLUT4 translocation to the plasma membrane in L6 myotubes. However, these effects were unlikely to be associated with activation of insulin, AMPK, ER, or cellular stress signalling cascades. Further studies revealed that AF increased the whole-body energy expenditure and physical activity. Taken together, our findings indicate that AF exerts a therapeutic potential for MetS and related diseases possibly by promoting physical activity associated whole-body energy expenditure and glucose uptake in muscle. These effects are possibly mediated by a new mechanism distinct from other therapeutics derived from Chinese medicine.
Collapse
Affiliation(s)
- Xiu Zhou
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen, China.,School of Health and Biomedical Sciences, RMIT University, Melbourne, VIC, Australia.,School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, China
| | - Sherouk Fouda
- School of Health and Biomedical Sciences, RMIT University, Melbourne, VIC, Australia
| | - Xiao-Yi Zeng
- School of Health and Biomedical Sciences, RMIT University, Melbourne, VIC, Australia
| | - Dongli Li
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen, China
| | - Kun Zhang
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen, China.,School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, China
| | - Jun Xu
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen, China
| | - Ji-Ming Ye
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen, China.,School of Health and Biomedical Sciences, RMIT University, Melbourne, VIC, Australia
| |
Collapse
|
55
|
Kearney AL, Cooke KC, Norris DM, Zadoorian A, Krycer JR, Fazakerley DJ, Burchfield JG, James DE. Serine 474 phosphorylation is essential for maximal Akt2 kinase activity in adipocytes. J Biol Chem 2019; 294:16729-16739. [PMID: 31548312 DOI: 10.1074/jbc.ra119.010036] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 09/15/2019] [Indexed: 01/06/2023] Open
Abstract
The Ser/Thr protein kinase Akt regulates essential biological processes such as cell survival, growth, and metabolism. Upon growth factor stimulation, Akt is phosphorylated at Ser474; however, how this phosphorylation contributes to Akt activation remains controversial. Previous studies, which induced loss of Ser474 phosphorylation by ablating its upstream kinase mTORC2, have implicated Ser474 phosphorylation as a driver of Akt substrate specificity. Here we directly studied the role of Akt2 Ser474 phosphorylation in 3T3-L1 adipocytes by preventing Ser474 phosphorylation without perturbing mTORC2 activity. This was achieved by utilizing a chemical genetics approach, where ectopically expressed S474A Akt2 was engineered with a W80A mutation to confer resistance to the Akt inhibitor MK2206, and thus allow its activation independent of endogenous Akt. We found that insulin-stimulated phosphorylation of four bona fide Akt substrates (TSC2, PRAS40, FOXO1/3a, and AS160) was reduced by ∼50% in the absence of Ser474 phosphorylation. Accordingly, insulin-stimulated mTORC1 activation, protein synthesis, FOXO nuclear exclusion, GLUT4 translocation, and glucose uptake were attenuated upon loss of Ser474 phosphorylation. We propose a model where Ser474 phosphorylation is required for maximal Akt2 kinase activity in adipocytes.
Collapse
Affiliation(s)
- Alison L Kearney
- Charles Perkins Centre, School of Life and Environmental Sciences, University of Sydney, Sydney, New South Wales 2006, Australia
| | - Kristen C Cooke
- Charles Perkins Centre, School of Life and Environmental Sciences, University of Sydney, Sydney, New South Wales 2006, Australia
| | - Dougall M Norris
- Charles Perkins Centre, School of Life and Environmental Sciences, University of Sydney, Sydney, New South Wales 2006, Australia
| | - Armella Zadoorian
- Charles Perkins Centre, School of Life and Environmental Sciences, University of Sydney, Sydney, New South Wales 2006, Australia
| | - James R Krycer
- Charles Perkins Centre, School of Life and Environmental Sciences, University of Sydney, Sydney, New South Wales 2006, Australia
| | - Daniel J Fazakerley
- Charles Perkins Centre, School of Life and Environmental Sciences, University of Sydney, Sydney, New South Wales 2006, Australia
| | - James G Burchfield
- Charles Perkins Centre, School of Life and Environmental Sciences, University of Sydney, Sydney, New South Wales 2006, Australia
| | - David E James
- Charles Perkins Centre, School of Life and Environmental Sciences, University of Sydney, Sydney, New South Wales 2006, Australia .,Sydney Medical School, University of Sydney, Sydney, New South Wales 2006, Australia
| |
Collapse
|
56
|
Wang Y, Zhou H, Palyha O, Mu J. Restoration of insulin receptor improves diabetic phenotype in T2DM mice. JCI Insight 2019; 4:124945. [PMID: 31391336 DOI: 10.1172/jci.insight.124945] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Accepted: 06/27/2019] [Indexed: 12/14/2022] Open
Abstract
Type 2 diabetes mellitus (T2DM), also known as adult-onset diabetes, is characterized by ineffective insulin action due to insulin resistance in key metabolic tissues. Insulin receptor (IR) plays an important role in insulin signal transduction, defect of which has been considered the fundamental cause of T2DM. IR content reduction in diabetes is one key contributor to the defective insulin signaling and diabetes progression. Rescuing IR levels by transgenic complementation has not been considered as a treatment option because it is limited by uncontrollable expression level, tissue selectivity, or developmental defects. In the current study, we demonstrated that single-dose adeno-associated virus (AAV) vector delivered expression of human IR (hIR) in the liver of inducible IR-knockout mice and significantly improved the diabetic phenotype caused by IR deletion during adulthood. Such an approach was also applied, for the first time to our knowledge, to treating ob/ob mice, a model of severe T2DM attributed to superfluous calorie intake and insulin resistance. Interestingly, similar treatment with AAV-hIR had no obvious effect in healthy animals, indicative of low hypoglycemic risk as a consequence of potential excessive insulin action. The results described here support restoration of IR expression as a safe and effective T2DM therapeutic with a long-lasting profile.
Collapse
MESH Headings
- Animals
- Antigens, CD/genetics
- Antigens, CD/metabolism
- Blood Glucose/analysis
- Dependovirus/genetics
- Diabetes Mellitus, Type 2/blood
- Diabetes Mellitus, Type 2/genetics
- Diabetes Mellitus, Type 2/pathology
- Diabetes Mellitus, Type 2/therapy
- Disease Models, Animal
- Genetic Therapy/adverse effects
- Genetic Therapy/methods
- Genetic Vectors/administration & dosage
- Genetic Vectors/adverse effects
- Genetic Vectors/genetics
- Humans
- Hypoglycemia/blood
- Hypoglycemia/diagnosis
- Hypoglycemia/genetics
- Insulin/metabolism
- Male
- Mice
- Mice, Knockout
- Receptor, Insulin/genetics
- Receptor, Insulin/metabolism
- Treatment Outcome
Collapse
|
57
|
Jaiswal N, Gavin MG, Quinn WJ, Luongo TS, Gelfer RG, Baur JA, Titchenell PM. The role of skeletal muscle Akt in the regulation of muscle mass and glucose homeostasis. Mol Metab 2019; 28:1-13. [PMID: 31444134 PMCID: PMC6822261 DOI: 10.1016/j.molmet.2019.08.001] [Citation(s) in RCA: 115] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 07/29/2019] [Accepted: 08/01/2019] [Indexed: 12/15/2022] Open
Abstract
OBJECTIVE Skeletal muscle insulin signaling is a major determinant of muscle growth and glucose homeostasis. Protein kinase B/Akt plays a prominent role in mediating many of the metabolic effects of insulin. Mice and humans harboring systemic loss-of-function mutations in Akt2, the most abundant Akt isoform in metabolic tissues, are glucose intolerant and insulin resistant. Since the skeletal muscle accounts for a significant amount of postprandial glucose disposal, a popular hypothesis in the diabetes field suggests that a reduction in Akt, specifically in skeletal muscle, leads to systemic glucose intolerance and insulin resistance. Despite this common belief, the specific role of skeletal muscle Akt in muscle growth and insulin sensitivity remains undefined. METHODS We generated multiple mouse models of skeletal muscle Akt deficiency to evaluate the role of muscle Akt signaling in vivo. The effects of these genetic perturbations on muscle mass, glucose homeostasis and insulin sensitivity were assessed using both in vivo and ex vivo assays. RESULTS Surprisingly, mice lacking Akt2 alone in skeletal muscle displayed normal skeletal muscle insulin signaling, glucose tolerance, and insulin sensitivity despite a dramatic reduction in phosphorylated Akt. In contrast, deletion of both Akt isoforms (M-AktDKO) prevented downstream signaling and resulted in muscle atrophy. Despite the absence of Akt signaling, in vivo and ex vivo insulin-stimulated glucose uptake were normal in M-AktDKO mice. Similar effects on insulin sensitivity were observed in mice with prolonged deletion (4 weeks) of both skeletal muscle Akt isoforms selectively in adulthood. Conversely, short term deletion (2 weeks) of skeletal muscle specific Akt in adult muscles impaired insulin tolerance paralleling the effect observed by acute pharmacological inhibition of Akt in vitro. Mechanistically, chronic ablation of Akt induced mitochondrial dysfunction and activation of AMPK, which was required for insulin-stimulated glucose uptake in the absence of Akt. CONCLUSIONS Together, these data indicate that chronic reduction in Akt activity alone in skeletal muscle is not sufficient to induce insulin resistance or prevent glucose uptake in all conditions. Therefore, since insulin-stimulated glucose disposal in skeletal muscle is markedly impaired in insulin-resistant states, we hypothesize that alterations in signaling molecules in addition to skeletal muscle Akt are necessary to perturb glucose tolerance and insulin sensitivity in vivo.
Collapse
Affiliation(s)
- N Jaiswal
- Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - M G Gavin
- Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - W J Quinn
- Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - T S Luongo
- Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - R G Gelfer
- Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - J A Baur
- Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA; Department of Physiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - P M Titchenell
- Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA; Department of Physiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
58
|
Suzawa M, Muhammad NM, Joseph BS, Bland ML. The Toll Signaling Pathway Targets the Insulin-like Peptide Dilp6 to Inhibit Growth in Drosophila. Cell Rep 2019; 28:1439-1446.e5. [DOI: 10.1016/j.celrep.2019.07.015] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 05/28/2019] [Accepted: 07/03/2019] [Indexed: 01/08/2023] Open
|
59
|
Grice BA, Barton KJ, Covert JD, Kreilach AM, Tackett L, Brozinick JT, Elmendorf JS. Excess membrane cholesterol is an early contributing reversible aspect of skeletal muscle insulin resistance in C57BL/6NJ mice fed a Western-style high-fat diet. Am J Physiol Endocrinol Metab 2019; 317:E362-E373. [PMID: 31237447 PMCID: PMC6732462 DOI: 10.1152/ajpendo.00396.2018] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Skeletal muscle insulin resistance manifests shortly after high-fat feeding, yet mechanisms are not known. Here we set out to determine whether excess skeletal muscle membrane cholesterol and cytoskeletal derangement known to compromise glucose transporter (GLUT)4 regulation occurs early after high-fat feeding. We fed 6-wk-old male C57BL/6NJ mice either a low-fat (LF, 10% kcal) or a high-fat (HF, 45% kcal) diet for 1 wk. This HF feeding challenge was associated with an increase, albeit slight, in body mass, glucose intolerance, and hyperinsulinemia. Liver analyses did not reveal signs of hepatic insulin resistance; however, skeletal muscle immunoblots of triad-enriched regions containing transverse tubule membrane showed a marked loss of stimulated GLUT4 recruitment. An increase in cholesterol was also found in these fractions from HF-fed mice. These derangements were associated with a marked loss of cortical filamentous actin (F-actin) that is essential for GLUT4 regulation and known to be compromised by increases in membrane cholesterol. Both the withdrawal of the HF diet and two subcutaneous injections of the cholesterol-lowering agent methyl-β-cyclodextrin at 3 and 6 days during the 1-wk HF feeding intervention completely mitigated cholesterol accumulation, cortical F-actin loss, and GLUT4 dysregulation. Moreover, these beneficial membrane/cytoskeletal changes occurred concomitant with a full restoration of metabolic responses. These results identify skeletal muscle membrane cholesterol accumulation as an early, reversible, feature of insulin resistance and suggest cortical F-actin loss as an early derangement of skeletal muscle insulin resistance.
Collapse
Affiliation(s)
- Brian A Grice
- Department of Anatomy, Cell Biology, and Physiology, Indiana University School of Medicine, Indianapolis, Indiana
- Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, Indiana
| | - Kelly J Barton
- Department of Anatomy, Cell Biology, and Physiology, Indiana University School of Medicine, Indianapolis, Indiana
- Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, Indiana
| | - Jacob D Covert
- Department of Anatomy, Cell Biology, and Physiology, Indiana University School of Medicine, Indianapolis, Indiana
- Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, Indiana
| | - Alec M Kreilach
- Department of Anatomy, Cell Biology, and Physiology, Indiana University School of Medicine, Indianapolis, Indiana
- Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, Indiana
| | - Lixuan Tackett
- Department of Anatomy, Cell Biology, and Physiology, Indiana University School of Medicine, Indianapolis, Indiana
- Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, Indiana
| | - Joseph T Brozinick
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana
- Eli Lilly and Company, Indianapolis, Indiana
| | - Jeffrey S Elmendorf
- Department of Anatomy, Cell Biology, and Physiology, Indiana University School of Medicine, Indianapolis, Indiana
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana
- Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, Indiana
| |
Collapse
|
60
|
Roth SW, Bitterman MD, Birnbaum MJ, Bland ML. Innate Immune Signaling in Drosophila Blocks Insulin Signaling by Uncoupling PI(3,4,5)P 3 Production and Akt Activation. Cell Rep 2019. [PMID: 29514084 PMCID: PMC5866056 DOI: 10.1016/j.celrep.2018.02.033] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
In obese adipose tissue, Toll-like receptor signaling in macrophages leads to insulin resistance in adipocytes. Similarly, Toll signaling in the Drosophila larval fat body blocks insulin-dependent growth and nutrient storage. We find that Toll acts cell autonomously to block growth but not PI(3,4,5)P3 production in fat body cells expressing constitutively active PI3K. Fat body Toll signaling blocks whole-animal growth in rictor mutants lacking TORC2 activity, but not in larvae lacking Pdk1. Phosphorylation of Akt on the Pdk1 site, Thr342, is significantly reduced by Toll signaling, and expression of mutant AktT342D rescues cell and animal growth, nutrient storage, and viability in animals with active Toll signaling. Altogether, these data show that innate immune signaling blocks insulin signaling at a more distal level than previously appreciated, and they suggest that manipulations affecting the Pdk1 arm of the pathway may have profound effects on insulin sensitivity in inflamed tissues.
