1
|
Zhang L, Liu X, Hu J, Quan H, Lee SK, Korivi M, Wang L, Li T, Li W. Aerobic exercise attenuates high-fat diet-induced glycometabolism impairments in skeletal muscle of rat: role of EGR-1/PTP1B signaling pathway. Nutr Metab (Lond) 2024; 21:113. [PMID: 39741281 DOI: 10.1186/s12986-024-00888-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Accepted: 12/15/2024] [Indexed: 01/02/2025] Open
Abstract
OBJECTIVE Impaired skeletal muscle glycogen synthesis contributes to insulin resistance (IR). Aerobic exercise reported to ameliorate IR by augmenting insulin signaling, however the detailed mechanism behind this improvement remains unclear. This study investigated whether aerobic exercise enhances glycogen anabolism and insulin sensitivity via EGR-1/PTP1B signaling pathway in skeletal muscle of rats. METHODS Sprague-Dawley rats fed a high-fat diet (HFD), and performed treadmill exercise training for 6-week. Oral glucose tolerance test was conducted to confirm the IR. Periodic Acid-Schiff (PAS) staining and anthrone colorimetry were used to assess the skeletal muscle glycogen. RT-qPCR, western blot, and immunofluorescence were used to detect the EGR-1/PTP1B pathway and associated signaling molecules. RESULTS We found that exercise training significantly decreased blood glucose, insulin, and homeostasis model assessment for IR (HOMA-IR) against HFD-induced elevation. Decreased muscle glycogen content due to HFD was significantly restored after exercise training. Exercise training promoted mRNA expressions of Irs1, Akt, and Glut4, while inhibited Gsk-3β expression against HFD. Next, the decreased IRS1 (phosphorylated/total), AKT (phosphorylated/total), and GLUT4, and increased GSK-3β proteins with HFD were significantly reversed by exercise. Furthermore, HFD-induced overexpression of EGR-1 and PTP1B evidenced by mRNA, protein, and immunofluorescence intensity, were substantially inhibited by exercise, which may contribute to promote insulin sensitivity and glycogen anabolism. CONCLUSIONS Aerobic exercise training promotes insulin sensitivity and skeletal muscle glycogen synthesis in HFD-fed rats. The beneficial effects of exercise might be mediated by EGR-1/PTP1B signaling pathway in skeletal muscle, however further studies are necessary to confirm this mechanism.
Collapse
Affiliation(s)
- Liangzhi Zhang
- College of Physical Education and Health Sciences, Zhejiang Normal University, 688 Yingbin Road, Jinhua, 321004, Zhejiang Province, China
| | - Xiaojie Liu
- College of Physical Education and Health Sciences, Zhejiang Normal University, 688 Yingbin Road, Jinhua, 321004, Zhejiang Province, China
| | - Jing Hu
- Department of Clinical Medicine, Medical College, Jinhua University of Vocational Technology, Jinhua, Zhejiang, China
| | - Helong Quan
- Exercise Capacity Assessment and Promotion Research Center, School of Physical Education, Northeast Normal University, Changchun, Jilin, China
| | - Sang Ki Lee
- Department of Sport Science, College of Natural Science, Chungnam National University, Deajeon, Korea
| | - Mallikarjuna Korivi
- College of Physical Education and Health Sciences, Zhejiang Normal University, 688 Yingbin Road, Jinhua, 321004, Zhejiang Province, China
| | - Lifeng Wang
- College of Physical Education and Health Sciences, Zhejiang Normal University, 688 Yingbin Road, Jinhua, 321004, Zhejiang Province, China
| | - Ting Li
- College of Physical Education and Health Sciences, Zhejiang Normal University, 688 Yingbin Road, Jinhua, 321004, Zhejiang Province, China.
| | - Wei Li
- College of Physical Education and Health Sciences, Zhejiang Normal University, 688 Yingbin Road, Jinhua, 321004, Zhejiang Province, China.
| |
Collapse
|
2
|
Borozdina NA, Dyachenko IA, Popkova DV. Promising Directions for Regulating Signaling Pathways Involved in the Type 2 Diabetes Mellitus Development (A Review). RUSSIAN JOURNAL OF BIOORGANIC CHEMISTRY 2024; 50:1263-1284. [DOI: 10.1134/s1068162024040137] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 11/11/2023] [Accepted: 11/12/2023] [Indexed: 01/04/2025]
|
3
|
Ge X, Hu M, Zhou M, Fang X, Chen X, Geng D, Wang L, Yang X, An H, Zhang M, Lin D, Zheng M, Cui X, Wang Q, Wu Y, Zheng K, Huang XF, Yu Y. Overexpression of forebrain PTP1B leads to synaptic and cognitive impairments in obesity. Brain Behav Immun 2024; 117:456-470. [PMID: 38336024 DOI: 10.1016/j.bbi.2024.02.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 01/23/2024] [Accepted: 02/04/2024] [Indexed: 02/12/2024] Open
Abstract
Obesity has reached pandemic proportions and is a risk factor for neurodegenerative diseases, including Alzheimer's disease. Chronic inflammation is common in obese patients, but the mechanism between inflammation and cognitive impairment in obesity remains unclear. Accumulative evidence shows that protein-tyrosine phosphatase 1B (PTP1B), a neuroinflammatory and negative synaptic regulator, is involved in the pathogenesis of neurodegenerative processes. We investigated the causal role of PTP1B in obesity-induced cognitive impairment and the beneficial effect of PTP1B inhibitors in counteracting impairments of cognition, neural morphology, and signaling. We showed that obese individuals had negative relationship between serum PTP1B levels and cognitive function. Furthermore, the PTP1B level in the forebrain increased in patients with neurodegenerative diseases and obese cognitive impairment mice with the expansion of white matter, neuroinflammation and brain atrophy. PTP1B globally or forebrain-specific knockout mice on an obesogenic high-fat diet showed enhanced cognition and improved synaptic ultrastructure and proteins in the forebrain. Specifically, deleting PTP1B in leptin receptor-expressing cells improved leptin synaptic signaling and increased BDNF expression in the forebrain of obese mice. Importantly, we found that various PTP1B allosteric inhibitors (e.g., MSI-1436, well-tolerated in Phase 1 and 1b clinical trials for obesity and type II diabetes) prevented these alterations, including improving cognition, neurite outgrowth, leptin synaptic signaling and BDNF in both obese cognitive impairment mice and a neural cell model of PTP1B overexpression. These findings suggest that increased forebrain PTP1B is associated with cognitive decline in obesity, whereas inhibition of PTP1B could be a promising strategy for preventing neurodegeneration induced by obesity.
Collapse
Affiliation(s)
- Xing Ge
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Minmin Hu
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Menglu Zhou
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Xiaoli Fang
- Department of Neurology, Affiliated Hospital of Xuzhou Medical University, Jiangsu 221006, China
| | - Xi Chen
- Illawarra Health and Medical Research Institute (IHMRI) and School of Medical, Indigenous, and Health, University of Wollongong, NSW 2522, Australia
| | - Deqin Geng
- Department of Neurology, Affiliated Hospital of Xuzhou Medical University, Jiangsu 221006, China
| | - Li Wang
- Affiliated Hospital of Liaoning University of Traditional Chinese Medicine, Shenyang, Liaoning 110032, China
| | - Xiaoying Yang
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Huimei An
- HuiLongGuan Clinical Medical School, Beijing HuiLongGuan Hospital, Peking University, Beijing 10096, China
| | - Meng Zhang
- HuiLongGuan Clinical Medical School, Beijing HuiLongGuan Hospital, Peking University, Beijing 10096, China
| | - Danhong Lin
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Mingxuan Zheng
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Xiaoying Cui
- Queensland Brain Institute, The University of Queensland, St Lucia, QLD 4113, Australia; Queensland Centre for Mental Health Research, Wacol, QLD, 4076, Australia
| | - Qing Wang
- Department of Neurology, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong 510280, China
| | - Yuqing Wu
- Jiangsu Province Key Laboratory of Anesthesiology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou 221004, China.
| | - Kuiyang Zheng
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China.
| | - Xu-Feng Huang
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China; Illawarra Health and Medical Research Institute (IHMRI) and School of Medical, Indigenous, and Health, University of Wollongong, NSW 2522, Australia.
| | - Yinghua Yu
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China.
| |
Collapse
|
4
|
The Antidiabetic Activities of Neocryptotanshinone: Screened by Molecular Docking and Related to the Modulation of PTP1B. Nutrients 2022; 14:nu14153031. [PMID: 35893885 PMCID: PMC9330310 DOI: 10.3390/nu14153031] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 07/19/2022] [Accepted: 07/20/2022] [Indexed: 01/27/2023] Open
Abstract
The aim of this study was to provide a practical experimental basis for the development of Neocryptotanshinone (NCTS) as an effective hypoglycemic drug and a theoretical method for the rapid screening of natural compounds with hypoglycemic effects. Molecular docking was used to screen the most suitable ligand. Hematoxylin and eosin, immunohistochemical staining, enzyme-linked immunosorbent assay and Western Blotting approved the hypoglycemic effect of NCTS. According to the free energy of binding, among 180 active compounds from the Traditional Chinese Medicine Integrated Database, NCTS was finally chose for investigation its hypoglycemic effects. In db/db mice, NCTS significantly reduced body weight and plasma glucose, improved glucose tolerance and levels of fasting plasma glucose and glycated hemoglobin A1c, and decreased insulin resistance after six-week administration. NCTS restored the pathological state in the liver of db/db mice and significantly decreased protein tyrosine phosphatase 1B (PTP1B) expression in the liver and muscle of db/db mice, which is related to the regulatory effect of NCTS on insulin receptor substrate 1. In conclusion, we successfully explored the hypoglycemic effect of NCTS in db/db mice via regulating the expression of PTP1B.
