1
|
Ahmed M, Biswas T, Mondal S. The strategic involvement of IRS in cancer progression. Biochem Biophys Res Commun 2023; 680:141-160. [PMID: 37738904 DOI: 10.1016/j.bbrc.2023.09.036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 09/13/2023] [Accepted: 09/15/2023] [Indexed: 09/24/2023]
Abstract
Insulin Receptor Substrate (IRS), an intracellular molecule devoid of an intrinsic kinase activity, is activated upon binding to IR which thereby works as a scaffold, organizing all signaling complexes and initiating the signaling process downstream. The level of IRS proteins and their stability in the cell is mostly maintained through the phosphorylation status of their tyrosine and serine residues. IRS is positively regulated by phosphorylation of its Tyr residues whereas a Ser residue phosphorylation attenuates it, although there exist some exceptions as well. Other post-translational modifications like O-linked glycosylation, N-linked glycosylation and acetylation also play a prominent role in IRS regulation. Since the discovery of the Warburg effect, people have been curious to find out all possible signaling networks and molecules that could lead to cancer and no doubt, the insulin signaling pathway is identified as one such pathway, which is highly deregulated in cancers. Eminent studies reveal that IRS is a pertinent regulator of cancer and is highly overexpressed in the five most commonly occurring cancers namely- Prostate, Ovarian, Breast, Colon and Lung cancers. IRS1 and IRS2 family members are actively involved in the progression, invasion and metastasis of these cancers. Recently, less studied IRS4 has also emerged as a contributor in ovarian, breast, colorectal and lung cancer, but no such studies related to IRS4 are found in Prostate cancer. The involvement of other IRS family members in cancer is still undiscovered and so paves the way for further exploration. This review is a time-lapse study of IRSs in the context of cancer done over the past two decades and it highlights all the major discoveries made till date, in these cancers from the perspective of IRS.
Collapse
Affiliation(s)
- Mehnaz Ahmed
- Department of Life Sciences, Presidency University, 86/1 College Street, Kolkata, 700073, West Bengal, India
| | - Tannishtha Biswas
- Department of Life Sciences, Presidency University, 86/1 College Street, Kolkata, 700073, West Bengal, India
| | - Susmita Mondal
- Department of Life Sciences, Presidency University, 86/1 College Street, Kolkata, 700073, West Bengal, India.
| |
Collapse
|
2
|
Hassouna MM, Moustafa MS, Hamdy M, Abdelsameea E, Abbasy M, Naguib M. Study of transcription factor 7-like 2 (TCF7L2) gene polymorphism in cirrhotic patients with diabetes. EGYPTIAN LIVER JOURNAL 2023; 13:54. [DOI: 10.1186/s43066-023-00285-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 09/21/2023] [Indexed: 01/04/2025] Open
Abstract
AbstractPatients with chronic liver disease (CLD) as chronic hepatitis C (CHC) are at high risk of diabetes type 2 (T2D). Genetic factors are suggested to modulate diabetes development in cirrhotic patients. TCF7L2 gene has been reported to be associated with type 2 diabetes, but the association of TCF7L2 with cirrhotic patients with diabetes is unclear. We aimed to study the TCF7L2 gene polymorphisms (rs 290487) in cirrhotic patients with diabetes.Method The study was assessed on 25 cirrhotic patients with type 2 diabetes who were compared to 25 cirrhotic HCV patients (nondiabetic), 25 diabetic type 2 patients, and 25 age- and gender-matched healthy control groups. After the collection of relevant clinical data and basic laboratory tests, single-nucleotide polymorphism (SNP) in the TCF7L2 gene (rs290487) was performed by a real-time PCR technique.Results Cirrhotic patients with diabetes presented significantly poorer liver function, higher incidence of cirrhotic complications, and higher glucose levels compared with cirrhotic nondiabetic patients. The TCF7L2 rs290487 TT variant showed significantly increased diabetes risk in cirrhotic patients compared with CC and CT genotypes.Conclusions TCF7L2 rs290487 polymorphism could be associated with increased diabetic risk in cirrhotic patients.
Collapse
|
3
|
Dhanya R. Quercetin for managing type 2 diabetes and its complications, an insight into multitarget therapy. Biomed Pharmacother 2021; 146:112560. [PMID: 34953390 DOI: 10.1016/j.biopha.2021.112560] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 12/17/2021] [Accepted: 12/19/2021] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Quercetin, a bioflavonoid abundant in grapefruit, onion, berries, etc., has vast therapeutic potential, especially against Type 2 diabetes and its complications. Quercetin showed similar effects as that of metformin, (widely prescribed antidiabetic drug) in cell lines models (Sajan et al., 2010; Dhanya et al., 2017). In vivo findings also showcase it as a promising agent against diabetes and its pathophysiological complications. SCOPE AND APPROACH Quercetin can be produced on a large scale through a novel fermentation-based glycosylation strategy from cheap substrates and can be utilized as a dietary supplement. The review focuses on the mounting evidence pointing to Quercetin as a promising candidate for managing type 2 diabetes and its oxidative stress mediated pathophysiological complications. CONCLUSION Quercetin acts on multiple targets of diabetes and regulates key signalling pathways which improve the symptoms as well as the complications of Type 2 diabetes. However further studies are needed to improve the bioavailability and to establish a dosing regimen for Quercetin.
Collapse
Affiliation(s)
- R Dhanya
- Cardiovascular Diseases and Diabetes Biology Division, Rajiv Gandhi Centre for Biotechnology (RGCB), Thycaud Post, Poojappura, Trivandrum 695014, Kerala, India.
| |
Collapse
|
4
|
Karlsson HKR, Kasahara A, Ikeda M, Chibalin AV, Harada J, Rydén M, Krook A, Kato M, Kubota K, Zierath JR. Quantitative phosphoproteomic analysis of IRS1 in skeletal muscle from men with normal glucose tolerance or type 2 diabetes: A case-control study. Metabolism 2021; 118:154726. [PMID: 33581131 DOI: 10.1016/j.metabol.2021.154726] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 01/23/2021] [Accepted: 02/08/2021] [Indexed: 11/20/2022]
Abstract
BACKGROUND & AIMS The physiological regulation and contribution of the multiple phosphorylation sites of insulin receptor substrate 1 (IRS1) to the pathogenesis of insulin resistance is unknown. Our aims were to map the phosphorylated motifs of IRS1 in skeletal muscle from people with normal glucose tolerance (NGT; n = 11) or type 2 diabetes mellitus (T2DM; n = 11). METHODS Skeletal muscle biopsies were obtained under fasted conditions or during a euglycemic clamp and IRS1 phosphorylation sites were identified by mass spectrometry. RESULTS We identified 33 phosphorylation sites in biopsies from fasted individuals, including 2 previously unreported sites ([Ser393] and [Thr1017]). In men with NGT and T2DM, insulin increased phosphorylation of 5 peptides covering 10 serine or threonine sites and decreased phosphorylation of 6 peptides covering 9 serine, threonine or tyrosine sites. Insulin-stimulation increased phosphorylation of 2 peptides, and decreased phosphorylation of 2 peptides only in men with NGT. Insulin increased phosphorylation of 2 peptides only in men with T2DM. CONCLUSIONS Despite severe skeletal muscle insulin resistance, the pattern of IRS1 phosphorylation was not uniformly altered in T2DM. Our results contribute to the evolving understanding of the physiological regulation of insulin signaling and complement the comprehensive map of IRS1 phosphorylation in T2DM.
Collapse
Affiliation(s)
- Håkan K R Karlsson
- Department of Molecular Medicine and Surgery, Integrative Physiology, Karolinska Institutet, Stockholm 171 77, Sweden
| | - Akiko Kasahara
- Discovery Science and Technology Department, Daiichi Sankyo RD Novare Co., Ltd., 1-16-13 Kitakasai, Edogawa-ku, Tokyo 134-8630, Japan
| | - Mika Ikeda
- Cardiovascular-Metabolics Research Laboratories, Daiichi Sankyo Co., Ltd., 1-2-58, Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan
| | - Alexander V Chibalin
- Department of Molecular Medicine and Surgery, Integrative Physiology, Karolinska Institutet, Stockholm 171 77, Sweden
| | - Jun Harada
- Cardiovascular-Metabolics Research Laboratories, Daiichi Sankyo Co., Ltd., 1-2-58, Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan
| | - Mikael Rydén
- Department of Medicine, Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Anna Krook
- Department of Physiology and Pharmacology, Integrative Physiology, Karolinska Institutet, Stockholm 171 77, Sweden
| | - Mitsunori Kato
- Discovery Science and Technology Department, Daiichi Sankyo RD Novare Co., Ltd., 1-16-13 Kitakasai, Edogawa-ku, Tokyo 134-8630, Japan
| | - Kazuishi Kubota
- Discovery Science and Technology Department, Daiichi Sankyo RD Novare Co., Ltd., 1-16-13 Kitakasai, Edogawa-ku, Tokyo 134-8630, Japan
| | - Juleen R Zierath
- Department of Molecular Medicine and Surgery, Integrative Physiology, Karolinska Institutet, Stockholm 171 77, Sweden; Department of Physiology and Pharmacology, Integrative Physiology, Karolinska Institutet, Stockholm 171 77, Sweden.
| |
Collapse
|
5
|
Mustapha S, Mohammed M, Azemi AK, Yunusa I, Shehu A, Mustapha L, Wada Y, Ahmad MH, Ahmad WANW, Rasool AHG, Mokhtar SS. Potential Roles of Endoplasmic Reticulum Stress and Cellular Proteins Implicated in Diabesity. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:8830880. [PMID: 33995826 PMCID: PMC8099518 DOI: 10.1155/2021/8830880] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Revised: 03/28/2021] [Accepted: 04/05/2021] [Indexed: 12/12/2022]
Abstract
The role of the endoplasmic reticulum (ER) has evolved from protein synthesis, processing, and other secretory pathways to forming a foundation for lipid biosynthesis and other metabolic functions. Maintaining ER homeostasis is essential for normal cellular function and survival. An imbalance in the ER implied stressful conditions such as metabolic distress, which activates a protective process called unfolded protein response (UPR). This response is activated through some canonical branches of ER stress, i.e., the protein kinase RNA-like endoplasmic reticulum kinase (PERK), inositol-requiring enzyme 1α (IRE1α), and activating transcription factor 6 (ATF6). Therefore, chronic hyperglycemia, hyperinsulinemia, increased proinflammatory cytokines, and free fatty acids (FFAs) found in diabesity (a pathophysiological link between obesity and diabetes) could lead to ER stress. However, limited data exist regarding ER stress and its association with diabesity, particularly the implicated proteins and molecular mechanisms. Thus, this review highlights the role of ER stress in relation to some proteins involved in diabesity pathogenesis and provides insight into possible pathways that could serve as novel targets for therapeutic intervention.
Collapse
Affiliation(s)
- Sagir Mustapha
- Department of Pharmacology, School of Medical Sciences, Universiti Sains Malaysia, 16150 Kota Bharu, Kelantan, Malaysia
- Department of Pharmacology and Therapeutics, Ahmadu Bello University Zaria, Kaduna, Nigeria
| | - Mustapha Mohammed
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, 11800 Penang, Pulau Pinang, Malaysia
- Department of Clinical Pharmacy and Pharmacy Practice, Ahmadu Bello University Zaria, Kaduna, Nigeria
| | - Ahmad Khusairi Azemi
- Department of Pharmacology, School of Medical Sciences, Universiti Sains Malaysia, 16150 Kota Bharu, Kelantan, Malaysia
| | - Ismaeel Yunusa
- Department of Clinical Pharmacy and Outcomes Sciences, University of South Carolina, College of Pharmacy, Columbia, SC, USA
| | - Aishatu Shehu
- Department of Pharmacology and Therapeutics, Ahmadu Bello University Zaria, Kaduna, Nigeria
| | - Lukman Mustapha
- Department of Pharmaceutical and Medicinal Chemistry, Kaduna State University, Kaduna, Nigeria
| | - Yusuf Wada
- Department of Medical Microbiology and Parasitology, School of Medical Sciences, Universiti Sains Malaysia, 16150 Kota Bharu, Kelantan, Malaysia
- Department of Zoology, Ahmadu Bello University Zaria, Kaduna, Nigeria
| | - Mubarak Hussaini Ahmad
- Department of Pharmacology and Therapeutics, Ahmadu Bello University Zaria, Kaduna, Nigeria
- School of Pharmacy Technician, Aminu Dabo College of Health Sciences and Technology, Kano, Nigeria
| | - Wan Amir Nizam Wan Ahmad
- Biomedicine Programme, School of Health Sciences, Universiti Sains Malaysia, 16150 Kota Bharu, Kelantan, Malaysia
| | - Aida Hanum Ghulam Rasool
- Department of Pharmacology, School of Medical Sciences, Universiti Sains Malaysia, 16150 Kota Bharu, Kelantan, Malaysia
- Hospital Universiti Sains Malaysia, 16150 Kota Bharu, Kelantan, Malaysia
| | - Siti Safiah Mokhtar
- Department of Pharmacology, School of Medical Sciences, Universiti Sains Malaysia, 16150 Kota Bharu, Kelantan, Malaysia
| |
Collapse
|
6
|
Zhang Y, Wu X, Zhao C, Li K, Zheng Y, Zhao J, Ge P. Integrative Analysis of Whole-genome Expression Profiling and Regulatory Network Identifies Novel Biomarkers for Insulin Resistance in Leptin Receptor-deficient Mice. Med Chem 2021; 16:635-642. [PMID: 31584376 DOI: 10.2174/1573406415666191004135450] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 05/11/2019] [Accepted: 08/23/2019] [Indexed: 11/22/2022]
Abstract
BACKGROUND Molecular characterization of insulin resistance, a growing health issue worldwide, will help to develop novel strategies and accurate biomarkers for disease diagnosis and treatment. OBJECTIVE Integrative analysis of gene expression profiling and gene regulatory network was exploited to identify potential biomarkers early in the development of insulin resistance. METHODS RNA was isolated from livers of animals at three weeks of age, and whole-genome expression profiling was performed and analyzed with Agilent mouse 4×44K microarrays. Differentially expressed genes were subsequently validated by qRT-PCR. Functional characterizations of genes and their interactions were performed by Gene Ontology (GO) analysis and gene regulatory network (GRN) analysis. RESULTS A total of 197 genes were found to be differentially expressed by fold change ≥2 and P < 0.05 in BKS-db +/+ mice relative to sex and age-matched controls. Functional analysis suggested that these differentially expressed genes were enriched in the regulation of phosphorylation and generation of precursor metabolites which are closely associated with insulin resistance. Then a gene regulatory network associated with insulin resistance (IRGRN) was constructed by integration of these differentially expressed genes and known human protein-protein interaction network. The principal component analysis demonstrated that 67 genes in IRGRN could clearly distinguish insulin resistance from the non-disease state. Some of these candidate genes were further experimentally validated by qRT-PCR, highlighting the predictive role as biomarkers in insulin resistance. CONCLUSION Our study provides new insight into the pathogenesis and treatment of insulin resistance and also reveals potential novel molecular targets and diagnostic biomarkers for insulin resistance.
