1
|
Oost LJ, Kurstjens S, Ma C, Hoenderop JGJ, Tack CJ, de Baaij JHF. Magnesium increases insulin-dependent glucose uptake in adipocytes. Front Endocrinol (Lausanne) 2022; 13:986616. [PMID: 36093068 PMCID: PMC9453642 DOI: 10.3389/fendo.2022.986616] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 08/03/2022] [Indexed: 01/12/2023] Open
Abstract
Background Type 2 diabetes (T2D) is characterized by a decreased insulin sensitivity. Magnesium (Mg2+) deficiency is common in people with T2D. However, the molecular consequences of low Mg2+ levels on insulin sensitivity and glucose handling have not been determined in adipocytes. The aim of this study is to determine the role of Mg2+ in the insulin-dependent glucose uptake. Methods First, the association of low plasma Mg2+ with markers of insulin resistance was assessed in a cohort of 395 people with T2D. Secondly, the molecular role of Mg2+ in insulin-dependent glucose uptake was studied by incubating 3T3-L1 adipocytes with 0 or 1 mmol/L Mg2+ for 24 hours followed by insulin stimulation. Radioactive-glucose labelling, enzymatic assays, immunocytochemistry and live microscopy imaging were used to analyze the insulin receptor phosphoinositide 3-kinases/Akt pathway. Energy metabolism was assessed by the Seahorse Extracellular Flux Analyzer. Results In people with T2D, plasma Mg2+ concentration was inversely associated with markers of insulin resistance; i.e., the lower Mg2+, the more insulin resistant. In Mg2+-deficient adipocytes, insulin-dependent glucose uptake was decreased by approximately 50% compared to control Mg2+condition. Insulin receptor phosphorylation Tyr1150/1151 and PIP3 mass were not decreased in Mg2+-deficient adipocytes. Live imaging microscopy of adipocytes transduced with an Akt sensor (FoxO1-Clover) demonstrated that FoxO1 translocation from the nucleus to the cytosol was reduced, indicting less Akt activation in Mg2+-deficient adipocytes. Immunocytochemistry using a Lectin membrane marker and at the membrane located Myc epitope-tagged glucose transporter 4 (GLUT4) demonstrated that GLUT4 translocation was diminished in insulin-stimulated Mg2+-deficient adipocytes compared to control conditions. Energy metabolism in Mg2+ deficient adipocytes was characterized by decreased glycolysis, upon insulin stimulation. Conclusions Mg2+ increases insulin-dependent glucose uptake in adipocytes and suggests that Mg2+ deficiency may contribute to insulin resistance in people with T2D.
Collapse
Affiliation(s)
- Lynette J. Oost
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Steef Kurstjens
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
- Laboratory of Clinical Chemistry and Hematology, Jeroen Bosch Hospital, ‘s-Hertogenbosch, Netherlands
| | - Chao Ma
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
- Beijing Tongren Hospital Beijing Institute of Ophthalmology, Beijing Ophthalmology and Visual Science Key Laboratory, Beijing Tongren Eye Center, Capital Medical University, Beijing, China
| | - Joost G. J. Hoenderop
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Cees J. Tack
- Department of Internal Medicine, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Jeroen H. F. de Baaij
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| |
Collapse
|
2
|
The Insulin Receptor: A Potential Target of Amarogentin Isolated from Gentiana rigescens Franch That Induces Neurogenesis in PC12 Cells. Biomedicines 2021; 9:biomedicines9050581. [PMID: 34065446 PMCID: PMC8160887 DOI: 10.3390/biomedicines9050581] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 05/10/2021] [Accepted: 05/11/2021] [Indexed: 01/19/2023] Open
Abstract
Amarogentin (AMA) is a secoiridoid glycoside isolated from the traditional Chinese medicine, Gentiana rigescens Franch. AMA exhibits nerve growth factor (NGF)-mimicking and NGF-enhancing activities in PC12 cells and in primary cortical neuron cells. In this study, a possible mechanism was found showing the remarkable induction of phosphorylation of the insulin receptor (INSR) and protein kinase B (AKT). The potential target of AMA was predicted by using a small-interfering RNA (siRNA) and the cellular thermal shift assay (CETSA). The AMA-induced neurite outgrowth was reduced by the siRNA against the INSR and the results of the CETSA suggested that the INSR showed a significant thermal stability-shifted effect upon AMA treatment. Other neurotrophic signaling pathways in PC12 cells were investigated using specific inhibitors, Western blotting and PC12(rasN17) and PC12(mtGAP) mutants. The inhibitors of the glucocorticoid receptor (GR), phospholipase C (PLC) and protein kinase C (PKC), Ras, Raf and mitogen-activated protein kinase (MEK) significantly reduced the neurite outgrowth induced by AMA in PC12 cells. Furthermore, the phosphorylation reactions of GR, PLC, PKC and an extracellular signal-regulated kinase (ERK) were significantly increased after inducing AMA and markedly decreased after treatment with the corresponding inhibitors. Collectively, these results suggested that AMA-induced neuritogenic activity in PC12 cells potentially depended on targeting the INSR and activating the downstream Ras/Raf/ERK and PI3K/AKT signaling pathways. In addition, the GR/PLC/PKC signaling pathway was found to be involved in the neurogenesis effect of AMA.