Collapse
Affiliation(s)
- Stephen W Roth
- Department of Pharmacology, University of Virginia, Charlottesville, VA 22908, USA
| | - Moshe D Bitterman
- Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Morris J Birnbaum
- Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Michelle L Bland
- Department of Pharmacology, University of Virginia, Charlottesville, VA 22908, USA.
| |
Collapse
|
61
|
Li J, Bai L, Wei F, Zhao J, Wang D, Xiao Y, Yan W, Wei J. Therapeutic Mechanisms of Herbal Medicines Against Insulin Resistance: A Review. Front Pharmacol 2019; 10:661. [PMID: 31258478 PMCID: PMC6587894 DOI: 10.3389/fphar.2019.00661] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Accepted: 05/23/2019] [Indexed: 12/16/2022] Open
Abstract
Insulin resistance is a condition in which insulin sensitivity is reduced and the insulin signaling pathway is impaired. Although often expressed as an increase in insulin concentration, the disease is characterized by a decrease in insulin action. This increased workload of the pancreas and the consequent decompensation are not only the main mechanisms for the development of type 2 diabetes (T2D), but also exacerbate the damage of metabolic diseases, including obesity, nonalcoholic fatty liver disease, polycystic ovary syndrome, metabolic syndrome, and others. Many clinical trials have suggested the potential role of herbs in the treatment of insulin resistance, although most of the clinical trials included in this review have certain flaws and bias risks in their methodological design, including the generation of randomization, the concealment of allocation, blinding, and inadequate reporting of sample size estimates. These studies involve not only the single-flavored herbs, but also herbal formulas, extracts, and active ingredients. Numerous of in vitro and in vivo studies have pointed out that the role of herbal medicine in improving insulin resistance is related to interventions in various aspects of the insulin signaling pathway. The targets involved in these studies include insulin receptor substrate, phosphatidylinositol 3-kinase, glucose transporter, AMP-activated protein kinase, glycogen synthase kinase 3, mitogen-activated protein kinases, c-Jun-N-terminal kinase, nuclear factor-kappaB, protein tyrosine phosphatase 1B, nuclear factor-E2-related factor 2, and peroxisome proliferator-activated receptors. Improved insulin sensitivity upon treatment with herbal medicine provides considerable prospects for treating insulin resistance. This article reviews studies of the target mechanisms of herbal treatments for insulin resistance.
Collapse
Affiliation(s)
- Jun Li
- Department of Endocrinology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China.,Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Litao Bai
- Department of Endocrinology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Fan Wei
- Department of Endocrinology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jing Zhao
- Department of Endocrinology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Danwei Wang
- Department of Endocrinology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yao Xiao
- Department of Endocrinology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Weitian Yan
- Department of Endocrinology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Junping Wei
- Department of Endocrinology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
62
|
Abstract
A pivotal metabolic function of insulin is the stimulation of glucose uptake into muscle and adipose tissues. The discovery of the insulin-responsive glucose transporter type 4 (GLUT4) protein in 1988 inspired its molecular cloning in the following year. It also spurred numerous cellular mechanistic studies laying the foundations for how insulin regulates glucose uptake by muscle and fat cells. Here, we reflect on the importance of the GLUT4 discovery and chronicle additional key findings made in the past 30 years. That exocytosis of a multispanning membrane protein regulates cellular glucose transport illuminated a novel adaptation of the secretory pathway, which is to transiently modulate the protein composition of the cellular plasma membrane. GLUT4 controls glucose transport into fat and muscle tissues in response to insulin and also into muscle during exercise. Thus, investigation of regulated GLUT4 trafficking provides a major means by which to map the essential signaling components that transmit the effects of insulin and exercise. Manipulation of the expression of GLUT4 or GLUT4-regulating molecules in mice has revealed the impact of glucose uptake on whole-body metabolism. Remaining gaps in our understanding of GLUT4 function and regulation are highlighted here, along with opportunities for future discoveries and for the development of therapeutic approaches to manage metabolic disease.
Collapse
Affiliation(s)
- Amira Klip
- Cell Biology Program, Hospital for Sick Children, Toronto, Ontario M5G 1X8, Canada
| | - Timothy E McGraw
- Department of Biochemistry, Weill Medical College of Cornell University, New York, New York 10065
| | - David E James
- Charles Perkins Centre, School of Life and Environmental Sciences, Sydney Medical School, University of Sydney, Camperdown, New South Wales 2050, Australia
| |
Collapse
|
63
|
Small L, Brandon AE, Parker BL, Deshpande V, Samsudeen AF, Kowalski GM, Reznick J, Wilks DL, Preston E, Bruce CR, James DE, Turner N, Cooney GJ. Reduced insulin action in muscle of high fat diet rats over the diurnal cycle is not associated with defective insulin signaling. Mol Metab 2019; 25:107-118. [PMID: 31029696 PMCID: PMC6600078 DOI: 10.1016/j.molmet.2019.04.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 04/04/2019] [Accepted: 04/08/2019] [Indexed: 12/02/2022] Open
Abstract
Objective Energy metabolism and insulin action follow a diurnal rhythm. It is therefore important that investigations into dysregulation of these pathways are relevant to the physiology of this diurnal rhythm. Methods We examined glucose uptake, markers of insulin action, and the phosphorylation of insulin signaling intermediates in muscle of chow and high fat, high sucrose (HFHS) diet-fed rats over the normal diurnal cycle. Results HFHS animals displayed hyperinsulinemia but had reduced systemic glucose disposal and lower muscle glucose uptake during the feeding period. Analysis of gene expression, enzyme activity, protein abundance and phosphorylation revealed a clear diurnal regulation of substrate oxidation pathways with no difference in Akt signaling in muscle. Transfection of a constitutively active Akt2 into the muscle of HFHS rats did not rescue diet-induced reductions in insulin-stimulated glucose uptake. Conclusions These studies suggest that reduced glucose uptake in muscle during the diurnal cycle induced by short-term HFHS-feeding is not the result of reduced insulin signaling. Investigating metabolism in rodents over the diurnal cycle more accurately models normal animal physiology. Diurnal regulation of substrate oxidation is altered in muscle of HFHS-fed rats. There is a disconnect between glucose uptake and canonical insulin signaling in muscle. Activation of Akt2 does not rescue diet-induced reductions in insulin-stimulated glucose uptake in muscle.
Collapse
Affiliation(s)
- Lewin Small
- Diabetes and Metabolism Division, The Garvan Institute of Medical Research, Sydney, NSW, Australia
| | - Amanda E Brandon
- Diabetes and Metabolism Division, The Garvan Institute of Medical Research, Sydney, NSW, Australia; The University of Sydney, School of Medical Science, Charles Perkins Centre D17, Sydney, NSW, Australia
| | - Benjamin L Parker
- The University of Sydney, School of Life and Environmental Science, Charles Perkins Centre D17, Sydney, NSW, Australia
| | - Vinita Deshpande
- The University of Sydney, School of Life and Environmental Science, Charles Perkins Centre D17, Sydney, NSW, Australia
| | - Azrah F Samsudeen
- Department of Pharmacology, School of Medical Science, University of New South Wales, Sydney, NSW, Australia
| | - Greg M Kowalski
- Deakin University, School of Exercise and Nutrition Sciences, Faculty of Health, Institute for Physical Activity and Nutrition, Geelong, Australia
| | - Jane Reznick
- Diabetes and Metabolism Division, The Garvan Institute of Medical Research, Sydney, NSW, Australia
| | - Donna L Wilks
- Diabetes and Metabolism Division, The Garvan Institute of Medical Research, Sydney, NSW, Australia
| | - Elaine Preston
- Diabetes and Metabolism Division, The Garvan Institute of Medical Research, Sydney, NSW, Australia
| | - Clinton R Bruce
- Deakin University, School of Exercise and Nutrition Sciences, Faculty of Health, Institute for Physical Activity and Nutrition, Geelong, Australia
| | - David E James
- The University of Sydney, School of Life and Environmental Science, Charles Perkins Centre D17, Sydney, NSW, Australia
| | - Nigel Turner
- Department of Pharmacology, School of Medical Science, University of New South Wales, Sydney, NSW, Australia
| | - Gregory J Cooney
- Diabetes and Metabolism Division, The Garvan Institute of Medical Research, Sydney, NSW, Australia; The University of Sydney, School of Medical Science, Charles Perkins Centre D17, Sydney, NSW, Australia.
| |
Collapse
|
64
|
Dalbram E, Basse AL, Zierath JR, Treebak JT. Voluntary wheel running in the late dark phase ameliorates diet-induced obesity in mice without altering insulin action. J Appl Physiol (1985) 2019; 126:993-1005. [DOI: 10.1152/japplphysiol.00737.2018] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Metabolic dysfunction and Type 2 diabetes are associated with perturbed circadian rhythms. However, exercise appears to ameliorate circadian disturbances, as it can phase-shift or reset the internal clock system. Evidence is emerging that exercise at a distinct time of day can correct misalignments of the circadian clock and influence energy metabolism. This suggests that timing of exercise training can be important for the prevention and management of metabolic dysfunction. In this study, obese, high-fat diet-fed mice were subjected to voluntary wheel running (VWR) at two different periods of the day to determine the effects of time-of-day-restricted VWR on basal and insulin-stimulated glucose disposal. VWR in the late dark phase reduced body weight gain compared with VWR in the beginning of the dark phase. Conversely, time-of-day-restricted VWR did not influence insulin action and glucose disposal, since skeletal muscle and adipose tissue glucose uptake and insulin signaling remained unaffected. Protein abundance of the core clock proteins, brain-muscle arnt-like 1 (BMAL1), and circadian locomotor output control kaput (CLOCK), were increased in skeletal muscle after VWR, independent of whether mice had access to running wheels in the early or late dark phase. Collectively, we provide evidence that VWR in the late dark phase ameliorates diet-induced obesity without altering insulin action or glucose homeostasis. NEW & NOTEWORTHY Exercise appears to ameliorate circadian disturbances as it can entrain the internal clock system. We provide evidence that voluntary wheel running increases core clock protein abundance and influences diet-induced obesity in mice in a time-of-day-dependent manner. However, the effect of time-of-day-restricted voluntary wheel running on body weight gain is not associated with enhanced basal- and insulin-stimulated glucose disposal, suggesting that time-of-day-restricted voluntary wheel running affects energy homeostasis rather than glucose homeostasis.
Collapse
Affiliation(s)
- Emilie Dalbram
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Astrid L. Basse
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Juleen R. Zierath
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
- Integrative Physiology, Department of Molecular Medicine and Surgery and Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Jonas T. Treebak
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
65
|
Kang BB, Chiang BH. EGCG regulation of non-insulin-responsive endosomal compartments in insulin-resistant skeletal muscle. FOOD BIOSCI 2019. [DOI: 10.1016/j.fbio.2018.12.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
66
|
Bakhtiyari S, Zaherara M, Haghani K, Khatami M, Rashidinejad A. The Phosphorylation of IRS1 S307 and Akt S473 Molecules in Insulin-Resistant C2C12 Cells Induced with Palmitate Is Influenced by Epigallocatechin Gallate from Green Tea. Lipids 2019; 54:141-148. [PMID: 30891789 DOI: 10.1002/lipd.12133] [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] [Received: 10/06/2018] [Revised: 01/03/2019] [Accepted: 01/22/2019] [Indexed: 12/25/2022]
Abstract
In the current investigation, the effect of epigallocatechin gallate (EGCG) on the phosphorylation of IRS1S307 and AktS473 molecules in insulin-resistant C2C12 muscle cells induced with palmitate was studied and compared with the effect of the antidiabetic drug, rosiglitazone. C2C12 myoblasts were cultured in Dulbecco's modified Eagle's medium and differentiated into myotubes using horse serum and the creatine kinase test was used to confirm their differentiation. The treatment of C2C12 myotubes was carried out with palmitate, where albumin was used as the conjugator. The Western blot technique was used to check the useful phosphorylation of IRS1S307 and AktS473 in C2C12 myotubes, in the presence or absence of palmitate. There was a significant (p < 0.00) and linear increase in the activity of creatine kinase over time (0 to 96 h after differentiation) with everyday myoblast formation. While neither EGCG nor rosiglitazone showed a significant (p > 0.05) effect on palmitate content during 96 h of incubation of IRS1S307 , EGCG alone or combined with rosiglitazone increased the phosphorylation of AktS473 , leading to the increase of glucose uptake into C2C12 cells. Thus, it can be concluded that EGCG alone or in combination with rosiglitazone may show some therapeutic effects for the prevention or treatment of Type 2 diabetes owing to its substantial effect on increasing the phosphorylation of AktS473 and the subsequent glucose uptake into the cells.
Collapse
Affiliation(s)
- Salar Bakhtiyari
- Department of Clinical Biochemistry, Ilam University of Medical Sciences, Pajouhesh Blv., Ilam 6939177143, Iran
| | - Motahareh Zaherara
- School of Medicine, Bam University of Medical Sciences, Khalije Fars, Bam 76617136699, Iran
| | - Karimeh Haghani
- Department of Clinical Biochemistry, Ilam University of Medical Sciences, Pajouhesh Blv., Ilam 6939177143, Iran
| | - Mehrdad Khatami
- Student Research Committee, School of Medicine, Bam University of Medical Sciences, Khalije Fars, Bam 76617136699, Iran.,NanoBioElectrochemistry Research Centre, Bam University of Medical Sciences, Bam, Iran
| | - Ali Rashidinejad
- Riddet Institute Centre of Research Excellence, Massey University, Tennent Drive, Palmerston North 4442, New Zealand
| |
Collapse
|
67
|
Liraglutide Ameliorates Hyperhomocysteinemia-Induced Alzheimer-Like Pathology and Memory Deficits in Rats via Multi-molecular Targeting. Neurosci Bull 2019; 35:724-734. [PMID: 30632006 DOI: 10.1007/s12264-018-00336-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Accepted: 09/17/2018] [Indexed: 01/06/2023] Open
Abstract
Hyperhomocysteinemia (Hhcy) is an independent risk factor for Alzheimer's disease (AD), and insulin-resistance is commonly seen in patients with Hhcy. Liraglutide (Lir), a glucagon-like peptide that increases the secretion and sensitivity of insulin, has a neurotrophic or neuroprotective effect. However, it is not known whether Lir ameliorates the AD-like pathology and memory deficit induced by Hhcy. By vena caudalis injection of homocysteine to produce the Hhcy model in rats, we found here that simultaneous administration of Lir for 2 weeks ameliorated the Hhcy-induced memory deficit, along with increased density of dendritic spines and up-regulation of synaptic proteins. Lir also attenuated the Hhcy-induced tau hyperphosphorylation and Aβ overproduction, and the molecular mechanisms involved the restoration of protein phosphatase-2A activity and inhibition of β- and γ-secretases. Phosphorylated insulin receptor substrate-1 also decreased after treatment with Lir. Our data reveal that Lir improves the Hhcy-induced AD-like spatial memory deficit and the mechanisms involve the modulation of insulin-resistance and the pathways generating abnormal tau and Aβ.