Collapse
Affiliation(s)
- Yuchi Zhang
- Department of Pharmacology, School of Basic Medical Sciences, Heilongjiang University of Chinese Medicine, Harbin 150040, China
| | - Xinyu Wu
- Department of Traditional Chinese Medicine, School of Basic Medical Sciences, Heilongjiang University of Chinese Medicine, Harbin 150040, China
| | - Cong Zhao
- Hongqi Hospital Affiliated to Mudanjiang Medical University, Mudanjiang 157011, China
| | - Kai Li
- Harbin Food and Drug Administration, Harbin 150016, China
| | - Yi Zheng
- Chinese People 's Liberation Army Military Economics Institute, Wuhan 430035, China
| | - Jing Zhao
- Department of Pharmacology, School of Basic Medical Sciences, Heilongjiang University of Chinese Medicine, Harbin 150040, China
| | - Pengling Ge
- Department of Pharmacology, School of Basic Medical Sciences, Heilongjiang University of Chinese Medicine, Harbin 150040, China
| |
Collapse
|
7
|
Acute Insulin Resistance and Rapid Alterations in Neuronal Derived Blood Exosome Concentration After Branched Endovascular Aortic Aneurysm Repair. Eur J Vasc Endovasc Surg 2020; 59:457-463. [DOI: 10.1016/j.ejvs.2019.10.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2019] [Revised: 09/26/2019] [Accepted: 10/11/2019] [Indexed: 12/23/2022]
|
8
|
Yu Q, Xia Z, Liong EC, Tipoe GL. Chronic aerobic exercise improves insulin sensitivity and modulates Nrf2 and NF‑κB/IκBα pathways in the skeletal muscle of rats fed with a high fat diet. Mol Med Rep 2019; 20:4963-4972. [PMID: 31702809 PMCID: PMC6854540 DOI: 10.3892/mmr.2019.10787] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 08/12/2019] [Indexed: 12/22/2022] Open
Abstract
The present study aimed to investigate the molecular mechanisms of the ameliorative effects of chronic aerobic exercise on non-alcoholic steatohepatitis (NASH) in rat skeletal muscle. Female Sprague-Dawley rats (n=6–9 per group) were divided into four groups: i) Rats fed with normal chow; ii) exercise rats fed with normal chow + exercise (run on a rotarod for 30 min per day from 9–12 weeks); iii) rats fed with a high-fat diet (HFD); iv) rats fed with an HFD + exercise. All HFD rats were fed with an HFD consisting of 30% fat from fish oil throughout the study for 12 weeks. Exercise decreased the levels of hepatic lipogenic markers carbohydrate-responsive element-binding protein, fat-specific protein 27 and liver X receptor and improved systemic glucose and insulin intolerance in the NASH animal model. The beneficial effects may have been mediated partly via the tripartite motif-containing family protein 72 (TRIM72)/PI3K/Akt/mTOR pathway, accompanied with an upregulation of glucose transporter 4 in the skeletal muscle. The exercise regimen activated the master regulator of antioxidant enzymes, nuclear factor erythroid 2-related factor 2, with upregulation of superoxide dismutase [Cu-Zn] expression and a corresponding decrease in kelch-like ECH-associated protein 1 expression, but failed to decrease the levels of the oxidative marker malondialdehyde in the HFD rat skeletal muscle. Chronic exercise decreased the expression of the inflammation marker NF-κB, followed by a decrease in interleukin-6 and tumor necrosis factor-α levels, as verified by a corresponding increase in the level of NF-κB inhibitor α expression. Exercise may exert its beneficial effects by improving muscle insulin sensitivity via the TRIM72/PI3K/Akt/mTOR pathway, contributing to the improvement of systemic insulin intolerance, and finally leading to decreased hepatic lipogenesis during NASH. The attenuation of insulin resistance by exercise may be partly achieved through a decrease in the level of inflammation and an increased antioxidant response.
Collapse
Affiliation(s)
- Qian Yu
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR, P.R. China
| | - Zhengyun Xia
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR, P.R. China
| | - Emily Chiu Liong
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR, P.R. China
| | - George Lim Tipoe
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR, P.R. China
| |
Collapse
|
9
|
Okatan EN, Olgar Y, Tuncay E, Turan B. Azoramide improves mitochondrial dysfunction in palmitate-induced insulin resistant H9c2 cells. Mol Cell Biochem 2019; 461:65-72. [PMID: 31327095 DOI: 10.1007/s11010-019-03590-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Accepted: 07/13/2019] [Indexed: 12/23/2022]
Abstract
Azoramide is identified as a new compound with the dual properties for the improvement of ER-folding capacity in various cells as well as for the treatment of T2DM. Although the effect of azoramide in glucose-homeostasis in mammalians is not known very well, a limited number of experimental studies showed that it could improve the insulin sensitivity in genetically obese mice. Therefore, here, we aimed to investigate the direct effect of azoramide on insulin signaling in insulin-resistant (IR) cardiomyocytes using IR-modelled ventricular cardiomyocytes. This model was established in H9c2 cells using palmitic acid incubation (50-μM for 24-h). The development of IR in cells was verified by monitoring the cellular 2-DG6P uptake assays in these treated cells. The 2-DG6P uptake was 50% less in the IR-cells compared to the control cells, while azoramide treatment (20-μM for 48-h) could prevent fully that decrease. In addition, azoramide treatment markedly preserved the IR-induced less ATP production and high-ROS production in these IR-cells. Furthermore, this treatment prevented the functional changes in mitochondria characterized by depolarized mitochondrial membrane potential and mitochondrial fusion or fusion-related protein levels as well as cellular ATP level. Moreover, this treatment provided marked protection against IR-associated changes in the insulin signaling pathway in cells, including recovery in the phosphorylation of IRS1 and Akt as well as the protein level of GLUT4 and Akt. Our present results, for the first time, demonstrated that azoramide plays an important protective role in IR-cardiomyocytes, at most, protective action on mitochondria. Therefore, one can suggest that azoramide, as a novel regulator, can provide direct cardioprotection in the IR-heart, at most, via affecting mitochondria and can be a good candidate as a new drug for the treatment of IR-associated cardiovascular disorders in mammalians with systemic IR.
Collapse
Affiliation(s)
- Esma Nur Okatan
- Department of Biophysics, Faculty of Medicine, Cumhuriyet University, Sivas, Turkey
| | - Yusuf Olgar
- Department of Biophysics, Faculty of Medicine, Ankara University, Ankara, Turkey
| | - Erkan Tuncay
- Department of Biophysics, Faculty of Medicine, Ankara University, Ankara, Turkey
| | - Belma Turan
- Department of Biophysics, Faculty of Medicine, Ankara University, Ankara, Turkey.
| |
Collapse
|
10
|
Parker SS, Krantz J, Kwak EA, Barker NK, Deer CG, Lee NY, Mouneimne G, Langlais PR. Insulin Induces Microtubule Stabilization and Regulates the Microtubule Plus-end Tracking Protein Network in Adipocytes. Mol Cell Proteomics 2019; 18:1363-1381. [PMID: 31018989 PMCID: PMC6601206 DOI: 10.1074/mcp.ra119.001450] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Indexed: 12/21/2022] Open
Abstract
Insulin-stimulated glucose uptake is known to involve microtubules, although the function of microtubules and the microtubule-regulating proteins involved in insulin action are poorly understood. CLASP2, a plus-end tracking microtubule-associated protein (+TIP) that controls microtubule dynamics, was recently implicated as the first +TIP associated with insulin-regulated glucose uptake. Here, using protein-specific targeted quantitative phosphoproteomics within 3T3-L1 adipocytes, we discovered that insulin regulates phosphorylation of the CLASP2 network members G2L1, MARK2, CLIP2, AGAP3, and CKAP5 as well as EB1, revealing the existence of a previously unknown microtubule-associated protein system that responds to insulin. To further investigate, G2L1 interactome studies within 3T3-L1 adipocytes revealed that G2L1 coimmunoprecipitates CLASP2 and CLIP2 as well as the master integrators of +TIP assembly, the end binding (EB) proteins. Live-cell total internal reflection fluorescence microscopy in adipocytes revealed G2L1 and CLASP2 colocalize on microtubule plus-ends. We found that although insulin increases the number of CLASP2-containing plus-ends, insulin treatment simultaneously decreases CLASP2-containing plus-end velocity. In addition, we discovered that insulin stimulates redistribution of CLASP2 and G2L1 from exclusive plus-end tracking to "trailing" behind the growing tip of the microtubule. Insulin treatment increases α-tubulin Lysine 40 acetylation, a mechanism that was observed to be regulated by a counterbalance between GSK3 and mTOR, and led to microtubule stabilization. Our studies introduce insulin-stimulated microtubule stabilization and plus-end trailing of +TIPs as new modes of insulin action and reveal the likelihood that a network of microtubule-associated proteins synergize to coordinate insulin-regulated microtubule dynamics.
Collapse
Affiliation(s)
- Sara S Parker
- From the ‡Department of Cellular & Molecular Medicine
| | - James Krantz
- §Department of Medicine, Division of Endocrinology
| | | | | | - Chris G Deer
- University of Arizona Research Computing, University of Arizona, Tucson, Arizona 85721
| | - Nam Y Lee
- ¶Department of Pharmacology,; ‖Department of Chemistry & Biochemistry, University of Arizona College of Medicine, Tucson, Arizona 85721
| | | | | |
Collapse
|
11
|
The Role of Mammalian Target of Rapamycin (mTOR) in Insulin Signaling. Nutrients 2017; 9:nu9111176. [PMID: 29077002 PMCID: PMC5707648 DOI: 10.3390/nu9111176] [Citation(s) in RCA: 187] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Revised: 10/23/2017] [Accepted: 10/24/2017] [Indexed: 12/15/2022] Open
Abstract
The mammalian target of rapamycin (mTOR) is a serine/threonine kinase that controls a wide spectrum of cellular processes, including cell growth, differentiation, and metabolism. mTOR forms two distinct multiprotein complexes known as mTOR complex 1 (mTORC1) and mTOR complex 2 (mTORC2), which are characterized by the presence of raptor and rictor, respectively. mTOR controls insulin signaling by regulating several downstream components such as growth factor receptor-bound protein 10 (Grb10), insulin receptor substrate (IRS-1), F-box/WD repeat-containing protein 8 (Fbw8), and insulin like growth factor 1 receptor/insulin receptor (IGF-IR/IR). In addition, mTORC1 and mTORC2 regulate each other through a feedback loop to control cell growth. This review outlines the current understanding of mTOR regulation in insulin signaling in the context of whole body metabolism.
Collapse
|
12
|
Parker L, Shaw CS, Stepto NK, Levinger I. Exercise and Glycemic Control: Focus on Redox Homeostasis and Redox-Sensitive Protein Signaling. Front Endocrinol (Lausanne) 2017; 8:87. [PMID: 28529499 PMCID: PMC5418238 DOI: 10.3389/fendo.2017.00087] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Accepted: 04/04/2017] [Indexed: 12/16/2022] Open
Abstract
Physical inactivity, excess energy consumption, and obesity are associated with elevated systemic oxidative stress and the sustained activation of redox-sensitive stress-activated protein kinase (SAPK) and mitogen-activated protein kinase signaling pathways. Sustained SAPK activation leads to aberrant insulin signaling, impaired glycemic control, and the development and progression of cardiometabolic disease. Paradoxically, acute exercise transiently increases oxidative stress and SAPK signaling, yet postexercise glycemic control and skeletal muscle function are enhanced. Furthermore, regular exercise leads to the upregulation of antioxidant defense, which likely assists in the mitigation of chronic oxidative stress-associated disease. In this review, we explore the complex spatiotemporal interplay between exercise, oxidative stress, and glycemic control, and highlight exercise-induced reactive oxygen species and redox-sensitive protein signaling as important regulators of glucose homeostasis.