Collapse
|
3
|
Karl K, Paul MD, Pasquale EB, Hristova K. Ligand bias in receptor tyrosine kinase signaling. J Biol Chem 2020; 295:18494-18507. [PMID: 33122191 PMCID: PMC7939482 DOI: 10.1074/jbc.rev120.015190] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Revised: 10/28/2020] [Indexed: 12/14/2022] Open
Abstract
Ligand bias is the ability of ligands to differentially activate certain receptor signaling responses compared with others. It reflects differences in the responses of a receptor to specific ligands and has implications for the development of highly specific therapeutics. Whereas ligand bias has been studied primarily for G protein-coupled receptors (GPCRs), there are also reports of ligand bias for receptor tyrosine kinases (RTKs). However, the understanding of RTK ligand bias is lagging behind the knowledge of GPCR ligand bias. In this review, we highlight how protocols that were developed to study GPCR signaling can be used to identify and quantify RTK ligand bias. We also introduce an operational model that can provide insights into the biophysical basis of RTK activation and ligand bias. Finally, we discuss possible mechanisms underpinning RTK ligand bias. Thus, this review serves as a primer for researchers interested in investigating ligand bias in RTK signaling.
Collapse
Affiliation(s)
- Kelly Karl
- Department of Materials Science and Engineering, Institute for NanoBioTechnology, and Program in Molecular Biophysics, Johns Hopkins University, Baltimore, Maryland, USA
| | - Michael D Paul
- Department of Materials Science and Engineering, Institute for NanoBioTechnology, and Program in Molecular Biophysics, Johns Hopkins University, Baltimore, Maryland, USA
| | - Elena B Pasquale
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, USA.
| | - Kalina Hristova
- Department of Materials Science and Engineering, Institute for NanoBioTechnology, and Program in Molecular Biophysics, Johns Hopkins University, Baltimore, Maryland, USA.
| |
Collapse
|
4
|
Bailey CJ, Day C. Treatment of type 2 diabetes: future approaches. Br Med Bull 2018; 126:123-137. [PMID: 29897499 DOI: 10.1093/brimed/ldy013] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2018] [Accepted: 03/26/2018] [Indexed: 01/13/2023]
Abstract
INTRODUCTION OR BACKGROUND Type 2 diabetes, which accounts for ~90% of all diabetes, is a heterogeneous and progressive disease with a variety of causative and potentiating factors. The hyperglycaemia of type 2 diabetes is often inadequately controlled, hence the need for a wider selection of glucose-lowering treatments. SOURCES OF DATA Medline, PubMed, Web of Science and Google Scholar. AREAS OF AGREEMENT Early, effective and sustained control of blood glucose defers the onset and reduces the severity of microvascular and neuropathic complications of type 2 diabetes and helps to reduce the risk of cardiovascular (CV) complications. AREAS OF CONTROVERSY Newer glucose-lowering agents require extensive long-term studies to confirm CV safety. The positioning of newer agents within therapeutic algorithms varies. GROWING POINTS In addition to their glucose-lowering efficacy, some new glucose-lowering agents may act independently to reduce CV and renal complications. AREAS TIMELY FOR DEVELOPING RESEARCH Studies of potential new glucose-lowering agents offer the opportunity to safely improve glycaemic control with prolonged efficacy and greater opportunity for therapeutic individualisation.
Collapse
Affiliation(s)
- Clifford J Bailey
- Department of Biomedical Sciences, School of Life and Health Sciences, Aston University, Birmingham, UK
| | - Caroline Day
- Department of Biomedical Sciences, School of Life and Health Sciences, Aston University, Birmingham, UK
| |
Collapse
|
5
|
Chatterjee S, Vashishta L, Waichale VS, Nayak VG, Melarkode R, Donnelly CM, Vallano PT, Chirmule N, Sengupta N. Development and validation of a cell based assay for the detection of neutralizing antibodies against recombinant insulins. J Immunol Methods 2017; 452:53-62. [PMID: 28935478 DOI: 10.1016/j.jim.2017.09.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 09/15/2017] [Accepted: 09/15/2017] [Indexed: 01/21/2023]
Abstract
Recombinant biopharmaceuticals can induce generation of anti-drug antibodies, which could potentially neutralize therapeutic drug activity. In this report, we describe development and validation of a cell-based assay for detection of neutralizing antibodies (Nab) against insulin and insulin analogues. In order to achieve clinically meaningful sensitivity the method used an early signalling event, insulin induced insulin receptor phosphorylation as the endpoint. Percentage insulin receptor phosphorylation in cell lysates was measured using ECL based ELISA. Presence of neutralizing antibodies (Nab) in samples will inhibit insulin induced receptor phosphorylation and consequently lead to a reduction in the percentage of phosphorylated insulin receptor. Additionally, usage of human insulin receptor overexpressing recombinant CHO cell line further improved the assay sensitivity by reducing the fixed drug (EC50) concentration used for induction of receptor phosphorylation. To ensure adequate free drug tolerance a pre-treatment step was introduced, where serum samples underwent acid dissociation and charcoal extraction before drug incubation. In order to distinguish ADA positive samples containing true Nab from samples containing non-antibody phosphorylation inhibitory serum factors, a confirmatory tier was integrated based on immunodepletion using protein AGL mix. Assay parameters including determination of screening and confirmatory cut-points, intra and inter assay precision, selectivity, specificity and stability were assessed during validation in accordance with recent regulatory guidelines and white papers. The advantage of selecting insulin receptor phosphorylation as assay endpoint made the assay capable of detecting Nab against insulin and insulin analogues.