Collapse
|
68
|
Brandon AE, Liao BM, Diakanastasis B, Parker BL, Raddatz K, McManus SA, O'Reilly L, Kimber E, van der Kraan AG, Hancock D, Henstridge DC, Meikle PJ, Cooney GJ, James DE, Reibe S, Febbraio MA, Biden TJ, Schmitz-Peiffer C. Protein Kinase C Epsilon Deletion in Adipose Tissue, but Not in Liver, Improves Glucose Tolerance. Cell Metab 2019; 29:183-191.e7. [PMID: 30318338 DOI: 10.1016/j.cmet.2018.09.013] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 07/16/2018] [Accepted: 09/12/2018] [Indexed: 02/02/2023]
Abstract
Protein kinase C epsilon (PKCɛ) activation in the liver is proposed to inhibit insulin action through phosphorylation of the insulin receptor. Here, however, we demonstrated that global, but not liver-specific, deletion of PKCɛ in mice protected against diet-induced glucose intolerance and insulin resistance. Furthermore, PKCɛ-dependent alterations in insulin receptor phosphorylation were not detected. Adipose-tissue-specific knockout mice did exhibit improved glucose tolerance, but phosphoproteomics revealed no PKCɛ-dependent effect on the activation of insulin signaling pathways. Altered phosphorylation of adipocyte proteins associated with cell junctions and endosomes was associated with changes in hepatic expression of several genes linked to glucose homeostasis and lipid metabolism. The primary effect of PKCɛ on glucose homeostasis is, therefore, not exerted directly in the liver as currently posited, and PKCɛ activation in this tissue should be interpreted with caution. However, PKCɛ activity in adipose tissue modulates glucose tolerance and is involved in crosstalk with the liver.
Collapse
Affiliation(s)
- Amanda E Brandon
- Diabetes and Metabolism Division, Garvan Institute of Medical Research, Sydney, NSW 2010, Australia; School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW 2006, Australia
| | - Bing M Liao
- Diabetes and Metabolism Division, Garvan Institute of Medical Research, Sydney, NSW 2010, Australia
| | - Barbara Diakanastasis
- Diabetes and Metabolism Division, Garvan Institute of Medical Research, Sydney, NSW 2010, Australia
| | - Benjamin L Parker
- School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW 2006, Australia
| | - Katy Raddatz
- Diabetes and Metabolism Division, Garvan Institute of Medical Research, Sydney, NSW 2010, Australia
| | - Sophie A McManus
- Diabetes and Metabolism Division, Garvan Institute of Medical Research, Sydney, NSW 2010, Australia
| | - Liam O'Reilly
- Diabetes and Metabolism Division, Garvan Institute of Medical Research, Sydney, NSW 2010, Australia
| | - Erica Kimber
- Diabetes and Metabolism Division, Garvan Institute of Medical Research, Sydney, NSW 2010, Australia
| | | | - Dale Hancock
- School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW 2006, Australia
| | | | - Peter J Meikle
- Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia
| | - Gregory J Cooney
- Diabetes and Metabolism Division, Garvan Institute of Medical Research, Sydney, NSW 2010, Australia; School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW 2006, Australia
| | - David E James
- School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW 2006, Australia
| | - Saskia Reibe
- Diabetes and Metabolism Division, Garvan Institute of Medical Research, Sydney, NSW 2010, Australia
| | - Mark A Febbraio
- Diabetes and Metabolism Division, Garvan Institute of Medical Research, Sydney, NSW 2010, Australia; St Vincent's Clinical School, University of New South Wales, Sydney, NSW 2010, Australia
| | - Trevor J Biden
- Diabetes and Metabolism Division, Garvan Institute of Medical Research, Sydney, NSW 2010, Australia; St Vincent's Clinical School, University of New South Wales, Sydney, NSW 2010, Australia
| | - Carsten Schmitz-Peiffer
- Diabetes and Metabolism Division, Garvan Institute of Medical Research, Sydney, NSW 2010, Australia; St Vincent's Clinical School, University of New South Wales, Sydney, NSW 2010, Australia.
| |
Collapse
|
69
|
Leptin Signaling in the Control of Metabolism and Appetite: Lessons from Animal Models. J Mol Neurosci 2018; 66:390-402. [PMID: 30284225 DOI: 10.1007/s12031-018-1185-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Accepted: 09/24/2018] [Indexed: 12/15/2022]
|
70
|
Fazakerley DJ, Krycer JR, Kearney AL, Hocking SL, James DE. Muscle and adipose tissue insulin resistance: malady without mechanism? J Lipid Res 2018; 60:1720-1732. [PMID: 30054342 DOI: 10.1194/jlr.r087510] [Citation(s) in RCA: 105] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Revised: 07/25/2018] [Indexed: 12/14/2022] Open
Abstract
Insulin resistance is a major risk factor for numerous diseases, including type 2 diabetes and cardiovascular disease. These disorders have dramatically increased in incidence with modern life, suggesting that excess nutrients and obesity are major causes of "common" insulin resistance. Despite considerable effort, the mechanisms that contribute to common insulin resistance are not resolved. There is universal agreement that extracellular perturbations, such as nutrient excess, hyperinsulinemia, glucocorticoids, or inflammation, trigger intracellular stress in key metabolic target tissues, such as muscle and adipose tissue, and this impairs the ability of insulin to initiate its normal metabolic actions in these cells. Here, we present evidence that the impairment in insulin action is independent of proximal elements of the insulin signaling pathway and is likely specific to the glucoregulatory branch of insulin signaling. We propose that many intracellular stress pathways act in concert to increase mitochondrial reactive oxygen species to trigger insulin resistance. We speculate that this may be a physiological pathway to conserve glucose during specific states, such as fasting, and that, in the presence of chronic nutrient excess, this pathway ultimately leads to disease. This review highlights key points in this pathway that require further research effort.
Collapse
Affiliation(s)
- Daniel J Fazakerley
- School of Life and Environmental Sciences, Central Clinical School, University of Sydney, Camperdown, New South Wales, Australia
| | - James R Krycer
- School of Life and Environmental Sciences, Central Clinical School, University of Sydney, Camperdown, New South Wales, Australia
| | - Alison L Kearney
- School of Life and Environmental Sciences, Central Clinical School, University of Sydney, Camperdown, New South Wales, Australia
| | - Samantha L Hocking
- Faculty of Medicine and Health, University of Sydney, Camperdown, New South Wales, Australia
| | - David E James
- School of Life and Environmental Sciences, Central Clinical School, University of Sydney, Camperdown, New South Wales, Australia .,Faculty of Medicine and Health, Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia
| |
Collapse
|
71
|
Regulation of Metabolic Disease-Associated Inflammation by Nutrient Sensors. Mediators Inflamm 2018; 2018:8261432. [PMID: 30116154 PMCID: PMC6079375 DOI: 10.1155/2018/8261432] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 05/21/2018] [Accepted: 06/14/2018] [Indexed: 12/15/2022] Open
Abstract
Visceral obesity is frequently associated with the development of type 2 diabetes (T2D), a highly prevalent chronic disease that features insulin resistance and pancreatic β-cell dysfunction as important hallmarks. Recent evidence indicates that the chronic, low-grade inflammation commonly associated with visceral obesity plays a major role connecting the excessive visceral fat deposition with the development of insulin resistance and pancreatic β-cell dysfunction. Herein, we review the mechanisms by which nutrients modulate obesity-associated inflammation.
Collapse
|
72
|
Tokarz VL, MacDonald PE, Klip A. The cell biology of systemic insulin function. J Cell Biol 2018; 217:2273-2289. [PMID: 29622564 PMCID: PMC6028526 DOI: 10.1083/jcb.201802095] [Citation(s) in RCA: 276] [Impact Index Per Article: 39.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Revised: 03/21/2018] [Accepted: 03/23/2018] [Indexed: 12/12/2022] Open
Abstract
Insulin is the paramount anabolic hormone, promoting carbon energy deposition in the body. Its synthesis, quality control, delivery, and action are exquisitely regulated by highly orchestrated intracellular mechanisms in different organs or "stations" of its bodily journey. In this Beyond the Cell review, we focus on these five stages of the journey of insulin through the body and the captivating cell biology that underlies the interaction of insulin with each organ. We first analyze insulin's biosynthesis in and export from the β-cells of the pancreas. Next, we focus on its first pass and partial clearance in the liver with its temporality and periodicity linked to secretion. Continuing the journey, we briefly describe insulin's action on the blood vasculature and its still-debated mechanisms of exit from the capillary beds. Once in the parenchymal interstitium of muscle and adipose tissue, insulin promotes glucose uptake into myofibers and adipocytes, and we elaborate on the intricate signaling and vesicle traffic mechanisms that underlie this fundamental function. Finally, we touch upon the renal degradation of insulin to end its action. Cellular discernment of insulin's availability and action should prove critical to understanding its pivotal physiological functions and how their failure leads to diabetes.
Collapse
Affiliation(s)
- Victoria L Tokarz
- Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Patrick E MacDonald
- Department of Pharmacology, University of Alberta, Edmonton, Alberta, Canada
| | - Amira Klip
- Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
73
|
Bandet CL, Mahfouz R, Véret J, Sotiropoulos A, Poirier M, Giussani P, Campana M, Philippe E, Blachnio-Zabielska A, Ballaire R, Le Liepvre X, Bourron O, Berkeš D, Górski J, Ferré P, Le Stunff H, Foufelle F, Hajduch E. Ceramide Transporter CERT Is Involved in Muscle Insulin Signaling Defects Under Lipotoxic Conditions. Diabetes 2018; 67:1258-1271. [PMID: 29759974 DOI: 10.2337/db17-0901] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Accepted: 04/29/2018] [Indexed: 11/13/2022]
Abstract
One main mechanism of insulin resistance (IR), a key feature of type 2 diabetes, is the accumulation of saturated fatty acids (FAs) in the muscles of obese patients with type 2 diabetes. Understanding the mechanism that underlies lipid-induced IR is an important challenge. Saturated FAs are metabolized into lipid derivatives called ceramides, and their accumulation plays a central role in the development of muscle IR. Ceramides are produced in the endoplasmic reticulum (ER) and transported to the Golgi apparatus through a transporter called CERT, where they are converted into various sphingolipid species. We show that CERT protein expression is reduced in all IR models studied because of a caspase-dependent cleavage. Inhibiting CERT activity in vitro potentiates the deleterious action of lipotoxicity on insulin signaling, whereas overexpression of CERT in vitro or in vivo decreases muscle ceramide content and improves insulin signaling. In addition, inhibition of caspase activity prevents ceramide-induced insulin signaling defects in C2C12 muscle cells. Altogether, these results demonstrate the importance of physiological ER-to-Golgi ceramide traffic to preserve muscle cell insulin signaling and identify CERT as a major actor in this process.