Collapse
Affiliation(s)
- Lewan Parker
- Institute of Sport, Exercise and Active Living (ISEAL), College of Sport and Exercise Science, Victoria University, Melbourne, VIC, Australia
- *Correspondence: Lewan Parker, ,
| | - Christopher S. Shaw
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Sciences, Deakin University, Geelong, VIC, Australia
| | - Nigel K. Stepto
- Institute of Sport, Exercise and Active Living (ISEAL), College of Sport and Exercise Science, Victoria University, Melbourne, VIC, Australia
- Monash Centre for Health Research and Implementation, School of Public Health and Preventative Medicine, Monash University, Clayton, VIC, Australia
- Australian Institute for Musculoskeletal Science (AIMSS), Victoria University and Western Health, St. Albans, VIC, Australia
| | - Itamar Levinger
- Institute of Sport, Exercise and Active Living (ISEAL), College of Sport and Exercise Science, Victoria University, Melbourne, VIC, Australia
- Australian Institute for Musculoskeletal Science (AIMSS), Victoria University and Western Health, St. Albans, VIC, Australia
| |
Collapse
|
13
|
White MF, Copps KD. The Mechanisms of Insulin Action. ENDOCRINOLOGY: ADULT AND PEDIATRIC 2016:556-585.e13. [DOI: 10.1016/b978-0-323-18907-1.00033-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
14
|
Endoplasmic reticulum stress impairs insulin receptor signaling in the brains of obese rats. PLoS One 2015; 10:e0126384. [PMID: 25978724 PMCID: PMC4433117 DOI: 10.1371/journal.pone.0126384] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Accepted: 04/01/2015] [Indexed: 12/11/2022] Open
Abstract
The incidence of obesity is increasing worldwide. It was reported that endoplasmic reticulum stress (ERS) could inhibit insulin receptor signaling by activating c-Jun N-terminal kinase (JNK) in the liver. However, the relationship between ERS and insulin receptor signaling in the brain during obesity remains unclear. The aim of the current study was to assess whether ERS alters insulin receptor signaling through the hyper-activation of JNK in the hippocampus and frontal cortex in the brains of obese rats. Obesity was induced using a high fat diet (HFD). The Morris water maze test was then performed to evaluate decreases in cognitive function, and western blot was used to verify whether abnormal insulin receptor signaling was induced by ERS in HFD rats exhibiting cognitive decline. In addition, to determine whether ERS activated JNK and consequently impaired insulin receptor signaling, SH-SY5Y cells were treated with the JNK inhibitor, SP600125, followed by tunicamycin or thapsigargin, and primary rat hippocampal and cortical neurons were transfected with siRNA against IRE1α and JNK. We found that the expression of phosphorylation of PKR-like kinase (PERK), phosphorylation of α subunit of translation initiation factor 2 (eIF2α), and phosphorylation of inositol-requiring kinase-1α (IRE-1α) were increased in the brains of rats with HFD when compared with control rats. The level of serine phosphorylation of insulin receptor substrate-1 (IRS-1) was also increased, while protein kinase B (PKB/Akt) was reduced. ERS was also found to inhibit insulin receptor signaling via the activation of JNK in SH-SY5Y cells, primary rat hippocampal, and cortical neurons. These results indicate that ERS was increased, thereby resulting in impaired insulin receptor signaling in the hippocampus and frontal cortex of obese rats.
Collapse
|
15
|
Wang J, Liu B, Han H, Yuan Q, Xue M, Xu F, Chen Y. Acute Hepatic Insulin Resistance Contributes to Hyperglycemia in Rats Following Myocardial Infarction. Mol Med 2015; 21:68-76. [PMID: 25730774 DOI: 10.2119/molmed.2014.00240] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Accepted: 02/23/2015] [Indexed: 12/27/2022] Open
Abstract
Although hyperglycemia is common in patients with acute myocardial infarction (MI), the underlying mechanisms are largely unknown. Insulin signaling plays a key role in the regulation of glucose homeostasis. In this study, we test the hypothesis that rapid alteration of insulin signaling pathways could be a potential contributor to acute hyperglycemia after MI. Male rats were used to produce MI by ligation of the left anterior descending coronary artery. Plasma glucose and insulin levels were significantly higher in MI rats than those in controls. Insulin-stimulated tyrosine phosphorylation of insulin receptor substrate 1 (IRS1) was reduced significantly in the liver tissue of MI rats compared with controls, followed by decreased attachment of phosphatidylinositol 3-kinase (PI3K) p85 subunit with IRS1 and Akt phosphorylation. However, insulin-stimulated signaling was not altered significantly in skeletal muscle after MI. The relative mRNA levels of phosphoenolpyruvate carboxykinase (PEPCK) and G6Pase were slightly higher in the liver tissue of MI rats than those in controls. Rosiglitazone (ROSI) markedly restored hepatic insulin signaling, inhibited gluconeogenesis and reduced plasma glucose levels in MI rats. Insulin resistance develops rapidly in liver but not skeletal muscle after MI, which contributes to acute hyperglycemia. Therapy aimed at potentiating hepatic insulin signaling may be beneficial for MI-induced hyperglycemia.
Collapse
Affiliation(s)
- Jiali Wang
- Department of Emergency, Shandong University, Jinan, China.,Chest Pain Center, Shandong University, Jinan, China.,Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Shandong University, Jinan, China.,Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Qilu Hospital, Shandong University, Jinan, China
| | - Baoshan Liu
- Department of Emergency, Shandong University, Jinan, China.,Chest Pain Center, Shandong University, Jinan, China.,Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Shandong University, Jinan, China.,Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Qilu Hospital, Shandong University, Jinan, China
| | - Hui Han
- Department of Emergency, Shandong University, Jinan, China.,Chest Pain Center, Shandong University, Jinan, China
| | - Qiuhuan Yuan
- Department of Emergency, Shandong University, Jinan, China.,Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Shandong University, Jinan, China.,Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Qilu Hospital, Shandong University, Jinan, China
| | - Mengyang Xue
- Department of Emergency, Shandong University, Jinan, China.,Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Shandong University, Jinan, China.,Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Qilu Hospital, Shandong University, Jinan, China
| | - Feng Xu
- Department of Emergency, Shandong University, Jinan, China.,Chest Pain Center, Shandong University, Jinan, China.,Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Shandong University, Jinan, China.,Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Qilu Hospital, Shandong University, Jinan, China
| | - Yuguo Chen
- Department of Emergency, Shandong University, Jinan, China.,Chest Pain Center, Shandong University, Jinan, China.,Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Shandong University, Jinan, China.,Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Qilu Hospital, Shandong University, Jinan, China
| |
Collapse
|
16
|
Stuart CA, Howell MEA, Cartwright BM, McCurry MP, Lee ML, Ramsey MW, Stone MH. Insulin resistance and muscle insulin receptor substrate-1 serine hyperphosphorylation. Physiol Rep 2014; 2:2/12/e12236. [PMID: 25472611 PMCID: PMC4332214 DOI: 10.14814/phy2.12236] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Insulin resistance in metabolic syndrome subjects is profound in spite of muscle insulin receptor and insulin-responsive glucose transporter (GLUT4) expression being nearly normal. Insulin receptor tyrosine kinase phosphorylation of insulin receptor substrate-1 (IRS-1) at Tyr896 is a necessary step in insulin stimulation of translocation of GLUT4 to the cell surface. Serine phosphorylation of IRS-1 by some kinases diminishes insulin action in mice. We evaluated the phosphorylation status of muscle IRS-1 in 33 subjects with the metabolic syndrome and seventeen lean controls. Each underwent euglycemic insulin clamps and a thigh muscle biopsy before and after 8 weeks of either strength or endurance training. Muscle IRS-1 phosphorylation at six sites was quantified by immunoblots. Metabolic syndrome muscle IRS-1 had excess phosphorylation at Ser337 and Ser636 but not at Ser307, Ser789, or Ser1101. Ser337 is a target for phosphorylation by glycogen synthase kinase 3 (GSK3) and Ser636 is phosphorylated by c-Jun N-terminal kinase 1 (JNK1). Exercise training without weight loss did not change the IRS-1 serine phosphorylation. These data suggest that baseline hyperphosphorylation of at least two key serines within muscle IRS-1 diminishes the transmission of the insulin signal and thereby decreases the insulin-stimulated translocation of GLUT4. Excess fasting phosphorylation of muscle IRS-1 at Ser636 may be a major cause of the insulin resistance seen in obesity and might prevent improvement in insulin responsiveness when exercise training is not accompanied by weight loss.
Collapse
Affiliation(s)
- Charles A Stuart
- Department of Internal Medicine, Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee
| | - Mary E A Howell
- Department of Internal Medicine, Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee
| | - Brian M Cartwright
- Department of Internal Medicine, Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee
| | - Melanie P McCurry
- Department of Internal Medicine, Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee
| | - Michelle L Lee
- Department of Allied Health, College of Clinical and Rehabilitative Health, East Tennessee State University, Johnson City, Tennessee
| | - Michael W Ramsey
- Department of Exercise and Sports Science, Clemmer College of Education, East Tennessee State University, Johnson City, Tennessee
| | - Michael H Stone
- Department of Exercise and Sports Science, Clemmer College of Education, East Tennessee State University, Johnson City, Tennessee
| |
Collapse
|
17
|
Kapogiannis D, Boxer A, Schwartz JB, Abner EL, Biragyn A, Masharani U, Frassetto L, Petersen RC, Miller BL, Goetzl EJ. Dysfunctionally phosphorylated type 1 insulin receptor substrate in neural-derived blood exosomes of preclinical Alzheimer's disease. FASEB J 2014; 29:589-96. [PMID: 25342129 DOI: 10.1096/fj.14-262048] [Citation(s) in RCA: 263] [Impact Index Per Article: 23.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Insulin resistance causes diminished glucose uptake in similar regions of the brain in Alzheimer's disease (AD) and type 2 diabetes mellitus (DM2). Brain tissue studies suggested that insulin resistance is caused by low insulin receptor signaling attributable to its abnormal association with more phospho (P)-serine-type 1 insulin receptor substrate (IRS-1) and less P-tyrosine-IRS-1. Plasma exosomes enriched for neural sources by immunoabsorption were obtained once from 26 patients with AD, 20 patients with DM2, 16 patients with frontotemporal dementia (FTD), and matched case control subjects. At 2 time points, they were obtained from 22 others when cognitively normal and 1 to 10 yr later when diagnosed with AD. Mean exosomal levels of extracted P-serine 312-IRS-1 and P-pan-tyrosine-IRS-1 by ELISA and the ratio of P-serine 312-IRS-1 to P-pan-tyrosine-IRS-1 (insulin resistance factor, R) for AD and DM2 and P-serine 312-IRS-1 and R for FTD were significantly different from those for case control subjects. The levels of R for AD were significantly higher than those for DM2 or FTD. Stepwise discriminant modeling showed correct classification of 100% of patients with AD, 97.5% of patients with DM2, and 84% of patients with FTD. In longitudinal studies of 22 patients with AD, exosomal levels of P-serine 312-IRS-1, P-pan-tyrosine-IRS-1, and R were significantly different 1 to 10 yr before and at the time of diagnosis compared with control subjects. Insulin resistance reflected in R values from this blood test is higher for patients with AD, DM2, and FTD than case control subjects; higher for patients with AD than patients with DM2 or FTD; and accurately predicts development of AD up to 10 yr prior to clinical onset.
Collapse
Affiliation(s)
| | - Adam Boxer
- Memory and Aging Center, Department of Neurology, and
| | - Janice B Schwartz
- Jewish Home of San Francisco, San Francisco, California, USA; Department of Medicine, University of California-San Francisco Medical Center, San Francisco, California, USA
| | - Erin L Abner
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky, USA; and
| | - Arya Biragyn
- Intramural Research Program, National Institute on Aging, Baltimore, Maryland, USA
| | - Umesh Masharani
- Jewish Home of San Francisco, San Francisco, California, USA
| | - Lynda Frassetto
- Jewish Home of San Francisco, San Francisco, California, USA
| | | | | | - Edward J Goetzl
- Intramural Research Program, National Institute on Aging, Baltimore, Maryland, USA; Jewish Home of San Francisco, San Francisco, California, USA; Department of Medicine, University of California-San Francisco Medical Center, San Francisco, California, USA;
| |
Collapse
|
18
|
Iqbal Z, Hoessli DC, Qazi WM, Ahmad M, Shakoori AR, Nasir-ud-Din. Exploring the sequence context of phosphorylatable amino acids: the contribution of the upgraded MAPRes tool. J Cell Biochem 2014; 116:370-9. [PMID: 25258092 DOI: 10.1002/jcb.24983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2014] [Accepted: 09/19/2014] [Indexed: 11/10/2022]
Abstract
Several models that predict where post-translational modifications are likely to occur and formulate the corresponding association rules are available to analyze the functional potential of a protein sequence, but an algorithm incorporating the functional groups of the involved amino acids in the sequence analyses process is not yet available. In its previous version, MAPRes was utilized to investigate the influence of the surrounding amino acids of post- translationally and co-translationally modifiable sites. The MAPRes has been upgraded to take into account the different biophysical and biochemical properties of the amino acids that have the potential to influence different post- translational modifications (PTMs). In the present study, the upgraded version of MAPRes was implemented on phosphorylated Ser/Thr/Tyr data by considering the polarity and charge of the surrounding amino acids. The patterns mined by MAPRes incorporating structural information on polarity and charge of amino acids suggest distinct structure-function relationships for phosphorylated serines in a multifunctional protein such as the insulin-receptor substrate-1 (IRS-1) protein. The new version of MAPRes is freely available at http://www.imsb.edu.pk/Database.htm.
Collapse
Affiliation(s)
- Zeeshan Iqbal
- Institute of Molecular Sciences and Bioinformatics, Lahore, Pakistan
| | | | | | | | | | | |
Collapse
|
19
|
Lasram MM, Dhouib IB, Annabi A, El Fazaa S, Gharbi N. A review on the molecular mechanisms involved in insulin resistance induced by organophosphorus pesticides. Toxicology 2014; 322:1-13. [DOI: 10.1016/j.tox.2014.04.009] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2014] [Revised: 04/23/2014] [Accepted: 04/24/2014] [Indexed: 02/06/2023]
|
20
|
Blesson CS, Sathishkumar K, Chinnathambi V, Yallampalli C. Gestational protein restriction impairs insulin-regulated glucose transport mechanisms in gastrocnemius muscles of adult male offspring. Endocrinology 2014; 155:3036-46. [PMID: 24797633 PMCID: PMC4098002 DOI: 10.1210/en.2014-1094] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Type II diabetes originates from various genetic and environmental factors. Recent studies showed that an adverse uterine environment such as that caused by a gestational low-protein (LP) diet can cause insulin resistance in adult offspring. The mechanism of insulin resistance induced by gestational protein restriction is not clearly understood. Our aim was to investigate the role of insulin signaling molecules in gastrocnemius muscles of gestational LP diet-exposed male offspring to understand their role in LP-induced insulin resistance. Pregnant Wistar rats were fed a control (20% protein) or isocaloric LP (6%) diet from gestational day 4 until delivery and a normal diet after weaning. Only male offspring were used in this study. Glucose and insulin responses were assessed after a glucose tolerance test. mRNA and protein levels of molecules involved in insulin signaling were assessed at 4 months in gastrocnemius muscles. Muscles were incubated ex vivo with insulin to evaluate insulin-induced phosphorylation of insulin receptor (IR), Insulin receptor substrate-1, Akt, and AS160. LP diet-fed rats gained less weight than controls during pregnancy. Male pups from LP diet-fed mothers were smaller but exhibited catch-up growth. Plasma glucose and insulin levels were elevated in LP offspring when subjected to a glucose tolerance test; however, fasting levels were comparable. LP offspring showed increased expression of IR and AS160 in gastrocnemius muscles. Ex vivo treatment of muscles with insulin showed increased phosphorylation of IR (Tyr972) in controls, but LP rats showed higher basal phosphorylation. Phosphorylation of Insulin receptor substrate-1 (Tyr608, Tyr895, Ser307, and Ser318) and AS160 (Thr642) were defective in LP offspring. Further, glucose transporter type 4 translocation in LP offspring was also impaired. A gestational LP diet leads to insulin resistance in adult offspring by a mechanism involving inefficient insulin-induced IR, Insulin receptor substrate-1, and AS160 phosphorylation and impaired glucose transporter type 4 translocation.