Collapse
Affiliation(s)
- Sanjukta Chatterjee
- Biocon Limited, Biocon Special Economic Zone, Plot Nos. 2 & 3, Phase IV-B.I.A. Bommasandra-Jigani Link Road, Bangalore 560099, India.
| | - Laxmikant Vashishta
- Biocon Limited, Biocon Special Economic Zone, Plot Nos. 2 & 3, Phase IV-B.I.A. Bommasandra-Jigani Link Road, Bangalore 560099, India
| | - Vinit S Waichale
- Biocon Limited, Biocon Special Economic Zone, Plot Nos. 2 & 3, Phase IV-B.I.A. Bommasandra-Jigani Link Road, Bangalore 560099, India
| | - Vivek G Nayak
- Biocon Limited, Biocon Special Economic Zone, Plot Nos. 2 & 3, Phase IV-B.I.A. Bommasandra-Jigani Link Road, Bangalore 560099, India
| | - Ramakrishnan Melarkode
- Biocon Limited, Biocon Special Economic Zone, Plot Nos. 2 & 3, Phase IV-B.I.A. Bommasandra-Jigani Link Road, Bangalore 560099, India
| | - Charles M Donnelly
- Mylan Pharmaceuticals, 3711 Collins Ferry Road, Morgantown, WV 26505, USA
| | - Patrick T Vallano
- Mylan Pharmaceuticals, 3711 Collins Ferry Road, Morgantown, WV 26505, USA
| | - Narendra Chirmule
- Biocon Limited, Biocon Special Economic Zone, Plot Nos. 2 & 3, Phase IV-B.I.A. Bommasandra-Jigani Link Road, Bangalore 560099, India
| | - Nilanjan Sengupta
- Biocon Limited, Biocon Special Economic Zone, Plot Nos. 2 & 3, Phase IV-B.I.A. Bommasandra-Jigani Link Road, Bangalore 560099, India.
| |
Collapse
|
6
|
Leist M, Rinné S, Datunashvili M, Aissaoui A, Pape HC, Decher N, Meuth SG, Budde T. Acetylcholine-dependent upregulation of TASK-1 channels in thalamic interneurons by a smooth muscle-like signalling pathway. J Physiol 2017; 595:5875-5893. [PMID: 28714121 DOI: 10.1113/jp274527] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Accepted: 07/10/2017] [Indexed: 12/13/2022] Open
Abstract
KEY POINTS The ascending brainstem transmitter acetylcholine depolarizes thalamocortical relay neurons while it induces hyperpolarization in local circuit inhibitory interneurons. Sustained K+ currents are modulated in thalamic neurons to control their activity modes; for the interneurons the molecular nature of the underlying ion channels is as yet unknown. Activation of TASK-1 K+ channels results in hyperpolarization of interneurons and suppression of their action potential firing. The modulation cascade involves a non-receptor tyrosine kinase, c-Src. The present study identifies a novel pathway for the activation of TASK-1 channels in CNS neurons that resembles cholinergic signalling and TASK-1 current modulation during hypoxia in smooth muscle cells. ABSTRACT The dorsal part of the lateral geniculate nucleus (dLGN) is the main thalamic site for state-dependent transmission of visual information. Non-retinal inputs from the ascending arousal system and inhibition provided by γ-aminobutyric acid (GABA)ergic local circuit interneurons (INs) control neuronal activity within the dLGN. In particular, acetylcholine (ACh) depolarizes thalamocortical relay neurons by inhibiting two-pore domain potassium (K2P ) channels. Conversely, ACh also hyperpolarizes INs via an as-yet-unknown mechanism. By using whole cell patch-clamp recordings in brain slices and appropriate pharmacological tools we here report that stimulation of type 2 muscarinic ACh receptors induces IN hyperpolarization by recruiting the G-protein βγ subunit (Gβγ), class-1A phosphatidylinositol-4,5-bisphosphate 3-kinase, and cellular and sarcoma (c-Src) tyrosine kinase, leading to activation of two-pore domain weakly inwardly rectifying K+ channel (TWIK)-related acid-sensitive K+ (TASK)-1 channels. The latter was confirmed by the use of TASK-1-deficient mice. Furthermore inhibition of phospholipase Cβ as well as an increase in the intracellular level of phosphatidylinositol-3,4,5-trisphosphate facilitated the muscarinic effect. Our results have uncovered a previously unknown role of c-Src tyrosine kinase in regulating IN function in the brain and identified a novel mechanism by which TASK-1 channels are activated in neurons.
