1
|
Boruah A, Afzal NU, Saha S, Mazumdar A, Ozah D, Bora T, Prabhakaran P, Manna P, Roy A. Enhanced glucose regulation potential of C-peptide mimics. Org Biomol Chem 2025. [PMID: 40309967 DOI: 10.1039/d5ob00318k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2025]
Abstract
The connecting peptide (C-peptide), once relegated as an epiphenomenon in insulin biosynthesis, has now been recognized for its capacity to instigate molecular effects along with important physiological functions. These findings suggest that C-peptide is a hormonally active molecule responsible for controlling a number of diabetes-related complications, in addition to proinsulin processing. Notably, it demonstrates cellular responsiveness and acts as a robust biomarker for beta cell functions, with a half-life longer than that of insulin. Herein, we investigated the effect of some synthetic C-peptide mimics on glucose homeostasis, specifically focusing on glucose uptake, GLUT4 translocation and associated membrane trafficking processes. The glucose utilization potential of some of the C-peptide mimics in L6 muscle myotubes was significantly better than that of the human C-peptide. One of the synthesized C-peptide mimics, CP8, showed glucose metabolism akin to insulin, including potential for Akt phosphorylation and IRβ autophosphorylation. Also, it showed the ability to mitigate intramolecular reactive oxygen species production. The results obtained here highlight the potential of C-peptide mimics on glucose metabolism, as well as interaction with Akt and IRβ, positioning them as promising candidates for future diabetes research.
Collapse
Affiliation(s)
- Alpana Boruah
- Chemical Sciences and Technology Division, CSIR - North East Institute of Science and Technology (CSIR-NEIST), Pulibor, Jorhat-785006, Assam, India.
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad-201002, India
| | - Nazim Uddin Afzal
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad-201002, India
- Center for Infectious Diseases, CSIR - North East Institute of Science and Technology (CSIR-NEIST), Pulibor, Jorhat-785006, Assam, India.
| | - Sayari Saha
- Chemical Sciences and Technology Division, CSIR - North East Institute of Science and Technology (CSIR-NEIST), Pulibor, Jorhat-785006, Assam, India.
| | - Anusmrita Mazumdar
- Chemical Sciences and Technology Division, CSIR - North East Institute of Science and Technology (CSIR-NEIST), Pulibor, Jorhat-785006, Assam, India.
| | - Dibyajyoti Ozah
- Clinical Center, CSIR - North East Institute of Science and Technology (CSIR-NEIST), Pulibor, Jorhat-785006, Assam, India
| | - Thaneswar Bora
- Clinical Center, CSIR - North East Institute of Science and Technology (CSIR-NEIST), Pulibor, Jorhat-785006, Assam, India
| | - Panchami Prabhakaran
- School of Chemical Sciences, Central University of Gujarat, Gandhinagar, 382030, Gujarat, India
| | - Prasenjit Manna
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad-201002, India
- Center for Infectious Diseases, CSIR - North East Institute of Science and Technology (CSIR-NEIST), Pulibor, Jorhat-785006, Assam, India.
| | - Arup Roy
- Chemical Sciences and Technology Division, CSIR - North East Institute of Science and Technology (CSIR-NEIST), Pulibor, Jorhat-785006, Assam, India.
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad-201002, India
| |
Collapse
|
2
|
Helmold BR, Ahrens A, Fitzgerald Z, Ozdinler PH. Spastin and alsin protein interactome analyses begin to reveal key canonical pathways and suggest novel druggable targets. Neural Regen Res 2025; 20:725-739. [PMID: 38886938 PMCID: PMC11433914 DOI: 10.4103/nrr.nrr-d-23-02068] [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: 12/21/2023] [Revised: 03/18/2024] [Accepted: 04/05/2024] [Indexed: 06/20/2024] Open
Abstract
Developing effective and long-term treatment strategies for rare and complex neurodegenerative diseases is challenging. One of the major roadblocks is the extensive heterogeneity among patients. This hinders understanding the underlying disease-causing mechanisms and building solutions that have implications for a broad spectrum of patients. One potential solution is to develop personalized medicine approaches based on strategies that target the most prevalent cellular events that are perturbed in patients. Especially in patients with a known genetic mutation, it may be possible to understand how these mutations contribute to problems that lead to neurodegeneration. Protein-protein interaction analyses offer great advantages for revealing how proteins interact, which cellular events are primarily involved in these interactions, and how they become affected when key genes are mutated in patients. This line of investigation also suggests novel druggable targets for patients with different mutations. Here, we focus on alsin and spastin, two proteins that are identified as "causative" for amyotrophic lateral sclerosis and hereditary spastic paraplegia, respectively, when mutated. Our review analyzes the protein interactome for alsin and spastin, the canonical pathways that are primarily important for each protein domain, as well as compounds that are either Food and Drug Administration-approved or are in active clinical trials concerning the affected cellular pathways. This line of research begins to pave the way for personalized medicine approaches that are desperately needed for rare neurodegenerative diseases that are complex and heterogeneous.
Collapse
Affiliation(s)
- Benjamin R. Helmold
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Angela Ahrens
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Zachary Fitzgerald
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - P. Hande Ozdinler
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Center for Molecular Innovation and Drug Discovery, Center for Developmental Therapeutics, Chemistry of Life Processes Institute, Northwestern University, Evanston, IL, USA
- Mesulam Center for Cognitive Neurology and Alzheimer’s Disease, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Feinberg School of Medicine, Les Turner ALS Center at Northwestern University, Chicago, IL, USA
| |
Collapse
|
3
|
Kondo M, Sawada K, Matsuda Y, Abe M, Sanechika N, Takanashi Y, Mori Y, Kimura M, Toyoda M. Study of the Effects of Deuterium-Depleted Water on the Expression of GLUT4 and Insulin Resistance in the Muscle Cell Line C2C12. Biomedicines 2024; 12:1771. [PMID: 39200235 PMCID: PMC11351524 DOI: 10.3390/biomedicines12081771] [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: 07/10/2024] [Revised: 07/29/2024] [Accepted: 08/04/2024] [Indexed: 09/02/2024] Open
Abstract
Deuterium-depleted water (DDW) is used in the treatment of many diseases, including cancer and diabetes. To detect the effect of DDW on gene expression and activation of the insulin-responsive transporter GLUT4 as a mechanism for improving the pathology of diabetes, we investigated the GLUT4 expression and glucose uptake at various concentrations of DDW using the myoblast cell line C2C12 differentiated into myotubes. GLUT4 gene expression significantly increased under deuterium depletion, reaching a maximum value at a deuterium concentration of approximately 50 ppm, which was approximately nine times that of natural water with a deuterium concentration of 150 ppm. GLUT4 protein also showed an increase at similar DDW concentrations. The membrane translocation of GLUT4 by insulin stimulation reached a maximum value at a deuterium concentration of approximately 50-75 ppm, which was approximately 2.2 times that in natural water. Accordingly, glucose uptake also increased by up to 2.2 times at a deuterium concentration of approximately 50 ppm. Drug-induced insulin resistance was attenuated, and the glucose uptake was four times higher in the presence of 10 ng/mL TNF-α and three times higher in the presence of 1 μg/mL resistin at a deuterium concentration of approximately 50 ppm relative to natural water. These results suggest that DDW promotes GLUT4 expression and insulin-stimulated activation in muscle cells and reduces insulin resistance, making it an effective treatment for diabetes.