Collapse
Affiliation(s)
- Cécile L Bandet
- INSERM UMRS 1138, Sorbonne Université, Sorbonne Paris Cité, Université Paris Descartes, Université Paris Diderot, Centre de Recherche des Cordeliers, Paris, France
- Institut Hospitalo-Universitaire ICAN, Paris, France
| | - Rana Mahfouz
- INSERM UMRS 1138, Sorbonne Université, Sorbonne Paris Cité, Université Paris Descartes, Université Paris Diderot, Centre de Recherche des Cordeliers, Paris, France
- Institut Hospitalo-Universitaire ICAN, Paris, France
| | - Julien Véret
- Université Paris-Diderot, Unité de biologie fonctionnelle et adaptative, CNRS UMR 8251, Paris, France
| | | | - Maxime Poirier
- INSERM UMRS 1138, Sorbonne Université, Sorbonne Paris Cité, Université Paris Descartes, Université Paris Diderot, Centre de Recherche des Cordeliers, Paris, France
- Institut Hospitalo-Universitaire ICAN, Paris, France
| | - Paola Giussani
- Department of Medical Biotechnology and Translational Medicine, Università di Milano, LITA Segrate, Milan, Italy
| | - Mélanie Campana
- Université Paris-Diderot, Unité de biologie fonctionnelle et adaptative, CNRS UMR 8251, Paris, France
| | - Erwann Philippe
- Université Paris-Diderot, Unité de biologie fonctionnelle et adaptative, CNRS UMR 8251, Paris, France
| | - Agnieszka Blachnio-Zabielska
- Departments of Physiology and Hygiene, Epidemiology and Metabolic Disorders, Medical University of Bialystok, Bialystok, Poland
| | - Raphaëlle Ballaire
- INSERM UMRS 1138, Sorbonne Université, Sorbonne Paris Cité, Université Paris Descartes, Université Paris Diderot, Centre de Recherche des Cordeliers, Paris, France
- Institut Hospitalo-Universitaire ICAN, Paris, France
| | - Xavier Le Liepvre
- INSERM UMRS 1138, Sorbonne Université, Sorbonne Paris Cité, Université Paris Descartes, Université Paris Diderot, Centre de Recherche des Cordeliers, Paris, France
- Institut Hospitalo-Universitaire ICAN, Paris, France
| | - Olivier Bourron
- INSERM UMRS 1138, Sorbonne Université, Sorbonne Paris Cité, Université Paris Descartes, Université Paris Diderot, Centre de Recherche des Cordeliers, Paris, France
- Institut Hospitalo-Universitaire ICAN, Paris, France
- Assistance Publique-Hôpitaux de Paris, Département de Diabétologie et Maladies métaboliques, Hôpital Pitié-Salpêtrière, Paris, France
| | - Dušan Berkeš
- Department of Organic Chemistry, Slovak University of Technology, Bratislava, Slovakia
| | - Jan Górski
- Departments of Physiology and Hygiene, Epidemiology and Metabolic Disorders, Medical University of Bialystok, Bialystok, Poland
| | - Pascal Ferré
- INSERM UMRS 1138, Sorbonne Université, Sorbonne Paris Cité, Université Paris Descartes, Université Paris Diderot, Centre de Recherche des Cordeliers, Paris, France
- Institut Hospitalo-Universitaire ICAN, Paris, France
| | - Hervé Le Stunff
- Université Paris-Diderot, Unité de biologie fonctionnelle et adaptative, CNRS UMR 8251, Paris, France
- UMR 9197 Institut des Neurosciences Paris Saclay (Neuro-PSI), Université Paris-Saclay, Saclay, France
| | - Fabienne Foufelle
- INSERM UMRS 1138, Sorbonne Université, Sorbonne Paris Cité, Université Paris Descartes, Université Paris Diderot, Centre de Recherche des Cordeliers, Paris, France
- Institut Hospitalo-Universitaire ICAN, Paris, France
| | - Eric Hajduch
- INSERM UMRS 1138, Sorbonne Université, Sorbonne Paris Cité, Université Paris Descartes, Université Paris Diderot, Centre de Recherche des Cordeliers, Paris, France
- Institut Hospitalo-Universitaire ICAN, Paris, France
| |
Collapse
|
74
|
Yoneyama Y, Inamitsu T, Chida K, Iemura SI, Natsume T, Maeda T, Hakuno F, Takahashi SI. Serine Phosphorylation by mTORC1 Promotes IRS-1 Degradation through SCFβ-TRCP E3 Ubiquitin Ligase. iScience 2018; 5:1-18. [PMID: 30240640 PMCID: PMC6123863 DOI: 10.1016/j.isci.2018.06.006] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Revised: 05/03/2018] [Accepted: 06/13/2018] [Indexed: 12/21/2022] Open
Abstract
The insulin receptor substrate IRS-1 is a key substrate of insulin and insulin-like growth factor (IGF) receptor tyrosine kinases that mediates their metabolic and growth-promoting actions. Proteasomal degradation of IRS-1 is induced following activation of the downstream kinase mTOR complex 1 (mTORC1) to constitute a negative feedback loop. However, the underlying mechanism remains poorly understood. Here we report that Ser 422 of IRS-1 is phosphorylated by mTORC1 and required for IRS-1 degradation induced by prolonged IGF stimulation. Phosphorylation of Ser 422 then recruits the SCFβ-TRCP E3 ligase complex, which catalyzes IRS-1 ubiquitination. Phosphorylation-dependent IRS-1 degradation contributes to impaired growth and survival responses to IGF in cells lacking TSC2, a negative regulator of mTORC1. Inhibition of IRS-1 degradation promotes sustained Akt activation in IGF-stimulated cells. Our work clarifies the nature of the IRS-1-mTORC1 feedback loop and elucidates its role in temporal regulation of IGF signaling. Ser 422 of IRS-1 is identified as a phosphorylation site by mTORC1 Phosphorylation of Ser 422 induces the binding of SCF β-TRCP E3 ligase to IRS-1 Ser 422 phosphorylation triggers the SCF β-TRCP-mediated degradation of IRS-1 The IRS-1-mTORC1 negative feedback loop determines the duration of IGF signaling
Collapse
Affiliation(s)
- Yosuke Yoneyama
- Department of Animal Resource Sciences and Applied Biological Chemistry, Graduate School of Agriculture and Life Sciences, The University of Tokyo, Bunkyo-ku, Tokyo 113-8657, Japan
| | - Tomomi Inamitsu
- Department of Animal Resource Sciences and Applied Biological Chemistry, Graduate School of Agriculture and Life Sciences, The University of Tokyo, Bunkyo-ku, Tokyo 113-8657, Japan
| | - Kazuhiro Chida
- Department of Animal Resource Sciences and Applied Biological Chemistry, Graduate School of Agriculture and Life Sciences, The University of Tokyo, Bunkyo-ku, Tokyo 113-8657, Japan
| | - Shun-Ichiro Iemura
- Translational Research Center, Fukushima Medical University, Fukushima-city, Fukushima 960-8031, Japan
| | - Tohru Natsume
- Molecular Profiling Research Center for Drug Discovery (molprof), National Institute of Advanced Industrial Science and Technology (AIST), Koto-ku, Tokyo 135-0064, Japan
| | - Tatsuya Maeda
- Institute for Quantitative Biosciences, The University of Tokyo, Bunkyo-ku, Tokyo 113-0032, Japan; Department of Integrated Human Sciences, Faculty of Medicine, Hamamatsu University School of Medicine, Hamamatsu-city, Shizuoka 431-3192, Japan
| | - Fumihiko Hakuno
- Department of Animal Resource Sciences and Applied Biological Chemistry, Graduate School of Agriculture and Life Sciences, The University of Tokyo, Bunkyo-ku, Tokyo 113-8657, Japan
| | - Shin-Ichiro Takahashi
- Department of Animal Resource Sciences and Applied Biological Chemistry, Graduate School of Agriculture and Life Sciences, The University of Tokyo, Bunkyo-ku, Tokyo 113-8657, Japan.
| |
Collapse
|
75
|
Ambery AG, Tackett L, Penque BA, Brozinick JT, Elmendorf JS. Exercise training prevents skeletal muscle plasma membrane cholesterol accumulation, cortical actin filament loss, and insulin resistance in C57BL/6J mice fed a western-style high-fat diet. Physiol Rep 2018; 5:5/16/e13363. [PMID: 28811359 PMCID: PMC5582260 DOI: 10.14814/phy2.13363] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Revised: 06/20/2017] [Accepted: 06/27/2017] [Indexed: 12/21/2022] Open
Abstract
Insulin action and glucose disposal are enhanced by exercise, yet the mechanisms involved remain imperfectly understood. While the causes of skeletal muscle insulin resistance also remain poorly understood, new evidence suggest excess plasma membrane (PM) cholesterol may contribute by damaging the cortical filamentous actin (F-actin) structure essential for GLUT4 glucose transporter redistribution to the PM upon insulin stimulation. Here, we investigated whether PM cholesterol toxicity was mitigated by exercise. Male C57BL/6J mice were placed on low-fat (LF, 10% kCal) or high-fat (HF, 45% kCal) diets for a total of 8 weeks. During the last 3 weeks of this LF/HF diet intervention, all mice were familiarized with a treadmill for 1 week and then either sham-exercised (0 m/min, 10% grade, 50 min) or exercised (13.5 m/min, 10% grade, 50 min) daily for 2 weeks. HF-feeding induced a significant gain in body mass by 3 weeks. Sham or chronic exercise did not affect food consumption, water intake, or body mass gain. Prior to sham and chronic exercise, "pre-intervention" glucose tolerance tests were performed on all animals and demonstrated that HF-fed mice were glucose intolerant. While sham exercise did not affect glucose tolerance in the LF or HF mice, exercised mice showed an improvement in glucose tolerance. Muscle from sham-exercised HF-fed mice showed a significant increase in PM cholesterol, loss of cortical F-actin, and decrease in insulin-stimulated glucose transport compared to sham-exercised LF-fed mice. These HF-fed skeletal muscle membrane/cytoskeletal abnormalities and insulin resistance were improved in exercised mice. These data reveal a new therapeutic aspect of exercise being regulation of skeletal muscle PM cholesterol homeostasis. Further studies on this mechanism of insulin resistance and the benefits of exercise on its prevention are needed.
Collapse
Affiliation(s)
- Ashley G Ambery
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, Indiana.,Center for Diabetes Metabolic Disease Indiana University School of Medicine, Indianapolis, Indiana
| | - Lixuan Tackett
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, Indiana.,Center for Diabetes Metabolic Disease Indiana University School of Medicine, Indianapolis, Indiana
| | - Brent A Penque
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, Indiana.,Center for Diabetes Metabolic Disease Indiana University School of Medicine, Indianapolis, Indiana
| | - Joseph T Brozinick
- Department Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana.,Eli Lilly and Company, Indianapolis, Indiana
| | - Jeffrey S Elmendorf
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, Indiana .,Center for Diabetes Metabolic Disease Indiana University School of Medicine, Indianapolis, Indiana.,Department Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana
| |
Collapse
|
76
|
Fazakerley DJ, Minard AY, Krycer JR, Thomas KC, Stöckli J, Harney DJ, Burchfield JG, Maghzal GJ, Caldwell ST, Hartley RC, Stocker R, Murphy MP, James DE. Mitochondrial oxidative stress causes insulin resistance without disrupting oxidative phosphorylation. J Biol Chem 2018; 293:7315-7328. [PMID: 29599292 PMCID: PMC5950018 DOI: 10.1074/jbc.ra117.001254] [Citation(s) in RCA: 96] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Revised: 03/19/2018] [Indexed: 01/02/2023] Open
Abstract
Mitochondrial oxidative stress, mitochondrial dysfunction, or both have been implicated in insulin resistance. However, disentangling the individual roles of these processes in insulin resistance has been difficult because they often occur in tandem, and tools that selectively increase oxidant production without impairing mitochondrial respiration have been lacking. Using the dimer/monomer status of peroxiredoxin isoforms as an indicator of compartmental hydrogen peroxide burden, we provide evidence that oxidative stress is localized to mitochondria in insulin-resistant 3T3-L1 adipocytes and adipose tissue from mice. To dissociate oxidative stress from impaired oxidative phosphorylation and study whether mitochondrial oxidative stress per se can cause insulin resistance, we used mitochondria-targeted paraquat (MitoPQ) to generate superoxide within mitochondria without directly disrupting the respiratory chain. At ≤10 μm, MitoPQ specifically increased mitochondrial superoxide and hydrogen peroxide without altering mitochondrial respiration in intact cells. Under these conditions, MitoPQ impaired insulin-stimulated glucose uptake and glucose transporter 4 (GLUT4) translocation to the plasma membrane in both adipocytes and myotubes. MitoPQ recapitulated many features of insulin resistance found in other experimental models, including increased oxidants in mitochondria but not cytosol; a more profound effect on glucose transport than on other insulin-regulated processes, such as protein synthesis and lipolysis; an absence of overt defects in insulin signaling; and defective insulin- but not AMP-activated protein kinase (AMPK)-regulated GLUT4 translocation. We conclude that elevated mitochondrial oxidants rapidly impair insulin-regulated GLUT4 translocation and significantly contribute to insulin resistance and that MitoPQ is an ideal tool for studying the link between mitochondrial oxidative stress and regulated GLUT4 trafficking.
Collapse
Affiliation(s)
- Daniel J Fazakerley
- Charles Perkins Centre, School of Life and Environmental Sciences, Camperdown, New South Wales 2006, Australia
| | - Annabel Y Minard
- Charles Perkins Centre, School of Life and Environmental Sciences, Camperdown, New South Wales 2006, Australia
| | - James R Krycer
- Charles Perkins Centre, School of Life and Environmental Sciences, Camperdown, New South Wales 2006, Australia
| | - Kristen C Thomas
- Charles Perkins Centre, School of Life and Environmental Sciences, Camperdown, New South Wales 2006, Australia
| | - Jacqueline Stöckli
- Charles Perkins Centre, School of Life and Environmental Sciences, Camperdown, New South Wales 2006, Australia
| | - Dylan J Harney
- Charles Perkins Centre, School of Life and Environmental Sciences, Camperdown, New South Wales 2006, Australia
| | - James G Burchfield
- Charles Perkins Centre, School of Life and Environmental Sciences, Camperdown, New South Wales 2006, Australia
| | - Ghassan J Maghzal
- Vascular Biology Division, Victor Chang Cardiac Research Institute, Darlinghurst, New South Wales 2010, Australia; St. Vincent's Clinical School, University of New South Wales, Sydney, New South Wales 2052, Australia
| | - Stuart T Caldwell
- School of Chemistry, University of Glasgow, Glasgow G12 8QQ, United Kingdom
| | - Richard C Hartley
- School of Chemistry, University of Glasgow, Glasgow G12 8QQ, United Kingdom
| | - Roland Stocker
- Vascular Biology Division, Victor Chang Cardiac Research Institute, Darlinghurst, New South Wales 2010, Australia; St. Vincent's Clinical School, University of New South Wales, Sydney, New South Wales 2052, Australia
| | - Michael P Murphy
- MRC Mitochondrial Biology Unit, Hills Road, University of Cambridge, Cambridge CB2 0XY, United Kingdom
| | - David E James
- Charles Perkins Centre, School of Life and Environmental Sciences, Camperdown, New South Wales 2006, Australia; Charles Perkins Centre, Sydney Medical School, University of Sydney, Camperdown, New South Wales 2006, Australia.
| |
Collapse
|
77
|
Small L, Brandon AE, Turner N, Cooney GJ. Modeling insulin resistance in rodents by alterations in diet: what have high-fat and high-calorie diets revealed? Am J Physiol Endocrinol Metab 2018; 314:E251-E265. [PMID: 29118016 DOI: 10.1152/ajpendo.00337.2017] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
For over half a century, researchers have been feeding different diets to rodents to examine the effects of macronutrients on whole body and tissue insulin action. During this period, the number of different diets and the source of macronutrients employed have grown dramatically. Because of the large heterogeneity in both the source and percentage of different macronutrients used for studies, it is not surprising that different high-calorie diets do not produce the same changes in insulin action. Despite this, diverse high-calorie diets continue to be employed in an attempt to generate a "generic" insulin resistance. The high-fat diet in particular varies greatly between studies with regard to the source, complexity, and ratio of dietary fat, carbohydrate, and protein. This review examines the range of rodent dietary models and methods for assessing insulin action. In almost all studies reviewed, rodents fed diets that had more than 45% of dietary energy as fat or simple carbohydrates had reduced whole body insulin action compared with chow. However, different high-calorie diets produced significantly different effects in liver, muscle, and whole body insulin action when insulin action was measured by the hyperinsulinemic-euglycemic clamp method. Rodent dietary models remain an important tool for exploring potential mechanisms of insulin resistance, but more attention needs to be given to the total macronutrient content and composition when interpreting dietary effects on insulin action.