Collapse
Affiliation(s)
- Michael Leist
- Institut für Physiologie I, Westfälische Wilhelms-Universität, Robert-Koch-Str. 27a, D-48149, Münster, Germany
| | - Susanne Rinné
- Institut für Physiologie und Pathophysiologie, AG Vegetative Physiologie, Philipps-Universität, Deutschhausstraße 1-2, D-35037, Marburg, Germany
| | - Maia Datunashvili
- Institut für Physiologie I, Westfälische Wilhelms-Universität, Robert-Koch-Str. 27a, D-48149, Münster, Germany
| | - Ania Aissaoui
- Institut für Physiologie I, Westfälische Wilhelms-Universität, Robert-Koch-Str. 27a, D-48149, Münster, Germany
| | - Hans-Christian Pape
- Institut für Physiologie I, Westfälische Wilhelms-Universität, Robert-Koch-Str. 27a, D-48149, Münster, Germany
| | - Niels Decher
- Institut für Physiologie und Pathophysiologie, AG Vegetative Physiologie, Philipps-Universität, Deutschhausstraße 1-2, D-35037, Marburg, Germany
| | - Sven G Meuth
- Department of Neurology, Westfälische Wilhelms-Universität, Albert-Schweitzer-Campus 1, D-48149, Münster, Germany
| | - Thomas Budde
- Institut für Physiologie I, Westfälische Wilhelms-Universität, Robert-Koch-Str. 27a, D-48149, Münster, Germany
| |
Collapse
|
7
|
Pinheiro BS, Lemos C, Neutzling Kaufmann F, Marques JM, da Silva-Santos CS, Carvalho E, Mackie K, Rodrigues RJ, Cunha RA, Köfalvi A. Hierarchical glucocorticoid-endocannabinoid interplay regulates the activation of the nucleus accumbens by insulin. Brain Res Bull 2016; 124:222-30. [PMID: 27208730 DOI: 10.1016/j.brainresbull.2016.05.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Revised: 05/16/2016] [Accepted: 05/17/2016] [Indexed: 12/28/2022]
Abstract
Here we asked if insulin activation of the nucleus accumbens in vitro is reflected by an increase in (3)H-deoxyglucose ([(3)H]DG) uptake, thus subserving a new model to study molecular mechanisms of central insulin actions. Additionally, we investigated the dependence of this insulin effect on endocannabinoids and corticosteroids, two major culprits in insulin resistance. We found that in acute accumbal slices, insulin (3 and 300nM but not at 0.3nM) produced an increase in [(3)H]DG uptake. The synthetic cannabinoid agonist, WIN55212-2 (500nM) and the glucocorticoid dexamethasone (10μM), impaired insulin (300nM) action on [(3)H]DG uptake. The glucocorticoid receptor (GcR) antagonist, mifepristone (10μM) prevented dexamethasone from inhibiting insulin's action. Strikingly, this anti-insulin action of dexamethasone was also blocked by two CB1 cannabinoid receptor (CB1R) antagonists, O-2050 (500nM) and SR141716A (500nM), as well as by tetrahydrolipstatin (10μM), an inhibitor of diacylglycerol lipases-the enzymes responsible for the synthesis of the endocannabinoid, 2-arachidonoyl-glycerol (2-AG). On the other hand, the blockade of the post-synaptic 2-AG metabolizing enzymes, α,β-serine hydrolase domain 6/12 by WWL70 (1μM) also prevented the action of insulin, probably via increasing endogenous 2-AG tone. Additionally, an anti-insulin receptor (InsR) antibody immunoprecipitated CB1Rs from accumbal homogenates, indicating a physical complexing of CB1Rs with InsRs that supports their functional interaction. Altogether, insulin stimulates glucose uptake in the nucleus accumbens. Accumbal GcR activation triggers the synthesis of 2-AG that in turn binds to the known CB1R-InsR heteromer, thus impeding insulin signaling.
Collapse
Affiliation(s)
- Bárbara S Pinheiro
- CNC, Center for Neuroscience and Cell Biology of Coimbra, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Cristina Lemos
- CNC, Center for Neuroscience and Cell Biology of Coimbra, University of Coimbra, 3004-504 Coimbra, Portugal
| | | | - Joana M Marques
- CNC, Center for Neuroscience and Cell Biology of Coimbra, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Carla S da Silva-Santos
- CNC, Center for Neuroscience and Cell Biology of Coimbra, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Eugénia Carvalho
- CNC, Center for Neuroscience and Cell Biology of Coimbra, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Ken Mackie
- Department of Psychological and Brain Sciences, Gill Center for Biomolecular Science, Indiana University, Bloomington, IN, USA
| | - Ricardo J Rodrigues
- CNC, Center for Neuroscience and Cell Biology of Coimbra, University of Coimbra, 3004-504 Coimbra, Portugal; Institute for Interdisciplinary Research, University of Coimbra, 3030-789 Coimbra, Portugal
| | - Rodrigo A Cunha
- CNC, Center for Neuroscience and Cell Biology of Coimbra, University of Coimbra, 3004-504 Coimbra, Portugal; FMUC, Faculty of Medicine, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Attila Köfalvi
- CNC, Center for Neuroscience and Cell Biology of Coimbra, University of Coimbra, 3004-504 Coimbra, Portugal; Institute for Interdisciplinary Research, University of Coimbra, 3030-789 Coimbra, Portugal.