Collapse
Affiliation(s)
- Masumi Kondo
- Division of Nephrology, Endocrinology and Metabolism, Department of Medicine, Tokai University Hachioji Hospital, 1838 Ishikawacho, Hachioji 192-0032, Japan; (M.K.); (M.A.)
| | - Kaichiro Sawada
- Division of Nephrology, Endocrinology and Metabolism, Department of Medicine, Tokai University School of Medicine, 143 Shimokasuya, Isehara 259-1193, Japan; (K.S.); (Y.M.); (N.S.); (Y.T.); (Y.M.); (M.K.)
| | - Yosuke Matsuda
- Division of Nephrology, Endocrinology and Metabolism, Department of Medicine, Tokai University School of Medicine, 143 Shimokasuya, Isehara 259-1193, Japan; (K.S.); (Y.M.); (N.S.); (Y.T.); (Y.M.); (M.K.)
| | - Makiko Abe
- Division of Nephrology, Endocrinology and Metabolism, Department of Medicine, Tokai University Hachioji Hospital, 1838 Ishikawacho, Hachioji 192-0032, Japan; (M.K.); (M.A.)
| | - Noriyuki Sanechika
- Division of Nephrology, Endocrinology and Metabolism, Department of Medicine, Tokai University School of Medicine, 143 Shimokasuya, Isehara 259-1193, Japan; (K.S.); (Y.M.); (N.S.); (Y.T.); (Y.M.); (M.K.)
| | - Yumi Takanashi
- Division of Nephrology, Endocrinology and Metabolism, Department of Medicine, Tokai University School of Medicine, 143 Shimokasuya, Isehara 259-1193, Japan; (K.S.); (Y.M.); (N.S.); (Y.T.); (Y.M.); (M.K.)
| | - Yoshitaka Mori
- Division of Nephrology, Endocrinology and Metabolism, Department of Medicine, Tokai University School of Medicine, 143 Shimokasuya, Isehara 259-1193, Japan; (K.S.); (Y.M.); (N.S.); (Y.T.); (Y.M.); (M.K.)
| | - Moritsugu Kimura
- Division of Nephrology, Endocrinology and Metabolism, Department of Medicine, Tokai University School of Medicine, 143 Shimokasuya, Isehara 259-1193, Japan; (K.S.); (Y.M.); (N.S.); (Y.T.); (Y.M.); (M.K.)
| | - Masao Toyoda
- Division of Nephrology, Endocrinology and Metabolism, Department of Medicine, Tokai University School of Medicine, 143 Shimokasuya, Isehara 259-1193, Japan; (K.S.); (Y.M.); (N.S.); (Y.T.); (Y.M.); (M.K.)
| |
Collapse
|
4
|
Sherlock BD, Boon MAA, Vlasiou M, Coster ACF. The Distance Between: An Algorithmic Approach to Comparing Stochastic Models to Time-Series Data. Bull Math Biol 2024; 86:111. [PMID: 39060776 PMCID: PMC11282162 DOI: 10.1007/s11538-024-01331-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 06/21/2024] [Indexed: 07/28/2024]
Abstract
While mean-field models of cellular operations have identified dominant processes at the macroscopic scale, stochastic models may provide further insight into mechanisms at the molecular scale. In order to identify plausible stochastic models, quantitative comparisons between the models and the experimental data are required. The data for these systems have small sample sizes and time-evolving distributions. The aim of this study is to identify appropriate distance metrics for the quantitative comparison of stochastic model outputs and time-evolving stochastic measurements of a system. We identify distance metrics with features suitable for driving parameter inference, model comparison, and model validation, constrained by data from multiple experimental protocols. In this study, stochastic model outputs are compared to synthetic data across three scales: that of the data at the points the system is sampled during the time course of each type of experiment; a combined distance across the time course of each experiment; and a combined distance across all the experiments. Two broad categories of comparators at each point were considered, based on the empirical cumulative distribution function (ECDF) of the data and of the model outputs: discrete based measures such as the Kolmogorov-Smirnov distance, and integrated measures such as the Wasserstein-1 distance between the ECDFs. It was found that the discrete based measures were highly sensitive to parameter changes near the synthetic data parameters, but were largely insensitive otherwise, whereas the integrated distances had smoother transitions as the parameters approached the true values. The integrated measures were also found to be robust to noise added to the synthetic data, replicating experimental error. The characteristics of the identified distances provides the basis for the design of an algorithm suitable for fitting stochastic models to real world stochastic data.
Collapse
Affiliation(s)
- Brock D Sherlock
- School of Mathematics and Statistics, University of New South Wales, Sydney, NSW, 2052, Australia
- Department of Mathematics and Computer Science, Eindhoven University of Technology, P.O. Box 513, 5600 MB, Eindhoven, The Netherlands
| | - Marko A A Boon
- Department of Mathematics and Computer Science, Eindhoven University of Technology, P.O. Box 513, 5600 MB, Eindhoven, The Netherlands
| | - Maria Vlasiou
- Faculty of Electrical Engineering, Mathematics and Computer Science, University of Twente, P.O. Box 217, 7500 AE, Enschede, The Netherlands
| | - Adelle C F Coster
- School of Mathematics and Statistics, University of New South Wales, Sydney, NSW, 2052, Australia.
| |
Collapse
|
5
|
Zanni-Ruiz E, Mayorga L, Pavarotti M. Flow cytometry protocol for GLUT4-myc detection on cell surfaces. Biosci Rep 2024; 44:BSR20231987. [PMID: 38533799 PMCID: PMC11016532 DOI: 10.1042/bsr20231987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 03/05/2024] [Accepted: 03/26/2024] [Indexed: 03/28/2024] Open
Abstract
Insulin and muscle contraction trigger GLUT4 translocation to the plasma membrane, which increases glucose uptake by muscle cells. Insulin resistance and Type 2 diabetes are the result of impaired GLUT4 translocation. Quantifying GLUT4 translocation is essential for comprehending the intricacies of both physiological and pathophysiological processes involved in glucose metabolism. The most commonly used methods for measuring GLUT4 translocation are the ELISA-type assay and the immunofluorescence assay. While some reports suggest that flow cytometry could be useful in quantifying GLUT4 translocation, this technique is not frequently used. Much of our current understanding of the regulation of GLUT4 has been based on experiments using the rat myoblast cell line (L6 cell) which expresses GLUT4 with a myc epitope on the exofacial loop. In the present study, we use the L6-GLUT4myc cell line to develop a flow cytometry-based approach to detect GLUT4 translocation. Flow cytometry offers the advantages of both immunofluorescence and ELISA-based assays. It allows easy identification of separate cell populations in the sample, similar to immunofluorescence, while providing results based on a population-level analysis of multiple individual cells, like an ELISA-based assay. Our results demonstrate a 0.6-fold increase with insulin stimulation compared with basal conditions. Finally, flow cytometry consistently yielded results across different experiments and exhibited sensitivity under the tested conditions.