Collapse
|
5
|
Exploring New Drug Targets for Type 2 Diabetes: Success, Challenges and Opportunities. Biomedicines 2022; 10:biomedicines10020331. [PMID: 35203540 PMCID: PMC8869656 DOI: 10.3390/biomedicines10020331] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 01/24/2022] [Accepted: 01/26/2022] [Indexed: 01/02/2023] Open
Abstract
There are substantial shortcomings in the drugs currently available for treatment of type 2 diabetes mellitus. The global diabetic crisis has not abated despite the introduction of new types of drugs and targets. Persistent unaddressed patient needs remain a significant factor in the quest for new leads in routine studies. Drug discovery methods in this area have followed developments in the market, contributing to a recent rise in the number of molecules. Nevertheless, troubling developments and fresh challenges are still evident. Recently, metformin, the most widely used first-line drug for diabetes, was found to contain a carcinogenic contaminant known as N-nitroso dimethylamine (NDMA). Therefore, purity and toxicity are also a big challenge for drug discovery and development. Moreover, newer drug classes against SGLT-2 illustrate both progress and difficulties. The same was true previously in the case of glucagon-like peptide-1 receptor agonists and dipeptidyl peptidase-4 inhibitors. Furthermore, researchers must study the importance of mechanistic characteristics of novel compounds, as well as exposure-related hazardous aspects of current and newly identified protein targets, in order to identify new pharmacological molecules with improved selectivity and specificity.
Collapse
|
6
|
Teimouri M, Hosseini H, ArabSadeghabadi Z, Babaei-Khorzoughi R, Gorgani-Firuzjaee S, Meshkani R. The role of protein tyrosine phosphatase 1B (PTP1B) in the pathogenesis of type 2 diabetes mellitus and its complications. J Physiol Biochem 2022; 78:307-322. [PMID: 34988903 DOI: 10.1007/s13105-021-00860-7] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 11/16/2021] [Indexed: 01/16/2023]
Abstract
Insulin resistance, the most important characteristic of the type 2 diabetes mellitus (T2DM), is mostly caused by impairment in the insulin receptor (IR) signal transduction pathway. Protein tyrosine phosphatase 1B (PTP1B), one of the main negative regulators of the IR signaling pathway, is broadly expressed in various cells and tissues. PTP1B decreases the phosphorylation of the IR resulting in insulin resistance in various tissues. The evidence for the physiological role of PTP1B in regulation of metabolic pathways came from whole-body PTP1B-knockout mice. Whole-body and tissue-specific PTP1B-knockout mice showed improvement in adiposity, insulin resistance, and glucose tolerance. In addition, the key role of PTP1B in the pathogenesis of T2DM and its complications was further investigated in mice models of PTP1B deficient/overexpression. In recent years, targeting PTP1B using PTP1B inhibitors is being considered an attractive target to treat T2DM. PTP1B inhibitors improve the sensitivity of the insulin receptor and have the ability to cure insulin resistance-related diseases. We herein summarized the biological functions of PTP1B in different tissues in vivo and in vitro. We also describe the effectiveness of potent PTP1B inhibitors as pharmaceutical agents to treat T2DM.
Collapse
Affiliation(s)
- Maryam Teimouri
- Department of Clinical Biochemistry, School of Allied Medical Sciences, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Hossein Hosseini
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Zahra ArabSadeghabadi
- Department of Clinical Sciences, Faculty of Veterinary Science, Bu-Ali Sina University, Hamedan, Iran
| | - Reyhaneh Babaei-Khorzoughi
- Department of Clinical Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Sattar Gorgani-Firuzjaee
- Department of Medical Laboratory Sciences, School of Allied Health Medicine, AJA University of Medical Sciences, Tehran, Iran
| | - Reza Meshkani
- Department of Clinical Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
7
|
Dinda B, Dinda M. Natural Products, a Potential Source of New Drugs Discovery to Combat Obesity and Diabetes: Their Efficacy and Multi-targets Actions in Treatment of These Diseases. NATURAL PRODUCTS IN OBESITY AND DIABETES 2022:101-275. [DOI: 10.1007/978-3-030-92196-5_4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
8
|
Kornicka-Garbowska K, Bourebaba L, Röcken M, Marycz K. Inhibition of protein tyrosine phosphatase improves mitochondrial bioenergetics and dynamics, reduces oxidative stress, and enhances adipogenic differentiation potential in metabolically impaired progenitor stem cells. Cell Commun Signal 2021; 19:106. [PMID: 34732209 PMCID: PMC8565043 DOI: 10.1186/s12964-021-00772-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 07/30/2021] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND Protein tyrosine phosphatase 1B (PTP1B) and low molecular weight protein tyrosine phosphatase (LMPTP) are implicated in the development of metabolic disorders. Yet, their role in progenitor stem cell adipogenic differentiation and modulation of mitochondrial dynamics remains elusive. METHODS In this study, we decided to investigate whether inhibition of PTP1B and LMPTP enhance adipogenic differentiation of metabolically impaired progenitor stem cells via modulation of mitochondrial bioenergetics and dynamics. Cells were cultured under adipogenic conditions in the presence of PTP1B and LMPTP inhibitors, and were subjected to the analysis of the main adipogenic-related and mitochondrial-related genes using RT-qPCR. Protein levels were established with western blot while mitochondrial morphology with MicroP software. RESULTS Selective inhibitors of both PTP1B and MPTP enhanced adipogenic differentiation of metabolically impaired progenitor stem cells. We have observed enhanced expression of PPARy and adiponectin in treated cells. What is more, increased antioxidative defence and alternations in mitochondrial bioenergetics were observed. We have found that inhibition of PTP1B as well as C23 activates oxidative phosphorylation and enhances mitochondrial fusion contributing to enhanced adipogenesis. CONCLUSIONS The presented data provides evidence that the application of PTP1B and LMPTP inhibitors enhances adipogenesis through the modulation of mitochondrial dynamics. Video abstract.
Collapse
Affiliation(s)
- Katarzyna Kornicka-Garbowska
- Department of Experimental Biology, Wroclaw University of Environmental and Life Sciences, Norwida 27B Street, A7 building, 50-375 Wroclaw, Poland
- International Institute of Translational Medicine, Malin, Jesionowa 11, 55-114 Wisznia Mała, Poland
| | - Lynda Bourebaba
- Department of Experimental Biology, Wroclaw University of Environmental and Life Sciences, Norwida 27B Street, A7 building, 50-375 Wroclaw, Poland
| | - Michael Röcken
- Faculty of Veterinary Medicine, Equine Clinic-Equine Surgery, Justus-Liebig University, 35392 Giessen, Germany
| | - Krzysztof Marycz
- Department of Experimental Biology, Wroclaw University of Environmental and Life Sciences, Norwida 27B Street, A7 building, 50-375 Wroclaw, Poland
- International Institute of Translational Medicine, Malin, Jesionowa 11, 55-114 Wisznia Mała, Poland
| |
Collapse
|
9
|
Shirif AZ, Kovačević S, Brkljačić J, Teofilović A, Elaković I, Djordjevic A, Matić G. Decreased Glucocorticoid Signaling Potentiates Lipid-Induced Inflammation and Contributes to Insulin Resistance in the Skeletal Muscle of Fructose-Fed Male Rats Exposed to Stress. Int J Mol Sci 2021; 22:ijms22137206. [PMID: 34281257 PMCID: PMC8269441 DOI: 10.3390/ijms22137206] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 06/25/2021] [Accepted: 06/28/2021] [Indexed: 01/03/2023] Open
Abstract
The modern lifestyle brings both excessive fructose consumption and daily exposure to stress which could lead to metabolic disturbances and type 2 diabetes. Muscles are important points of glucose and lipid metabolism, with a crucial role in the maintenance of systemic energy homeostasis. We investigated whether 9-week fructose-enriched diet, with and without exposure to 4-week unpredictable stress, disturbs insulin signaling in the skeletal muscle of male rats and evaluated potential contributory roles of muscle lipid metabolism, glucocorticoid signaling and inflammation. The combination of fructose-enriched diet and stress increased peroxisome proliferator-activated receptors-α and -δ and stimulated lipid uptake, lipolysis and β-oxidation in the muscle of fructose-fed stressed rats. Combination of treatment also decreased systemic insulin sensitivity judged by lower R-QUICKI, and lowered muscle protein content and stimulatory phosphorylations of insulin receptor supstrate-1 and Akt, as well as the level of 11β-hydroxysteroid dehydrogenase type 1 and glucocorticoid receptor. At the same time, increased levels of protein tyrosine phosphatase-1B, nuclear factor-κB, tumor necrosis factor-α, were observed in the muscle of fructose-fed stressed rats. Based on these results, we propose that decreased glucocorticoid signaling in the skeletal muscle can make a setting for lipid-induced inflammation and the development of insulin resistance in fructose-fed stressed rats.