| |
Collapse
|
8
|
Bailey CJ, Tahrani AA, Barnett AH. Future glucose-lowering drugs for type 2 diabetes. Lancet Diabetes Endocrinol 2016; 4:350-9. [PMID: 26809680 DOI: 10.1016/s2213-8587(15)00462-3] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Revised: 11/13/2015] [Accepted: 11/18/2015] [Indexed: 12/15/2022]
Abstract
The multivariable and progressive natural history of type 2 diabetes limits the effectiveness of available glucose-lowering drugs. Constraints imposed by comorbidities (notably cardiovascular disease and renal impairment) and the need to avoid hypoglycaemia, weight gain, and drug interactions further complicate the treatment process. These challenges have prompted the development of new formulations and delivery methods for existing drugs alongside research into novel pharmacological entities. Advances in incretin-based therapies include a miniature implantable osmotic pump to give continuous delivery of a glucagon-like peptide-1 receptor agonist for 6-12 months and once-weekly tablets of dipeptidyl peptidase-4 inhibitors. Hybrid molecules that combine the properties of selected incretins and other peptides are at early stages of development, and proof of concept has been shown for small non-peptide molecules to activate glucagon-like peptide-1 receptors. Additional sodium-glucose co-transporter inhibitors are progressing in development as well as possible new insulin-releasing biological agents and small-molecule inhibitors of glucagon action. Adiponectin receptor agonists, selective peroxisome proliferator-activated receptor modulators, cellular glucocorticoid inhibitors, and analogues of fibroblast growth factor 21 are being considered as potential new approaches to glucose lowering. Compounds that can enhance insulin receptor and post-receptor signalling cascades or directly promote selected pathways of glucose metabolism have suggested opportunities for future treatments. However, pharmacological interventions that are able to restore normal β-cell function and β-cell mass, normalise insulin action, and fully correct glucose homoeostasis are a distant vision.
Collapse
Affiliation(s)
- Clifford J Bailey
- School of Life and Health Sciences, Aston University, Birmingham, UK.
| | - Abd A Tahrani
- Department of Diabetes and Endocrinology, Heart of England NHS Foundation Trust, Birmingham, UK
| | - Anthony H Barnett
- Department of Diabetes and Endocrinology, Heart of England NHS Foundation Trust, Birmingham, UK; Centre for Endocrinology, Diabetes and Metabolism, University of Birmingham, Birmingham, UK
| |
Collapse
|
9
|
Altaf QA, Barnett AH, Tahrani AA. Novel therapeutics for type 2 diabetes: insulin resistance. Diabetes Obes Metab 2015; 17:319-34. [PMID: 25308775 DOI: 10.1111/dom.12400] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2014] [Revised: 09/23/2014] [Accepted: 10/04/2014] [Indexed: 12/19/2022]
Abstract
Insulin resistance (IR) plays an important role in the pathogenesis of type 2 diabetes (T2D) and cardiovascular disease. Hence improving IR is a major target of treatment in patients with T2D. Obesity and lack of exercise are major causes of IR. However, recent evidence implicates sleep disorders and disorders of the circadian rhythm in the pathogenesis of IR. Weight loss and lifestyle changes are the cornerstone and most effective treatments of IR, but adherence and patient's acceptability are poor. Bariatric surgery results in significant and sustainable long-term weight loss associated with beneficial impact on IR and glucose metabolism, making this an attractive treatment option for patients with T2D. Currently available pharmacological options targeting IR (such as metformin and thiazolidinediones) do not maintain glycaemic measures within targets long term and can be associated with significant side effects. Over the last two decades, many pharmacological agents targeting different aspects of the insulin signalling pathway were developed to improve IR, but only a minority reached clinical trials. Such treatments need to be specific and reversible as many of the components of the insulin signalling pathway are involved in other cellular functions such as apoptosis. Recent evidence highlighted the role of circadian rhythm and sleep-related disorders in the pathogenesis of IR. In this article, we review the latest developments in the pharmacological and non-pharmacological interventions targeting IR including bariatric surgery. We will also review the role of circadian rhythm and sleep-related disorders in the development and treatment of IR.
Collapse
Affiliation(s)
- Q-A Altaf
- Department of Diabetes and Endocrinology, Heart of England NHS Foundation Trust, Birmingham, UK; Centre of Endocrinology, Diabetes and Metabolism, University of Birmingham, Birmingham, UK
| | | | | |
Collapse
|
10
|
Non-peptidyl insulin mimetics as a potential antidiabetic agent. Drug Discov Today 2013; 18:748-55. [DOI: 10.1016/j.drudis.2013.04.005] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2013] [Revised: 03/21/2013] [Accepted: 04/11/2013] [Indexed: 12/29/2022]
|
11
|
Johnson KF, Van Zeeland R, Stanley LM. Palladium-catalyzed synthesis of N-tert-prenylindoles. Org Lett 2013; 15:2798-801. [PMID: 23714013 DOI: 10.1021/ol4011344] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Palladium-catalyzed N-tert-prenylations of indoles, tricarbonylchromium-activated indoles, and indolines that occur in high yields (up to 94%) with high tert-prenyl-to-n-prenyl selectivity (up to 12:1) are reported.