Collapse
Affiliation(s)
- Lewin Small
- Diabetes and Metabolism Division, Garvan Institute , Sydney, New South Wales , Australia
| | - Amanda E Brandon
- Diabetes and Metabolism Division, Garvan Institute , Sydney, New South Wales , Australia
- Sydney Medical School, Charles Perkins Centre, The University of Sydney , New South Wales , Australia
| | - Nigel Turner
- Department of Pharmacology, School of Medical Science, University of New South Wales , Sydney, New South Wales , Australia
| | - Gregory J Cooney
- Diabetes and Metabolism Division, Garvan Institute , Sydney, New South Wales , Australia
- Sydney Medical School, Charles Perkins Centre, The University of Sydney , New South Wales , Australia
| |
Collapse
|
78
|
Fazakerley DJ, Chaudhuri R, Yang P, Maghzal GJ, Thomas KC, Krycer JR, Humphrey SJ, Parker BL, Fisher-Wellman KH, Meoli CC, Hoffman NJ, Diskin C, Burchfield JG, Cowley MJ, Kaplan W, Modrusan Z, Kolumam G, Yang JY, Chen DL, Samocha-Bonet D, Greenfield JR, Hoehn KL, Stocker R, James DE. Mitochondrial CoQ deficiency is a common driver of mitochondrial oxidants and insulin resistance. eLife 2018; 7:32111. [PMID: 29402381 PMCID: PMC5800848 DOI: 10.7554/elife.32111] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Accepted: 01/02/2018] [Indexed: 12/11/2022] Open
Abstract
Insulin resistance in muscle, adipocytes and liver is a gateway to a number of metabolic diseases. Here, we show a selective deficiency in mitochondrial coenzyme Q (CoQ) in insulin-resistant adipose and muscle tissue. This defect was observed in a range of in vitro insulin resistance models and adipose tissue from insulin-resistant humans and was concomitant with lower expression of mevalonate/CoQ biosynthesis pathway proteins in most models. Pharmacologic or genetic manipulations that decreased mitochondrial CoQ triggered mitochondrial oxidants and insulin resistance while CoQ supplementation in either insulin-resistant cell models or mice restored normal insulin sensitivity. Specifically, lowering of mitochondrial CoQ caused insulin resistance in adipocytes as a result of increased superoxide/hydrogen peroxide production via complex II. These data suggest that mitochondrial CoQ is a proximal driver of mitochondrial oxidants and insulin resistance, and that mechanisms that restore mitochondrial CoQ may be effective therapeutic targets for treating insulin resistance. After we eat, our blood sugar levels increase. To counteract this, the pancreas releases a hormone called insulin. Part of insulin’s effect is to promote the uptake of sugar from the blood into muscle and fat tissue for storage. Under certain conditions, such as obesity, this process can become defective, leading to a condition known as insulin resistance. This condition makes a number of human diseases more likely to develop, including type 2 diabetes. Working out how insulin resistance develops could therefore unveil new treatment strategies for these diseases. Mitochondria – structures that produce most of a cell’s energy supply – appear to play a role in the development of insulin resistance. Mitochondria convert nutrients such as fats and sugars into molecules called ATP that fuel the many processes required for life. However, ATP production can also generate potentially harmful intermediates often referred to as ‘reactive oxygen species’ or ‘oxidants’. Previous studies have suggested that an increase in the amount of oxidants produced in mitochondria can cause insulin resistance. Fazakerley et al. therefore set out to identify the reason for increased oxidants in mitochondria, and did so by analysing the levels of proteins and oxidants found in cells grown in the laboratory, and mouse and human tissue samples. This led them to find that concentrations of a molecule called coenzyme Q (CoQ), an essential component of mitochondria that helps to produce ATP, were lower in mitochondria from insulin-resistant fat and muscle tissue. Further experiments suggested a link between the lower levels of CoQ and the higher levels of oxidants in the mitochondria. Replenishing the mitochondria of the lab-grown cells and insulin-resistant mice with CoQ restored ‘normal’ oxidant levels and prevented the development of insulin resistance. Strategies that aim to increase mitochondria CoQ levels may therefore prevent or reverse insulin resistance. Although CoQ supplements are readily available, swallowing CoQ does not efficiently deliver CoQ to mitochondria in humans, so alternative treatment methods must be found. It is also of interest that statins, common drugs taken by millions of people around the world to lower cholesterol, also lower CoQ and have been reported to increase the risk of developing type 2 diabetes. Further research is therefore needed to investigate whether CoQ might provide the link between statins and type 2 diabetes.
Collapse
Affiliation(s)
- Daniel J Fazakerley
- Charles Perkins Centre, School of Life and Environmental Sciences, University of Sydney, Camperdown, Australia
| | - Rima Chaudhuri
- Charles Perkins Centre, School of Life and Environmental Sciences, University of Sydney, Camperdown, Australia
| | - Pengyi Yang
- School of Mathematics and Statistics, University of Sydney, Camperdown, Australia
| | - Ghassan J Maghzal
- Vascular Biology Division, Victor Chang Cardiac Research Institute, Darlinghurst, Australia
| | - Kristen C Thomas
- Charles Perkins Centre, School of Life and Environmental Sciences, University of Sydney, Camperdown, Australia
| | - James R Krycer
- Charles Perkins Centre, School of Life and Environmental Sciences, University of Sydney, Camperdown, Australia
| | - Sean J Humphrey
- Charles Perkins Centre, School of Life and Environmental Sciences, University of Sydney, Camperdown, Australia
| | - Benjamin L Parker
- Charles Perkins Centre, School of Life and Environmental Sciences, University of Sydney, Camperdown, Australia
| | - Kelsey H Fisher-Wellman
- Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, United States
| | - Christopher C Meoli
- Charles Perkins Centre, School of Life and Environmental Sciences, University of Sydney, Camperdown, Australia
| | - Nolan J Hoffman
- Charles Perkins Centre, School of Life and Environmental Sciences, University of Sydney, Camperdown, Australia
| | - Ciana Diskin
- Charles Perkins Centre, School of Life and Environmental Sciences, University of Sydney, Camperdown, Australia
| | - James G Burchfield
- Charles Perkins Centre, School of Life and Environmental Sciences, University of Sydney, Camperdown, Australia
| | - Mark J Cowley
- The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Darlinghurst, Australia
| | - Warren Kaplan
- Peter Wills Bioinformatics Centre, Garvan Institute of Medical Research, Darlinghurst, Australia
| | | | | | - Jean Yh Yang
- School of Mathematics and Statistics, University of Sydney, Camperdown, Australia
| | - Daniel L Chen
- Garvan Institute of Medical Research, Darlinghurst, Australia
| | | | | | - Kyle L Hoehn
- School of Biotechnology and Biomedical Sciences, University of New South Wales, Sydney, Australia
| | - Roland Stocker
- Vascular Biology Division, Victor Chang Cardiac Research Institute, Darlinghurst, Australia.,St Vincent's Clinical School, University of New South Wales, Sydney, Australia
| | - David E James
- Charles Perkins Centre, School of Life and Environmental Sciences, University of Sydney, Camperdown, Australia.,Charles Perkins Centre, Sydney Medical School, University of Sydney, Camperdown NSW, Australia
| |
Collapse
|
79
|
Chaudhuri R, Krycer JR, Fazakerley DJ, Fisher-Wellman KH, Su Z, Hoehn KL, Yang JYH, Kuncic Z, Vafaee F, James DE. The transcriptional response to oxidative stress is part of, but not sufficient for, insulin resistance in adipocytes. Sci Rep 2018; 8:1774. [PMID: 29379070 PMCID: PMC5789081 DOI: 10.1038/s41598-018-20104-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Accepted: 01/12/2018] [Indexed: 02/06/2023] Open
Abstract
Insulin resistance is a major risk factor for metabolic diseases such as Type 2 diabetes. Although the underlying mechanisms of insulin resistance remain elusive, oxidative stress is a unifying driver by which numerous extrinsic signals and cellular stresses trigger insulin resistance. Consequently, we sought to understand the cellular response to oxidative stress and its role in insulin resistance. Using cultured 3T3-L1 adipocytes, we established a model of physiologically-derived oxidative stress by inhibiting the cycling of glutathione and thioredoxin, which induced insulin resistance as measured by impaired insulin-stimulated 2-deoxyglucose uptake. Using time-resolved transcriptomics, we found > 2000 genes differentially-expressed over 24 hours, with specific metabolic and signalling pathways enriched at different times. We explored this coordination using a knowledge-based hierarchical-clustering approach to generate a temporal transcriptional cascade and identify key transcription factors responding to oxidative stress. This response shared many similarities with changes observed in distinct insulin resistance models. However, an anti-oxidant reversed insulin resistance phenotypically but not transcriptionally, implying that the transcriptional response to oxidative stress is insufficient for insulin resistance. This suggests that the primary site by which oxidative stress impairs insulin action occurs post-transcriptionally, warranting a multi-level ‘trans-omic’ approach when studying time-resolved responses to cellular perturbations.
Collapse
Affiliation(s)
- Rima Chaudhuri
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, 2006, Australia.,School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW 2006, Australia
| | - James R Krycer
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, 2006, Australia.,School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW 2006, Australia
| | - Daniel J Fazakerley
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, 2006, Australia.,School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW 2006, Australia
| | | | - Zhiduan Su
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, 2006, Australia.,School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW 2006, Australia
| | - Kyle L Hoehn
- School of Biotechnology and Biomolecular Sciences, The University of New South Wales, Sydney, NSW, 2052, Australia
| | - Jean Yee Hwa Yang
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, 2006, Australia.,School of Mathematics and Statistics, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Zdenka Kuncic
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, 2006, Australia.,School of Physics and Australian Institute for Nanoscale Science and Technology, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Fatemeh Vafaee
- School of Biotechnology and Biomolecular Sciences, The University of New South Wales, Sydney, NSW, 2052, Australia.
| | - David E James
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, 2006, Australia. .,School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW 2006, Australia. .,Sydney Medical School, The University of Sydney, Sydney, NSW, 2006, Australia.
| |
Collapse
|
80
|
Haeusler RA, McGraw TE, Accili D. Biochemical and cellular properties of insulin receptor signalling. Nat Rev Mol Cell Biol 2018; 19:31-44. [PMID: 28974775 PMCID: PMC5894887 DOI: 10.1038/nrm.2017.89] [Citation(s) in RCA: 475] [Impact Index Per Article: 67.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The mechanism of insulin action is a central theme in biology and medicine. In addition to the rather rare condition of insulin deficiency caused by autoimmune destruction of pancreatic β-cells, genetic and acquired abnormalities of insulin action underlie the far more common conditions of type 2 diabetes, obesity and insulin resistance. The latter predisposes to diseases ranging from hypertension to Alzheimer disease and cancer. Hence, understanding the biochemical and cellular properties of insulin receptor signalling is arguably a priority in biomedical research. In the past decade, major progress has led to the delineation of mechanisms of glucose transport, lipid synthesis, storage and mobilization. In addition to direct effects of insulin on signalling kinases and metabolic enzymes, the discovery of mechanisms of insulin-regulated gene transcription has led to a reassessment of the general principles of insulin action. These advances will accelerate the discovery of new treatment modalities for diabetes.
Collapse
Affiliation(s)
- Rebecca A Haeusler
- Columbia University College of Physicians and Surgeons, Department of Pathology and Cell Biology, New York, New York 10032, USA
| | - Timothy E McGraw
- Weill Cornell Medicine, Departments of Biochemistry and Cardiothoracic Surgery, New York, New York 10065, USA
| | - Domenico Accili
- Columbia University College of Physicians & Surgeons, Department of Medicine, New York, New York 10032, USA
| |
Collapse
|
81
|
Translocation and Redistribution of GLUT4 Using a Dual-Labeled Reporter Assay. Methods Mol Biol 2017. [PMID: 29218525 DOI: 10.1007/978-1-4939-7507-5_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
It is crucial to determine the regulation of GLUT4 translocation and redistribution to the plasma membrane. The HA-GLUT4-GFP dual-reporter construct has become an important tool in the assessment of GLUT4 recycling in cultured adipocytes and myocytes. Through the use of light microscopy, this reporter construct allows for visualization of GLUT4 specifically at the cell surface or GLUT4 that has recycled from the cell surface while simultaneously marking the total GLUT4 pool. Here, we discuss and outline the general application of this reporter construct and its use in evaluating GLUT4 translocation within cultured adipocytes.
Collapse
|
82
|
mTORC1 Is a Major Regulatory Node in the FGF21 Signaling Network in Adipocytes. Cell Rep 2017; 17:29-36. [PMID: 27681418 DOI: 10.1016/j.celrep.2016.08.086] [Citation(s) in RCA: 90] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Revised: 06/01/2016] [Accepted: 08/24/2016] [Indexed: 11/22/2022] Open
Abstract
FGF21 improves the metabolic profile of obese animals through its actions on adipocytes. To elucidate the signaling network responsible for mediating these effects, we quantified dynamic changes in the adipocyte phosphoproteome following acute exposure to FGF21. FGF21 regulated a network of 821 phosphosites on 542 proteins. A major FGF21-regulated signaling node was mTORC1/S6K. In contrast to insulin, FGF21 activated mTORC1 via MAPK rather than through the canonical PI3K/AKT pathway. Activation of mTORC1/S6K by FGF21 was surprising because this is thought to contribute to deleterious metabolic effects such as obesity and insulin resistance. Rather, mTORC1 mediated many of the beneficial actions of FGF21 in vitro, including UCP1 and FGF21 induction, increased adiponectin secretion, and enhanced glucose uptake without any adverse effects on insulin action. This study provides a global view of FGF21 signaling and suggests that mTORC1 may act to facilitate FGF21-mediated health benefits in vivo.