Collapse
|
10
|
Kazemi M, Kim JY, Parry SA, Azziz R, Lujan ME. Disparities in cardio metabolic risk between Black and White women with polycystic ovary syndrome: a systematic review and meta-analysis. Am J Obstet Gynecol 2021; 224:428-444.e8. [PMID: 33316275 DOI: 10.1016/j.ajog.2020.12.019] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 11/26/2020] [Accepted: 12/05/2020] [Indexed: 12/26/2022]
Abstract
OBJECTIVE We conducted a systematic review and meta-analysis to summarize and quantitatively pool evidence on cardiometabolic health disparities between Black and White women with polycystic ovary syndrome in the United States in response to the call for further delineation of these disparities in the international evidence-based guideline for the assessment and management of polycystic ovary syndrome. DATA SOURCES Databases of MEDLINE, Web of Science, and Scopus were searched initially through March 05, 2020, and confirmed on September 11, 2020. STUDY ELIGIBILITY CRITERIA Observational studies documenting cardiometabolic risk profile (glucoregulatory, lipid profile, anthropometric, and blood pressure status) in Black and White women with polycystic ovary syndrome were included. Studies on children (<17 years old) and pregnant or menopausal-aged women (>50 years) were excluded. The primary outcome was fasting glucose. Furthermore, data on major cardiovascular events (stroke, coronary heart disease, heart failure) and mortality rate (cardiovascular death, total mortality) were evaluated. METHODS Data were pooled by random-effects models and expressed as mean differences and 95% confidence intervals. Studies were weighted based on the inverse of the variance. Heterogeneity was evaluated by Cochran Q and I2 statistics. Study methodologic quality was assessed by the Newcastle-Ottawa scale. RESULTS A total of 11 studies (N=2851 [652 Black and 2199 White]) evaluated cardiometabolic risk profile and all had high quality (Newcastle-Ottawa scale score of ≥8). No studies reported on cardiovascular events and mortality rate. Black women had comparable fasting glucose (-0.61 [-1.69 to 2.92] mg/dL; I2=62.5%), yet exhibited increased fasting insulin (6.76 [4.97-8.56] μIU/mL; I2=59.0%); homeostatic model assessment of insulin resistance (1.47 [0.86-2.08]; I2=83.2%); systolic blood pressure (3.32 [0.34-6.30] mm Hg; I2=52.0%); and decreased triglyceride (-32.56 [-54.69 to -10.42] mg/dL; I2=68.0%) compared with White women (all, P≤.03). Groups exhibited comparable total cholesterol, low-density lipoprotein cholesterol, high-density lipoprotein cholesterol, and diastolic blood pressure (all, P≥.06). CONCLUSIONS Black women with polycystic ovary syndrome have a greater tendency for an adverse cardiometabolic risk profile (increased insulin, homeostatic model assessment of insulin resistance, and systolic blood pressure) despite lower triglycerides than White women. Our observations support the consideration of these disparities for diagnostic, monitoring, and management practices in Black women and for future guideline recommendations. Given the heterogeneity among studies, future research should address the relative contributions of biologic, environmental, socioeconomic, and healthcare factors to the observed disparities. Furthermore, longitudinal research is required to address patient-pressing complications, including cardiovascular events and mortality rate in Black women with polycystic ovary syndrome as a high-risk yet understudied population.
Collapse
Affiliation(s)
- Maryam Kazemi
- Human Metabolic Research Unit, Division of Nutritional Sciences, Cornell University, Ithaca, NY.
| | - Joy Y Kim
- Human Metabolic Research Unit, Division of Nutritional Sciences, Cornell University, Ithaca, NY
| | - Stephen A Parry
- Cornell Statistical Consulting Unit, Cornell University, Ithaca, NY
| | - Ricardo Azziz
- Department of Obstetrics and Gynecology, University of Alabama at Birmingham, Birmingham, AL; Department of Obstetrics and Gynecology, David Geffen School of Medicine at UCLA, Los Angeles, CA; Department of Health Policy, Management, and Behavior, School of Public Health, University at Albany, Albany, NY
| | - Marla E Lujan
- Human Metabolic Research Unit, Division of Nutritional Sciences, Cornell University, Ithaca, NY.
| |
Collapse
|
11
|
Chen S, Sbuh N, Veedu RN. Antisense Oligonucleotides as Potential Therapeutics for Type 2 Diabetes. Nucleic Acid Ther 2020; 31:39-57. [PMID: 33026966 DOI: 10.1089/nat.2020.0891] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Type 2 diabetes (T2D) is a chronic metabolic disorder characterized by persistent hyperglycemia resulting from inefficient signaling and insufficient production of insulin. Conventional management of T2D has largely relied on small molecule-based oral hypoglycemic medicines, which do not halt the progression of the disease due to limited efficacy and induce adverse effects as well. To this end, antisense oligonucleotide has attracted immense attention in developing antidiabetic agents because of their ability to downregulate the expression of disease-causing genes at the RNA and protein level. To date, seven antisense agents have been approved by the United States Food and Drug Administration for therapies of a variety of human maladies, including genetic disorders. Herein, we provide a comprehensive review of antisense molecules developed for suppressing the causative genes believed to be responsible for insulin resistance and hyperglycemia toward preventing and treating T2D.
Collapse
Affiliation(s)
- Suxiang Chen
- Centre for Molecular Medicine and Innovative Therapeutics, Murdoch University, Perth, Australia.,Perron Institute for Neurological and Translational Science, Perth, Australia
| | - Nabayet Sbuh
- Centre for Molecular Medicine and Innovative Therapeutics, Murdoch University, Perth, Australia.,Perron Institute for Neurological and Translational Science, Perth, Australia
| | - Rakesh N Veedu
- Centre for Molecular Medicine and Innovative Therapeutics, Murdoch University, Perth, Australia.,Perron Institute for Neurological and Translational Science, Perth, Australia
| |
Collapse
|
12
|
Oses M, Margareto Sanchez J, Portillo MP, Aguilera CM, Labayen I. Circulating miRNAs as Biomarkers of Obesity and Obesity-Associated Comorbidities in Children and Adolescents: A Systematic Review. Nutrients 2019; 11:nu11122890. [PMID: 31783635 PMCID: PMC6950354 DOI: 10.3390/nu11122890] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Revised: 11/11/2019] [Accepted: 11/25/2019] [Indexed: 01/13/2023] Open
Abstract
Early detection of obesity and its associated comorbidities in children needs priority for the development of effective therapeutic intervention. Circulating miRNAs (microRNAs) have been proposed as biomarkers for obesity and its comorbidities; therefore, we conducted a systematic review to summarize results of studies that have quantified the profile of miRNAs in children and adolescents with obesity and/or associated disorders. Nine studies aiming to examine differences in miRNA expression levels between children with normal weight and obesity or between obese children with or without cardiometabolic diseases were included in this review. We identified four miRNAs overexpressed in obesity (miR-222, miR-142-3, miR-140-5p, and miR-143) and two miRNAs (miR-122 and miR-34a) overexpressed in children with obesity and nonalcoholic fatty liver disease (NAFLD) and/or insulin resistance. In conclusion, circulating miRNAs are promising diagnostic biomarkers of obesity-associated diseases such as NAFLD and type 2 diabetes already in childhood. However, more studies in children, using massive search technology and with larger sample sizes, are required to draw any firm conclusions.
Collapse
Affiliation(s)
- Maddi Oses
- Institute for Innovation & Sustainable Development in Food Chain, Public University of Navarre, Jeronimo de Ayanz Building, Campus de Arrosadia, 31006 Pamplona, Spain;
- Correspondence: ; Tel.: +34-636818502
| | | | - Maria P. Portillo
- Nutrition and Obesity Group, Department of Nutrition and Food Science, Public University of the Basque Country (UPV/EHU) and Lucio Lascaray Research Institute, 01006 Vitoria, Spain;
- CIBEROBN (Physiopathology of Obesity and Nutrition Network CB12/03/30038), Health Institute Carlos III (ISCIII), 28029 Madrid, Spain;
- Instituto de Investigación Sanitaria BIOARABA, 01006 Vitoria, Spain
| | - Concepción María Aguilera
- CIBEROBN (Physiopathology of Obesity and Nutrition Network CB12/03/30038), Health Institute Carlos III (ISCIII), 28029 Madrid, Spain;
- Department of Biochemistry and Molecular Biology II, Institute of Nutrition and Food Technology (INYTA), Biomedical Research Centre (CIBM), University of Granada, Avda. del Conocimiento s/n, 18016 Armilla, Spain
- Instituto de Investigación Biosanitaria IBS.GRANADA, Complejo Hospitalario Universitario de Granada, 18014 Granada, Spain
| | - Idoia Labayen
- Institute for Innovation & Sustainable Development in Food Chain, Public University of Navarre, Jeronimo de Ayanz Building, Campus de Arrosadia, 31006 Pamplona, Spain;
| |
Collapse
|
13
|
Clavier T, Grangé S, Pressat-Laffouilhere T, Besnier E, Renet S, Fraineau S, Thiebaut PA, Richard V, Veber B, Tamion F. Gene Expression of Protein Tyrosine Phosphatase 1B and Endoplasmic Reticulum Stress During Septic Shock. Front Med (Lausanne) 2019; 6:240. [PMID: 31737637 PMCID: PMC6839276 DOI: 10.3389/fmed.2019.00240] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Accepted: 10/14/2019] [Indexed: 12/17/2022] Open
Abstract
Introduction: Protein Tyrosine Phosphatase 1B (PTP1B) and endoplasmic reticulum stress (ERS) are involved in the septic inflammatory response. Their inhibition is associated with improved survival in murine models of sepsis. The objective was to describe PTP1B and ERS expression during septic shock in human. Material and Methods: Prospective study including patients admitted to intensive care unit (ICU) for septic shock. Blood samples were collected on days 1 (D1), 3 and 5 (D5). Quantitative PCR (performed from whole blood) evaluated the expression of genes coding for PTP1B (PTPN1) and key elements of ERS (GRP78, ATF6, CHOP) or for endothelial dysfunction-related markers (ICAM1 and ET1). We analyzed gene variation between D5 and D1, collected glycemic parameters, insulin resistance and organ failure was evaluated by Sequential Organ Failure Assessment (SOFA) score. Results: We included 44 patients with a mean SAPS II 50 ± 16 and a mortality rate of 13.6%. Between D1 and D5, there was a significant decrease of PTPN1 (p < 0.001) and ATF6 (p < 0.001) expressions. Their variations of expression were correlated with SOFA variation (PTPN1, r = 0.35, CI 95% [0.05; 0.54], p = 0.03 and ATF6, r = 0.45 CI 95% [0.20; 0.65], p < 0.001). We did not find any correlation between PTPN1 expression and insulin resistance or glycemic parameters. Between D1 and D5, ATF6 and PTPN1 expressions were correlated with that of ET1. Conclusions: Our study has evaluated for the first time the expression of PTP1B and ERS in patients with septic shock, revealing that gene expression variation of PTPN1 and ATF6 are partly correlated with the evolution of septic organ failure and with endothelial dysfunction markers expression.