Collapse
Affiliation(s)
- Kirsten F Johnson
- Department of Chemistry, Iowa State University, Ames, Iowa 50011, USA
| | | | | |
Collapse
|
12
|
Lai MC, Lo YS, Yang C. The effect of demethylasterriquinone B-1 on insulin secretion in rat pancreas. ACTA ACUST UNITED AC 2013. [DOI: 10.4236/jdm.2013.33017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
13
|
Takakusagi Y, Takakusagi K, Sugawara F, Sakaguchi K. Use of phage display technology for the determination of the targets for small-molecule therapeutics. Expert Opin Drug Discov 2012; 5:361-89. [PMID: 22823088 DOI: 10.1517/17460441003653155] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
IMPORTANCE OF THE FIELD Target discovery of drug-like small-molecules contributes to our understanding of biological phenomena at the molecular level as well as elucidating the mode of action of bioactive compounds. Research in this field is of high value because, in addition to basic observations, the data can be used to directly identify molecular targets or investigate pharmacokinetic characteristics of drugs in clinical use. AREAS COVERED IN THIS REVIEW In addition to providing a brief overview of phage display (PD) technology, we discuss screening platforms, different types of phage libraries and the application of this method to the determination of targets for small-molecule therapeutics over the past decade. WHAT THE READER WILL GAIN Readers will gain an understanding of the basis of PD technology through successful examples of the use of this method for the determination of targets for small-molecule therapeutics. They will learn what kinds of small-molecules were used to identify their binding partner, what characteristics and drawbacks are present in the use of small-molecule as bait, and what kinds of approaches were introduced in order to improve the technique to overcome the limitations of conventional strategies. TAKE HOME MESSAGE A suitable combination of diverse technologies from various different fields can act synergistically to increase throughput and enhance the efficiency of PD technology for the determination of targets for small-molecule therapeutics. The most suitable method for successful target identification of small-molecules of interest using PD technology can often be determined by referring to past examples.
Collapse
Affiliation(s)
- Yoichi Takakusagi
- Tokyo University of Science, Faculty of Science and Technology, Department of Applied Biological Science, 2641 Yamazaki, Noda, Chiba 278-8510, Japan +81 4 7124 1501 ext. 3409 ; +81 4 7123 9767 ; ;
| | | | | | | |
Collapse
|
14
|
Sengul IF, Wood K, Bowyer PK, Bhadbhade M, Chen R, Kumar N, Black DS. Synthesis of new di-(3-indolyl)arenes. Tetrahedron 2012. [DOI: 10.1016/j.tet.2012.06.074] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
|
15
|
Baus D, Yan Y, Li Z, Garyantes T, de Hoop M, Tennagels N. A robust assay measuring GLUT4 translocation in rat myoblasts overexpressing GLUT4-myc and AS160_v2. Anal Biochem 2009; 397:233-40. [PMID: 19854150 DOI: 10.1016/j.ab.2009.10.036] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2009] [Revised: 10/16/2009] [Accepted: 10/20/2009] [Indexed: 11/30/2022]
Abstract
Muscle and fat cells translocate GLUT4 (glucose transporter 4) to the plasma membrane when stimulated by insulin. Usually, this event is measured in differentiated adipocytes, myotubes, or cell lines overexpressing tagged GLUT4 by immunostaining. However, measurement of the translocation in differentiated adipocytes or myotubes or GLUT4 overexpressing cell lines is difficult because of high assay variability caused by either the differentiation protocol or low assay sensitivity. We recently reported the identification of a novel splice variant of AS160 (substrate of 160kDa), namely AS160_v2, and showed that its coexpression with GLUT4 in L6 myoblasts increased the insulin-stimulated glucose uptake rate due to an increased amount of GLUT4 on the cell surface. L6 cells, which coexpress myc-tagged GLUT4 and AS160_v2, can be efficiently used to generate an assay useful for identifying compounds that affect cellular responses to insulin. We compared the EC(50) values for radioactive glucose uptake and GLUT4 translocation of different insulins and several small molecules to validate the assay. The use of L6 cells overexpressing AS160_v2 can be considered as a novel tool for the characterization of molecules modulating insulin signaling and GLUT4 translocation, and an image-based assay increases our confidence in the mode of action of the compounds identified.