Collapse
|
83
|
Gupta A, Beg M, Kumar D, Shankar K, Varshney S, Rajan S, Srivastava A, Singh K, Sonkar S, Mahdi AA, Dikshit M, Gaikwad AN. Chronic hyper-leptinemia induces insulin signaling disruption in adipocytes: Implications of NOS2. Free Radic Biol Med 2017; 112:93-108. [PMID: 28739528 DOI: 10.1016/j.freeradbiomed.2017.07.016] [Citation(s) in RCA: 11] [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: 11/03/2016] [Revised: 07/11/2017] [Accepted: 07/20/2017] [Indexed: 01/12/2023]
Abstract
Leptin, following its discovery, has developed a formidable interest in the scientific community to delineate its contribution towards overall metabolic homeostasis. Contradictory reports have been published on leptin administration effects on whole body insulin sensitivity. Following late reports, we surveyed human serum leptin levels along with other metabolic parameters including BMI and HOMA-IR. We found a positive correlation between leptin levels and insulin resistance parameters. Considering the presence of the long form of leptin receptor on adipocytes, we explored the effects of chronic physiological hyper-leptinemic exposure on adipocyte insulin sensitivity. Chronic leptin (50ng/ml) treatment in 3T3-L1 adipocytes decreased insulin-induced phosphorylation of nodal insulin signaling proteins along with reduced glucose uptake. Metabolic flux studies indicated mitochondrial dysfunction and reduced oxygen consumption rate. Leptin treatment also increased both cellular and mitochondrial superoxide levels concomitant to increased expression of nitric oxide synthase-2 (NOS2). Further, pharmacological depletion of NOS2 reversed leptin mediated effects on insulin signaling. In-vivo implantation of leptin osmotic pumps in C57BL/6 mice also decreased insulin responsiveness. Interestingly, these effects were lacking in NOS2 knockout strain. In conclusion, our studies put forward a potential link between leptin and adipocyte insulin responsiveness in an NOS2 dependent manner.
Collapse
Affiliation(s)
- Abhishek Gupta
- Division of Pharmacology, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Muheeb Beg
- Division of Pharmacology, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Durgesh Kumar
- Division of Pharmacology, CSIR-Central Drug Research Institute, Lucknow 226031, India; Academy of Scientific and Innovative Research, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Kripa Shankar
- Division of Pharmacology, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Salil Varshney
- Division of Pharmacology, CSIR-Central Drug Research Institute, Lucknow 226031, India; Academy of Scientific and Innovative Research, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Sujith Rajan
- Division of Pharmacology, CSIR-Central Drug Research Institute, Lucknow 226031, India; Academy of Scientific and Innovative Research, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Ankita Srivastava
- Division of Pharmacology, CSIR-Central Drug Research Institute, Lucknow 226031, India; Academy of Scientific and Innovative Research, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Kalpana Singh
- Department of Biochemistry, King George's Medical University, Lucknow 226003, India
| | - Satyendra Sonkar
- Department of Internal Medicine, King George's Medical University, Lucknow 226003, India
| | - Abbas Ali Mahdi
- Department of Biochemistry, King George's Medical University, Lucknow 226003, India
| | - Madhu Dikshit
- Division of Pharmacology, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Anil Nilkanth Gaikwad
- Division of Pharmacology, CSIR-Central Drug Research Institute, Lucknow 226031, India.
| |
Collapse
|
84
|
Zhang Y, Wu YT, Zheng W, Han XX, Jiang YH, Hu PL, Tang ZX, Shi LE. The antibacterial activity and antibacterial mechanism of a polysaccharide from Cordyceps cicadae. J Funct Foods 2017. [DOI: 10.1016/j.jff.2017.09.047] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
|
85
|
Insulin action and resistance in obesity and type 2 diabetes. Nat Med 2017; 23:804-814. [PMID: 28697184 DOI: 10.1038/nm.4350] [Citation(s) in RCA: 841] [Impact Index Per Article: 105.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Accepted: 05/11/2017] [Indexed: 12/12/2022]
Abstract
Nutritional excess is a major forerunner of type 2 diabetes. It enhances the secretion of insulin, but attenuates insulin's metabolic actions in the liver, skeletal muscle and adipose tissue. However, conflicting evidence indicates a lack of knowledge of the timing of these events during the development of obesity and diabetes, pointing to a key gap in our understanding of metabolic disease. This Perspective reviews alternate viewpoints and recent results on the temporal and mechanistic connections between hyperinsulinemia, obesity and insulin resistance. Although much attention has addressed early steps in the insulin signaling cascade, insulin resistance in obesity seems to be largely elicited downstream of these steps. New findings also connect insulin resistance to extensive metabolic cross-talk between the liver, adipose tissue, pancreas and skeletal muscle. These and other advances over the past 5 years offer exciting opportunities and daunting challenges for the development of new therapeutic strategies for the treatment of type 2 diabetes.
Collapse
|
86
|
Gaur V, Connor T, Venardos K, Henstridge DC, Martin SD, Swinton C, Morrison S, Aston-Mourney K, Gehrig SM, van Ewijk R, Lynch GS, Febbraio MA, Steinberg GR, Hargreaves M, Walder KR, McGee SL. Scriptaid enhances skeletal muscle insulin action and cardiac function in obese mice. Diabetes Obes Metab 2017; 19:936-943. [PMID: 28155245 DOI: 10.1111/dom.12896] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Revised: 01/26/2017] [Accepted: 01/28/2017] [Indexed: 12/20/2022]
Abstract
AIM To determine the effect of Scriptaid, a compound that can replicate aspects of the exercise adaptive response through disruption of the class IIa histone deacetylase (HDAC) corepressor complex, on muscle insulin action in obesity. MATERIALS AND METHODS Diet-induced obese mice were administered Scriptaid (1 mg/kg) via daily intraperitoneal injection for 4 weeks. Whole-body and skeletal muscle metabolic phenotyping of mice was performed, in addition to echocardiography, to assess cardiac morphology and function. RESULTS Scriptaid treatment had no effect on body weight or composition, but did increase energy expenditure, supported by increased lipid oxidation, while food intake was also increased. Scriptaid enhanced the expression of oxidative genes and proteins, increased fatty acid oxidation and reduced triglycerides and diacylglycerides in skeletal muscle. Furthermore, ex vivo insulin-stimulated glucose uptake by skeletal muscle was enhanced. Surprisingly, heart weight was reduced in Scriptaid-treated mice and was associated with enhanced expression of genes involved in oxidative metabolism in the heart. Scriptaid also improved indices of both diastolic and systolic cardiac function. CONCLUSION These data show that pharmacological targeting of the class IIa HDAC corepressor complex with Scriptaid could be used to enhance muscle insulin action and cardiac function in obesity.
Collapse
Affiliation(s)
- Vidhi Gaur
- Metabolic Research Unit, School of Medicine, Deakin University, Geelong, Australia
| | - Timothy Connor
- Metabolic Research Unit, School of Medicine, Deakin University, Geelong, Australia
| | - Kylie Venardos
- Metabolic Research Unit, School of Medicine, Deakin University, Geelong, Australia
| | - Darren C Henstridge
- Metabolism and Inflammation Program, Baker IDI Heart and Diabetes Institute, Melbourne, Australia
| | - Sheree D Martin
- Metabolic Research Unit, School of Medicine, Deakin University, Geelong, Australia
| | - Courtney Swinton
- Metabolic Research Unit, School of Medicine, Deakin University, Geelong, Australia
| | - Shona Morrison
- Metabolic Research Unit, School of Medicine, Deakin University, Geelong, Australia
| | | | - Stefan M Gehrig
- Department of Physiology, The University of Melbourne, Parkville, Australia
| | - Roelof van Ewijk
- Department of Physiology, The University of Melbourne, Parkville, Australia
| | - Gordon S Lynch
- Department of Physiology, The University of Melbourne, Parkville, Australia
| | - Mark A Febbraio
- Metabolism and Inflammation Program, Baker IDI Heart and Diabetes Institute, Melbourne, Australia
- Division of Diabetes and Metabolism, Garvan Institute of Medical Research, Darlinghurst, Australia
| | - Gregory R Steinberg
- Division of Endocrinology and Metabolism, Department of Medicine, McMaster University, Hamilton, Canada
| | - Mark Hargreaves
- Department of Physiology, The University of Melbourne, Parkville, Australia
| | - Ken R Walder
- Metabolic Research Unit, School of Medicine, Deakin University, Geelong, Australia
| | - Sean L McGee
- Metabolic Research Unit, School of Medicine, Deakin University, Geelong, Australia
- Metabolism and Inflammation Program, Baker IDI Heart and Diabetes Institute, Melbourne, Australia
| |
Collapse
|
87
|
Norris DM, Yang P, Krycer JR, Fazakerley DJ, James DE, Burchfield JG. An improved Akt reporter reveals intra- and inter-cellular heterogeneity and oscillations in signal transduction. J Cell Sci 2017; 130:2757-2766. [PMID: 28663386 DOI: 10.1242/jcs.205369] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Accepted: 06/26/2017] [Indexed: 11/20/2022] Open
Abstract
Akt is a key node in a range of signal transduction cascades and play a critical role in diseases such as cancer and diabetes. Fluorescently-tagged Akt reporters have been used to discern Akt localisation, yet it has not been clear how well these tools recapitulate the behaviour of endogenous Akt proteins. Here, we observed that fusion of eGFP to Akt2 impaired both its insulin-stimulated plasma membrane recruitment and its phosphorylation. Endogenous-like responses were restored by replacing eGFP with TagRFP-T. The improved response magnitude and sensitivity afforded by TagRFP-T-Akt2 over eGFP-Akt2 enabled monitoring of signalling outcomes in single cells at physiological doses of insulin with subcellular resolution and revealed two previously unreported features of Akt biology. In 3T3-L1 adipocytes, stimulation with insulin resulted in recruitment of Akt2 to the plasma membrane in a polarised fashion. Additionally, we observed oscillations in plasma membrane localised Akt2 in the presence of insulin with a consistent periodicity of 2 min. Our studies highlight the importance of fluorophore choice when generating reporter constructs and shed light on new Akt signalling responses that may encode complex signalling information.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Dougall M Norris
- Charles Perkins Centre, School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW 2006, Australia
| | - Pengyi Yang
- Charles Perkins Centre, School of Mathematics and Statistics, The University of Sydney, Sydney, NSW 2006, Australia
| | - James R Krycer
- Charles Perkins Centre, School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW 2006, Australia
| | - Daniel J Fazakerley
- Charles Perkins Centre, School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW 2006, Australia
| | - David E James
- Charles Perkins Centre, School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW 2006, Australia.,Sydney Medical School, The University of Sydney, Sydney, NSW 2006, Australia
| | - James G Burchfield
- Charles Perkins Centre, School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW 2006, Australia
| |
Collapse
|
88
|
Miotto PM, Horbatuk M, Proudfoot R, Matravadia S, Bakovic M, Chabowski A, Holloway GP. α-Linolenic acid supplementation and exercise training reveal independent and additive responses on hepatic lipid accumulation in obese rats. Am J Physiol Endocrinol Metab 2017; 312:E461-E470. [PMID: 28270444 PMCID: PMC5494579 DOI: 10.1152/ajpendo.00438.2016] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Revised: 03/02/2017] [Accepted: 03/03/2017] [Indexed: 02/06/2023]
Abstract
α-Linolenic acid (ALA) supplementation or exercise training can independently prevent hepatic lipid accumulation and reduced insulin signaling; however, this may occur through different mechanisms of action. In the current study, obese Zucker rats displayed decreased phospholipid (PL) content in association with hepatic lipid abundance, and therefore, we examined whether ALA and exercise training would prevent these abnormalities differently to reveal additive effects on the liver. To achieve this aim, obese Zucker rats were fed control diet alone or supplemented with ALA and were sedentary or exercise trained for 4 wk (C-Sed, ALA-Sed, C-Ex, and ALA-Ex). ALA-Sed rats had increased microsomal-triglyceride transfer protein (MTTP), a protein required for lipoprotein assembly/secretion, as well as modestly increased PL content in the absence of improvements in mitochondrial content, lipid accumulation, or insulin sensitivity. In contrast, C-Ex rats had increased mitochondrial content and insulin sensitivity; however, this corresponded with minimal improvements in PL content and hepatic lipid accumulation. Importantly, ALA-Ex rats demonstrated additive improvements in PL content and hepatic steatosis, which corresponded with increased mitochondrial content, MTTP and apolipoprotein B100 content, greater serum triacylglyceride, and insulin sensitivity. Overall, these data demonstrate additive effects of ALA and exercise training on hepatic lipid accumulation, as exercise training preferentially increased mitochondrial content, while ALA promoted an environment conducive for lipid secretion. These data highlight the potential for combination therapy to mitigate liver disease progression.
Collapse
Affiliation(s)
- Paula M Miotto
- Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario, Canada; and
| | - Meaghan Horbatuk
- Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario, Canada; and
| | - Ross Proudfoot
- Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario, Canada; and
| | - Sarthak Matravadia
- Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario, Canada; and
| | - Marica Bakovic
- Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario, Canada; and
| | - Adrian Chabowski
- Department of Physiology, Medical University of Bialystok, Bialystok, Poland
| | - Graham P Holloway
- Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario, Canada; and
| |
Collapse
|
89
|
Irimia JM, Meyer CM, Segvich DM, Surendran S, DePaoli-Roach AA, Morral N, Roach PJ. Lack of liver glycogen causes hepatic insulin resistance and steatosis in mice. J Biol Chem 2017; 292:10455-10464. [PMID: 28483921 DOI: 10.1074/jbc.m117.786525] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Revised: 05/06/2017] [Indexed: 01/16/2023] Open
Abstract
Disruption of the Gys2 gene encoding the liver isoform of glycogen synthase generates a mouse strain (LGSKO) that almost completely lacks hepatic glycogen, has impaired glucose disposal, and is pre-disposed to entering the fasted state. This study investigated how the lack of liver glycogen increases fat accumulation and the development of liver insulin resistance. Insulin signaling in LGSKO mice was reduced in liver, but not muscle, suggesting an organ-specific defect. Phosphorylation of components of the hepatic insulin-signaling pathway, namely IRS1, Akt, and GSK3, was decreased in LGSKO mice. Moreover, insulin stimulation of their phosphorylation was significantly suppressed, both temporally and in an insulin dose response. Phosphorylation of the insulin-regulated transcription factor FoxO1 was somewhat reduced and insulin treatment did not elicit normal translocation of FoxO1 out of the nucleus. Fat overaccumulated in LGSKO livers, showing an aberrant distribution in the acinus, an increase not explained by a reduction in hepatic triglyceride export. Rather, when administered orally to fasted mice, glucose was directed toward hepatic lipogenesis as judged by the activity, protein levels, and expression of several fatty acid synthesis genes, namely, acetyl-CoA carboxylase, fatty acid synthase, SREBP1c, chREBP, glucokinase, and pyruvate kinase. Furthermore, using cultured primary hepatocytes, we found that lipogenesis was increased by 40% in LGSKO cells compared with controls. Of note, the hepatic insulin resistance was not associated with increased levels of pro-inflammatory markers. Our results suggest that loss of liver glycogen synthesis diverts glucose toward fat synthesis, correlating with impaired hepatic insulin signaling and glucose disposal.