Collapse
Affiliation(s)
- Chellakkan S Blesson
- Department of Obstetrics and Gynecology (C.S.B., C.Y.), Baylor College of Medicine, Houston, Texas 77030; and Division of Reproductive Endocrinology (K.S., V.C.), Department of Obstetrics and Gynecology, University of Texas Medical Branch, Galveston, Texas 77555
| | | | | | | |
Collapse
|
21
|
Caruso M, Ma D, Msallaty Z, Lewis M, Seyoum B, Al-janabi W, Diamond M, Abou-Samra AB, Højlund K, Tagett R, Draghici S, Zhang X, Horowitz JF, Yi Z. Increased interaction with insulin receptor substrate 1, a novel abnormality in insulin resistance and type 2 diabetes. Diabetes 2014; 63:1933-47. [PMID: 24584551 PMCID: PMC4030113 DOI: 10.2337/db13-1872] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Insulin receptor substrate 1 (IRS1) is a key mediator of insulin signal transduction. Perturbations involving IRS1 complexes may lead to the development of insulin resistance and type 2 diabetes (T2D). Surprisingly little is known about the proteins that interact with IRS1 in humans under health and disease conditions. We used a proteomic approach to assess IRS1 interaction partners in skeletal muscle from lean healthy control subjects (LCs), obese insulin-resistant nondiabetic control subjects (OCs), and participants with T2D before and after insulin infusion. We identified 113 novel endogenous IRS1 interaction partners, which represents the largest IRS1 interactome in humans and provides new targets for studies of IRS1 complexes in various diseases. Furthermore, we generated the first global picture of IRS1 interaction partners in LCs, and how they differ in OCs and T2D patients. Interestingly, dozens of proteins in OCs and/or T2D patients exhibited increased associations with IRS1 compared with LCs under the basal and/or insulin-stimulated conditions, revealing multiple new dysfunctional IRS1 pathways in OCs and T2D patients. This novel abnormality, increased interaction of multiple proteins with IRS1 in obesity and T2D in humans, provides new insights into the molecular mechanism of insulin resistance and identifies new targets for T2D drug development.
Collapse
Affiliation(s)
- Michael Caruso
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy/Health Sciences, Wayne State University, Detroit, MI
| | - Danjun Ma
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy/Health Sciences, Wayne State University, Detroit, MI
| | - Zaher Msallaty
- Division of Endocrinology, Wayne State University School of Medicine, Wayne State University, Detroit, MI
| | - Monique Lewis
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy/Health Sciences, Wayne State University, Detroit, MI
| | - Berhane Seyoum
- Division of Endocrinology, Wayne State University School of Medicine, Wayne State University, Detroit, MI
| | - Wissam Al-janabi
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy/Health Sciences, Wayne State University, Detroit, MI
| | - Michael Diamond
- Department of Obstetrics and Gynecology, Wayne State University, Detroit, MIDepartment of Obstetrics and Gynecology, Georgia Regents University, Augusta, GA
| | - Abdul B Abou-Samra
- Division of Endocrinology, Wayne State University School of Medicine, Wayne State University, Detroit, MIDepartment of Medicine, Hamad Medical Corporation, Doha, Qatar
| | - Kurt Højlund
- Diabetes Research Centre, Department of Endocrinology, Odense University Hospital, Odense, Denmark
| | - Rebecca Tagett
- Department of Computer Science, College of Engineering, Wayne State University, Detroit, MI
| | - Sorin Draghici
- Department of Computer Science, College of Engineering, Wayne State University, Detroit, MI
| | - Xiangmin Zhang
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy/Health Sciences, Wayne State University, Detroit, MI
| | | | - Zhengping Yi
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy/Health Sciences, Wayne State University, Detroit, MI
| |
Collapse
|
22
|
Hançer NJ, Qiu W, Cherella C, Li Y, Copps KD, White MF. Insulin and metabolic stress stimulate multisite serine/threonine phosphorylation of insulin receptor substrate 1 and inhibit tyrosine phosphorylation. J Biol Chem 2014; 289:12467-84. [PMID: 24652289 PMCID: PMC4007441 DOI: 10.1074/jbc.m114.554162] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2014] [Revised: 03/18/2014] [Indexed: 12/27/2022] Open
Abstract
IRS1 and IRS2 are key substrates of the insulin receptor tyrosine kinase. Mass spectrometry reveals more than 50 phosphorylated IRS1 serine and threonine residues (Ser(P)/Thr(P) residues) in IRS1 from insulin-stimulated cells or human tissues. We investigated a subset of IRS1 Ser(P)/Thr(P) residues using a newly developed panel of 25 phospho-specific monoclonal antibodies (αpS/TmAb(Irs1)). CHO cells overexpressing the human insulin receptor and rat IRS1 were stimulated with insulin in the absence or presence of inhibitors of the PI3K → Akt → mechanistic target of rapamycin (mTOR) → S6 kinase or MEK pathways. Nearly all IRS1 Ser(P)/Thr(P) residues were stimulated by insulin and significantly suppressed by PI3K inhibition; fewer were suppressed by Akt or mTOR inhibition, and none were suppressed by MEK inhibition. Insulin-stimulated Irs1 tyrosine phosphorylation (Tyr(P)(Irs1)) was enhanced by inhibition of the PI3K → Akt → mTOR pathway and correlated with decreased Ser(P)-302(Irs1), Ser(P)-307(Irs1), Ser(P)-318(Irs1), Ser(P)-325(Irs1), and Ser(P)-346(Irs1). Metabolic stress modeled by anisomycin, thapsigargin, or tunicamycin increased many of the same Ser(P)/Thr(P) residues as insulin, some of which (Ser(P)-302(Irs1), Ser(P)-307(Irs1), and four others) correlated significantly with impaired insulin-stimulated Tyr(P)(Irs1). Thus, IRS1 Ser(P)/Thr(P) is an integrated response to insulin stimulation and metabolic stress, which associates with reduced Tyr(P)(Irs1) in CHO(IR)/IRS1 cells.
Collapse
Affiliation(s)
- Nancy J. Hançer
- From the Division of Endocrinology, Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts 02115
| | - Wei Qiu
- From the Division of Endocrinology, Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts 02115
| | - Christine Cherella
- From the Division of Endocrinology, Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts 02115
| | - Yedan Li
- From the Division of Endocrinology, Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts 02115
| | - Kyle D. Copps
- From the Division of Endocrinology, Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts 02115
| | - Morris F. White
- From the Division of Endocrinology, Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts 02115
| |
Collapse
|
23
|
Matos MAD, Ottone VDO, Duarte TC, Sampaio PFDM, Costa KB, Fonseca CA, Neves MPC, Schneider SM, Moseley P, Coimbra CC, Magalhães FDC, Rocha-Vieira E, Amorim FT. Exercise reduces cellular stress related to skeletal muscle insulin resistance. Cell Stress Chaperones 2014; 19:263-70. [PMID: 23975543 PMCID: PMC3933613 DOI: 10.1007/s12192-013-0453-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2013] [Revised: 08/02/2013] [Accepted: 08/05/2013] [Indexed: 01/09/2023] Open
Abstract
This study sought to evaluate the effects of a single session of exercise on the expression of Hsp70, of c-jun N-terminal kinase (JNK), and insulin receptor substrate 1 serine 612 (IRS(ser612)) phosphorylation in the skeletal muscle of obese and obese insulin-resistant patients. Twenty-seven volunteers were divided into three experimental groups (eutrophic insulin-sensitive, obese insulin-sensitive, and obese insulin-resistant) according to their body mass index and the presence of insulin resistance. The volunteers performed 60 min of aerobic exercise on a cycle ergometer at 60 % of peak oxygen consumption. M. vastus lateralis samples were obtained before and after exercise. The protein expressions were evaluated by Western blot. Our findings show that compared with paired eutrophic controls, obese subjects have higher basal levels of p-JNK (100 ± 23 % vs. 227 ± 67 %, p = 0.03) and p-IRS-1(ser612) (100 ± 23 % vs. 340 ± 67 %, p < 0.001) and reduced HSP70 (100 ± 16 % vs. 63 ± 12 %, p < 0.001). The presence of insulin resistance results in a further increase in p-JNK (460 ± 107 %, p < 0.001) and a decrease in Hsp70 (46 ± 5 %, p = 0.006), but p-IRS-1(ser612) levels did not differ from obese subjects (312 ± 73 %, p > 0.05). Exercise reduced p-JNK in obese insulin-resistant subjects (328 ± 33 %, p = 0.001), but not in controls or obese subjects. Furthermore, exercise reduced p-IRS-1(ser612) for both obese (122 ± 44 %) and obese insulin-resistant (185 ± 36 %) subjects. A main effect of exercise was observed in HSP70 (p = 0.007). We demonstrated that a single session of exercise promotes changes that characterize a reduction in cellular stress that may contribute to exercise-induced increase in insulin sensitivity.
Collapse
Affiliation(s)
- Mariana Aguiar de Matos
- />Exercise Physiology Laboratory, Healthy and Biological Sciences Faculty, Federal University of Jequitinhonha and Mucuri Valleys, Rua da Glória 187, Diamantina, 39100-100 Minas Gerais Brazil
| | - Vinícius de Oliveira Ottone
- />Exercise Physiology Laboratory, Healthy and Biological Sciences Faculty, Federal University of Jequitinhonha and Mucuri Valleys, Rua da Glória 187, Diamantina, 39100-100 Minas Gerais Brazil
| | - Tamiris Campos Duarte
- />Exercise Physiology Laboratory, Healthy and Biological Sciences Faculty, Federal University of Jequitinhonha and Mucuri Valleys, Rua da Glória 187, Diamantina, 39100-100 Minas Gerais Brazil
| | - Pâmela Fiche da Matta Sampaio
- />Exercise Physiology Laboratory, Healthy and Biological Sciences Faculty, Federal University of Jequitinhonha and Mucuri Valleys, Rua da Glória 187, Diamantina, 39100-100 Minas Gerais Brazil
| | - Karine Beatriz Costa
- />Exercise Physiology Laboratory, Healthy and Biological Sciences Faculty, Federal University of Jequitinhonha and Mucuri Valleys, Rua da Glória 187, Diamantina, 39100-100 Minas Gerais Brazil
| | - Cheyenne Alves Fonseca
- />Exercise Physiology Laboratory, Healthy and Biological Sciences Faculty, Federal University of Jequitinhonha and Mucuri Valleys, Rua da Glória 187, Diamantina, 39100-100 Minas Gerais Brazil
| | - Miguel Pontes Correa Neves
- />Exercise Physiology Laboratory, Healthy and Biological Sciences Faculty, Federal University of Jequitinhonha and Mucuri Valleys, Rua da Glória 187, Diamantina, 39100-100 Minas Gerais Brazil
| | | | | | - Cândido Celso Coimbra
- />Endocrinology Laboratory, Biological Sciences Institute, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Flávio de Castro Magalhães
- />Exercise Physiology Laboratory, Healthy and Biological Sciences Faculty, Federal University of Jequitinhonha and Mucuri Valleys, Rua da Glória 187, Diamantina, 39100-100 Minas Gerais Brazil
| | - Etel Rocha-Vieira
- />Exercise Physiology Laboratory, Healthy and Biological Sciences Faculty, Federal University of Jequitinhonha and Mucuri Valleys, Rua da Glória 187, Diamantina, 39100-100 Minas Gerais Brazil
| | - Fabiano Trigueiro Amorim
- />Exercise Physiology Laboratory, Healthy and Biological Sciences Faculty, Federal University of Jequitinhonha and Mucuri Valleys, Rua da Glória 187, Diamantina, 39100-100 Minas Gerais Brazil
| |
Collapse
|
24
|
Boucher J, Kleinridders A, Kahn CR. Insulin receptor signaling in normal and insulin-resistant states. Cold Spring Harb Perspect Biol 2014; 6:6/1/a009191. [PMID: 24384568 DOI: 10.1101/cshperspect.a009191] [Citation(s) in RCA: 971] [Impact Index Per Article: 88.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
In the wake of the worldwide increase in type-2 diabetes, a major focus of research is understanding the signaling pathways impacting this disease. Insulin signaling regulates glucose, lipid, and energy homeostasis, predominantly via action on liver, skeletal muscle, and adipose tissue. Precise modulation of this pathway is vital for adaption as the individual moves from the fed to the fasted state. The positive and negative modulators acting on different steps of the signaling pathway, as well as the diversity of protein isoform interaction, ensure a proper and coordinated biological response to insulin in different tissues. Whereas genetic mutations are causes of rare and severe insulin resistance, obesity can lead to insulin resistance through a variety of mechanisms. Understanding these pathways is essential for development of new drugs to treat diabetes, metabolic syndrome, and their complications.