Collapse
Affiliation(s)
- Daniela Baus
- Sanofi-Aventis Deutschland, 65926 Frankfurt am Main, Germany
| | | | | | | | | | | |
Collapse
|
16
|
Lin B, Li Z, Park K, Deng L, Pai A, Zhong L, Pirrung MC, Webster NJG. Identification of novel orally available small molecule insulin mimetics. J Pharmacol Exp Ther 2007; 323:579-85. [PMID: 17687071 DOI: 10.1124/jpet.107.126102] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Oral hypoglycemic agents have great potential for the treatment of both type 1 and type 2 diabetes. Here we report the identification of novel, small-molecule, insulin mimetics that activate the insulin receptor (IR) in vivo and in vitro, stimulate the Akt and extracellular signal-regulated kinase pathways downstream of the IR, and mimic the ability of insulin to stimulate glucose uptake, glycogen synthesis, and lipid synthesis in 3T3-L1 adipocytes. However, the compounds do not mimic the mitogenic effect of insulin. In animals, these compounds have oral hypoglycemic effects in both normal C57BL6 mice and diabetic db/db mice. Quantitative structure activity relationship modeling on data from a library of 60 compounds has highlighted structural features that are important for IR agonist activity that can be used to guide design of second and third generation compounds with greater potency and specificity.
Collapse
Affiliation(s)
- Bo Lin
- Veterans Affairs San Diego Healthcare System and Veterans Medical Research Foundation, San Diego, California, USA
| | | | | | | | | | | | | | | |
Collapse
|
17
|
Kim H, Deng L, Xiong X, Hunter WD, Long MC, Pirrung MC. Glyceraldehyde 3-phosphate dehydrogenase is a cellular target of the insulin mimic demethylasterriquinone B1. J Med Chem 2007; 50:3423-6. [PMID: 17595071 PMCID: PMC2556249 DOI: 10.1021/jm070437i] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
This study was undertaken to identify cellular proteins that bind an orally active natural product insulin mimic. Phage display cloning was used with a biotinylated derivative of this molecule as bait. Among the proteins identified was glyceraldehyde 3-phosphate dehydrogenase (GAPDH), which has recently been shown to affect insulin receptor signaling. Binding data support a role for human GAPDH as another target of the insulin mimic, which could explain its action as a selective insulin receptor modulator.
Collapse
Affiliation(s)
| | | | | | | | | | - Michael C. Pirrung
- To whom correspondence should be addressed: f: 951-827-2749; p: 951-827-2722; e:
| |
Collapse
|
18
|
Shah DI, Singh M. Effect of demethylasterriquinone b1 in hypertension associated vascular endothelial dysfunction. Int J Cardiol 2007; 120:317-24. [PMID: 17240464 DOI: 10.1016/j.ijcard.2006.10.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2006] [Revised: 10/08/2006] [Accepted: 10/13/2006] [Indexed: 12/21/2022]
Abstract
BACKGROUND Activation of Akt stimulates phosphorylation of eNOS, production of nitric oxide and reduces oxidative stress. The study has been designed to investigate the effect of DAQ B1, an activator of Akt, in hypertension associated vascular endothelial dysfunction. METHODS Rats were uninephroctomized and DOCA (40 mg kg(-1), s.c.) was administered to rats to produce hypertension (MABP>140 mm Hg). Vascular endothelial dysfunction was assessed using isolated aortic ring preparation, electron microscopy of thoracic aorta and serum concentration of nitrite/nitrate. The expression of messenger RNA for p22phox and eNOS was assessed by reverse transcription-polymerase chain reaction. Serum TBARS and aortic superoxide anion were estimated to assess oxidative stress. RESULTS DAQ B1 (5 mg kg(-1), p.o.) or atorvastatin (30 mg kg(-1), p.o.) markedly improved acetylcholine induced endothelium dependent relaxation, vascular endothelial lining, expression of mRNA for eNOS and p22phox, serum nitrite/nitrate concentration and serum TBARS in hypertensive rats. However, this ameliorative effect of DAQ B1 has been prevented by L-NAME (25 mg kg(-1), i.p.), an inhibitor of eNOS. CONCLUSION Therefore, it may be concluded that DAQ B1 induced activation of Akt may activate eNOS and consequently reduce oxidative stress to improve hypertension associated vascular endothelial dysfunction.
Collapse
Affiliation(s)
- Dhvanit I Shah
- Department of Pharmaceutical Sciences and Drug Research, Faculty of Medicine, Punjabi University, Patiala-147002 Punjab India.
| | | |
Collapse
|
19
|
Cole GM, Frautschy SA. The role of insulin and neurotrophic factor signaling in brain aging and Alzheimer’s Disease. Exp Gerontol 2007; 42:10-21. [PMID: 17049785 DOI: 10.1016/j.exger.2006.08.009] [Citation(s) in RCA: 127] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2006] [Revised: 08/23/2006] [Accepted: 08/23/2006] [Indexed: 12/15/2022]
Abstract
Although increased lifespan is associated with reduced insulin signaling, insulin signaling is essential for neuronal development and survival. Insulin resistance is central to Type II diabetes and is also implicated in the pathogenesis of Alzheimer's Disease (AD). This has prompted ongoing clinical trials in AD patients to test the efficacy of improving insulin - like signaling with dietary omega-3 fatty acids or insulin - sensitizing drugs as well as exercise regimens. Here we review the role of insulin signaling in brain aging and AD, concluding that the signaling pathways downstream to neurotrophic and insulin signaling are defective and coincident with aberrant phosphorylation and translocation of key components, notably AKT and GSK3beta, but also rac> PAK signaling. These responses are likely to contribute to defects in synaptic plasticity, learning and memory. Both oligomers of beta-amyloid (which are elevated in the AD brain) and pro-inflammatory cytokines (which are elevated in the aged or AD brain) can be used to mimic the trophic factor/insulin resistance observed in AD, but details on other factors and mechanisms contributing to this resistance remain elusive. A better understanding of the precise mechanisms underlying alterations in the insulin/neurotrophic factor signal transduction pathways should aid the search for better AD therapeutic and prevention strategies.