Collapse
Affiliation(s)
- Jose M Irimia
- From the Departments of Biochemistry and Molecular Biology and
| | | | - Dyann M Segvich
- From the Departments of Biochemistry and Molecular Biology and
| | - Sneha Surendran
- Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | | | - Nuria Morral
- Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - Peter J Roach
- From the Departments of Biochemistry and Molecular Biology and
| |
Collapse
|
90
|
Di Meo S, Iossa S, Venditti P. Skeletal muscle insulin resistance: role of mitochondria and other ROS sources. J Endocrinol 2017; 233:R15-R42. [PMID: 28232636 DOI: 10.1530/joe-16-0598] [Citation(s) in RCA: 205] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Accepted: 01/31/2017] [Indexed: 12/12/2022]
Abstract
At present, obesity is one of the most important public health problems in the world because it causes several diseases and reduces life expectancy. Although it is well known that insulin resistance plays a pivotal role in the development of type 2 diabetes mellitus (the more frequent disease in obese people) the link between obesity and insulin resistance is yet a matter of debate. One of the most deleterious effects of obesity is the deposition of lipids in non-adipose tissues when the capacity of adipose tissue is overwhelmed. During the last decade, reduced mitochondrial function has been considered as an important contributor to 'toxic' lipid metabolite accumulation and consequent insulin resistance. More recent reports suggest that mitochondrial dysfunction is not an early event in the development of insulin resistance, but rather a complication of the hyperlipidemia-induced reactive oxygen species (ROS) production in skeletal muscle, which might promote mitochondrial alterations, lipid accumulation and inhibition of insulin action. Here, we review the literature dealing with the mitochondria-centered mechanisms proposed to explain the onset of obesity-linked IR in skeletal muscle. We conclude that the different pathways leading to insulin resistance may act synergistically because ROS production by mitochondria and other sources can result in mitochondrial dysfunction, which in turn can further increase ROS production leading to the establishment of a harmful positive feedback loop.
Collapse
Affiliation(s)
- Sergio Di Meo
- Department of BiologyUniversity of Naples 'Federico II', Naples, Italy
| | - Susanna Iossa
- Department of BiologyUniversity of Naples 'Federico II', Naples, Italy
| | - Paola Venditti
- Department of BiologyUniversity of Naples 'Federico II', Naples, Italy
| |
Collapse
|
91
|
Taddeo EP, Hargett SR, Lahiri S, Nelson ME, Liao JA, Li C, Slack-Davis JK, Tomsig JL, Lynch KR, Yan Z, Harris TE, Hoehn KL. Lysophosphatidic acid counteracts glucagon-induced hepatocyte glucose production via STAT3. Sci Rep 2017; 7:127. [PMID: 28273928 PMCID: PMC5428006 DOI: 10.1038/s41598-017-00210-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Accepted: 02/14/2017] [Indexed: 01/25/2023] Open
Abstract
Hepatic glucose production (HGP) is required to maintain normoglycemia during fasting. Glucagon is the primary hormone responsible for increasing HGP; however, there are many additional hormone and metabolic factors that influence glucagon sensitivity. In this study we report that the bioactive lipid lysophosphatidic acid (LPA) regulates hepatocyte glucose production by antagonizing glucagon-induced expression of the gluconeogenic enzyme phosphoenolpyruvate carboxykinase (PEPCK). Treatment of primary hepatocytes with exogenous LPA blunted glucagon-induced PEPCK expression and glucose production. Similarly, knockout mice lacking the LPA-degrading enzyme phospholipid phosphate phosphatase type 1 (PLPP1) had a 2-fold increase in endogenous LPA levels, reduced PEPCK levels during fasting, and decreased hepatic gluconeogenesis in response to a pyruvate challenge. Mechanistically, LPA antagonized glucagon-mediated inhibition of STAT3, a transcriptional repressor of PEPCK. Importantly, LPA did not blunt glucagon-stimulated glucose production or PEPCK expression in hepatocytes lacking STAT3. These data identify a novel role for PLPP1 activity and hepatocyte LPA levels in glucagon sensitivity via a mechanism involving STAT3.
Collapse
Affiliation(s)
- Evan P Taddeo
- Department of Pharmacology, School of Medicine, University of Virginia, Charlottesville, VA, 22908, USA
| | - Stefan R Hargett
- Department of Pharmacology, School of Medicine, University of Virginia, Charlottesville, VA, 22908, USA
| | - Sujoy Lahiri
- Department of Pharmacology, School of Medicine, University of Virginia, Charlottesville, VA, 22908, USA
| | - Marin E Nelson
- Department of Pharmacology, School of Medicine, University of Virginia, Charlottesville, VA, 22908, USA
| | - Jason A Liao
- Department of Pharmacology, School of Medicine, University of Virginia, Charlottesville, VA, 22908, USA
| | - Chien Li
- Department of Pharmacology, School of Medicine, University of Virginia, Charlottesville, VA, 22908, USA
| | - Jill K Slack-Davis
- Department of Microbiology, Immunology and Cancer Biology, School of Medicine, University of Virginia, Charlottesville, VA, 22908, USA
| | - Jose L Tomsig
- Department of Toxicology, School of Medicine, University of Virginia, Charlottesville, VA, 22908, USA
| | - Kevin R Lynch
- Department of Pharmacology, School of Medicine, University of Virginia, Charlottesville, VA, 22908, USA
| | - Zhen Yan
- Department of Pharmacology, School of Medicine, University of Virginia, Charlottesville, VA, 22908, USA.,Robert M. Berne Cardiovascular Research Center, School of Medicine, University of Virginia, Charlottesville, VA, 22908, USA
| | - Thurl E Harris
- Department of Pharmacology, School of Medicine, University of Virginia, Charlottesville, VA, 22908, USA
| | - Kyle L Hoehn
- Department of Pharmacology, School of Medicine, University of Virginia, Charlottesville, VA, 22908, USA. .,School of Biotechnology and Biomolecular Sciences, University of New South Wales, Kensington, Sydney, NSW, 2052, Australia.
| |
Collapse
|
92
|
Mullins RJ, Mustapic M, Goetzl EJ, Kapogiannis D. Exosomal biomarkers of brain insulin resistance associated with regional atrophy in Alzheimer's disease. Hum Brain Mapp 2017; 38:1933-1940. [PMID: 28105773 DOI: 10.1002/hbm.23494] [Citation(s) in RCA: 101] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Revised: 11/30/2016] [Accepted: 12/01/2016] [Indexed: 01/03/2023] Open
Abstract
Brain insulin resistance (IR), which depends on insulin-receptor-substrate-1 (IRS-1) phosphorylation, is characteristic of Alzheimer's disease (AD). Previously, we demonstrated higher pSer312-IRS-1 (ineffective insulin signaling) and lower p-panTyr-IRS-1 (effective insulin signaling) in neural origin-enriched plasma exosomes of AD patients vs. CONTROLS Here, we hypothesized that these exosomal biomarkers associate with brain atrophy in AD. We studied 24 subjects with biomarker-supported probable AD (low CSF Aβ42 ). Exosomes were isolated from plasma, enriched for neural origin using immunoprecipitation for L1CAM, and measured for pSer312 - and p-panTyr-IRS-1 phosphotypes. MPRAGE images were segmented by brain tissue type and voxel-based morphometry (VBM) analysis for gray matter against pSer312 - and p-panTyr-IRS-1 was conducted. Given the regionally variable brain expression of IRS-1, we used the Allen Brain Atlas to make spatial comparisons between VBM results and IRS-1 expression. Brain volume was positively associated with P-panTyr-IRS-1 and negatively associated with pSer312 -IRS-1 in a strikingly similar regional pattern (bilateral parietal-occipital junction, R middle temporal gyrus). This volumetric association pattern was spatially correlated with Allen Human Brain atlas normal brain IRS-1 expression. Exosomal biomarkers of brain IR are thus associated with atrophy in AD as could be expected by their pathophysiological roles and do so in a pattern that reflects regional IRS-1 expression. Furthermore, neural-origin plasma exosomes may recover molecular signals from specific brain regions. Hum Brain Mapp 38:1933-1940, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Roger J Mullins
- Laboratory of Neurosciences, Intramural Research Program, National Institute on Aging/National Institutes of Health (NIA/NIH)
| | - Maja Mustapic
- Laboratory of Neurosciences, Intramural Research Program, National Institute on Aging/National Institutes of Health (NIA/NIH)
| | - Edward J Goetzl
- Department of Medicine, University of California, San Francisco
| | - Dimitrios Kapogiannis
- Laboratory of Neurosciences, Intramural Research Program, National Institute on Aging/National Institutes of Health (NIA/NIH)
| |
Collapse
|
93
|
Abstract
Insulin resistance is one of the defining features of type 2 diabetes and the metabolic syndrome and accompanies many other clinical conditions, ranging from obesity to lipodystrophy to glucocorticoid excess. Extraordinary efforts have gone into defining the mechanisms that underlie insulin resistance, with most attention focused on altered signalling as well as mitochondrial and endoplasmic reticulum stress. Here, nuclear mechanisms of insulin resistance, including transcriptional and epigenomic effects, will be discussed. Three levels of control involving transcription factors, transcriptional cofactors, and chromatin-modifying enzymes will be considered. Well-studied examples of the first include PPAR-γ in adipose tissue and the glucocorticoid receptor and FoxO1 in a variety of insulin-sensitive tissues. These proteins work in concert with cofactors such as PGC-1α and CRTC2, and chromatin-modifying enzymes including DNA methyltransferases and histone acetyltransferases, to regulate key genes that promote insulin-stimulated glucose uptake, gluconeogenesis or other pathways that affect systemic insulin action. Furthermore, genetic variation associated with increased risk of type 2 diabetes is often related to altered transcription factor binding, either by affecting the transcription factor itself, or more commonly by changing the binding affinity of a noncoding regulatory region. Finally, several avenues for therapeutic exploitation in the battle against metabolic disease will be discussed, including small-molecule inhibitors and activators of these factors and their related pathways.
Collapse
Affiliation(s)
- E D Rosen
- Division of Endocrinology and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
94
|
Minard AY, Wong MKL, Chaudhuri R, Tan SX, Humphrey SJ, Parker BL, Yang JY, Laybutt DR, Cooney GJ, Coster ACF, Stöckli J, James DE. Hyperactivation of the Insulin Signaling Pathway Improves Intracellular Proteostasis by Coordinately Up-regulating the Proteostatic Machinery in Adipocytes. J Biol Chem 2016; 291:25629-25640. [PMID: 27738101 DOI: 10.1074/jbc.m116.741140] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Revised: 10/11/2016] [Indexed: 01/25/2023] Open
Abstract
Hyperinsulinemia, which is associated with aging and metabolic disease, may lead to defective protein homeostasis (proteostasis) due to hyperactivation of insulin-sensitive pathways such as protein synthesis. We investigated the effect of chronic hyperinsulinemia on proteostasis by generating a time-resolved map of insulin-regulated protein turnover in adipocytes using metabolic pulse-chase labeling and high resolution mass spectrometry. Hyperinsulinemia increased the synthesis of nearly half of all detected proteins and did not affect protein degradation despite suppressing autophagy. Unexpectedly, this marked elevation in protein synthesis was accompanied by enhanced protein stability and folding and not by markers of proteostasis stress such as protein carbonylation and aggregation. The improvement in proteostasis was attributed to a coordinate up-regulation of proteins in the global proteostasis network, including ribosomal, proteasomal, chaperone, and endoplasmic reticulum/mitochondrial unfolded protein response proteins. We conclude that defects associated with hyperactivation of the insulin signaling pathway are unlikely attributed to defective proteostasis because up-regulation of protein synthesis by insulin is accompanied by up-regulation of proteostatic machinery.
Collapse
Affiliation(s)
- Annabel Y Minard
- From The Garvan Institute of Medical Research, Sydney, New South Wales 2010, Australia.,Charles Perkins Centre, School of Life Environmental Sciences
| | - Martin K L Wong
- From The Garvan Institute of Medical Research, Sydney, New South Wales 2010, Australia.,Charles Perkins Centre, School of Life Environmental Sciences.,School of Physics
| | - Rima Chaudhuri
- Charles Perkins Centre, School of Life Environmental Sciences
| | - Shi-Xiong Tan
- From The Garvan Institute of Medical Research, Sydney, New South Wales 2010, Australia
| | - Sean J Humphrey
- Charles Perkins Centre, School of Life Environmental Sciences
| | | | | | - D Ross Laybutt
- From The Garvan Institute of Medical Research, Sydney, New South Wales 2010, Australia
| | | | - Adelle C F Coster
- Department of Applied Mathematics, School of Mathematics and Statistics, University of New South Wales, Sydney, New South Wales 2052, Australia
| | | | - David E James
- Charles Perkins Centre, School of Life Environmental Sciences, .,School of Medicine, University of Sydney, Sydney, New South Wales 2006, Australia, and
| |
Collapse
|
95
|
Jansson D, Scotter EL, Rustenhoven J, Coppieters N, Smyth LCD, Oldfield RL, Bergin PS, Mee EW, Graham ES, Faull RLM, Dragunow M. Interferon-γ blocks signalling through PDGFRβ in human brain pericytes. J Neuroinflammation 2016; 13:249. [PMID: 27654972 PMCID: PMC5031293 DOI: 10.1186/s12974-016-0722-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Accepted: 09/13/2016] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Neuroinflammation and blood-brain barrier (BBB) disruption are common features of many brain disorders, including Alzheimer's disease, epilepsy, and motor neuron disease. Inflammation is thought to be a driver of BBB breakdown, but the underlying mechanisms for this are unclear. Brain pericytes are critical cells for maintaining the BBB and are immunologically active. We sought to test the hypothesis that inflammation regulates the BBB by altering pericyte biology. METHODS We exposed primary adult human brain pericytes to chronic interferon-gamma (IFNγ) for 4 days and measured associated functional aspects of pericyte biology. Specifically, we examined the influence of inflammation on platelet-derived growth factor receptor-beta (PDGFRβ) expression and signalling, as well as pericyte proliferation and migration by qRT-PCR, immunocytochemistry, flow cytometry, and western blotting. RESULTS Chronic IFNγ treatment had marked effects on pericyte biology most notably through the PDGFRβ, by enhancing agonist (PDGF-BB)-induced receptor phosphorylation, internalization, and subsequent degradation. Functionally, chronic IFNγ prevented PDGF-BB-mediated pericyte proliferation and migration. CONCLUSIONS Because PDGFRβ is critical for pericyte function and its removal leads to BBB leakage, our results pinpoint a mechanism linking chronic brain inflammation to BBB dysfunction.