Collapse
Affiliation(s)
- Jérémie Boucher
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center and Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts 02115
| | | | | |
Collapse
|
25
|
O'Neill HM, Holloway GP, Steinberg GR. AMPK regulation of fatty acid metabolism and mitochondrial biogenesis: implications for obesity. Mol Cell Endocrinol 2013; 366:135-51. [PMID: 22750049 DOI: 10.1016/j.mce.2012.06.019] [Citation(s) in RCA: 241] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2011] [Revised: 03/13/2012] [Accepted: 06/21/2012] [Indexed: 12/25/2022]
Abstract
Skeletal muscle plays an important role in regulating whole-body energy expenditure given it is a major site for glucose and lipid oxidation. Obesity and type 2 diabetes are causally linked through their association with skeletal muscle insulin resistance, while conversely exercise is known to improve whole body glucose homeostasis simultaneously with muscle insulin sensitivity. Exercise activates skeletal muscle AMP-activated protein kinase (AMPK). AMPK plays a role in regulating exercise capacity, skeletal muscle mitochondrial content and contraction-stimulated glucose uptake. Skeletal muscle AMPK is also thought to be important for regulating fatty acid metabolism; however, direct genetic evidence in this area is currently lacking. This review will discuss the current paradigms regarding the influence of AMPK in regulating skeletal muscle fatty acid metabolism and mitochondrial biogenesis at rest and during exercise, and highlight the potential implications in the development of insulin resistance.
Collapse
Affiliation(s)
- Hayley M O'Neill
- University of Melbourne, Department of Medicine, St. Vincent's Institute of Medical Research, Melbourne, Victoria, Australia.
| | | | | |
Collapse
|
26
|
High-fat load: mechanism(s) of insulin resistance in skeletal muscle. INTERNATIONAL JOURNAL OF OBESITY SUPPLEMENTS 2012; 2:S31-S36. [PMID: 26052434 DOI: 10.1038/ijosup.2012.20] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Skeletal muscle from sedentary obese patients is characterized by depressed electron transport activity, reduced expression of genes required for oxidative metabolism, altered mitochondrial morphology and lower overall mitochondrial content. These findings imply that obesity, or more likely the metabolic imbalance that causes obesity, leads to a progressive decline in mitochondrial function, eventually culminating in mitochondrial dissolution or mitoptosis. A decrease in the sensitivity of skeletal muscle to insulin represents one of the earliest maladies associated with high dietary fat intake and weight gain. Considerable evidence has accumulated to suggest that the cytosolic ectopic accumulation of fatty acid metabolites, including diacylglycerol and ceramides, underlies the development of insulin resistance in skeletal muscle. However, an alternative mechanism has recently been evolving, which places the etiology of insulin resistance in the context of cellular/mitochondrial bioenergetics and redox systems biology. Overnutrition, particularly from high-fat diets, generates fuel overload within the mitochondria, resulting in the accumulation of partially oxidized acylcarnitines, increased mitochondrial hydrogen peroxide (H2O2) emission and a shift to a more oxidized intracellular redox environment. Blocking H2O2 emission prevents the shift in redox environment and preserves insulin sensitivity, providing evidence that the mitochondrial respiratory system is able to sense and respond to cellular metabolic imbalance. Mitochondrial H2O2 emission is a major regulator of protein redox state, as well as the overall cellular redox environment, raising the intriguing possibility that elevated H2O2 emission from nutrient overload may represent the underlying basis for the development of insulin resistance due to disruption of normal redox control mechanisms regulating protein function, including the insulin signaling and glucose transport processes.
Collapse
|
27
|
Copps KD, White MF. Regulation of insulin sensitivity by serine/threonine phosphorylation of insulin receptor substrate proteins IRS1 and IRS2. Diabetologia 2012; 55:2565-2582. [PMID: 22869320 PMCID: PMC4011499 DOI: 10.1007/s00125-012-2644-8] [Citation(s) in RCA: 718] [Impact Index Per Article: 55.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2012] [Accepted: 04/23/2012] [Indexed: 12/11/2022]
Abstract
The insulin receptor substrate proteins IRS1 and IRS2 are key targets of the insulin receptor tyrosine kinase and are required for hormonal control of metabolism. Tissues from insulin-resistant and diabetic humans exhibit defects in IRS-dependent signalling, implicating their dysregulation in the initiation and progression of metabolic disease. However, IRS1 and IRS2 are regulated through a complex mechanism involving phosphorylation of >50 serine/threonine residues (S/T) within their long, unstructured tail regions. In cultured cells, insulin-stimulated kinases (including atypical PKC, AKT, SIK2, mTOR, S6K1, ERK1/2 and ROCK1) mediate feedback (autologous) S/T phosphorylation of IRS, with both positive and negative effects on insulin sensitivity. Additionally, insulin-independent (heterologous) kinases can phosphorylate IRS1/2 under basal conditions (AMPK, GSK3) or in response to sympathetic activation and lipid/inflammatory mediators, which are present at elevated levels in metabolic disease (GRK2, novel and conventional PKCs, JNK, IKKβ, mPLK). An emerging view is that the positive/negative regulation of IRS by autologous pathways is subverted/co-opted in disease by increased basal and other temporally inappropriate S/T phosphorylation. Compensatory hyperinsulinaemia may contribute strongly to this dysregulation. Here, we examine the links between altered patterns of IRS S/T phosphorylation and the emergence of insulin resistance and diabetes.
Collapse
Affiliation(s)
- K D Copps
- Howard Hughes Medical Institute, Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, CLS 16020, 300 Longwood Avenue, Boston, MA, 02115, USA
| | - M F White
- Howard Hughes Medical Institute, Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, CLS 16020, 300 Longwood Avenue, Boston, MA, 02115, USA.
| |
Collapse
|
28
|
Pham K, Langlais P, Zhang X, Chao A, Zingsheim M, Yi Z. Insulin-stimulated phosphorylation of protein phosphatase 1 regulatory subunit 12B revealed by HPLC-ESI-MS/MS. Proteome Sci 2012; 10:52. [PMID: 22937917 PMCID: PMC3546068 DOI: 10.1186/1477-5956-10-52] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2012] [Accepted: 07/31/2012] [Indexed: 11/10/2022] Open
Abstract
UNLABELLED BACKGROUND Protein phosphatase 1 (PP1) is one of the major phosphatases responsible for protein dephosphorylation in eukaryotes. Protein phosphatase 1 regulatory subunit 12B (PPP1R12B), one of the regulatory subunits of PP1, can bind to PP1cδ, one of the catalytic subunits of PP1, and modulate the specificity and activity of PP1cδ against its substrates. Phosphorylation of PPP1R12B on threonine 646 by Rho kinase inhibits the activity of the PP1c-PPP1R12B complex. However, it is not currently known whether PPP1R12B phosphorylation at threonine 646 and other sites is regulated by insulin. We set out to identify phosphorylation sites in PPP1R12B and to quantify the effect of insulin on PPP1R12B phosphorylation by using high-performance liquid chromatography-electrospray ionization-tandem mass spectrometry. RESULTS 14 PPP1R12B phosphorylation sites were identified, 7 of which were previously unreported. Potential kinases were predicted for these sites. Furthermore, relative quantification of PPP1R12B phosphorylation sites for basal and insulin-treated samples was obtained by using peak area-based label-free mass spectrometry of fragment ions. The results indicate that insulin stimulates the phosphorylation of PPP1R12B significantly at serine 29 (3.02 ± 0.94 fold), serine 504 (11.67 ± 3.33 fold), and serine 645/threonine 646 (2.34 ± 0.58 fold). CONCLUSION PPP1R12B was identified as a phosphatase subunit that undergoes insulin-stimulated phosphorylation, suggesting that PPP1R12B might play a role in insulin signaling. This study also identified novel targets for future investigation of the regulation of PPP1R12B not only in insulin signaling in cell models, animal models, and in humans, but also in other signaling pathways.
Collapse
Affiliation(s)
- Kimberly Pham
- Center for Metabolic and Vascular Biology, Arizona State University, Tempe, AZ, USA
| | - Paul Langlais
- Center for Metabolic and Vascular Biology, Arizona State University, Tempe, AZ, USA
| | - Xiangmin Zhang
- Center for Metabolic and Vascular Biology, Arizona State University, Tempe, AZ, USA.,Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy/Health Sciences, Wayne State University, 259 Mack Ave., Detroit, MI, USA
| | - Alex Chao
- Center for Metabolic and Vascular Biology, Arizona State University, Tempe, AZ, USA
| | - Morgan Zingsheim
- Center for Metabolic and Vascular Biology, Arizona State University, Tempe, AZ, USA
| | - Zhengping Yi
- Center for Metabolic and Vascular Biology, Arizona State University, Tempe, AZ, USA.,Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy/Health Sciences, Wayne State University, 259 Mack Ave., Detroit, MI, USA
| |
Collapse
|
29
|
Identification of the amino acids 300-600 of IRS-2 as 14-3-3 binding region with the importance of IGF-1/insulin-regulated phosphorylation of Ser-573. PLoS One 2012; 7:e43296. [PMID: 22912850 PMCID: PMC3422239 DOI: 10.1371/journal.pone.0043296] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2012] [Accepted: 07/19/2012] [Indexed: 11/20/2022] Open
Abstract
Phosphorylation of insulin receptor substrate (IRS)-2 on tyrosine residues is a key event in IGF-1/insulin signaling and leads to activation of the PI 3-kinase and the Ras/MAPK pathway. Furthermore, phosphorylated serine/threonine residues on IRS-2 can induce 14-3-3 binding. In this study we searched IRS-2 for novel phosphorylation sites and investigated the interaction between IRS-2 and 14-3-3. Mass spectrometry identified a total of 24 serine/threonine residues on IRS-2 with 12 sites unique for IRS-2 while the other residues are conserved in IRS-1 and IRS-2. IGF-1 stimulation led to increased binding of 14-3-3 to IRS-2 in transfected HEK293 cells and this binding was prevented by inhibition of the PI 3-kinase pathway and an Akt/PKB inhibitor. Insulin-stimulated interaction between endogenous IRS-2 and 14-3-3 was observed in rat hepatoma cells and in mice liver after an acute insulin stimulus and refeeding. Using different IRS-2 fragments enabled localization of the IGF-1-dependent 14-3-3 binding region spanning amino acids 300–600. The 24 identified residues on IRS-2 included several 14-3-3 binding candidates in the region 300–600. Single alanine mutants of these candidates led to the identification of serine 573 as 14-3-3 binding site. A phospho-site specific antibody was generated to further characterize serine 573. IGF-1-dependent phosphorylation of serine 573 was reduced by inhibition of PI 3-kinase and Akt/PKB. A negative role of this phosphorylation site was implicated by the alanine mutant of serine 573 which led to enhanced phosphorylation of Akt/PKB in an IGF-1 time course experiment. To conclude, our data suggest a physiologically relevant role for IGF-1/insulin-dependent 14-3-3 binding to IRS-2 involving serine 573.
Collapse
|
30
|
Chao A, Zhang X, Ma D, Langlais P, Luo M, Mandarino LJ, Zingsheim M, Pham K, Dillon J, Yi Z. Site-specific phosphorylation of protein phosphatase 1 regulatory subunit 12A stimulated or suppressed by insulin. J Proteomics 2012; 75:3342-50. [PMID: 22516431 DOI: 10.1016/j.jprot.2012.03.043] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2011] [Revised: 03/08/2012] [Accepted: 03/26/2012] [Indexed: 10/28/2022]
Abstract
Protein phosphatase 1 (PP1) is one of the major phosphatases responsible for protein dephosphorylation in eukaryotes. So far, only few specific phosphorylation sites of PP1 regulatory subunit 12A (PPP1R12A) have been shown to regulate the PP1 activity. The effect of insulin on PPP1R12A phosphorylation is largely unknown. Utilizing a mass spectrometry based phosphorylation identification and quantification approach, we identified 21 PPP1R12A phosphorylation sites (7 novel sites, including Ser20, Thr22, Thr453, Ser478, Thr671, Ser678, and Ser680) and quantified 16 of them under basal and insulin stimulated conditions in hamster ovary cells overexpressing the insulin receptor (CHO/IR), an insulin sensitive cell model. Insulin stimulated the phosphorylation of PPP1R12A significantly at Ser477, Ser478, Ser507, Ser668, and Ser695, while simultaneously suppressing the phosphorylation of PPP1R12A at Ser509 (more than 2-fold increase or decrease compared to basal). Our data demonstrate that PPP1R12A undergoes insulin stimulated/suppressed phosphorylation, suggesting that PPP1R12A phosphorylation may play a role in insulin signal transduction. The novel PPP1R12A phosphorylation sites as well as the new insulin-responsive phosphorylation sites of PPP1R12A in CHO/IR cells provide targets for investigation of the regulation of PPP1R12A and the PPP1R12A-PP1cδ complex in insulin action and other signaling pathways in other cell models, animal models, and humans.
Collapse
Affiliation(s)
- Alex Chao
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy/Health Sciences, Wayne State University, Detroit, MI, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Hepatitis C virus activates the mTOR/S6K1 signaling pathway in inhibiting IRS-1 function for insulin resistance. J Virol 2012; 86:6315-22. [PMID: 22457523 DOI: 10.1128/jvi.00050-12] [Citation(s) in RCA: 101] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Hepatitis C virus (HCV) infection significantly increases the prevalence of type 2 diabetes mellitus (T2DM). Insulin receptor substrate 1 (IRS-1) plays a key role in insulin signaling, thus enabling metabolic regulation in mammalian cells. We have previously shown that HCV infection modulates phosphorylation of Akt, a downstream target of IRS-1. In this study, we further examined the status of total IRS-1 and the downstream regulation of the Akt pathway in understanding mTOR/S6K1 signaling using HCV genotype 2a (clone JFH1)-infected hepatocytes. Inhibition of IRS-1 expression was observed in HCV-infected hepatocytes compared to that in a mock-infected control. The status of the tuberous sclerosis complex (TSC-1/TSC-2) was significantly decreased after HCV infection of human hepatocytes, showing a modulation of the downstream Akt pathway. Subsequent study indicated an increased level of Rheb and mTOR expression in HCV-infected hepatocytes. Interestingly, the phosphoS6K1 level was higher in HCV-infected hepatocytes, suggesting a novel mechanism for IRS-1 inhibition. Ectopic expression of TSC-1/TSC-2 significantly recovered the IRS-1 protein expression level in HCV-infected hepatocytes. Further analyses indicated that HCV core protein plays a significant role in modulating the mTOR/S6K1 signaling pathway. Proteasome inhibitor MG 132 recovered IRS-1 and TSC1/2 expression, suggesting that degradation occurred via the ubiquitin proteasome pathway. A functional consequence of IRS-1 inhibition was reflected in a decrease in GLUT4 protein expression and upregulation of the gluconeogenic enzyme PCK2 in HCV-infected hepatocytes. Together, these observations suggested that HCV infection activates the mTOR/S6K1 pathway in inhibiting IRS-1 function and perturbs glucose metabolism via downregulation of GLUT4 and upregulation of PCK2 for insulin resistance.