Collapse
Affiliation(s)
- Thomas Clavier
- Department of Anesthesiology and Critical Care, Rouen University Hospital, Rouen, France.,Normandie Univ, UNIROUEN, INSERM U1096, FHU REMOD-VHF, Rouen, France.,Department of Medical Critical Care, Rouen University Hospital, Rouen, France
| | - Steven Grangé
- Department of Medical Critical Care, Rouen University Hospital, Rouen, France
| | | | - Emmanuel Besnier
- Department of Anesthesiology and Critical Care, Rouen University Hospital, Rouen, France.,Normandie Univ, UNIROUEN, INSERM U1096, FHU REMOD-VHF, Rouen, France
| | - Sylvanie Renet
- Normandie Univ, UNIROUEN, INSERM U1096, FHU REMOD-VHF, Rouen, France
| | - Sylvain Fraineau
- Normandie Univ, UNIROUEN, INSERM U1096, FHU REMOD-VHF, Rouen, France
| | | | - Vincent Richard
- Normandie Univ, UNIROUEN, INSERM U1096, FHU REMOD-VHF, Rouen, France
| | - Benoit Veber
- Department of Anesthesiology and Critical Care, Rouen University Hospital, Rouen, France
| | - Fabienne Tamion
- Normandie Univ, UNIROUEN, INSERM U1096, FHU REMOD-VHF, Rouen, France.,Department of Medical Critical Care, Rouen University Hospital, Rouen, France
| |
Collapse
|
14
|
Angiotensin II induces apoptosis of cardiac microvascular endothelial cells via regulating PTP1B/PI3K/Akt pathway. In Vitro Cell Dev Biol Anim 2019; 55:801-811. [PMID: 31502193 DOI: 10.1007/s11626-019-00395-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Accepted: 08/02/2019] [Indexed: 12/28/2022]
Abstract
Endothelial cell apoptosis and renin-angiotensin-aldosterone system (RAAS) activation are the major pathological mechanisms for cardiovascular disease and heart failure; however, the interaction and mechanism between them remain unclear. Investigating the role of PTP1B in angiotensin II (Ang II)-induced apoptosis of primary cardiac microvascular endothelial cells (CMECs) may provide direct evidence of the link between endothelial cell apoptosis and RAAS. Isolated rat CMECs were treated with different concentrations of Ang II to induce apoptosis, and an Ang II concentration of 4 nM was selected as the effective dose for the subsequent studies. The CMECs were cultured for 48 h with or without Ang II (4 nM) in the absence or presence of the PTP1B inhibitor TCS 401 (8 μM) and the PI3K inhibitor LY294002 (10 μM). The level of CMEC apoptosis was assessed by TUNEL staining and caspase-3 activity. The protein expressions of PTP1B, PI3K, Akt, p-Akt, Bcl-2, Bax, caspase-3, and cleaved caspase-3 were determined by Western blot (WB). The results showed that Ang II increased apoptosis of CMECs, upregulated PTP1B expression, and inhibited the PI3K/Akt pathway. Furthermore, cotreatment with PTP1B inhibitor significantly decreased the number of apoptotic CMECs induced by Ang II, along with increased PI3K expression, phosphorylation of Akt and the ratio of Bcl-2/Bax, decreased caspase-3 activity, and a cleaved caspase-3/caspase-3 ratio, while treatment with LY294002 partly inhibited the anti-apoptotic effect of the PTP1B inhibitor. Ang II induces apoptosis of primary rat CMECs via regulating the PTP1B/PI3K/Akt pathway.
Collapse
|
15
|
Ito Y, Fukui M, Kanda M, Morishita K, Shoji Y, Kitao T, Hinoi E, Shirahase H. Therapeutic effects of the allosteric protein tyrosine phosphatase 1B inhibitor KY-226 on experimental diabetes and obesity via enhancements in insulin and leptin signaling in mice. J Pharmacol Sci 2018; 137:38-46. [DOI: 10.1016/j.jphs.2018.03.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Revised: 02/13/2018] [Accepted: 02/26/2018] [Indexed: 01/14/2023] Open
|
16
|
Yang Z, Wu F, He Y, Zhang Q, Zhang Y, Zhou G, Yang H, Zhou P. A novel PTP1B inhibitor extracted fromGanoderma lucidumameliorates insulin resistance by regulating IRS1-GLUT4 cascades in the insulin signaling pathway. Food Funct 2018; 9:397-406. [PMID: 29215104 DOI: 10.1039/c7fo01489a] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
A schematic diagram showing the IRS1-GLUT4 insulin signaling pathway influenced by PTP1B and FYGL in L6 cells.
Collapse
Affiliation(s)
- Zhou Yang
- State Key Laboratory of Molecular Engineering of Polymers
- Department of Macromolecular Science
- Fudan University
- Shanghai 200433
- P. R. China
| | - Fan Wu
- State Key Laboratory of Molecular Engineering of Polymers
- Department of Macromolecular Science
- Fudan University
- Shanghai 200433
- P. R. China
| | - Yanming He
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine
- Shanghai University of Traditional Chinese Medicine
- Shanghai 200437
- P. R. China
| | - Qiang Zhang
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine
- Shanghai University of Traditional Chinese Medicine
- Shanghai 200437
- P. R. China
| | - Yuan Zhang
- Department of Medicine
- St Vincent's Hospital
- University of Melbourne
- Fitzroy
- Australia
| | - Guangrong Zhou
- State Key Laboratory of Molecular Engineering of Polymers
- Department of Macromolecular Science
- Fudan University
- Shanghai 200433
- P. R. China
| | - Hongjie Yang
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine
- Shanghai University of Traditional Chinese Medicine
- Shanghai 200437
- P. R. China
| | - Ping Zhou
- State Key Laboratory of Molecular Engineering of Polymers
- Department of Macromolecular Science
- Fudan University
- Shanghai 200433
- P. R. China
| |
Collapse
|
17
|
Perez-Suarez I, Ponce-González JG, de La Calle-Herrero J, Losa-Reyna J, Martin-Rincon M, Morales-Alamo D, Santana A, Holmberg HC, Calbet JAL. Severe energy deficit upregulates leptin receptors, leptin signaling, and PTP1B in human skeletal muscle. J Appl Physiol (1985) 2017; 123:1276-1287. [DOI: 10.1152/japplphysiol.00454.2017] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Revised: 06/22/2017] [Accepted: 07/19/2017] [Indexed: 11/22/2022] Open
Abstract
In obesity, leptin receptors (OBR) and leptin signaling in skeletal muscle are downregulated. To determine whether OBR and leptin signaling are upregulated with a severe energy deficit, 15 overweight men were assessed before the intervention (PRE), after 4 days of caloric restriction (3.2 kcal·kg body wt−1·day−1) in combination with prolonged exercise (CRE; 8 h walking + 45 min single-arm cranking/day) to induce an energy deficit of ~5,500 kcal/day, and following 3 days of control diet (isoenergetic) and reduced exercise (CD). During CRE, the diet consisted solely of whey protein ( n = 8) or sucrose ( n = 7; 0.8 g·kg body wt−1·day−1). Muscle biopsies were obtained from the exercised and the nonexercised deltoid muscles and from the vastus lateralis. From PRE to CRE, serum glucose, insulin, and leptin were reduced. OBR expression was augmented in all examined muscles associated with increased maximal fat oxidation. Compared with PRE, after CD, phospho-Tyr1141OBR, phospho-Tyr985OBR, JAK2, and phospho-Tyr1007/1008JAK2 protein expression were increased in all muscles, whereas STAT3 and phospho-Tyr705STAT3 were increased only in the arms. The expression of protein tyrosine phosphatase 1B (PTP1B) in skeletal muscle was increased by 18 and 45% after CRE and CD, respectively ( P < 0.05). Suppressor of cytokine signaling 3 (SOCS3) tended to increase in the legs and decrease in the arm muscles (ANOVA interaction: P < 0.05). Myosin heavy chain I isoform was associated with OBR protein expression ( r = −0.75), phospho-Tyr985OBR ( r = 0.88), and phospho-Tyr705STAT3/STAT3 ( r = 0.74). In summary, despite increased PTP1B expression, skeletal muscle OBR and signaling are upregulated by a severe energy deficit with greater response in the arm than in the legs likely due to SOCS3 upregulation in the leg muscles. NEW & NOTEWORTHY This study shows that the skeletal muscle leptin receptors and their corresponding signaling cascade are upregulated in response to a severe energy deficit, contributing to increase maximal fat oxidation. The responses are more prominent in the arm muscles than in the legs but partly blunted by whey protein ingestion and high volume of exercise. This occurs despite an increase of protein tyrosine phosphatase 1B protein expression, a known inhibitor of insulin and leptin signaling.