Collapse
Affiliation(s)
- Emilia Zanni-Ruiz
- Laboratorio de Transporte Intracelular, Instituto de Histología y Embriología de Mendoza Dr. Mario H Burgos, Mendoza, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
- Universidad Nacional de Cuyo, Mendoza, Argentina
| | - Luis Segundo Mayorga
- Laboratorio de Transporte Intracelular, Instituto de Histología y Embriología de Mendoza Dr. Mario H Burgos, Mendoza, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
- Facultad de Ciencias Exactas y Naturales, Universidad Nacional de Cuyo, Mendoza, Argentina
- Universidad Nacional de Cuyo, Mendoza, Argentina
| | - Martin Alejandro Pavarotti
- Laboratorio de Transporte Intracelular, Instituto de Histología y Embriología de Mendoza Dr. Mario H Burgos, Mendoza, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
- Universidad Nacional de Cuyo, Mendoza, Argentina
| |
Collapse
|
6
|
Gao Y, Li QY, Finni T, Pesola AJ. Enhanced muscle activity during interrupted sitting improves glycemic control in overweight and obese men. Scand J Med Sci Sports 2024; 34:e14628. [PMID: 38629807 DOI: 10.1111/sms.14628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Revised: 03/28/2024] [Accepted: 04/04/2024] [Indexed: 04/19/2024]
Abstract
The efficacy of interrupting prolonged sitting may be influenced by muscle activity patterns. This study examined the effects of interrupting prolonged sitting time with different muscle activity patterns on continuously monitored postprandial glycemic response. Eighteen overweight and obese men (21.0 ± 1.2 years; 28.8 ± 2.2 kg/m2) participated in this randomized four-arm crossover study, including uninterrupted sitting for 8.5 h (SIT) and interruptions in sitting with matched energy expenditure and duration but varying muscle activity: 30-min walking at 4 km/h (ONE), sitting with 3-min walking at 4 km/h (WALK) or squatting (SQUAT) every 45 min for 10 times. Net incremental area under the curve (netiAUC) for glucose was compared between conditions. Quadriceps, hamstring, and gluteal muscles electromyogram (EMG) patterns including averaged muscle EMG amplitude (aEMG) and EMG activity duration were used to predict the effects on glucose netiAUC. Compared with SIT (10.2 mmol/L/h [95%CI 6.3 to 11.7]), glucose netiAUC was lower during sitting interrupted with any countermeasure (ONE 9.2 mmol/L/h [8.0 to 10.4], WALK 7.9 mmol/L/h [6.4 to 9.3], and SQUAT 7.9 mmol/L/h [6.4 to 9.3], all p < 0.05). Furthermore, WALK and SQUAT resulted in a lower glucose netiAUC compared with ONE (both p < 0.05). Only increased aEMG in quadriceps (-0.383 mmol/L/h [-0.581 to -0.184], p < 0.001) and gluteal muscles (-0.322 mmol/L/h [-0.593 to -0.051], p = 0.022) was associated with a reduction in postprandial glycemic response. Collectively, short, frequent walking or squatting breaks effectively enhance glycemic control in overweight and obese men compared to a single bout of walking within prolonged sitting. These superior benefits seem to be associated with increased muscle activity intensity in the targeted muscle groups during frequent transitions from sitting to activity.
Collapse
Affiliation(s)
- Ying Gao
- Department of Sports Science, College of Education, Zhejiang University, Hangzhou, China
| | - Qing-Yang Li
- Department of Sports Science, College of Education, Zhejiang University, Hangzhou, China
| | - Taija Finni
- Faculty of Sport and Health Sciences, Neuromuscular Research Center, University of Jyväskylä, Jyväskylä, Finland
| | - Arto J Pesola
- Active Life Lab, South-Eastern Finland University of Applied Sciences, Mikkeli, Finland
| |
Collapse
|
7
|
Tranter JD, Kumar A, Nair VK, Sah R. Mechanosensing in Metabolism. Compr Physiol 2023; 14:5269-5290. [PMID: 38158369 PMCID: PMC11681368 DOI: 10.1002/cphy.c230005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2024]
Abstract
Electrical mechanosensing is a process mediated by specialized ion channels, gated directly or indirectly by mechanical forces, which allows cells to detect and subsequently respond to mechanical stimuli. The activation of mechanosensitive (MS) ion channels, intrinsically gated by mechanical forces, or mechanoresponsive (MR) ion channels, indirectly gated by mechanical forces, results in electrical signaling across lipid bilayers, such as the plasma membrane. While the functions of mechanically gated channels within a sensory context (e.g., proprioception and touch) are well described, there is emerging data demonstrating functions beyond touch and proprioception, including mechanoregulation of intracellular signaling and cellular/systemic metabolism. Both MR and MS ion channel signaling have been shown to contribute to the regulation of metabolic dysfunction, including obesity, insulin resistance, impaired insulin secretion, and inflammation. This review summarizes our current understanding of the contributions of several MS/MR ion channels in cell types implicated in metabolic dysfunction, namely, adipocytes, pancreatic β-cells, hepatocytes, and skeletal muscle cells, and discusses MS/MR ion channels as possible therapeutic targets. © 2024 American Physiological Society. Compr Physiol 14:5269-5290, 2024.
Collapse
Affiliation(s)
- John D. Tranter
- Department of Internal Medicine, Cardiovascular Division, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Ashutosh Kumar
- Department of Internal Medicine, Cardiovascular Division, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Vinayak K. Nair
- Department of Internal Medicine, Cardiovascular Division, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Rajan Sah
- Department of Internal Medicine, Cardiovascular Division, Washington University School of Medicine, St. Louis, Missouri, USA
- Center for Cardiovascular Research, Washington University, St. Louis, Missouri, USA
- St. Louis VA Medical Center, St. Louis, Missouri, USA
| |
Collapse
|
8
|
Widjaja NA, Caesar LA, Nova S, Ardianah E. Beyond the Scale: Investigating Adiponectin, ICAM-1, and VCAM-1 as Metabolic Markers in Obese Adolescents with Metabolic Syndrome. J Obes 2023; 2023:4574042. [PMID: 37822716 PMCID: PMC10564580 DOI: 10.1155/2023/4574042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 08/04/2023] [Accepted: 08/23/2023] [Indexed: 10/13/2023] Open
Abstract
Background Adiponectin acts to prevent vascular dysfunction due to obesity by inhibiting ICAM-1 and VCAM-1 expressions. Objective We investigate adiponectin ICAM-1, VCAM-1, and metabolic syndrome (MetS) in obese adolescents. Methods A cross-sectional study with healthy obese adolescents aged 13 to 18 years was conducted from October 2019 to January 2020. Statistical analysis conducted was a test of normality and homogeneity tests, ANOVA/Kruskal-Wallis, independent sample T-test/Mann-Whitney U test, and Spearman correlation and determined as significant if p value <0.05. Results 125 obese adolescents were recruited. 42 (33.6%) were obese with MetS (we grouped as MetS) and 83 (66.4%) subjects without MetS (non-MetS group). VCAM-1 was significantly higher on boys with MetS compared to girls with MetS, and even girls with MetS had lower levels of VCAM-1 than boys with non-MetS. ICAM-1 was significantly higher in boys with low-level HDL-c (p < 0.05) and correlated weakly with HDL-c, while adiponectin levels were significantly lower in girls with central obesity and hypertriglyceridemia. Path analysis showed that triglyceride had a direct effect on ICAM-1 but not VCAM-1 in both obese boys and girls. Adiponectin had a negative direct effect on ICAM-1 and VCAM-1 in girls. However, on boys, diastole blood pressure had a negative direct effect, which might be the role of sex hormones indirectly. Conclusion VCAM-1 was significantly higher in boys than girls, which showed that boys had a higher risk of atherosclerosis. ICAM-1 showed no significant difference in both gender and metabolic states. Adiponectin showed a protective effect by lowering ICAM-1 and VCAM-1 directly on girls.