Collapse
|
32
|
Langlais P, Yi Z, Finlayson J, Luo M, Mapes R, De Filippis E, Meyer C, Plummer E, Tongchinsub P, Mattern M, Mandarino LJ. Global IRS-1 phosphorylation analysis in insulin resistance. Diabetologia 2011; 54:2878-89. [PMID: 21850561 PMCID: PMC3882165 DOI: 10.1007/s00125-011-2271-9] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2011] [Accepted: 07/08/2011] [Indexed: 01/25/2023]
Abstract
AIMS/HYPOTHESIS IRS-1 serine phosphorylation is often elevated in insulin resistance models, but confirmation in vivo in humans is lacking. We therefore analysed IRS-1 phosphorylation in human muscle in vivo. METHODS We used HPLC-electrospray ionisation (ESI)-MS/MS to quantify IRS-1 phosphorylation basally and after insulin infusion in vastus lateralis muscle from lean healthy, obese non-diabetic and type 2 diabetic volunteers. RESULTS Basal Ser323 phosphorylation was increased in type 2 diabetic patients (2.1 ± 0.43, p ≤ 0.05, fold change vs lean controls). Thr495 phosphorylation was decreased in type 2 diabetic patients (p ≤ 0.05). Insulin increased IRS-1 phosphorylation at Ser527 (1.4 ± 0.17, p ≤ 0.01, fold change, 60 min after insulin infusion vs basal) and Ser531 (1.3 ± 0.16, p ≤ 0.01, fold change, 60 min after insulin infusion vs basal) in the lean controls and suppressed phosphorylation at Ser348 (0.56 ± 0.11, p ≤ 0.01, fold change, 240 min after insulin infusion vs basal), Thr446 (0.64 ± 0.16, p ≤ 0.05, fold change, 60 min after insulin infusion vs basal), Ser1100 (0.77 ± 0.22, p ≤ 0.05, fold change, 240 min after insulin infusion vs basal) and Ser1142 (1.3 ± 0.2, p ≤ 0.05, fold change, 60 min after insulin infusion vs basal). CONCLUSIONS/INTERPRETATION We conclude that, unlike some aspects of insulin signalling, the ability of insulin to increase or suppress certain IRS-1 phosphorylation sites is intact in insulin resistance. However, some IRS-1 phosphorylation sites do not respond to insulin, whereas other Ser/Thr phosphorylation sites are either increased or decreased in insulin resistance.
Collapse
Affiliation(s)
- P. Langlais
- Center for Metabolic and Vascular Biology, School of Life Science, Arizona State University, ISTB1, 550 E. Orange St, Tempe, AZ 85287, USA
| | - Z. Yi
- Center for Metabolic and Vascular Biology, School of Life Science, Arizona State University, ISTB1, 550 E. Orange St, Tempe, AZ 85287, USA; Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy/Health Sciences, Wayne State University, Detroit, MI, USA
| | - J. Finlayson
- Center for Metabolic and Vascular Biology, School of Life Science, Arizona State wwUniversity, ISTB1, 550 E. Orange St, Tempe, AZ 85287, USA
| | - M. Luo
- Center for Metabolic and Vascular Biology, School of Life Science, Arizona State University, ISTB1, 550 E. Orange St, Tempe, AZ 85287, USA
| | - R. Mapes
- Center for Metabolic and Vascular Biology, School of Life Science, Arizona State University, ISTB1, 550 E. Orange St, Tempe, AZ 85287, USA
| | - E. De Filippis
- Center for Metabolic and Vascular Biology, School of Life Science, Arizona State University, ISTB1, 550 E. Orange St, Tempe, AZ 85287, USA
| | - C. Meyer
- Center for Metabolic and Vascular Biology, School of Life Science, Arizona State University, ISTB1, 550 E. Orange St, Tempe, AZ 85287, USA; Division of Endocrinology, Carl T. Hayden VA Medical Center, Mayo Clinic in Arizona, Phoenix, AZ, USA
| | - E. Plummer
- Division of Endocrinology, Carl T. Hayden VA Medical Center, Mayo Clinic in Arizona, Phoenix, AZ, USA
| | - P. Tongchinsub
- Center for Metabolic and Vascular Biology, School of Life Science, Arizona State University, ISTB1, 550 E. Orange St, Tempe, AZ 85287, USA
| | - M. Mattern
- Center for Metabolic and Vascular Biology, School of Life Science, Arizona State University, ISTB1, 550 E. Orange St, Tempe, AZ 85287, USA
| | - L. J. Mandarino
- Center for Metabolic and Vascular Biology, School of Life Science, Arizona State University, ISTB1, 550 E. Orange St, Tempe, AZ 85287, USA; Department of Medicine, Mayo Clinic in Arizona, Scottsdale, AZ, USA
| |
Collapse
|
33
|
Everman S, Yi Z, Langlais P, Mandarino LJ, Luo M, Roberts C, Katsanos CS. Reproducibility of an HPLC-ESI-MS/MS method for the measurement of stable-isotope enrichment of in vivo-labeled muscle ATP synthase beta subunit. PLoS One 2011; 6:e26171. [PMID: 22022551 PMCID: PMC3192170 DOI: 10.1371/journal.pone.0026171] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2011] [Accepted: 09/21/2011] [Indexed: 01/06/2023] Open
Abstract
We sought to evaluate the reproducibility of a liquid chromatography-tandem mass spectrometry (LC-MS/MS)-based approach to measure the stable-isotope enrichment of in vivo-labeled muscle ATP synthase β subunit (β-F1-ATPase), a protein most directly involved in ATP production, and whose abundance is reduced under a variety of circumstances. Muscle was obtained from a rat infused with stable-isotope-labeled leucine. The muscle was homogenized, β-F1-ATPase immunoprecipitated, and the protein was resolved using 1D-SDS PAGE. Following trypsin digestion of the isolated protein, the resultant peptide mixtures were subjected to analysis by HPLC-ESI-MS/MS, which resulted in the detection of multiple β-F1-ATPase peptides. There were three β-F1-ATPase unique peptides with a leucine residue in the amino acid sequence, and which were detected with high intensity relative to other peptides and assigned with >95% probability to β-F1-ATPase. These peptides were specifically targeted for fragmentation to access their stable-isotope enrichment based on MS/MS peak areas calculated from extracted ion chromatographs for selected labeled and unlabeled fragment ions. Results showed best linearity (R(2) = 0.99) in the detection of MS/MS peak areas for both labeled and unlabeled fragment ions, over a wide range of amounts of injected protein, specifically for the β-F1-ATPase(134-143) peptide. Measured stable-isotope enrichment was highly reproducible for the β-F1-ATPase(134-143) peptide (CV = 2.9%). Further, using mixtures of synthetic labeled and unlabeled peptides we determined that there is an excellent linear relationship (R(2) = 0.99) between measured and predicted enrichment for percent enrichments ranging between 0.009% and 8.185% for the β-F1-ATPase(134-143) peptide. The described approach provides a reliable approach to measure the stable-isotope enrichment of in-vivo-labeled muscle β-F1-ATPase based on the determination of the enrichment of the β-F1-ATPase(134-143) peptide.
Collapse
Affiliation(s)
- Sarah Everman
- Center for Metabolic and Vascular Biology, School of Life Sciences, Arizona State University, Tempe, Arizona, United States of America
- Mayo Clinic Arizona, Scottsdale, Arizona, United States of America
| | - Zhengping Yi
- Center for Metabolic and Vascular Biology, School of Life Sciences, Arizona State University, Tempe, Arizona, United States of America
- Mayo Clinic Arizona, Scottsdale, Arizona, United States of America
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy/Health Sciences, Wayne State University, Detroit, Michigan, United States of America
| | - Paul Langlais
- Center for Metabolic and Vascular Biology, School of Life Sciences, Arizona State University, Tempe, Arizona, United States of America
- Mayo Clinic Arizona, Scottsdale, Arizona, United States of America
| | - Lawrence J. Mandarino
- Center for Metabolic and Vascular Biology, School of Life Sciences, Arizona State University, Tempe, Arizona, United States of America
- Mayo Clinic Arizona, Scottsdale, Arizona, United States of America
| | - Moulun Luo
- Center for Metabolic and Vascular Biology, School of Life Sciences, Arizona State University, Tempe, Arizona, United States of America
- Mayo Clinic Arizona, Scottsdale, Arizona, United States of America
| | - Christine Roberts
- Center for Metabolic and Vascular Biology, School of Life Sciences, Arizona State University, Tempe, Arizona, United States of America
- Mayo Clinic Arizona, Scottsdale, Arizona, United States of America
| | - Christos S. Katsanos
- Center for Metabolic and Vascular Biology, School of Life Sciences, Arizona State University, Tempe, Arizona, United States of America
- Mayo Clinic Arizona, Scottsdale, Arizona, United States of America
- * E-mail:
| |
Collapse
|
34
|
Hung CH, Lee CM, Lu SN. Hepatitis C virus-associated insulin resistance: pathogenic mechanisms and clinical implications. Expert Rev Anti Infect Ther 2011; 9:525-33. [PMID: 21609264 DOI: 10.1586/eri.11.33] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
It is now widely recognized that chronic hepatitis C is a metabolic disease, strongly associated with Type 2 diabetic mellitus and insulin resistance (IR). Chronic hepatitis C virus (HCV) infection promotes IR mainly through interfering with the insulin signaling pathway in hepatocytes, increasing the inflammatory response with production of cytokines such as TNF-α and IL-6, and increasing oxidative stress. Accumulated evidence indicates that HCV-associated IR may lead to fibrosis progression, resistance to antiviral therapy, hepatocarcinogenesis and extrahepatic manifestations. Thus, HCV-associated IR is a therapeutic target at any stage of HCV infection. However, specific pharmaceutical treatments of IR are still being evaluated in clinical trials, but available data do not warrant their use in all chronic hepatitis C patients with IR.
Collapse
Affiliation(s)
- Chao-Hung Hung
- Division of Hepatogastroenterology, Department of Internal Medicine, Chang Gung Memorial Hospital-Kaohsiung Medical Center, Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | | | | |
Collapse
|
35
|
Evaluation of organ-specific glucose metabolism by 18F-FDG in insulin receptor substrate-1 (IRS-1) knockout mice as a model of insulin resistance. Ann Nucl Med 2011; 25:755-61. [DOI: 10.1007/s12149-011-0522-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2011] [Accepted: 07/20/2011] [Indexed: 10/18/2022]
|
36
|
Holmes RM, Yi Z, De Filippis E, Berria R, Shahani S, Sathyanarayana P, Sherman V, Fujiwara K, Meyer C, Christ-Roberts C, Hwang H, Finlayson J, Dong LQ, Mandarino LJ, Bajaj M. Increased abundance of the adaptor protein containing pleckstrin homology domain, phosphotyrosine binding domain and leucine zipper motif (APPL1) in patients with obesity and type 2 diabetes: evidence for altered adiponectin signalling. Diabetologia 2011; 54:2122-31. [PMID: 21562756 PMCID: PMC3131511 DOI: 10.1007/s00125-011-2173-x] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2011] [Accepted: 03/29/2011] [Indexed: 12/14/2022]
Abstract
AIMS/HYPOTHESIS The adiponectin signalling pathway is largely unknown, but recently the adaptor protein containing pleckstrin homology domain, phosphotyrosine binding domain and leucine zipper motif (APPL1), has been shown to interact directly with adiponectin receptor (ADIPOR)1. APPL1 is present in C2C12 myoblasts and mouse skeletal muscle, but its presence in human skeletal muscle has not been investigated. METHODS Samples from type 2 diabetic, and lean and non-diabetic obese participants were analysed by: immunoprecipitation and western blot; HPLC-electrospray ionisation (ESI)-mass spectrometry (MS) analysis; peak area analysis by MS; HPLC-ESI-MS/MS/MS analysis; and RT-PCR analysis of APPL1 mRNA. RESULTS Immunoprecipitation and western blot indicated a band specific to APPL1. Tryptic digestion and HPLC-ESI-MS analysis of whole-muscle homogenate APPL1 unambiguously identified APPL1 with 56% sequence coverage. Peak area analysis by MS validated western blot results, showing APPL1 levels to be significantly increased in type 2 diabetic and obese as compared with lean participants. Targeted phosphopeptide analysis by HPLC-ESI-MS/MS/MS showed that APPL1 was phosphorylated specifically on Ser(401). APPL1 mRNA expression was significantly increased in obese and type 2 diabetic participants as compared with lean participants. After bariatric surgery in morbidly obese participants with subsequent weight loss, skeletal muscle APPL1 abundance was significantly reduced (p < 0.05) in association with an increase in plasma adiponectin (p < 0.01), increased levels of ADIPOR1 (p < 0.05) and increased muscle AMP-activated protein kinase (AMPK) phosphorylation (p < 0.05). CONCLUSIONS/INTERPRETATION APPL1 abundance is significantly higher in type 2 diabetic muscle; APPL1 is phosphorylated in vivo on Ser(401). Improvements in hyperglycaemia and hypoadiponectinaemia following weight loss are associated with reduced skeletal muscle APPL1, and increased plasma adiponectin levels and muscle AMPK phosphorylation.