Collapse
Affiliation(s)
- Ismael Perez-Suarez
- Department of Physical Education, University of Las Palmas de Gran Canaria, Las Palmas de Gran Canaria, Spain
- Research Institute of Biomedical and Health Sciences, Las Palmas de Gran Canaria, Spain; and
| | | | - Jaime de La Calle-Herrero
- Department of Physical Education, University of Las Palmas de Gran Canaria, Las Palmas de Gran Canaria, Spain
| | - Jose Losa-Reyna
- Department of Physical Education, University of Las Palmas de Gran Canaria, Las Palmas de Gran Canaria, Spain
| | - Marcos Martin-Rincon
- Department of Physical Education, University of Las Palmas de Gran Canaria, Las Palmas de Gran Canaria, Spain
- Research Institute of Biomedical and Health Sciences, Las Palmas de Gran Canaria, Spain; and
| | - David Morales-Alamo
- Department of Physical Education, University of Las Palmas de Gran Canaria, Las Palmas de Gran Canaria, Spain
- Research Institute of Biomedical and Health Sciences, Las Palmas de Gran Canaria, Spain; and
| | - Alfredo Santana
- Clinical Genetics Unit, Complejo Hospitalario Universitario Insular-Materno Infantil de Las Palmas de Gran Canaria, Las Palmas de Gran Canaria, Spain
- Research Institute of Biomedical and Health Sciences, Las Palmas de Gran Canaria, Spain; and
| | - Hans-Christer Holmberg
- Swedish Winter Sports Research Centre, Department of Health Sciences, Mid Sweden University, Östersund, Sweden
| | - Jose A. L. Calbet
- Department of Physical Education, University of Las Palmas de Gran Canaria, Las Palmas de Gran Canaria, Spain
- Research Institute of Biomedical and Health Sciences, Las Palmas de Gran Canaria, Spain; and
| |
Collapse
|
18
|
IRE1α links Nck1 deficiency to attenuated PTP1B expression in HepG2 cells. Cell Signal 2017; 36:79-90. [DOI: 10.1016/j.cellsig.2017.04.017] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Revised: 04/11/2017] [Accepted: 04/23/2017] [Indexed: 12/23/2022]
|
19
|
Shah R, Murthy V, Pacold M, Danielson K, Tanriverdi K, Larson MG, Hanspers K, Pico A, Mick E, Reis J, de Ferranti S, Freinkman E, Levy D, Hoffmann U, Osganian S, Das S, Freedman JE. Extracellular RNAs Are Associated With Insulin Resistance and Metabolic Phenotypes. Diabetes Care 2017; 40:546-553. [PMID: 28183786 PMCID: PMC5360281 DOI: 10.2337/dc16-1354] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Accepted: 01/07/2017] [Indexed: 02/03/2023]
Abstract
OBJECTIVE Insulin resistance (IR) is a hallmark of obesity and metabolic disease. Circulating extracellular RNAs (ex-RNAs), stable RNA molecules in plasma, may play a role in IR, though most studies on ex-RNAs in IR are small. We sought to characterize the relationship between ex-RNAs and metabolic phenotypes in a large community-based human cohort. RESEARCH DESIGN AND METHODS We measured circulating plasma ex-RNAs in 2,317 participants without diabetes in the Framingham Heart Study (FHS) Offspring Cohort at cycle 8 and defined associations between ex-RNAs and IR (measured by circulating insulin level). We measured association between candidate ex-RNAs and markers of adiposity. Sensitivity analyses included individuals with diabetes. In a separate cohort of 90 overweight/obese youth, we measured selected ex-RNAs and metabolites. Biology of candidate microRNAs was investigated in silico. RESULTS The mean age of FHS participants was 65.8 years (56% female), with average BMI 27.7 kg/m2; participants in the youth cohort had a mean age of 15.5 years (60% female), with mean BMI 33.8 kg/m2. In age-, sex-, and BMI-adjusted models across 391 ex-RNAs in FHS, 18 ex-RNAs were associated with IR (of which 16 were microRNAs). miR-122 was associated with IR and regional adiposity in adults and IR in children (independent of metabolites). Pathway analysis revealed metabolic regulatory roles for miR-122, including regulation of IR pathways (AMPK, target of rapamycin signaling, and mitogen-activated protein kinase). CONCLUSIONS These results provide translational evidence in support of an important role of ex-RNAs as novel circulating factors implicated in IR.
Collapse
Affiliation(s)
- Ravi Shah
- Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Venkatesh Murthy
- Department of Medicine and Radiology, University of Michigan-Ann Arbor, Ann Arbor, MI
| | - Michael Pacold
- Metabolomics Core, Whitehead Institute, Massachusetts Institute of Technology, Boston, MA
| | - Kirsty Danielson
- Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | | | - Martin G Larson
- Biostatistics Department, Boston University School of Public Health, Boston, MA
| | | | | | - Eric Mick
- University of Massachusetts at Worcester, Worcester, MA
| | - Jared Reis
- Division of Cardiovascular Sciences, National Heart, Lung, and Blood Institute, Bethesda, MD
| | - Sarah de Ferranti
- Preventative Cardiology, Department of Medicine and Cardiology, Boston Children's Hospital, Boston, MA
| | - Elizaveta Freinkman
- Metabolomics Core, Whitehead Institute, Massachusetts Institute of Technology, Boston, MA
| | - Daniel Levy
- Division of Cardiovascular Sciences, National Heart, Lung, and Blood Institute, Bethesda, MD
| | - Udo Hoffmann
- Department of Radiology, Massachusetts General Hospital, Boston, MA
| | - Stavroula Osganian
- Department of Medicine, Division of General Pediatrics, Boston Children's Hospital, Boston, MA
| | - Saumya Das
- Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | | |
Collapse
|
20
|
Promising Inhibitory Effects of Anthraquinones, Naphthopyrone, and Naphthalene Glycosides, from Cassia obtusifolia on α-Glucosidase and Human Protein Tyrosine Phosphatases 1B. Molecules 2016; 22:molecules22010028. [PMID: 28035984 PMCID: PMC6155831 DOI: 10.3390/molecules22010028] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Revised: 12/22/2016] [Accepted: 12/23/2016] [Indexed: 11/18/2022] Open
Abstract
The present work aims to evaluate the anti-diabetic potentials of 16 anthraquinones, two naphthopyrone glycosides, and one naphthalene glycoside from Cassia obtusifolia via inhibition against the protein tyrosine phosphatases 1B (PTP1B) and α-glucosidase. Among them, anthraquinones emodin and alaternin exhibited the highest inhibitory activities on PTP1B and α-glucosidase, respectively. Moreover, we examined the effects of alaternin and emodin on stimulation of glucose uptake by insulin-resistant human HepG2 cells. The results showed that alaternin and emodin significantly increased the insulin-provoked glucose uptake. In addition, our kinetic study revealed that alaternin competitively inhibited PTP1B, and showed mixed-type inhibition against α-glucosidase. In order to confirm enzyme inhibition, we predicted the 3D structure of PTP1B using Autodock 4.2 to simulate the binding of alaternin. The docking simulation results demonstrated that four residues of PTP1B (Gly183, Arg221, Ile219, Gly220) interact with three hydroxyl groups of alaternin and that the binding energy was negative (−6.30 kcal/mol), indicating that the four hydrogen bonds stabilize the open form of the enzyme and potentiate tight binding of the active site of PTP1B, resulting in more effective PTP1B inhibition. The results of the present study clearly demonstrate that C. obtusifolia and its constituents have potential anti-diabetic activity and can be used as a functional food for the treatment of diabetes and associated complications.
Collapse
|
21
|
Protamine zinc insulin combined with sodium selenite improves glycometabolism in the diabetic KKAy mice. Sci Rep 2016; 6:26563. [PMID: 27212152 PMCID: PMC4876423 DOI: 10.1038/srep26563] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2016] [Accepted: 05/05/2016] [Indexed: 12/22/2022] Open
Abstract
Long-term, high dosage protamine zinc insulin (PZI) treatments produce adverse reactions. The trace element selenium (Se) is a candidate for the prevention of diabetes due to anti-oxidative stress activity and the regulation of glycometabolism. In this study, we aimed to investigate the anti-diabetic effects of a combination of PZI and Se on type 2 diabetes. Diabetic KKAy mice were randomized into the following groups: model group and groups that were subcutaneously injected with PZI, Se, high or low dose PZI + Se for 6 weeks. PZI combined with Se decreased the body weight and fasting blood glucose levels. Moreover, this treatment also improved insulin tolerance, as determined by the reduced values from the oral glucose tolerance test and insulin tolerance test, and increased insulin levels and insulin sensitivity index. PZI combined with Se ameliorated skeletal muscle and β-cell damage and the impaired mitochondrial morphology. Oxidative stress was also reduced. Furthermore, PZI combined with Se upregulated phosphatidylinositol 3-kinase (PI3K) and downregulated protein tyrosine phosphatase 1B (PTP1B). Importantly, the low dosage combination produced effects similar to PZI alone. In conclusion, PZI combined with Se improved glycometabolism and ameliorated the tissue and mitochondrial damage, which might be associated with the PI3K and PTP1B pathways.