Collapse
Affiliation(s)
- Nur Aisiyah Widjaja
- Faculty of Medicine, Child Health Department, Universitas Airlangga, Jl. Mayjen Prof. Dr. Moestopo No. 47, Surabaya 60132, Indonesia
| | - Leonardo Alexander Caesar
- Faculty of Medicine, Child Health Department, Universitas Airlangga, Jl. Mayjen Prof. Dr. Moestopo No. 47, Surabaya 60132, Indonesia
| | - Suhasta Nova
- Faculty of Medicine, Child Health Department, Universitas Airlangga, Jl. Mayjen Prof. Dr. Moestopo No. 47, Surabaya 60132, Indonesia
| | - Eva Ardianah
- Ikatan Dokter Indonesia Surabaya, Jl. Mayjen Prof. Dr. Moestopo No. 117, Surabaya 60132, Indonesia
| |
Collapse
|
9
|
Wang AJ, Ren J, Wang A, Hascall VC. Heparin and calreticulin interact on the surface of early G0/G1 dividing rat mesangial cells to regulate hyperglycemic glucose-induced responses. J Biol Chem 2023; 299:103074. [PMID: 36858200 PMCID: PMC10060746 DOI: 10.1016/j.jbc.2023.103074] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 02/17/2023] [Accepted: 02/18/2023] [Indexed: 03/02/2023] Open
Abstract
Heparin can block pathological responses associated with diabetic nephropathy in animal models and human patients. Our previous studies showed that the interaction of heparin on the surface of rat mesangial cells (RMCs) entering G1 of cell division in hyperglycemic glucose: 1) blocked glucose uptake by glucose transporter 4; 2) inhibited cytosolic uridine diphosphate-glucose elevation that would occur within 6 h from G0/G1; and 3) prevented subsequent activation of hyaluronan synthesis in intracellular compartments and subsequent inflammatory responses. However, specific proteins that interact with heparin are unresolved. Here, we showed by live cell imaging that fluorescent heparin was rapidly internalized into the cytoplasm and then into the endoplasmic reticulum, Golgi, and nuclei compartments. Biotinylated-heparin was applied onto the surface of growth arrested G0/G1 RMCs in order to extract heparin-binding protein(s). SDS-PAGE gels showed two bands at ∼70 kDa in the extract that were absent when unlabeled heparin was used to compete. Trypsin digests of the bands were analyzed by MS and identified as calreticulin and prelamin A/C. Immunostaining with their antibodies identified the presence of calreticulin on the G0/G1 RMC cell surface. Previous studies have shown that calreticulin can be on the cell surface and can interact with the LDL receptor-related protein, which has been implicated in glucose transport by interaction with glucose transporter 4. Thus, cell surface calreticulin can act as a heparin receptor through a mechanism involving LRP1, which prevents the intracellular responses in high glucose and reprograms the cells to synthesize an extracellular hyaluronan matrix after division.
Collapse
Affiliation(s)
- Andrew Jun Wang
- Department of Biomedical Engineering, Cleveland Clinic, Cleveland, Ohio, USA
| | - Juan Ren
- Department of Biomedical Engineering, Cleveland Clinic, Cleveland, Ohio, USA
| | - Aimin Wang
- Department of Biomedical Engineering, Cleveland Clinic, Cleveland, Ohio, USA
| | - Vincent C Hascall
- Department of Biomedical Engineering, Cleveland Clinic, Cleveland, Ohio, USA.
| |
Collapse
|
10
|
Lee SR, Jeong SH, Mukae M, Jeong KJ, Kwun HJ, Hong EJ. GLUT4 degradation by GLUTFOURINH® in mice resembles moderate-obese diabetes of human with hyperglycemia and low lipid accumulation. Biochim Biophys Acta Mol Basis Dis 2023; 1869:166668. [PMID: 36822448 DOI: 10.1016/j.bbadis.2023.166668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Revised: 01/28/2023] [Accepted: 02/14/2023] [Indexed: 02/23/2023]
Abstract
BACKGROUNDS AND AIMS Type 2 diabetes mellitus (T2D) is a chronic disease characterized by insulin resistance and hyperglycemia. To investigate T2D, genetic and chemical induced hyper-obese rodent models have been experimentally developed. However, establishment of moderate-obese diabetes model will confer diverse opportunities for translational studies. In this study, we found the chemical, GLUTFOURINH® (GFI), induces post-translational degradation of glucose transporter 4 (GLUT4). We aimed to establish novel diabetic model by using GFI. METHODS AND RESULTS Low plasma membrane GLUT4 (pmGLUT4) levels by GFI resulted in reduction of intracellular glucose uptake and TG, and increase of intracellular FFA in A204 cells. Likewise, GFI treatment decreased intracellular TG and increased intracellular FFA levels in Hep3B and 3T3-L1 cells. Mice were administered with GFI (16 mg/kg) for short-term (3-day) and long-term (28- and 31-day) to compared with vehicle injection, HFD model, and T2D model, respectively. Short-term and long-term GFI treatments induced hyperglycemia and hyperinsulinemia with low pmGLUT4 levels. Compared to HFD model, long-term GFI with HFD reduced adipose weight and intracellular TG accumulation, but increased plasma FFA. GFI treatment resulted in insulin resistance by showing low QUICKI and high HOMA-IR values, and low insulin response during insulin tolerance test. Additionally, low pmGLUT4 by GFI heightened hyperglycemia, hyperinsulinemia, and insulin resistance compared to T2D model. CONCLUSIONS In summary, we report GLUT4 degradation by novel chemical (GFI) induces moderate-obese diabetes representing hyperglycemia, insulin resistance and low intracellular lipid accumulation. The GLUT4 degradation by GFI has translational value for studying diseases related to moderate-obese diabetes.