Collapse
Affiliation(s)
- Greg M Cole
- Greater Los Angeles Veterans Affairs Healthcare System, Geriatric Research, Education and Clinical Center, 16111 Plummer Street, Sepulveda, CA 91343, USA.
| | | |
Collapse
|
20
|
Shah DI, Singh M. Possible role of Akt to improve vascular endothelial dysfunction in diabetic and hyperhomocysteinemic rats. Mol Cell Biochem 2006; 295:65-74. [PMID: 16841179 DOI: 10.1007/s11010-006-9273-9] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2006] [Accepted: 06/26/2006] [Indexed: 10/24/2022]
Abstract
The study has been designed to investigate the effect of demethylasterroquinone B1 (DAQ B1), an activator of Akt, in diabetes mellitus (DM) and hyperhomocysteinemia (HHcy)-induced vascular endothelial dysfunction. Streptozotocin (55 mg kg(-1), i.v.) and methionine (1.7% w/w, p.o., 4 weeks) were administered to rats to produce DM (serum glucose >140 mg dl(-1)) and HHcy (serum homocysteine >10 microM), respectively. Vascular endothelial dysfunction was assessed using isolated aortic ring preparation, electron microscopy of thoracic aorta and serum concentration of nitrite/nitrate. The expression of messenger RNA for p22phox and eNOS was assessed by reverse transcription-polymerase chain reaction. Serum thiobarbituric acid reactive substances (TBARS) and aortic superoxide anion were estimated to assess oxidative stress. DAQ B1 (5 mg kg(-1), p.o.) or atorvastatin (30 mg kg(-1), p.o.) in diabetic and hyperhomocysteinemic rats significantly reduced serum glucose and homocysteine concentration. DAQ B1 or atorvastatin markedly improved acetylcholine-induced endothelium-dependent relaxation, vascular endothelial lining, serum nitrite/nitrate concentration and serum TBARS in diabetic and hyperhomocysteinemic rats. However, this ameliorative effect of DAQ B1 has been prevented by L-NAME (25 mg kg(-1), i.p.), an inhibitor of eNOS. Therefore, it may be concluded that DAQ B1-induced activation of Akt may activate eNOS and consequently reduce oxidative stress to improve vascular endothelial dysfunction.
Collapse
MESH Headings
- Acetylcholine/pharmacology
- Animals
- Aorta/drug effects
- Aorta/enzymology
- Aorta/ultrastructure
- Blood Glucose/metabolism
- Diabetes Mellitus, Experimental/chemically induced
- Diabetes Mellitus, Experimental/enzymology
- Diabetes Mellitus, Experimental/physiopathology
- Endothelium, Vascular/enzymology
- Endothelium, Vascular/physiopathology
- Endothelium, Vascular/ultrastructure
- Gene Expression Regulation, Enzymologic/drug effects
- Homocysteine/blood
- Hyperhomocysteinemia/chemically induced
- Hyperhomocysteinemia/enzymology
- Hyperhomocysteinemia/physiopathology
- Male
- NADPH Oxidases/genetics
- NADPH Oxidases/metabolism
- Nitrates/blood
- Nitric Oxide Synthase Type III/genetics
- Nitric Oxide Synthase Type III/metabolism
- Nitrites/blood
- Proto-Oncogene Proteins c-akt/agonists
- Proto-Oncogene Proteins c-akt/metabolism
- Quinones/pharmacology
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Rats
- Rats, Wistar
- Superoxides/metabolism
- Thiobarbituric Acid Reactive Substances/analysis
- Vasodilation/drug effects
Collapse
Affiliation(s)
- Dhvanit I Shah
- Department of Pharmaceutical Sciences & Drug Research, Faculty of Medicine, Punjabi University, Patiala, 147002
| | | |
Collapse
|
21
|
Pirrung MC, Fujita K, Park K. Organometallic routes to 2,5-dihydroxy-3-(indol-3-yl)benzoquinones. Synthesis of demethylasterriquinone B4. J Org Chem 2005; 70:2537-42. [PMID: 15787540 DOI: 10.1021/jo048126s] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
A method has been developed to sequentially add indole-3-mercurials to dichlorinated quinones using palladium catalysis. These reactions can be used in the modular assembly of bis(indol-3-yl)benzoquinones, a significant natural product family.
Collapse
Affiliation(s)
- Michael C Pirrung
- Department of Chemistry, Levine Science Research Center, Duke University, Durham, North Carolina 27708-0317, USA.
| | | | | |
Collapse
|