Collapse
Affiliation(s)
- R. M. Holmes
- Department of Physiology, University of Texas Health Science Center, San Antonio, TX USA
- Center for Metabolic and Vascular Biology, College of Liberal Arts and Sciences, PO Box 873704, Tempe, AZ 85287-3704 USA
| | - Z. Yi
- Center for Metabolic and Vascular Biology, College of Liberal Arts and Sciences, PO Box 873704, Tempe, AZ 85287-3704 USA
| | - E. De Filippis
- Department of Medicine, University of Texas Health Science Center, San Antonio, TX USA
- Center for Metabolic and Vascular Biology, College of Liberal Arts and Sciences, PO Box 873704, Tempe, AZ 85287-3704 USA
| | - R. Berria
- Department of Medicine, University of Texas Health Science Center, San Antonio, TX USA
| | - S. Shahani
- Endocrinology Division, Baylor College of Medicine and St Luke’s Hospital, 1709 Dryden Street, Houston, TX 77030 USA
| | - P. Sathyanarayana
- Endocrinology Division, Baylor College of Medicine and St Luke’s Hospital, 1709 Dryden Street, Houston, TX 77030 USA
| | - V. Sherman
- Endocrinology Division, Baylor College of Medicine and St Luke’s Hospital, 1709 Dryden Street, Houston, TX 77030 USA
| | - K. Fujiwara
- Department of Medicine, University of Texas Health Science Center, San Antonio, TX USA
- Center for Metabolic and Vascular Biology, College of Liberal Arts and Sciences, PO Box 873704, Tempe, AZ 85287-3704 USA
| | - C. Meyer
- Center for Metabolic and Vascular Biology, College of Liberal Arts and Sciences, PO Box 873704, Tempe, AZ 85287-3704 USA
| | - C. Christ-Roberts
- Center for Metabolic and Vascular Biology, College of Liberal Arts and Sciences, PO Box 873704, Tempe, AZ 85287-3704 USA
| | - H. Hwang
- Center for Metabolic and Vascular Biology, College of Liberal Arts and Sciences, PO Box 873704, Tempe, AZ 85287-3704 USA
| | - J. Finlayson
- Center for Metabolic and Vascular Biology, College of Liberal Arts and Sciences, PO Box 873704, Tempe, AZ 85287-3704 USA
| | - L. Q. Dong
- Department of Cellular and Structural Biology, University of Texas Health Science Center, San Antonio, TX USA
- Department of Pharmacology, University of Texas Health Science Center, San Antonio, TX USA
| | - L. J. Mandarino
- Center for Metabolic and Vascular Biology, College of Liberal Arts and Sciences, PO Box 873704, Tempe, AZ 85287-3704 USA
- Department of Medicine, Mayo Clinic-Mayo Clinic Arizona, 13400 East Shea Boulevard, Scottsdale, AZ 85259 USA
| | - M. Bajaj
- Endocrinology Division, Baylor College of Medicine and St Luke’s Hospital, 1709 Dryden Street, Houston, TX 77030 USA
| |
Collapse
|
37
|
Fritsche L, Neukamm SS, Lehmann R, Kremmer E, Hennige AM, Hunder-Gugel A, Schenk M, Häring HU, Schleicher ED, Weigert C. Insulin-induced serine phosphorylation of IRS-2 via ERK1/2 and mTOR: studies on the function of Ser675 and Ser907. Am J Physiol Endocrinol Metab 2011; 300:E824-36. [PMID: 21098738 DOI: 10.1152/ajpendo.00409.2010] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
The identity of specific serine phosphorylation residues of insulin receptor substrate (IRS)-2 and their impact on insulin signal transduction are largely unknown. Ser(675) and Ser(907) of mouse IRS-2 are adjacent to PI 3-kinase or Grb2 binding domains, respectively. Using monoclonal phosphosite-specific antibodies, we demonstrated the phosphorylation of both serines after stimulation of Fao hepatoma cells with insulin, anisomycin, or phorbol esters. Phosphorylation of both sites was a late and prolonged event during insulin treatment and was also detected in liver tissue of insulin-treated as well as refed mice. Inhibition and siRNA-mediated knockdown of ERK1/2 indicated that the insulin-induced phosphorylation of Ser(907) was ERK dependent. Phosphorylation of Ser(907) did not prevent the insulin-induced association of IRS-2 with Grb2, but phosphorylation of the adjacent Tyr(911) was proved to be crucial in HEK 293 cells expressing IRS-2 Ala mutants. The insulin-induced phosphorylation of Ser(675) was prevented by inhibition and siRNA-mediated knockdown of mTOR but not of p70(S6K1). Mutation of Ser(675) to Ala did not affect downstream insulin signaling but increased the half-life of the protein, suggesting an involvement of phospho-Ser(675) in an accelerated degradation of IRS-2. Moreover, the insulin-induced degradation of IRS-2 was blocked by inhibition of mTOR. We conclude that the two novel insulin-dependent serine phosphorylation sites of IRS-2 were not involved in the regulation of the adjacent PI 3-kinase and Grb2 binding domains but might be implicated in the ERK- and mTOR-mediated negative feedback control.
Collapse
Affiliation(s)
- Louise Fritsche
- Dept. of Internal Medicine, Div. of Pathobiochemistry and Clinical Chemistry, Univ. of Tuebingen, Otfried-Mueller-Straße 10, 72076 Tuebingen, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Abstract
The adaptor protein raptor is the functional identifier for mammalian target of rapamycin (mTOR) complex 1 (mTORC1), acting to target mTOR to specific substrates for phosphorylation and regulation. Using HPLC-electrospray ionization tandem mass spectrometry, we confirmed the phosphorylation of raptor at Ser696, Thr706, Ser721, Ser722, Ser855, Ser859, Ser863, Thr865, Ser877, Ser881, Ser883, and Ser884 and identified Tyr692, Ser699, Thr700, Ser704, Ser854, Ser857, Ser882, Ser886, Ser887, and Thr889 as new, previously unidentified raptor phosphorylation sites. Treatment of cells with insulin increased the phosphorylation of raptor at Ser696, Ser855, Ser863, and Thr865 and suppressed the phosphorylation of Ser722. Ser696 phosphorylation was insensitive to mTOR inhibition with rapamycin, whereas treatment of cells with the MAPK inhibitor PD98059 inhibited the insulin-stimulated phosphorylation of raptor at Ser696. In vitro incubation of raptor with p42 MAPK significantly increased raptor phosphorylation (P < 0.01), whereas phosphorylation of a Ser696Ala mutant was decreased (P < 0.05), suggesting MAPK is capable of directly phosphorylating raptor at Ser696. Mutation of Ser696 to alanine interfered with insulin-stimulated phosphorylation of the mTOR downstream substrate p70S6 kinase. Incubation of cells with the MAPK inhibitor PD98059 and the phosphatidylinositol 3-kinase inhibitor wortmannin decreased the insulin stimulated phosphorylation of raptor, suggesting that the MAPK and phosphatidylinositol 3-kinase pathways may merge at mTORC1.
Collapse
Affiliation(s)
- Paul Langlais
- Center for Metabolic and Vascular Biology, Arizona State University, P.O. Box 874501, Tempe, Arizona 85287-4501, USA
| | | | | |
Collapse
|
39
|
Langlais P, Mandarino LJ, Yi Z. Label-free relative quantification of co-eluting isobaric phosphopeptides of insulin receptor substrate-1 by HPLC-ESI-MS/MS. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2010; 21:1490-9. [PMID: 20594869 PMCID: PMC2995262 DOI: 10.1016/j.jasms.2010.05.009] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2009] [Revised: 04/30/2010] [Accepted: 05/22/2010] [Indexed: 05/18/2023]
Abstract
Intracellular signal transduction is often regulated by transient protein phosphorylation in response to external stimuli. Insulin signaling is dependent on specific protein phosphorylation events, and analysis of insulin receptor substrate-1 (IRS-1) phosphorylation reveals a complex interplay between tyrosine, serine, and threonine phosphorylation. The phospho-specific antibody-based quantification approach for analyzing changes in site-specific phosphorylation of IRS-1 is difficult due to the dearth of phospho-antibodies compared with the large number of known IRS-1 phosphorylation sites. We previously published a method detailing a peak area-based mass spectrometry approach, using precursor ions for peptides, to quantify the relative abundance of site-specific phosphorylation in the absence or presence of insulin. We now present an improvement wherein site-specific phosphorylation is quantified by determining the peak area of fragment ions respective to the phospho-site of interest. This provides the advantage of being able to quantify co-eluting isobaric phosphopeptides (differentially phosphorylated versions of the same peptide), allowing for a more comprehensive analysis of protein phosphorylation. Quantifying human IRS-1 phosphorylation sites at Ser303, Ser323, Ser330, Ser348, Ser527, and Ser531 shows that this method is linear (n = 3; r(2) = 0.85 +/- 0.05, 0.96 +/- 0.01, 0.96 +/- 0.02, 0.86 +/- 0.07, 0.90 +/- 0.03, 0.91 +/- 0.04, respectively) over an approximate 10-fold range of concentrations and reproducible (n = 4; coefficient of variation = 0.12, 0.14, 0.29, 0.30, 0.12, 0.06, respectively). This application of label-free, fragment ion-based quantification to assess relative phosphorylation changes of specific proteins will prove useful for understanding how various cell stimuli regulate protein function by phosphorylation.
Collapse
Affiliation(s)
- Paul Langlais
- ASU/Mayo Center for Metabolic and Vascular Biology, Arizona State University, Tempe, Arizona 85287-4501, USA
| | | | | |
Collapse
|
40
|
cJun NH2-terminal kinase 1 (JNK1): roles in metabolic regulation of insulin resistance. Trends Biochem Sci 2010; 35:490-6. [PMID: 20452774 DOI: 10.1016/j.tibs.2010.04.004] [Citation(s) in RCA: 119] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2010] [Revised: 04/08/2010] [Accepted: 04/08/2010] [Indexed: 01/12/2023]
Abstract
The cJun NH(2)-terminal kinase isoform JNK1 is implicated in the mechanism of obesity-induced insulin resistance. Feeding a high-fat diet causes activation of the JNK1 signaling pathway, insulin resistance, and obesity in mice. Germ-line ablation of Jnk1 prevents both diet-induced obesity and insulin resistance. Genetic analysis indicates that the effects of JNK1 on insulin resistance can be separated from effects of JNK1 on obesity. Emerging research indicates that JNK1 plays multiple roles in the regulation of insulin resistance, including altered gene expression, hormone/cytokine production, and lipid metabolism. Together, these studies establish JNK1 as a potential pharmacological target for the development of drugs that might be useful for the treatment of insulin resistance, metabolic syndrome, and type 2 diabetes.
Collapse
|
41
|
Regulation of skeletal muscle oxidative capacity and insulin signaling by the mitochondrial rhomboid protease PARL. Cell Metab 2010; 11:412-26. [PMID: 20444421 PMCID: PMC3835349 DOI: 10.1016/j.cmet.2010.04.004] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2008] [Revised: 06/15/2009] [Accepted: 04/07/2010] [Indexed: 12/25/2022]
Abstract
Type 2 diabetes mellitus (T2DM) and aging are characterized by insulin resistance and impaired mitochondrial energetics. In lower organisms, remodeling by the protease pcp1 (PARL ortholog) maintains the function and lifecycle of mitochondria. We examined whether variation in PARL protein content is associated with mitochondrial abnormalities and insulin resistance. PARL mRNA and mitochondrial mass were both reduced in elderly subjects and in subjects with T2DM. Muscle knockdown of PARL in mice resulted in malformed mitochondrial cristae, lower mitochondrial content, decreased PGC1alpha protein levels, and impaired insulin signaling. Suppression of PARL protein in healthy myotubes lowered mitochondrial mass and insulin-stimulated glycogen synthesis and increased reactive oxygen species production. We propose that lower PARL expression may contribute to the mitochondrial abnormalities seen in aging and T2DM.
Collapse
|
42
|
Højlund K, Yi Z, Lefort N, Langlais P, Bowen B, Levin K, Beck-Nielsen H, Mandarino LJ. Human ATP synthase beta is phosphorylated at multiple sites and shows abnormal phosphorylation at specific sites in insulin-resistant muscle. Diabetologia 2010; 53:541-51. [PMID: 20012595 DOI: 10.1007/s00125-009-1624-0] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2009] [Accepted: 11/10/2009] [Indexed: 10/20/2022]
Abstract
AIMS/HYPOTHESIS Insulin resistance in skeletal muscle is linked to mitochondrial dysfunction in obesity and type 2 diabetes. Emerging evidence indicates that reversible phosphorylation regulates oxidative phosphorylation (OxPhos) proteins. The aim of this study was to identify and quantify site-specific phosphorylation of the catalytic beta subunit of ATP synthase (ATPsyn-beta) and determine protein abundance of ATPsyn-beta and other OxPhos components in skeletal muscle from healthy and insulin-resistant individuals. METHODS Skeletal muscle biopsies were obtained from lean, healthy, obese, non-diabetic and type 2 diabetic volunteers (each group n = 10) for immunoblotting of proteins, and hypothesis-driven identification and quantification of phosphorylation sites on ATPsyn-beta using targeted nanospray tandem mass spectrometry. Volunteers were metabolically characterised by euglycaemic-hyperinsulinaemic clamps. RESULTS Seven phosphorylation sites were identified on ATPsyn-beta purified from human skeletal muscle. Obese individuals with and without type 2 diabetes were characterised by impaired insulin-stimulated glucose disposal rates, and showed a approximately 30% higher phosphorylation of ATPsyn-beta at Tyr361 and Thr213 (within the nucleotide-binding region of ATP synthase) as well as a coordinated downregulation of ATPsyn-beta protein and other OxPhos components. Insulin increased Tyr361 phosphorylation of ATPsyn-beta by approximately 50% in lean and healthy, but not insulin-resistant, individuals. CONCLUSIONS/INTERPRETATION These data demonstrate that ATPsyn-beta is phosphorylated at multiple sites in human skeletal muscle, and suggest that abnormal site-specific phosphorylation of ATPsyn-beta together with reduced content of OxPhos proteins contributes to mitochondrial dysfunction in insulin resistance. Further characterisation of phosphorylation of ATPsyn-beta may offer novel targets of treatment in human diseases with mitochondrial dysfunction, such as diabetes.