Collapse
Affiliation(s)
- Sang R Lee
- College of Veterinary Medicine, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Su Hee Jeong
- College of Veterinary Medicine, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Moeka Mukae
- College of Veterinary Medicine, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Kang Joo Jeong
- College of Veterinary Medicine, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Hyo-Jung Kwun
- College of Veterinary Medicine, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Eui-Ju Hong
- College of Veterinary Medicine, Chungnam National University, Daejeon 34134, Republic of Korea.
| |
Collapse
|
11
|
Casertano M, Genovese M, Santi A, Pranzini E, Balestri F, Piazza L, Del Corso A, Avunduk S, Imperatore C, Menna M, Paoli P. Evidence of Insulin-Sensitizing and Mimetic Activity of the Sesquiterpene Quinone Avarone, a Protein Tyrosine Phosphatase 1B and Aldose Reductase Dual Targeting Agent from the Marine Sponge Dysidea avara. Pharmaceutics 2023; 15:pharmaceutics15020528. [PMID: 36839851 PMCID: PMC9964544 DOI: 10.3390/pharmaceutics15020528] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 01/18/2023] [Accepted: 01/31/2023] [Indexed: 02/08/2023] Open
Abstract
Type 2 diabetes mellitus (T2DM) is a complex disease characterized by impaired glucose homeostasis and serious long-term complications. First-line therapeutic options for T2DM treatment are monodrug therapies, often replaced by multidrug therapies to ensure that non-responding patients maintain target glycemia levels. The use of multitarget drugs instead of mono- or multidrug therapies has been emerging as a main strategy to treat multifactorial diseases, including T2DM. Therefore, modern drug discovery in its early stages aims to identify potential modulators for multiple targets; for this purpose, exploration of the chemical space of natural products represents a powerful tool. Our study demonstrates that avarone, a sesquiterpene quinone obtained from the sponge Dysidea avara, is capable of inhibiting in vitro PTP1B, the main negative regulator of the insulin receptor, while it improves insulin sensitivity, and mitochondria activity in C2C12 cells. We observe that when avarone is administered alone, it acts as an insulin-mimetic agent. In addition, we show that avarone acts as a tight binding inhibitor of aldose reductase (AKR1B1), the enzyme involved in the development of diabetic complications. Overall, avarone could be proposed as a novel natural hit to be developed as a multitarget drug for diabetes and its pathological complications.
Collapse
Affiliation(s)
- Marcello Casertano
- Department of Pharmacy, University of Naples “Federico II”, Via D. Montesano 49, 80131 Naples, Italy
| | - Massimo Genovese
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale Morgagni 50, 50134 Florence, Italy
| | - Alice Santi
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale Morgagni 50, 50134 Florence, Italy
| | - Erica Pranzini
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale Morgagni 50, 50134 Florence, Italy
| | - Francesco Balestri
- Biochemistry Unit, Department of Biology, University of Pisa, Via S. Zeno 51, 56123 Pisa, Italy
- Interdepartmental Research Center for Marine Pharmacology, Via Bonanno 6, 56126 Pisa, Italy
| | - Lucia Piazza
- Biochemistry Unit, Department of Biology, University of Pisa, Via S. Zeno 51, 56123 Pisa, Italy
| | - Antonella Del Corso
- Biochemistry Unit, Department of Biology, University of Pisa, Via S. Zeno 51, 56123 Pisa, Italy
- Interdepartmental Research Center for Marine Pharmacology, Via Bonanno 6, 56126 Pisa, Italy
| | - Sibel Avunduk
- Medical Laboratory Programme, Vocational School of Health Care, Mugla University, Marmaris 48187, Turkey
| | - Concetta Imperatore
- Department of Pharmacy, University of Naples “Federico II”, Via D. Montesano 49, 80131 Naples, Italy
| | - Marialuisa Menna
- Department of Pharmacy, University of Naples “Federico II”, Via D. Montesano 49, 80131 Naples, Italy
- Correspondence: (M.M.); (P.P.); Tel.: +39-081678518 (M.M.); +39-0552751248 (P.P.)
| | - Paolo Paoli
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale Morgagni 50, 50134 Florence, Italy
- Correspondence: (M.M.); (P.P.); Tel.: +39-081678518 (M.M.); +39-0552751248 (P.P.)
| |
Collapse
|
12
|
Yudhani RD, Sari Y, Nugrahaningsih DAA, Sholikhah EN, Rochmanti M, Purba AKR, Khotimah H, Nugrahenny D, Mustofa M. In Vitro Insulin Resistance Model: A Recent Update. J Obes 2023; 2023:1964732. [PMID: 36714242 PMCID: PMC9876677 DOI: 10.1155/2023/1964732] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 12/28/2022] [Accepted: 01/07/2023] [Indexed: 01/20/2023] Open
Abstract
Insulin resistance, which affects insulin-sensitive tissues, including adipose tissues, skeletal muscle, and the liver, is the central pathophysiological mechanism underlying type 2 diabetes progression. Decreased glucose uptake in insulin-sensitive tissues disrupts insulin signaling pathways, particularly the PI3K/Akt pathway. An in vitro model is appropriate for studying the cellular and molecular mechanisms underlying insulin resistance because it is easy to maintain and the results can be easily reproduced. The application of cell-based models for exploring the pathogenesis of diabetes and insulin resistance as well as for developing drugs for these conditions is well known. However, a comprehensive review of in vitro insulin resistance models is lacking. Therefore, this review was conducted to provide a comprehensive overview and summary of the latest in vitro insulin resistance models, particularly 3T3-L1 (preadipocyte), C2C12 (skeletal muscle), and HepG2 (liver) cell lines induced with palmitic acid, high glucose, or chronic exposure to insulin.
Collapse
Affiliation(s)
- Ratih D. Yudhani
- Department of Pharmacology, Faculty of Medicine, Universitas Sebelas Maret, Jl. Ir. Sutami No. 36A, Surakarta, Central Java 57126, Indonesia
| | - Yulia Sari
- Department of Parasitology, Faculty of Medicine, Universitas Sebelas Maret, Jl. Ir. Sutami No. 36A, Surakarta, Central Java 57126, Indonesia
| | - Dwi A. A. Nugrahaningsih
- Department of Pharmacology and Therapy, Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada, Jl. Farmako, Sekip Utara, Sleman, Daerah Istimewa Yogyakarta 55281, Indonesia
| | - Eti N. Sholikhah
- Department of Pharmacology and Therapy, Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada, Jl. Farmako, Sekip Utara, Sleman, Daerah Istimewa Yogyakarta 55281, Indonesia
| | - Maftuchah Rochmanti
- Department of Anatomy, Histology and Pharmacology, Faculty of Medicine, Universitas Airlangga, Jl Mayjen Prof. Dr. Moestopo 47, Surabaya, East Java 60131, Indonesia
| | - Abdul K. R. Purba
- Department of Anatomy, Histology and Pharmacology, Faculty of Medicine, Universitas Airlangga, Jl Mayjen Prof. Dr. Moestopo 47, Surabaya, East Java 60131, Indonesia
| | - Husnul Khotimah
- Department of Pharmacology, Faculty of Medicine, Universitas Brawijaya, Jl. Veteran, Malang, East Java 65145, Indonesia
| | - Dian Nugrahenny
- Department of Pharmacology, Faculty of Medicine, Universitas Brawijaya, Jl. Veteran, Malang, East Java 65145, Indonesia
| | - Mustofa Mustofa
- Department of Pharmacology and Therapy, Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada, Jl. Farmako, Sekip Utara, Sleman, Daerah Istimewa Yogyakarta 55281, Indonesia
| |
Collapse
|
13
|
GLUT4 translocation and dispersal operate in multiple cell types and are negatively correlated with cell size in adipocytes. Sci Rep 2022; 12:20535. [PMID: 36446811 PMCID: PMC9708847 DOI: 10.1038/s41598-022-24736-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 11/18/2022] [Indexed: 11/30/2022] Open
Abstract
The regulated translocation of the glucose transporter, GLUT4, to the surface of adipocytes and muscle is a key action of insulin. This is underpinned by the delivery and fusion of GLUT4-containing vesicles with the plasma membrane. Recent studies have revealed that a further action of insulin is to mediate the dispersal of GLUT4 molecules away from the site of GLUT4 vesicle fusion with the plasma membrane. Although shown in adipocytes, whether insulin-stimulated dispersal occurs in other cells and/or is exhibited by other proteins remains a matter of debate. Here we show that insulin stimulates GLUT4 dispersal in the plasma membrane of adipocytes, induced pluripotent stem cell-derived cardiomyocytes and HeLa cells, suggesting that this phenomenon is specific to GLUT4 expressed in all cell types. By contrast, insulin-stimulated dispersal of TfR was not observed in HeLa cells, suggesting that the mechanism may be unique to GLUT4. Consistent with dispersal being an important physiological mechanism, we observed that insulin-stimulated GLUT4 dispersal is reduced under conditions of insulin resistance. Adipocytes of different sizes have been shown to exhibit distinct metabolic properties: larger adipocytes exhibit reduced insulin-stimulated glucose transport compared to smaller cells. Here we show that both GLUT4 delivery to the plasma membrane and GLUT4 dispersal are reduced in larger adipocytes, supporting the hypothesis that larger adipocytes are refractory to insulin challenge compared to their smaller counterparts, even within a supposedly homogeneous population of cells.