Collapse
|
22
|
Eleftheriou P, Petrou A, Geronikaki A, Liaras K, Dirnali S, Anna M. Prediction of enzyme inhibition and mode of inhibitory action based on calculation of distances between hydrogen bond donor/acceptor groups of the molecule and docking analysis: An application on the discovery of novel effective PTP1B inhibitors. SAR AND QSAR IN ENVIRONMENTAL RESEARCH 2015; 26:557-576. [PMID: 26294069 DOI: 10.1080/1062936x.2015.1074939] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2015] [Accepted: 07/16/2015] [Indexed: 06/04/2023]
Abstract
PTP1B is a protein tyrosine phosphatase involved in insulin receptor desensitization. PTP1B inhibition prolongs the activated state of the receptor, practically enhancing the effect of insulin. Thus PTP1B has become a drug target for the treatment of type II diabetes. PTP1b is an enzyme with multiple binding sites for competitive and allosteric inhibitors. Prediction of inhibitory action using docking analysis has limited success in case of enzymes with multiple binding sites, since the selection of the right crystal structure depends on the kind of inhibitor. In the present study, a two-step strategy for the prediction of PTP1b inhibitory action was applied to 12 compounds. Based on the study of known inhibitors, we isolated the structural characteristics required for binding to each binding site. As a first step, 3D-structures of the molecules were produced and their structural parameters were measured and used for prediction of the binding site of the compound. These results were used for the selection of the appropriate crystal structure for docking analysis of each compound, and the final prediction was based on the estimated binding energies. This strategy effectively predicted the activity of all compounds. A linear correlation was found between estimated binding energy and inhibition measured in vitro (r = -0.894).
Collapse
Affiliation(s)
- P Eleftheriou
- a Department of Medical Laboratory Studies, School of Health and Medical Care , Alexander Technological Educational Institute of Thessaloniki , Greece
| | - A Petrou
- b Department of Medicinal Chemistry, School of Medicine , Aristotle University of Thessaloniki , Greece
| | - A Geronikaki
- b Department of Medicinal Chemistry, School of Medicine , Aristotle University of Thessaloniki , Greece
| | - K Liaras
- b Department of Medicinal Chemistry, School of Medicine , Aristotle University of Thessaloniki , Greece
| | - S Dirnali
- a Department of Medical Laboratory Studies, School of Health and Medical Care , Alexander Technological Educational Institute of Thessaloniki , Greece
| | - M Anna
- a Department of Medical Laboratory Studies, School of Health and Medical Care , Alexander Technological Educational Institute of Thessaloniki , Greece
| |
Collapse
|
23
|
Herre DJ, Norman JB, Anderson R, Tremblay ML, Huby AC, Belin de Chantemèle EJ. Deletion of Protein Tyrosine Phosphatase 1B (PTP1B) Enhances Endothelial Cyclooxygenase 2 Expression and Protects Mice from Type 1 Diabetes-Induced Endothelial Dysfunction. PLoS One 2015; 10:e0126866. [PMID: 25974252 PMCID: PMC4431674 DOI: 10.1371/journal.pone.0126866] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Accepted: 04/08/2015] [Indexed: 01/13/2023] Open
Abstract
Protein tyrosine phosphatase 1B (PTP1B) dephosphorylates receptors tyrosine kinase and acts as a molecular brake on insulin signaling pathway. Conditions of metabolic dysfunction increase PTP1B, when deletion of PTP1B protects against metabolic disorders by increasing insulin signaling. Although vascular insulin signaling contributes to the control of glucose disposal, little is known regarding the direct role of PTP1B in the control of endothelial function. We hypothesized that metabolic dysfunctions increase PTP1B expression in endothelial cells and that PTP1B deletion prevents endothelial dysfunction in situation of diminished insulin secretion. Type I diabetes (T1DM) was induced in wild-type (WT) and PTP1B-deficient mice (KO) with streptozotocin (STZ) injection. After 28 days of T1DM, KO mice exhibited a similar reduction in body weight and plasma insulin levels and a comparable increase in glycemia (WT: 384±20 vs. Ko: 432±29 mg/dL), cholesterol and triglycerides, as WT mice. T1DM increased PTP1B expression and impaired endothelial NO-dependent relaxation, in mouse aorta. PTP1B deletion did not affect baseline endothelial function, but preserved endothelium-dependent relaxation, in T1DM mice. NO synthase inhibition with L-NAME abolished endothelial relaxation in control and T1DM WT mice, whereas L-NAME and the cyclooxygenases inhibitor indomethacin were required to abolish endothelium relaxation in T1DM KO mice. PTP1B deletion increased COX-2 expression and PGI2 levels, in mouse aorta and plasma respectively, in T1DM mice. In parallel, simulation of diabetic conditions increased PTP1B expression and knockdown of PTP1B increased COX-2 but not COX-1 expression, in primary human aortic endothelial cells. Taken together these data indicate that deletion of PTP1B protected endothelial function by compensating the reduction in NO bioavailability by increasing COX-2-mediated release of the vasodilator prostanoid PGI2, in T1DM mice.
Collapse
Affiliation(s)
- David J. Herre
- Physiology Department, Medical College of Georgia at Georgia Regents University, Augusta, GA, United States of America
| | - J. Blake Norman
- Physiology Department, Medical College of Georgia at Georgia Regents University, Augusta, GA, United States of America
| | - Ruchi Anderson
- Physiology Department, Medical College of Georgia at Georgia Regents University, Augusta, GA, United States of America
| | - Michel L. Tremblay
- Goodman Cancer Center and Department of Biochemistry, McGill University, Montreal, QC, Canada
| | - Anne-Cecile Huby
- Physiology Department, Medical College of Georgia at Georgia Regents University, Augusta, GA, United States of America
| | - Eric J. Belin de Chantemèle
- Physiology Department, Medical College of Georgia at Georgia Regents University, Augusta, GA, United States of America
- * E-mail:
| |
Collapse
|
24
|
Liu ZQ, Liu T, Chen C, Li MY, Wang ZY, Chen RS, Wei GX, Wang XY, Luo DQ. Fumosorinone, a novel PTP1B inhibitor, activates insulin signaling in insulin-resistance HepG2 cells and shows anti-diabetic effect in diabetic KKAy mice. Toxicol Appl Pharmacol 2015; 285:61-70. [PMID: 25796170 DOI: 10.1016/j.taap.2015.03.011] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Revised: 03/03/2015] [Accepted: 03/10/2015] [Indexed: 10/23/2022]
Abstract
Insulin resistance is a characteristic feature of type 2 diabetes mellitus (T2DM) and is characterized by defects in insulin signaling. Protein tyrosine phosphatase 1B (PTP1B) is a key negative regulator of the insulin signaling pathways, and its increased activity and expression are implicated in the pathogenesis of insulin resistance. Therefore, the inhibition of PTP1B is anticipated to become a potential therapeutic strategy to treat T2DM. Fumosorinone (FU), a new natural product isolated from insect fungi Isaria fumosorosea, was found to inhibit PTP1B activity in our previous study. Herein, the effects of FU on insulin resistance and mechanism in vitro and in vivo were investigated. FU increased the insulin-provoked glucose uptake in insulin-resistant HepG2 cells, and also reduced blood glucose and lipid levels of type 2 diabetic KKAy mice. FU decreased the expression of PTP1B both in insulin-resistant HepG2 cells and in liver tissues of diabetic KKAy mice. Furthermore, FU increased the phosphorylation of IRβ, IRS-2, Akt, GSK3β and Erk1/2 in insulin-resistant HepG2 cells, as well as the phosphorylation of IRβ, IRS-2, Akt in liver tissues of diabetic KKAy mice. These results showed that FU increased glucose uptake and improved insulin resistance by down-regulating the expression of PTP1B and activating the insulin signaling pathway, suggesting that it may possess antidiabetic properties.