Collapse
Affiliation(s)
- Deidre Jansson
- Department of Pharmacology and Clinical Pharmacology, The University of Auckland, 1023, Auckland, New Zealand.,Gravida National Centre for Growth and Development, The University of Auckland, 1023, Auckland, New Zealand.,Centre for Brain Research, The University of Auckland, 1023, Auckland, New Zealand
| | - Emma L Scotter
- Department of Pharmacology and Clinical Pharmacology, The University of Auckland, 1023, Auckland, New Zealand.,Centre for Brain Research, The University of Auckland, 1023, Auckland, New Zealand
| | - Justin Rustenhoven
- Department of Pharmacology and Clinical Pharmacology, The University of Auckland, 1023, Auckland, New Zealand.,Centre for Brain Research, The University of Auckland, 1023, Auckland, New Zealand
| | - Natacha Coppieters
- Department of Anatomy and Medical Imaging, The University of Auckland, 1023, Auckland, New Zealand.,Centre for Brain Research, The University of Auckland, 1023, Auckland, New Zealand
| | - Leon C D Smyth
- Department of Pharmacology and Clinical Pharmacology, The University of Auckland, 1023, Auckland, New Zealand.,Centre for Brain Research, The University of Auckland, 1023, Auckland, New Zealand
| | | | - Peter S Bergin
- Centre for Brain Research, The University of Auckland, 1023, Auckland, New Zealand.,Auckland City Hospital, 1023, Auckland, New Zealand
| | - Edward W Mee
- Centre for Brain Research, The University of Auckland, 1023, Auckland, New Zealand.,Auckland City Hospital, 1023, Auckland, New Zealand
| | - E Scott Graham
- Department of Pharmacology and Clinical Pharmacology, The University of Auckland, 1023, Auckland, New Zealand.,Centre for Brain Research, The University of Auckland, 1023, Auckland, New Zealand
| | - Richard L M Faull
- Department of Anatomy and Medical Imaging, The University of Auckland, 1023, Auckland, New Zealand.,Centre for Brain Research, The University of Auckland, 1023, Auckland, New Zealand
| | - Mike Dragunow
- Department of Pharmacology and Clinical Pharmacology, The University of Auckland, 1023, Auckland, New Zealand. .,Gravida National Centre for Growth and Development, The University of Auckland, 1023, Auckland, New Zealand. .,Centre for Brain Research, The University of Auckland, 1023, Auckland, New Zealand. .,Department of Pharmacology and Clinical Pharmacology, The University of Auckland, Private Bag 92019, 1142, Auckland, New Zealand.
| |
Collapse
|
96
|
Pierre N, Appriou Z, Gratas-Delamarche A, Derbré F. From physical inactivity to immobilization: Dissecting the role of oxidative stress in skeletal muscle insulin resistance and atrophy. Free Radic Biol Med 2016; 98:197-207. [PMID: 26744239 DOI: 10.1016/j.freeradbiomed.2015.12.028] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2015] [Revised: 12/23/2015] [Accepted: 12/24/2015] [Indexed: 12/16/2022]
Abstract
In the literature, the terms physical inactivity and immobilization are largely used as synonyms. The present review emphasizes the need to establish a clear distinction between these two situations. Physical inactivity is a behavior characterized by a lack of physical activity, whereas immobilization is a deprivation of movement for medical purpose. In agreement with these definitions, appropriate models exist to study either physical inactivity or immobilization, leading thereby to distinct conclusions. In this review, we examine the involvement of oxidative stress in skeletal muscle insulin resistance and atrophy induced by, respectively, physical inactivity and immobilization. A large body of evidence demonstrates that immobilization-induced atrophy depends on the chronic overproduction of reactive oxygen and nitrogen species (RONS). On the other hand, the involvement of RONS in physical inactivity-induced insulin resistance has not been investigated. This observation outlines the need to elucidate the mechanism by which physical inactivity promotes insulin resistance.
Collapse
Affiliation(s)
- Nicolas Pierre
- EA1274 Laboratory "Movement, Sport and Health Sciences" M2S, Rennes 2 University - ENS Rennes, Bruz, France
| | - Zephyra Appriou
- EA1274 Laboratory "Movement, Sport and Health Sciences" M2S, Rennes 2 University - ENS Rennes, Bruz, France
| | - Arlette Gratas-Delamarche
- EA1274 Laboratory "Movement, Sport and Health Sciences" M2S, Rennes 2 University - ENS Rennes, Bruz, France
| | - Frédéric Derbré
- EA1274 Laboratory "Movement, Sport and Health Sciences" M2S, Rennes 2 University - ENS Rennes, Bruz, France.
| |
Collapse
|
97
|
Mohamad M, Mitchell SJ, Wu LE, White MY, Cordwell SJ, Mach J, Solon‐Biet SM, Boyer D, Nines D, Das A, Catherine Li S, Warren A, Hilmer SN, Fraser R, Sinclair DA, Simpson SJ, Cabo R, Le Couteur DG, Cogger VC. Ultrastructure of the liver microcirculation influences hepatic and systemic insulin activity and provides a mechanism for age-related insulin resistance. Aging Cell 2016; 15:706-15. [PMID: 27095270 PMCID: PMC4933657 DOI: 10.1111/acel.12481] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/18/2016] [Indexed: 12/16/2022] Open
Abstract
While age‐related insulin resistance and hyperinsulinemia are usually considered to be secondary to changes in muscle, the liver also plays a key role in whole‐body insulin handling and its role in age‐related changes in insulin homeostasis is largely unknown. Here, we show that patent pores called ‘fenestrations’ are essential for insulin transfer across the liver sinusoidal endothelium and that age‐related loss of fenestrations causes an impaired insulin clearance and hyperinsulinemia, induces hepatic insulin resistance, impairs hepatic insulin signaling, and deranges glucose homeostasis. To further define the role of fenestrations in hepatic insulin signaling without any of the long‐term adaptive responses that occur with aging, we induced acute defenestration using poloxamer 407 (P407), and this replicated many of the age‐related changes in hepatic glucose and insulin handling. Loss of fenestrations in the liver sinusoidal endothelium is a hallmark of aging that has previously been shown to cause deficits in hepatic drug and lipoprotein metabolism and now insulin. Liver defenestration thus provides a new mechanism that potentially contributes to age‐related insulin resistance.
Collapse
Affiliation(s)
- Mashani Mohamad
- Ageing and Alzheimers Institute Centre for Education and Research on Ageing University of Sydney and Concord Hospital Sydney NSW Australia
- ANZAC Research Institute University of Sydney and Concord Hospital Sydney NSW Australia
- Faculty of Pharmacy Universiti Teknologi MARA Selangor Malaysia
| | - Sarah Jayne Mitchell
- Translational Gerontology Branch National Institute on Aging National Institutes of Health Baltimore MD USA
| | - Lindsay Edward Wu
- Laboratory for Ageing Research School of Medical Sciences University of New South Wales Sydney NSW Australia
| | | | | | - John Mach
- Kolling Institute of Medical Research Royal North Shore Hospital and University of Sydney Sydney NSW Australia
| | - Samantha Marie Solon‐Biet
- Ageing and Alzheimers Institute Centre for Education and Research on Ageing University of Sydney and Concord Hospital Sydney NSW Australia
- ANZAC Research Institute University of Sydney and Concord Hospital Sydney NSW Australia
- Charles Perkins Centre University of Sydney Sydney NSW Australia
| | - Dawn Boyer
- Translational Gerontology Branch National Institute on Aging National Institutes of Health Baltimore MD USA
| | - Dawn Nines
- Translational Gerontology Branch National Institute on Aging National Institutes of Health Baltimore MD USA
| | - Abhirup Das
- Laboratory for Ageing Research School of Medical Sciences University of New South Wales Sydney NSW Australia
| | - Shi‐Yun Catherine Li
- Laboratory for Ageing Research School of Medical Sciences University of New South Wales Sydney NSW Australia
| | - Alessandra Warren
- Ageing and Alzheimers Institute Centre for Education and Research on Ageing University of Sydney and Concord Hospital Sydney NSW Australia
- ANZAC Research Institute University of Sydney and Concord Hospital Sydney NSW Australia
| | - Sarah Nicole Hilmer
- Kolling Institute of Medical Research Royal North Shore Hospital and University of Sydney Sydney NSW Australia
| | - Robin Fraser
- Department of Pathology University of Otago Christchurch New Zealand
| | - David Andrew Sinclair
- Laboratory for Ageing Research School of Medical Sciences University of New South Wales Sydney NSW Australia
- Department of Genetics Harvard Medical School Boston MA USA
| | | | - Rafael Cabo
- Translational Gerontology Branch National Institute on Aging National Institutes of Health Baltimore MD USA
| | - David George Le Couteur
- Ageing and Alzheimers Institute Centre for Education and Research on Ageing University of Sydney and Concord Hospital Sydney NSW Australia
- ANZAC Research Institute University of Sydney and Concord Hospital Sydney NSW Australia
- Charles Perkins Centre University of Sydney Sydney NSW Australia
| | - Victoria Carroll Cogger
- Ageing and Alzheimers Institute Centre for Education and Research on Ageing University of Sydney and Concord Hospital Sydney NSW Australia
- ANZAC Research Institute University of Sydney and Concord Hospital Sydney NSW Australia
- Charles Perkins Centre University of Sydney Sydney NSW Australia
| |
Collapse
|
98
|
JNK Signaling: Regulation and Functions Based on Complex Protein-Protein Partnerships. Microbiol Mol Biol Rev 2016; 80:793-835. [PMID: 27466283 DOI: 10.1128/mmbr.00043-14] [Citation(s) in RCA: 378] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The c-Jun N-terminal kinases (JNKs), as members of the mitogen-activated protein kinase (MAPK) family, mediate eukaryotic cell responses to a wide range of abiotic and biotic stress insults. JNKs also regulate important physiological processes, including neuronal functions, immunological actions, and embryonic development, via their impact on gene expression, cytoskeletal protein dynamics, and cell death/survival pathways. Although the JNK pathway has been under study for >20 years, its complexity is still perplexing, with multiple protein partners of JNKs underlying the diversity of actions. Here we review the current knowledge of JNK structure and isoforms as well as the partnerships of JNKs with a range of intracellular proteins. Many of these proteins are direct substrates of the JNKs. We analyzed almost 100 of these target proteins in detail within a framework of their classification based on their regulation by JNKs. Examples of these JNK substrates include a diverse assortment of nuclear transcription factors (Jun, ATF2, Myc, Elk1), cytoplasmic proteins involved in cytoskeleton regulation (DCX, Tau, WDR62) or vesicular transport (JIP1, JIP3), cell membrane receptors (BMPR2), and mitochondrial proteins (Mcl1, Bim). In addition, because upstream signaling components impact JNK activity, we critically assessed the involvement of signaling scaffolds and the roles of feedback mechanisms in the JNK pathway. Despite a clarification of many regulatory events in JNK-dependent signaling during the past decade, many other structural and mechanistic insights are just beginning to be revealed. These advances open new opportunities to understand the role of JNK signaling in diverse physiological and pathophysiological states.
Collapse
|
99
|
Affourtit C. Mitochondrial involvement in skeletal muscle insulin resistance: A case of imbalanced bioenergetics. BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2016; 1857:1678-93. [PMID: 27473535 DOI: 10.1016/j.bbabio.2016.07.008] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Revised: 06/19/2016] [Accepted: 07/23/2016] [Indexed: 12/16/2022]
Abstract
Skeletal muscle insulin resistance in obesity associates with mitochondrial dysfunction, but the causality of this association is controversial. This review evaluates mitochondrial models of nutrient-induced muscle insulin resistance. It transpires that all models predict that insulin resistance arises as a result of imbalanced cellular bioenergetics. The nature and precise origin of the proposed insulin-numbing molecules differ between models but all species only accumulate when metabolic fuel supply outweighs energy demand. This observation suggests that mitochondrial deficiency in muscle insulin resistance is not merely owing to intrinsic functional defects, but could instead be an adaptation to nutrient-induced changes in energy expenditure. Such adaptive effects are likely because muscle ATP supply is fully driven by energy demand. This market-economic control of myocellular bioenergetics offers a mechanism by which insulin-signalling deficiency can cause apparent mitochondrial dysfunction, as insulin resistance lowers skeletal muscle anabolism and thus dampens ATP demand and, consequently, oxidative ATP synthesis.
Collapse
Affiliation(s)
- Charles Affourtit
- School of Biomedical and Healthcare Sciences, Plymouth University Peninsula Schools of Medicine and Dentistry, Plymouth University, Drake Circus, PL4 8AA Plymouth, UK.
| |
Collapse
|
100
|
Snook LA, Wright DC, Holloway GP. Postprandial control of fatty acid transport proteins' subcellular location is not dependent on insulin. FEBS Lett 2016; 590:2661-70. [PMID: 27311759 DOI: 10.1002/1873-3468.12260] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Revised: 04/28/2016] [Accepted: 06/14/2016] [Indexed: 01/09/2023]
Abstract
Fatty acid transport proteins rapidly translocate to the plasma membrane in response to various stimuli, including insulin, influencing lipid uptake into muscle. However, our understanding of the mechanisms regulating postprandial fatty acid transporter subcellular location remains limited. We demonstrate that the response of fatty acid transporters to insulin stimulation is extremely brief and not temporally matched in the postprandial state. We further show that high-fat diet-induced accumulation of fatty acid transporters on the plasma membrane can occur in the absence of insulin. Altogether, these data suggest that insulin is not the primary signal regulating fatty acid transporter relocation in vivo.
Collapse
Affiliation(s)
- Laelie A Snook
- Department of Human Health and Nutritional Sciences, University of Guelph, Canada
| | - David C Wright
- Department of Human Health and Nutritional Sciences, University of Guelph, Canada
| | - Graham P Holloway
- Department of Human Health and Nutritional Sciences, University of Guelph, Canada
| |
Collapse
|