Collapse
|
14
|
Huang CH, Lin CH, Huang HH, Tsai GJ. Development of Fermented Shrimp Shell Product with Hypoglycemic and Hypolipidemic Effects on Diabetic Rats. Metabolites 2022; 12:metabo12080695. [PMID: 35893262 PMCID: PMC9332839 DOI: 10.3390/metabo12080695] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 07/16/2022] [Accepted: 07/25/2022] [Indexed: 02/04/2023] Open
Abstract
In 2020, approximately 9.3 billion tons of crustaceans were consumed, and 45–48% of shrimp shell (SS) by-products were discarded as waste. In this study, the SS of Litopenaeus vannamei was fermented by Lactobacillus plantarum LV33204, Stenotrophomonas maltophilia LV2122 (strong proteolytic activity), and Aeromonas dhakensis LV1111 (chitin-degrading activity), and the optimal fermentation conditions of liquid-fermented SS was established. Contents of total peptide, astaxanthin, and total phenolic content of the fermented SS were significantly higher than that of unfermented SS. In the presence of fermented SS, glucose uptake and insulin resistance of TNF-α-stimulated FL83B hepatocytes were markedly improved. Furthermore, daily oral supplement of fermented SS to streptozotocin (STZ)/nicotinamide (NA)-induced diabetic rats for 7 weeks significantly reduced plasma glucose and insulin resistance. Meanwhile, ingestion of fermented SS might enhance hepatic catabolism of glucose by increasing hexokinase and glucose-6-phosphate dehydrogenase activity and decreasing glucose-6-phosphatase activity. In addition, the fermented SS downregulated plasma total cholesterol (TG), triglycerides (TCs), low-density lipoprotein cholesterol (LDL-C), liver TG, and TC and lipid peroxidation levels in diabetic rats. In conclusion, a biorefinery process for waste SS was established through mixed strain fermentation. The in vitro and in vivo data reveal that the fermented SS is a promising functional food for the management of diabetic hyperglycemia and hyperlipidemia.
Collapse
Affiliation(s)
- Chung-Hsiung Huang
- Department of Food Science, National Taiwan Ocean University, Keelung 20224, Taiwan; (C.-H.H.); (C.-H.L.); (H.-H.H.)
| | - Chih-Heng Lin
- Department of Food Science, National Taiwan Ocean University, Keelung 20224, Taiwan; (C.-H.H.); (C.-H.L.); (H.-H.H.)
| | - Hsiao-Han Huang
- Department of Food Science, National Taiwan Ocean University, Keelung 20224, Taiwan; (C.-H.H.); (C.-H.L.); (H.-H.H.)
| | - Guo-Jane Tsai
- Center for Marine Bioscience and Biotechnology, National Taiwan Ocean University, Keelung 20224, Taiwan
- Correspondence:
| |
Collapse
|
15
|
Larsen JK, Larsen MR, Birk JB, Steenberg DE, Hingst JR, Højlund K, Chadt A, Al-Hasani H, Deshmukh AS, Wojtaszewski JF, Kjøbsted R. Illumination of the Endogenous Insulin-Regulated TBC1D4 Interactome in Human Skeletal Muscle. Diabetes 2022; 71:906-920. [PMID: 35192682 PMCID: PMC9074744 DOI: 10.2337/db21-0855] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 02/15/2022] [Indexed: 11/13/2022]
Abstract
Insulin-stimulated muscle glucose uptake is a key process in glycemic control. This process depends on the redistribution of glucose transporters to the surface membrane, a process that involves regulatory proteins such as TBC1D1 and TBC1D4. Accordingly, a TBC1D4 loss-of-function mutation in human skeletal muscle is associated with an increased risk of type 2 diabetes, and observations from carriers of a TBC1D1 variant associate this protein to a severe obesity phenotype. Here, we identified interactors of the endogenous TBC1D4 protein in human skeletal muscle by an unbiased proteomics approach. We detected 76 proteins as candidate TBC1D4 interactors. The binding of 12 of these interactors was regulated by insulin, including proteins known to be involved in glucose metabolism (e.g., 14-3-3 proteins and α-actinin-4 [ACTN4]). TBC1D1 also coprecipitated with TBC1D4 and vice versa in both human and mouse skeletal muscle. This interaction was not regulated by insulin or exercise in young, healthy, lean individuals. Similarly, the exercise- and insulin-regulated phosphorylation of the TBC1D1-TBC1D4 complex was intact. In contrast, we observed an altered interaction as well as compromised insulin-stimulated phosphoregulation of the TBC1D1-TBC1D4 complex in muscle of obese individuals with type 2 diabetes. Altogether, we provide a repository of TBC1D4 interactors in human and mouse skeletal muscle that serve as potential regulators of TBC1D4 function and, thus, insulin-stimulated glucose uptake in human skeletal muscle.