Collapse
Affiliation(s)
- Zhi-Qin Liu
- College of Life Sciences, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, Hebei University, Baoding 071002, PR China; College of Pharmaceutical Sciences, key laboratory of pharmaceutical quality control of Hebei province, Hebei University, Baoding 071002, PR China
| | - Ting Liu
- College of Life Sciences, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, Hebei University, Baoding 071002, PR China
| | - Chuan Chen
- College of Life Sciences, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, Hebei University, Baoding 071002, PR China
| | - Ming-Yan Li
- College of Pharmaceutical Sciences, key laboratory of pharmaceutical quality control of Hebei province, Hebei University, Baoding 071002, PR China
| | - Zi-Yu Wang
- College of Pharmaceutical Sciences, key laboratory of pharmaceutical quality control of Hebei province, Hebei University, Baoding 071002, PR China
| | - Ruo-Song Chen
- College of Pharmaceutical Sciences, key laboratory of pharmaceutical quality control of Hebei province, Hebei University, Baoding 071002, PR China
| | - Gui-Xiang Wei
- College of Pharmaceutical Sciences, key laboratory of pharmaceutical quality control of Hebei province, Hebei University, Baoding 071002, PR China
| | - Xiao-Yi Wang
- College of Pharmaceutical Sciences, key laboratory of pharmaceutical quality control of Hebei province, Hebei University, Baoding 071002, PR China
| | - Du-Qiang Luo
- College of Life Sciences, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, Hebei University, Baoding 071002, PR China.
| |
Collapse
|
25
|
Wang LJ, Jiang B, Wu N, Wang SY, Shi DY. Natural and semisynthetic protein tyrosine phosphatase 1B (PTP1B) inhibitors as anti-diabetic agents. RSC Adv 2015. [DOI: 10.1039/c5ra01754h] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Natural products offered more opportunities to develop new drugs and leading compounds as potent PTP1B inhibitors for treating T2DM.
Collapse
Affiliation(s)
- Li-Jun Wang
- Institute of Oceanology
- Chinese Academy of Sciences
- Qingdao
- China
| | - Bo Jiang
- Institute of Oceanology
- Chinese Academy of Sciences
- Qingdao
- China
| | - Ning Wu
- Institute of Oceanology
- Chinese Academy of Sciences
- Qingdao
- China
| | - Shuai-Yu Wang
- Institute of Oceanology
- Chinese Academy of Sciences
- Qingdao
- China
| | - Da-Yong Shi
- Institute of Oceanology
- Chinese Academy of Sciences
- Qingdao
- China
| |
Collapse
|
26
|
Li H, Dusseault J, Larose L. Nck1 depletion induces activation of the PI3K/Akt pathway by attenuating PTP1B protein expression. Cell Commun Signal 2014; 12:71. [PMID: 25398386 PMCID: PMC4236421 DOI: 10.1186/s12964-014-0071-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Accepted: 10/19/2014] [Indexed: 12/17/2022] Open
Abstract
Background Activation of the PI3K/Akt pathway mediates crucial cellular functions regulated by receptor tyrosine kinases, such as cell growth, proliferation, survival and metabolism. Previously, we reported that the whole-body knockout of the Src homology domain-containing adaptor protein Nck1 improves overall glucose homeostasis and insulin-induced activation of the PI3K/Akt pathway in liver of obese mice. The aim of the current study is to elucidate the mechanism by which Nck1 depletion regulates hepatic insulin signaling. Results Here, we demonstrate that Nck1 regulates the activation of the PI3K/Akt pathway in a protein tyrosine phosphatase 1B (PTP1B)-dependent mechanism. Indeed, depletion of Nck1 by siRNA in HepG2 cells enhances PI3K-dependent basal and growth factor-induced Akt activation. In accordance, primary hepatocytes isolated from Nck1−/− mice also display enhanced Akt activation in response to insulin. Activation of the PI3K/Akt pathway in Nck1-depleted HepG2 cells relies on higher levels of tyrosine-phosphorylated proteins and correlates with decreased PTP1B levels. Interestingly, Nck1 and PTP1B in cells are found in a common molecular complex and their interaction is dependent on the SH3 domains of Nck1. Finally, Nck1 depletion in HepG2 cells neither affects PTP1B gene transcription nor PTP1B protein stability, suggesting that Nck1 modulates PTP1B expression at the translational level. Conclusion Our study provides strong evidence supporting that the adaptor protein Nck1 interacts with PTP1B and also regulates PTP1B expression. In this manner, Nck1 plays a role in regulating the PI3K/Akt pathway.
Collapse
Affiliation(s)
- Hui Li
- Department of Medicine, Polypeptide Laboratory, McGill University and The Research Institute of McGill University Health Centre, Montreal, QC, Canada.
| | - Julie Dusseault
- Department of Medicine, Polypeptide Laboratory, McGill University and The Research Institute of McGill University Health Centre, Montreal, QC, Canada.
| | - Louise Larose
- Department of Medicine, Polypeptide Laboratory, McGill University and The Research Institute of McGill University Health Centre, Montreal, QC, Canada.
| |
Collapse
|
27
|
Guadalupe-Grau A, Larsen S, Guerra B, Calbet JAL, Dela F, Helge JW. Influence of age on leptin induced skeletal muscle signalling. Acta Physiol (Oxf) 2014; 211:214-28. [PMID: 24605926 DOI: 10.1111/apha.12273] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2013] [Revised: 06/03/2013] [Accepted: 03/04/2014] [Indexed: 12/27/2022]
Abstract
AIM Age associated fat mass accumulation could be because of dysregulation of leptin signalling in skeletal muscle. Thus, we investigated total protein expression and phosphorylation levels of the long isoform of the leptin receptor (OB-Rb), and leptin signalling through janus kinase 2 (JAK2)/signal transducer and activator of transcription 3 (STAT3), insulin receptor substrate 1 (IRS-1), AMP-activated protein kinase (AMPK) and acetyl-coenzyme A carboxylase (ACC), combined with the leptin signalling inhibitors suppressor of cytokine signalling 3 (SOCS3) and protein tyrosine phosphatase 1B (PTP1B) in human skeletal muscle of different age. METHODS Vastus lateralis muscle biopsies were obtained from 39 men matched for BMI < 30 kg m(-2) and separated into three groups: 13 young (Y, 24 ± 4 years); 14 middle aged (MA, 44 ± 5 years) and 12 aged (A, 58 ± 8 years) subjects. RESULTS Whole body fat percentage and plasma leptin were higher (P < 0.05), whereas lean mass, plasma free testosterone and total testosterone were lower (P < 0.05) in A compared to Y. Skeletal muscle OB-Rb (170 KDa) protein expression and pTyr(1141) -OB-R170 were comparable between groups, whereas pTyr(985) -OB-R170 was lower in A compared to Y (P < 0.05). pSTAT3 levels tended (P = 0.09) to be lower (50%) in A compared to Y. In A, muscle PTP1B was greater and IRS-1 lower than Y and MA respectively (P < 0.05). PTyr(612) -IRS-1 tended to be lower in A than in Y (P = 0.09). Suppressor of cytokine signalling 3 (SOCS3) protein expression, pJAK2, pSer(1101) -IRS-1, pAMPKα and pACCβ were similar between groups. CONCLUSION Age is associated with dysregulation of the leptin signalling and increased PTP1B protein expression in skeletal muscle.
Collapse
Affiliation(s)
- A. Guadalupe-Grau
- Department of Physical Education; University of Las Palmas de Gran Canaria; Las Palmas de Gran Canaria Spain
- Xlab; Department of Biomedical Sciences; Faculty of Health Sciences; Center for Healthy Aging; University of Copenhagen; Copenhagen Denmark
| | - S. Larsen
- Xlab; Department of Biomedical Sciences; Faculty of Health Sciences; Center for Healthy Aging; University of Copenhagen; Copenhagen Denmark
| | - B. Guerra
- Department of Clinical Sciences; Molecular and Translational Endocrinology Group; Associate Unit of University of Las Palmas de Gran Canaria and Biomedical Institute “Alberto Sols” - CSIC; Las Palmas de Gran Canaria Spain
- ICIC; Cancer Research Institute of the Canary Islands; Las Palmas de Gran Canaria Spain
- Instituto Universitario de Investigaciones Biomédicas y Sanitarias; Las Palmas de Gran Canaria Spain
| | - J. A. L. Calbet
- Department of Physical Education; University of Las Palmas de Gran Canaria; Las Palmas de Gran Canaria Spain
- Instituto Universitario de Investigaciones Biomédicas y Sanitarias; Las Palmas de Gran Canaria Spain
| | - F. Dela
- Xlab; Department of Biomedical Sciences; Faculty of Health Sciences; Center for Healthy Aging; University of Copenhagen; Copenhagen Denmark
| | - J. W. Helge
- Xlab; Department of Biomedical Sciences; Faculty of Health Sciences; Center for Healthy Aging; University of Copenhagen; Copenhagen Denmark
| |
Collapse
|
28
|
Maeda A, Kai K, Ishii M, Ishii T, Akagawa M. Safranal, a novel protein tyrosine phosphatase 1B inhibitor, activates insulin signaling in C2C12 myotubes and improves glucose tolerance in diabetic KK-Aymice. Mol Nutr Food Res 2014; 58:1177-89. [DOI: 10.1002/mnfr.201300675] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2013] [Revised: 12/24/2013] [Accepted: 01/02/2014] [Indexed: 02/05/2023]
Affiliation(s)
- Ayumi Maeda
- Department of Biological Chemistry; Division of Applied Life Science; Graduate School of Life and Environmental Sciences; Osaka Prefecture University; Sakai Japan
| | - Kenji Kai
- Department of Biological Chemistry; Division of Applied Life Science; Graduate School of Life and Environmental Sciences; Osaka Prefecture University; Sakai Japan
| | - Megumi Ishii
- Department of Biological Chemistry; Division of Applied Life Science; Graduate School of Life and Environmental Sciences; Osaka Prefecture University; Sakai Japan
| | - Takeshi Ishii
- Department of Food and Nutritional Sciences, and Global COE Program; University of Shizuoka; Shizuoka Japan
| | - Mitsugu Akagawa
- Department of Biological Chemistry; Division of Applied Life Science; Graduate School of Life and Environmental Sciences; Osaka Prefecture University; Sakai Japan
| |
Collapse
|
29
|
Ji W, Chen X, Lv J, Wang M, Ren S, Yuan B, Wang B, Chen L. Liraglutide Exerts Antidiabetic Effect via PTP1B and PI3K/Akt2 Signaling Pathway in Skeletal Muscle of KKAy Mice. Int J Endocrinol 2014; 2014:312452. [PMID: 25183970 PMCID: PMC4144308 DOI: 10.1155/2014/312452] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2014] [Revised: 07/22/2014] [Accepted: 07/22/2014] [Indexed: 02/06/2023] Open
Abstract
Background. Liraglutide (a glucagon-like peptide 1 analog) was used for the treatment of type 2 diabetes (T2DM) which could produce glucose-dependent insulin secretion. Aim. The aim was to investigate whether liraglutide could improve myofibril and mitochondria injury in skeletal muscle and the mechanisms in diabetic KKAy mice. Method. We divided the male KKAy mice into 2 groups: liraglutide group (250 μg/kg/day liraglutide subcutaneous injection) and model group; meanwhile, the male C57BL/6J mice were considered as the control. After 6 weeks, the ultrastructure of skeletal muscle was observed by electron microscope. The gene expressions of protein tyrosine phosphatase 1B (PTP1B), phosphatidylinositol 3-kinase (PI3K), and glucose transporter type 4 (GLUT4) were determined by real-time PCR. The protein levels of the above molecules and phospho-Akt2 (p-Akt2) were measured by Western blot. Results. Liraglutide significantly ameliorated the injury of mitochondria by increasing the number (+441%) and the area (+113%) of mitochondria and mitochondrial area/100 µm(2) (+396%) in skeletal muscle of KKAy mice. The results of real-time PCR and Western blot showed that liraglutide downregulated PTP1B while it upregulated PI3K and GLUT4 (P < 0.01). The protein level of p-Akt2/Akt2 was also increased (P < 0.01). Conclusion. These results revealed that liraglutide could improve myofibril and mitochondria injury in skeletal muscle against T2DM via PTP1B and PI3K/Akt2 signaling pathway.