Collapse
Affiliation(s)
- K Højlund
- Center for Metabolic Biology, School of Life Sciences, Arizona State University, PO Box 87370, Tempe, AZ 85287, USA.
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Højlund K, Bowen BP, Hwang H, Flynn CR, Madireddy L, Geetha T, Langlais P, Meyer C, Mandarino LJ, Yi Z. In vivo phosphoproteome of human skeletal muscle revealed by phosphopeptide enrichment and HPLC-ESI-MS/MS. J Proteome Res 2010; 8:4954-65. [PMID: 19764811 DOI: 10.1021/pr9007267] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Protein phosphorylation plays an essential role in signal transduction pathways that regulate substrate and energy metabolism, contractile function, and muscle mass in human skeletal muscle. Abnormal phosphorylation of signaling enzymes has been identified in insulin-resistant muscle using phosphoepitope-specific antibodies, but its role in other skeletal muscle disorders remains largely unknown. This may be in part due to insufficient knowledge of relevant targets. Here, we therefore present the first large-scale in vivo phosphoproteomic study of human skeletal muscle from 3 lean, healthy volunteers. Trypsin digestion of 3-5 mg human skeletal muscle protein was followed by phosphopeptide enrichment using SCX and TiO(2). The resulting phosphopeptides were analyzed by HPLC-ESI-MS/MS. Using this unbiased approach, we identified 306 distinct in vivo phosphorylation sites in 127 proteins, including 240 phosphoserines, 53 phosphothreonines, and 13 phosphotyrosines in at least 2 out of 3 subjects. In addition, 61 ambiguous phosphorylation sites were identified in at least 2 out of 3 subjects. The majority of phosphoproteins detected are involved in sarcomeric function, excitation-contraction coupling (the Ca(2+)-cycle), glycolysis, and glycogen metabolism. Of particular interest, we identified multiple novel phosphorylation sites on several sarcomeric Z-disk proteins known to be involved in signaling and muscle disorders. These results provide numerous new targets for the investigation of human skeletal muscle phosphoproteins in health and disease and demonstrate feasibility of phosphoproteomics research of human skeletal muscle in vivo.
Collapse
Affiliation(s)
- Kurt Højlund
- Center for Metabolic Biology, Arizona State University, Tempe, Arizona 85287-3704, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Copps KD, Hancer NJ, Opare-Ado L, Qiu W, Walsh C, White MF. Irs1 serine 307 promotes insulin sensitivity in mice. Cell Metab 2010; 11:84-92. [PMID: 20074531 PMCID: PMC3314336 DOI: 10.1016/j.cmet.2009.11.003] [Citation(s) in RCA: 155] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2009] [Revised: 09/03/2009] [Accepted: 11/03/2009] [Indexed: 12/11/2022]
Abstract
Phosphorylation of the insulin receptor substrates (Irs) on serine residues-typified by Ser307 of rodent Irs1-is thought to mediate insulin resistance. To determine whether Ser307 negatively regulates Irs1 in vivo, we generated knockin mice in which Ser307 (human Ser312) was replaced with alanine (A/A). Unexpectedly, A/A mice that were fed a high-fat diet developed more severe insulin resistance than control mice, accompanied by enhanced pancreatic compensation and impaired muscle insulin signaling. Chow-fed mice whose livers lacked Irs2 but retained a single knockin allele (A/lox::LKO2) were profoundly insulin resistant (versus +/lox::LKO2 mice), and their hepatocytes showed impaired insulin signaling ex vivo. Similarly, mutant A307 Irs1 adenovirus only partially restored the response to injected insulin in mice lacking hepatic Irs1 and Irs2. Thus, contrary to the results of cell-based experiments, Ser307 in mice is a positive regulatory site that moderates the severity of insulin resistance by maintaining proximal insulin signaling.
Collapse
Affiliation(s)
- Kyle D Copps
- Children's Hospital Boston, Harvard Medical School, MA 02115, USA
| | | | | | | | | | | |
Collapse
|
45
|
Klein AL, Berkaw MN, Buse MG, Ball LE. O-linked N-acetylglucosamine modification of insulin receptor substrate-1 occurs in close proximity to multiple SH2 domain binding motifs. Mol Cell Proteomics 2009; 8:2733-45. [PMID: 19671924 DOI: 10.1074/mcp.m900207-mcp200] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Insulin receptor substrate-1 (IRS-1) is a highly phosphorylated adaptor protein critical to insulin and IGF-1 receptor signaling. Ser/Thr kinases impact the metabolic and mitogenic effects elicited by insulin and IGF-1 through feedback and feed forward regulation at the level of IRS-1. Ser/Thr residues of IRS-1 are also O-GlcNAc-modified, which may influence the phosphorylation status of the protein. To facilitate the understanding of the functional effects of O-GlcNAc modification on IRS-1-mediated signaling, we identified the sites of O-GlcNAc modification of rat and human IRS-1. Tandem mass spectrometric analysis of IRS-1, exogenously expressed in HEK293 cells, revealed that the C terminus, which is rich in docking sites for SH2 domain-containing proteins, was O-GlcNAc-modified at multiple residues. Rat IRS-1 was O-GlcNAc-modified at Ser(914), Ser(1009), Ser(1036), and Ser(1041). Human IRS-1 was O-GlcNAc-modified at Ser(984) or Ser(985), at Ser(1011), and possibly at multiple sites within residues 1025-1045. O-GlcNAc modification at a conserved residue in rat (Ser(1009)) and human (Ser(1011)) IRS-1 is adjacent to a putative binding motif for the N-terminal SH2 domains of p85alpha and p85beta regulatory subunits of phosphatidylinositol 3-kinase and the tyrosine phosphatase SHP2 (PTPN11). Immunoblot analysis using an antibody generated against human IRS-1 Ser(1011) GlcNAc further confirmed the site of attachment and the identity of the +203.2-Da mass shift as beta-N-acetylglucosamine. The accumulation of IRS-1 Ser(1011) GlcNAc in HEPG2 liver cells and MC3T3-E1 preosteoblasts upon inhibition of O-GlcNAcase indicates that O-GlcNAcylation of endogenously expressed IRS-1 is a dynamic process that occurs at normal glucose concentrations (5 mm). O-GlcNAc modification did not occur at any known or newly identified Ser/Thr phosphorylation sites and in most cases occurred simultaneously with phosphorylation of nearby residues. These findings suggest that O-GlcNAc modification represents an additional layer of posttranslational regulation that may impact the specificity of effects elicited by insulin and IGF-1.
Collapse
Affiliation(s)
- Amanda L Klein
- Department of Molecular and Cellular Pharmacology, Medical University of South Carolina, Charleston, South Carolina 29425, USA
| | | | | | | |
Collapse
|
46
|
Abstract
Insulin signaling at target tissues is essential for growth and development and for normal homeostasis of glucose, fat, and protein metabolism. Control over this process is therefore tightly regulated. It can be achieved by a negative feedback control mechanism whereby downstream components inhibit upstream elements along the insulin-signaling pathway (autoregulation) or by signals from apparently unrelated pathways that inhibit insulin signaling thus leading to insulin resistance. Phosphorylation of insulin receptor substrate (IRS) proteins on serine residues has emerged as a key step in these control processes under both physiological and pathological conditions. The list of IRS kinases implicated in the development of insulin resistance is growing rapidly, concomitant with the list of potential Ser/Thr phosphorylation sites in IRS proteins. Here, we review a range of conditions that activate IRS kinases to phosphorylate IRS proteins on "hot spot" domains. The flexibility vs. specificity features of this reaction is discussed and its characteristic as an "array" phosphorylation is suggested. Finally, its implications on insulin signaling, insulin resistance and type 2 diabetes, an emerging epidemic of the 21st century are outlined.
Collapse
Affiliation(s)
- Sigalit Boura-Halfon
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, 76100 Israel
| | | |
Collapse
|
47
|
Alterations of insulin signaling in type 2 diabetes: A review of the current evidence from humans. Biochim Biophys Acta Mol Basis Dis 2009; 1792:83-92. [DOI: 10.1016/j.bbadis.2008.10.019] [Citation(s) in RCA: 153] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2008] [Revised: 10/21/2008] [Accepted: 10/21/2008] [Indexed: 01/03/2023]
|
48
|
Boura-Halfon S, Zick Y. Serine kinases of insulin receptor substrate proteins. VITAMINS AND HORMONES 2009; 80:313-49. [PMID: 19251043 DOI: 10.1016/s0083-6729(08)00612-2] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Signaling of insulin and insulin-like growth factor-I (IGF-1) at target tissues is essential for growth, development and for normal homeostasis of glucose, fat, and protein metabolism. Control over this process is therefore tightly regulated. It can be achieved by a negative-feedback control mechanism, whereby downstream components inhibit upstream elements along the insulin and IGF-1 signaling pathway or by signals from other pathways that inhibit insulin/IGF-1 signaling thus leading to insulin/IGF-1 resistance. Phosphorylation of insulin receptor substrates (IRS) proteins on serine residues has emerged as a key step in these control processes both under physiological and pathological conditions. The list of IRS kinases is growing rapidly, concomitant with the list of potential Ser/Thr phosphorylation sites in IRS proteins. Here we review a range of conditions that activate IRS kinases to phosphorylate IRS proteins on selected domains. The specificity of this reaction is discussed and its characteristic as an "array" phosphorylation is suggested. Finally, its implications on insulin/IGF-1 signaling, insulin/IGF-1 resistance and diabetes, an emerging epidemic of the twenty-first century are outlined.
Collapse
Affiliation(s)
- Sigalit Boura-Halfon
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| | | |
Collapse
|
49
|
Weigert C, Kron M, Kalbacher H, Pohl AK, Runge H, Häring HU, Schleicher E, Lehmann R. Interplay and effects of temporal changes in the phosphorylation state of serine-302, -307, and -318 of insulin receptor substrate-1 on insulin action in skeletal muscle cells. Mol Endocrinol 2008; 22:2729-40. [PMID: 18927238 DOI: 10.1210/me.2008-0102] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Transduction of the insulin signal is mediated by multisite Tyr and Ser/Thr phosphorylation of the insulin receptor substrates (IRSs). Previous studies on the function of single-site phosphorylation, particularly phosphorylation of Ser-302, -307, and -318 of IRS-1, showed attenuating as well as enhancing effects on insulin action. In this study we investigated a possible cross talk of these opposedly acting serine residues in insulin-stimulated skeletal muscle cells by monitoring phosphorylation kinetics, and applying loss of function, gain of function, and combination mutants of IRS-1. The phosphorylation at Ser-302 was rapid and transient, followed first by Ser-318 phosphorylation and later by phosphorylation of Ser-307, which remained elevated for 120 min. Mutation of Ser-302 to alanine clearly reduced the subsequent protein kinase C-zeta-mediated Ser-318 phosphorylation. The Ser-307 phosphorylation was independent of Ser-302 and/or Ser-318 phosphorylation status. The functional consequences of these phosphorylation patterns were studied by the expression of IRS-1 mutants. The E302A307E318 mutant simulating the early phosphorylation pattern resulted in a significant increase in Akt and glycogen synthase kinase 3 phosphorylation. Furthermore, glucose uptake was enhanced. Because the down-regulation of the insulin signal was not affected, this phosphorylation pattern seems to be involved in the enhancement but not in the termination of the insulin signal. This enhancing effect was completely absent when Ser-302 was unphosphorylated and Ser-307 was phosphorylated as simulated by the A302E307E318 mutant. Phospho-Ser-318, sequentially phosphorylated at least by protein kinase C-zeta and a mammalian target of rapamycin/raptor-dependent kinase, was part of the positive as well as of the subsequent negative phosphorylation pattern. Thus we conclude that insulin stimulation temporally generates different phosphorylation statuses of the same residues that exert different functions in insulin signaling.
Collapse
Affiliation(s)
- Cora Weigert
- Division of Clinical Chemistry and Pathobiochemistry, University Hospital of Tübingen, Tübingen, Germany
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Hepatitis C virus core protein upregulates serine phosphorylation of insulin receptor substrate-1 and impairs the downstream akt/protein kinase B signaling pathway for insulin resistance. J Virol 2007; 82:2606-12. [PMID: 18160431 DOI: 10.1128/jvi.01672-07] [Citation(s) in RCA: 138] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Chronic hepatitis C virus (HCV) infection has a significantly increased prevalence of type 2 diabetes mellitus (T2DM). Insulin resistance is a critical component of T2DM pathogenesis. Several mechanisms are likely to be involved in the pathogenesis of HCV-related insulin resistance. Since we and others have previously observed that HCV core protein activates c-Jun N-terminal kinase (JNK) and mitogen-activated protein kinase, we examined the contribution of these pathways to insulin resistance in hepatocytes. Our experimental findings suggest that HCV core protein alone or in the presence of other viral proteins increases Ser(312) phosphorylation of the insulin receptor substrate-1 (IRS-1). Hepatocytes infected with cell culture-grown HCV genotype 1a or 2a displayed a significant increase in the Ser(473) phosphorylation status of the Ser/Thr kinase protein kinase B (Akt/PKB), while Thr(308) phosphorylation was not significantly altered. HCV core protein-mediated Ser(312) phosphorylation of IRS-1 was inhibited by JNK (SP600125) and phosphatidylinositol-3 kinase (LY294002) inhibitors. A functional assay also suggested that hepatocytes expressing HCV core protein alone or infected with cell culture-grown HCV exhibited a suppression of 2-deoxy-d-[(3)H]glucose uptake. Inhibition of the JNK signaling pathway significantly restored glucose uptake despite HCV core expression in hepatocytes. Taken together, our results demonstrated that HCV core protein increases IRS-1 phosphorylation at Ser(312) which may contribute in part to the mechanism of insulin resistance.
Collapse
|