Collapse
Affiliation(s)
- Jeppe K. Larsen
- August Krogh Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Magnus R. Larsen
- August Krogh Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Jesper B. Birk
- August Krogh Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Dorte E. Steenberg
- August Krogh Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Janne R. Hingst
- August Krogh Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Kurt Højlund
- Steno Diabetes Center Odense, Odense University Hospital, Odense, Denmark
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Alexandra Chadt
- German Diabetes Center, Leibniz Center for Diabetes Research at the Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Hadi Al-Hasani
- German Diabetes Center, Leibniz Center for Diabetes Research at the Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Atul S. Deshmukh
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Jørgen F.P. Wojtaszewski
- August Krogh Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
- Corresponding authors: Rasmus Kjøbsted, , and Jørgen F.P. Wojtaszewski,
| | - Rasmus Kjøbsted
- August Krogh Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
- Corresponding authors: Rasmus Kjøbsted, , and Jørgen F.P. Wojtaszewski,
| |
Collapse
|
16
|
Roberts BS, Yang CQ, Neher SB. Characterization of lipoprotein lipase storage vesicles in 3T3-L1 adipocytes. J Cell Sci 2022; 135:jcs258734. [PMID: 34382637 PMCID: PMC8403984 DOI: 10.1242/jcs.258734] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 07/10/2021] [Indexed: 12/12/2022] Open
Abstract
Lipoprotein lipase (LPL) is a secreted triglyceride lipase involved in the clearance of very-low-density lipoproteins and chylomicrons from circulation. LPL is expressed primarily in adipose and muscle tissues and transported to the capillary lumen. LPL secretion is regulated by insulin in adipose tissue; however, few studies have examined the regulatory and trafficking steps involved in secretion. Here, we describe the intracellular localization and insulin-dependent trafficking of LPL in 3T3-L1 adipocytes. We compared LPL trafficking to the better characterized trafficking pathways taken by leptin and GLUT4 (also known as SLC2A4). We show that the LPL trafficking pathway shares some characteristics of these other pathways, but that LPL subcellular localization and trafficking are distinct from those of GLUT4 and leptin. LPL secretion occurs slowly in response to insulin and rapidly in response to the Ca2+ ionophore ionomycin. This regulated trafficking is dependent on Golgi protein kinase D and the ADP-ribosylation factor GTPase ARF1. Together, these data give support to a new trafficking pathway for soluble cargo that is active in adipocytes.
Collapse
Affiliation(s)
| | | | - Saskia B. Neher
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
17
|
Fazakerley DJ, Koumanov F, Holman GD. GLUT4 On the move. Biochem J 2022; 479:445-462. [PMID: 35147164 PMCID: PMC8883492 DOI: 10.1042/bcj20210073] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 01/11/2022] [Accepted: 01/14/2022] [Indexed: 12/16/2022]
Abstract
Insulin rapidly stimulates GLUT4 translocation and glucose transport in fat and muscle cells. Signals from the occupied insulin receptor are translated into downstream signalling changes in serine/threonine kinases within timescales of seconds, and this is followed by delivery and accumulation of the glucose transporter GLUT4 at the plasma membrane. Kinetic studies have led to realisation that there are distinct phases of this stimulation by insulin. There is a rapid initial burst of GLUT4 delivered to the cell surface from a subcellular reservoir compartment and this is followed by a steady-state level of continuing stimulation in which GLUT4 recycles through a large itinerary of subcellular locations. Here, we provide an overview of the phases of insulin stimulation of GLUT4 translocation and the molecules that are currently considered to activate these trafficking steps. Furthermore, we suggest how use of new experimental approaches together with phospho-proteomic data may help to further identify mechanisms for activation of these trafficking processes.
Collapse
Affiliation(s)
- Daniel J Fazakerley
- Metabolic Research Laboratories, Wellcome-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge CB2 0QQ, U.K
| | - Francoise Koumanov
- Department for Health, Centre for Nutrition, Exercise, and Metabolism, University of Bath, Bath, Somerset BA2 7AY, U.K
| | - Geoffrey D Holman
- Department of Biology and Biochemistry, University of Bath, Bath, Somerset BA2 7AY, U.K
| |
Collapse
|
18
|
Ma J, Zhang X, Song Y, Qin Y, Tan Y, Zheng L, Cheng B, Xi X. D609 inhibition of phosphatidylcholine-specific phospholipase C attenuates prolonged insulin stimulation-mediated GLUT4 downregulation in 3T3-L1 adipocytes. J Physiol Biochem 2022; 78:355-363. [PMID: 35048323 PMCID: PMC9242966 DOI: 10.1007/s13105-022-00872-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 01/06/2022] [Indexed: 11/30/2022]
Abstract
Glucose uptake is stimulated by insulin via stimulation of glucose transporter 4 (GLUT4) translocation to the plasma membrane from intracellular compartments in adipose tissue and muscles. Insulin stimulation for prolonged periods depletes GLUT4 protein, particularly in highly insulin-responsive GLUT4 storage vesicles. This depletion mainly occurs via H2O2-mediated retromer inhibition. However, the post-receptor mechanism of insulin activation of oxidative stress remains unknown. Here, we show that phosphatidylcholine-specific phospholipase C (PC-PLC) plays an important role in insulin-mediated downregulation of GLUT4. In the study, 3T3-L1 adipocytes were exposed to a PC-PLC inhibitor, tricyclodecan-9-yl-xanthogenate (D609), for 30 min prior to the stimulation with 500 nM insulin for 4 h, weakening the depletion of GLUT4. D609 also prevents insulin-driven H2O2 generation in 3T3-L1 adipocytes. Exogenous PC-PLC and its product, phosphocholine (PCho), also caused GLUT4 depletion and promoted H2O2 generation in 3T3-L1 adipocytes. Furthermore, insulin-mediated the increase in the cellular membrane PC-PLC activity was observed in Amplex Red assays. These results suggested that PC-PLC plays an important role in insulin-mediated downregulation of GLUT4 and that PCho may serve as a signaling molecule.
Collapse
Affiliation(s)
- Jinhui Ma
- Department of Endocrinology, Affiliated Hospital of Hebei University, Baoding, 071000, China
| | - Xu Zhang
- Baoding Maternal and Child Hospital, Baoding, 071000, China
| | - Yankun Song
- School of Medicine, Hebei University, Baoding, 071000, China
| | - Yan Qin
- Central Laboratory, Affiliated Hospital of Hebei University, Baoding, 071000, China
| | - Yinghui Tan
- Central Laboratory, Affiliated Hospital of Hebei University, Baoding, 071000, China
| | - Lishuang Zheng
- Central Laboratory, Affiliated Hospital of Hebei University, Baoding, 071000, China
| | - Baoqian Cheng
- School of Medicine, Hebei Medical University, Shijiazhuang, 050017, China
| | - Xin Xi
- Central Laboratory, Affiliated Hospital of Hebei University, Baoding, 071000, China.