Collapse
Affiliation(s)
- Wenjun Ji
- College of Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Xinlin Chen
- College of Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Juan Lv
- Xi'an No. 1 Hospital, Xi'an, Shaanxi 710002, China
| | - Meng Wang
- College of Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Shuting Ren
- College of Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Bingxiang Yuan
- College of Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Bing Wang
- College of Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Lina Chen
- College of Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
- *Lina Chen:
| |
Collapse
|
30
|
Halenova TI, Taras Shevchenko National University of Kyiv, Ukraine. ISOLATION AND CHARACTERIZATION OF INSULIN RECEPTOR OF PLASMA MEMBRANES OF RAT LIVER CELLS AT MODEL OF TYPE 2 DIABETES. BIOTECHNOLOGIA ACTA 2014. [DOI: 10.15407/biotech7.03.081] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
31
|
Owen C, Lees EK, Grant L, Zimmer DJ, Mody N, Bence KK, Delibegović M. Inducible liver-specific knockdown of protein tyrosine phosphatase 1B improves glucose and lipid homeostasis in adult mice. Diabetologia 2013; 56:2286-96. [PMID: 23832083 DOI: 10.1007/s00125-013-2992-z] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2013] [Accepted: 06/14/2013] [Indexed: 12/19/2022]
Abstract
AIMS/HYPOTHESIS Protein tyrosine phosphatase 1B (PTP1B) is a key negative regulator of insulin signalling. Hepatic PTP1B deficiency, using the Alb-Cre promoter to drive Ptp1b deletion from birth in mice, improves glucose homeostasis, insulin sensitivity and lipid metabolism. The aim of this study was to investigate the therapeutic potential of decreasing liver PTP1B levels in obese and insulin-resistant adult mice. METHODS Inducible Ptp1b liver-specific knockout mice were generated using SA-Cre-ER(T2) mice crossed with Ptp1b floxed (Ptp1b(fl/fl)) mice. Mice were fed a high-fat diet (HFD) for 12 weeks to induce obesity and insulin resistance. Tamoxifen was administered in the HFD to induce liver-specific deletion of Ptp1b (SA-Ptp1b(-/-) mice). Body weight, glucose homeostasis, lipid homeostasis, serum adipokines, insulin signalling and endoplasmic reticulum (ER) stress were examined. RESULTS Despite no significant change in body weight relative to HFD-fed Ptp1b(fl/fl) control mice, HFD-fed SA-Ptp1b(-/-) mice exhibited a reversal of glucose intolerance as determined by improved glucose and pyruvate tolerance tests, decreased fed and fasting blood glucose and insulin levels, lower HOMA of insulin resistance, circulating leptin, serum and liver triacylglycerols, serum NEFA and decreased HFD-induced ER stress. This was associated with decreased glycogen synthase, eukaryotic translation initiation factor-2α kinase 3, eukaryotic initiation factor 2α and c-Jun NH2-terminal kinase 2 phosphorylation, and decreased expression of Pepck. CONCLUSIONS/INTERPRETATION Inducible liver-specific PTP1B knockdown reverses glucose intolerance and improves lipid homeostasis in HFD-fed obese and insulin-resistant adult mice. This suggests that knockdown of liver PTP1B in individuals who are already obese/insulin resistant may have relatively rapid, beneficial therapeutic effects.
Collapse
Affiliation(s)
- C Owen
- Institute of Medical Sciences, School of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, UK
| | | | | | | | | | | | | |
Collapse
|
32
|
Krüger J, Trappiel M, Dagnell M, Stawowy P, Meyborg H, Böhm C, Bhanot S, Ostman A, Kintscher U, Kappert K. Targeting density-enhanced phosphatase-1 (DEP-1) with antisense oligonucleotides improves the metabolic phenotype in high-fat diet-fed mice. Cell Commun Signal 2013; 11:49. [PMID: 23889985 PMCID: PMC3734182 DOI: 10.1186/1478-811x-11-49] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2013] [Accepted: 07/08/2013] [Indexed: 01/27/2023] Open
Abstract
Background Insulin signaling is tightly controlled by tyrosine dephosphorylation of the insulin receptor through protein-tyrosine-phosphatases (PTPs). DEP-1 is a PTP dephosphorylating tyrosine residues in a variety of receptor tyrosine kinases. Here, we analyzed whether DEP-1 activity is differentially regulated in liver, skeletal muscle and adipose tissue under high-fat diet (HFD), examined the role of DEP-1 in insulin resistance in vivo, and its function in insulin signaling. Results Mice were fed an HFD for 10 weeks to induce obesity-associated insulin resistance. Thereafter, HFD mice were subjected to systemic administration of specific antisense oligonucleotides (ASOs), highly accumulating in hepatic tissue, against DEP-1 or control ASOs. Targeting DEP-1 led to improvement of insulin sensitivity, reduced basal glucose level, and significant reduction of body weight. This was accompanied by lower insulin and leptin serum levels. Suppression of DEP-1 in vivo also induced hyperphosphorylation in the insulin signaling cascade of the liver. Moreover, DEP-1 physically associated with the insulin receptor in situ, and recombinant DEP-1 dephosphorylated the insulin receptor in vitro. Conclusions These results indicate that DEP-1 acts as an endogenous antagonist of the insulin receptor, and downregulation of DEP-1 results in an improvement of insulin sensitivity. DEP-1 may therefore represent a novel target for attenuation of metabolic diseases.
Collapse
|
33
|
Mandavia C, Sowers JR. Phosphoprotein Phosphatase PP2A Regulation of Insulin Receptor Substrate 1 and Insulin Metabolic Signaling. Cardiorenal Med 2012; 2:308-313. [PMID: 23381670 DOI: 10.1159/000343889] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2012] [Accepted: 09/24/2012] [Indexed: 12/19/2022] Open
Abstract
Insulin (INS) metabolic signaling is important for normal cardiovascular and renal function as well as for exerting the classic actions of INS, such as glucose uptake in skeletal muscle tissue. There is emerging evidence that tyrosine phosphatases as well as protein kinases have important modulating roles in INS metabolic signaling in both cardiovascular and classically INS- sensitive tissues. For example, increases in phosphatase activity may partially explain how angiotensin II and aldosterone attenuate activation of the INS receptor substrate protein 1 (IRS-1)-phosphatidylinositol 3 kinase-protein kinase B pathway, thereby promoting INS resistance. On the other hand, phosphatase activation may also exert beneficial and cardiovascular protective effects in conditions such as overnutrition by blocking serine phosphorylation of IRS-1, thereby improving downstream INS metabolic signaling. Both the beneficial and the detrimental effects exerted by the activation of phosphatases will be covered in this report.
Collapse
Affiliation(s)
- Chirag Mandavia
- Department of Internal Medicine, University of Missouri-Columbia School of Medicine, Columbia, Mo., USA
| | | |
Collapse
|
34
|
Takada M, Sumi M, Maeda A, Watanabe F, Kamiya T, Ishii T, Nakano M, Akagawa M. Pyrroloquinoline quinone, a novel protein tyrosine phosphatase 1B inhibitor, activates insulin signaling in C2C12 myotubes and improves impaired glucose tolerance in diabetic KK-Ay mice. Biochem Biophys Res Commun 2012; 428:315-20. [DOI: 10.1016/j.bbrc.2012.10.055] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2012] [Accepted: 10/12/2012] [Indexed: 01/22/2023]
|