| |
Collapse
|
19
|
Pehlivanoglu S, Sahan OB, Pehlivanoglu S, Aktas Kont K. Epithelial mesenchymal transition regulator TWIST1 transcription factor stimulates glucose uptake through upregulation of GLUT1, GLUT3, and GLUT12 in vitro. In Vitro Cell Dev Biol Anim 2021; 57:933-943. [PMID: 34791627 DOI: 10.1007/s11626-021-00635-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 11/03/2021] [Indexed: 11/25/2022]
Abstract
TWIST1 is a major regulator of epithelial mesenchymal transition process, essential in cancer metastasis. Cancer cells increase glucose uptake capabilities to meet their high energy requirements. In this study, we explored the potential role of TWIST1 on glucose transport into the 293T cells in an insulin-dependent and insulin-independent manner. For this purpose, the ectopic expression of TWIST1 was successfully performed by electroporation. The altered mRNA expressions of GLUT-1, -3, -4, and -12, insulin receptor (InsR), and insulin receptor substrate (IRS)-1 and -2 were assessed in control and TWIST1-overexpressing cells. Glucose uptake rates of the cells were evaluated by fluorometric glucose uptake assay. Our findings showed that the transcriptional expression levels of GLUT-1, -3, and -12 genes were significantly upregulated by TWIST1. However, TWIST1 did not alter the mRNA and protein expressions of the InsR, its substrates (IRS-1 and -2), and GLUT-4 genes in 293T cells which are main factors for insulin-stimulated glucose uptake pathway. Also, the glucose transport activities were significantly increased in TWIST1-overexpressing cells compared to controls due to fetal bovine serum (FBS) stimulation, but there was a slight non-significant difference in insulin stimulation. Thus, our data suggest that TWIST1 could promote glucose uptake independently of insulin and is possible to be evaluated as a metabolic marker in cancer. Further investigations are needed to clarify the precise molecular mechanisms underlying the cells' glucose uptake and consumption during tumorigenesis.
Collapse
Affiliation(s)
- Suray Pehlivanoglu
- Department of Molecular Biology and Genetics, Faculty of Science, Necmettin Erbakan University, Konya, Turkey.
| | - Ozge Burcu Sahan
- Center for Stem Cells Research and Development, Hacettepe University, Ankara, Turkey
| | - Sebnem Pehlivanoglu
- Aziz Sancar Research Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey
| | - Kadriye Aktas Kont
- Department of Molecular Biology and Genetics, Faculty of Science, Necmettin Erbakan University, Konya, Turkey
| |
Collapse
|
20
|
Attie AD, Tang QQ, Bornfeldt KE. The insulin centennial-100 years of milestones in biochemistry. J Biol Chem 2021; 297:101278. [PMID: 34717954 PMCID: PMC8605089 DOI: 10.1016/j.jbc.2021.101278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/13/2021] [Indexed: 11/21/2022] Open
|
21
|
Mitochondrial Antioxidant SkQ1 Has a Beneficial Effect in Experimental Diabetes as Based on the Analysis of Expression of microRNAs and mRNAs for the Oxidative Metabolism Regulators. Antioxidants (Basel) 2021; 10:antiox10111749. [PMID: 34829620 PMCID: PMC8615282 DOI: 10.3390/antiox10111749] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 10/28/2021] [Accepted: 10/28/2021] [Indexed: 12/24/2022] Open
Abstract
Diabetes mellitus and related complications are among the most important problems of the world-leading healthcare systems. Despite their priority, molecular and genetic aspects of diabetes pathogenesis are poorly understood; however, the involvement of oxidative stress in this process is undoubted. Rats with experimental diabetes induced by the intraperitoneal injection of alloxan were subjected to the antioxidant pre-therapy with a series of mitochondria-targeted 10-(6’-plastoquinonyl)decyltriphenylphosphonium (SkQ1) injections and analyzed for the expression of mRNAs and microRNAs by real-time quantitative polymerase chain reaction to identify potential predictors of diabetes. Animals that received SkQ1 before diabetes induction demonstrated lower blood glucose levels compared to the diabetic animals not subjected to the therapy. SkQ1 caused changes in the mRNA levels of genes involved in the cellular defense against free radicals, which indicates a beneficial effect of the pre-therapy. Moreover, similar changes were observed on the epigenetic level, as the microRNA expression patterns not only proved the SkQ1 efficacy but also correlated with the expression levels of their mRNA targets. Oxidative stress and macromolecule damage by free radicals are determining factors in diabetes, which suggests that strategies aimed at restoring the antioxidant status of the cell can be beneficial. Mitochondria-targeted antioxidant SkQ1 demonstrates positive effects on several levels, from the normalization of the blood glucose content to genetic and epigenetic changes. Our results can serve as a basis for the development of novel therapeutic and diagnostic strategies.
Collapse
|
22
|
Attie AD, Tang QQ, Bornfeldt KE. The insulin centennial-100 years of milestones in biochemistry. J Lipid Res 2021; 62:100132. [PMID: 34717951 PMCID: PMC8721491 DOI: 10.1016/j.jlr.2021.100132] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/13/2021] [Indexed: 11/05/2022] Open
|
23
|
Batty SR, Langlais PR. Microtubules in insulin action: what's on the tube? Trends Endocrinol Metab 2021; 32:776-789. [PMID: 34462181 PMCID: PMC8446328 DOI: 10.1016/j.tem.2021.07.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 07/23/2021] [Accepted: 07/26/2021] [Indexed: 11/17/2022]
Abstract
Microtubules (MT) have a role in the intracellular response to insulin stimulation and subsequent glucose transport by glucose transporter 4 (GLUT4), which resides in specialized storage vesicles that travel through the cell. Before GLUT4 is inserted into the plasma membrane for glucose transport, it undergoes complex trafficking through the cell via the integration of cytoskeletal networks. In this review, we highlight the importance of MT elements in insulin action in adipocytes through a summary of MT depolymerization studies, MT-based GLUT4 movement, molecular motor proteins involved in GLUT4 trafficking, as well as MT-related phenomena in response to insulin and links between insulin action and MT-associated proteins.
Collapse
Affiliation(s)
- Skylar R Batty
- Department of Molecular and Cellular Biology, University of Arizona, Tucson, AZ, USA
| | - Paul R Langlais
- Department of Medicine, Division of Endocrinology, University of Arizona College of Medicine, Tucson, AZ, USA.
| |
Collapse
|
24
|
Orphan GPR116 mediates the insulin sensitizing effects of the hepatokine FNDC4 in adipose tissue. Nat Commun 2021; 12:2999. [PMID: 34016966 PMCID: PMC8137956 DOI: 10.1038/s41467-021-22579-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 03/12/2021] [Indexed: 12/22/2022] Open
Abstract
The proper functional interaction between different tissues represents a key component in systemic metabolic control. Indeed, disruption of endocrine inter-tissue communication is a hallmark of severe metabolic dysfunction in obesity and diabetes. Here, we show that the FNDC4-GPR116, liver-white adipose tissue endocrine axis controls glucose homeostasis. We found that the liver primarily controlled the circulating levels of soluble FNDC4 (sFNDC4) and lowering of the hepatokine FNDC4 led to prediabetes in mice. Further, we identified the orphan adhesion GPCR GPR116 as a receptor of sFNDC4 in the white adipose tissue. Upon direct and high affinity binding of sFNDC4 to GPR116, sFNDC4 promoted insulin signaling and insulin-mediated glucose uptake in white adipocytes. Indeed, supplementation with FcsFNDC4 in prediabetic mice improved glucose tolerance and inflammatory markers in a white-adipocyte selective and GPR116-dependent manner. Of note, the sFNDC4-GPR116, liver-adipose tissue axis was dampened in (pre) diabetic human patients. Thus our findings will now allow for harnessing this endocrine circuit for alternative therapeutic strategies in obesity-related pre-diabetes. The soluble bioactive form of the transmembrane protein fibronectin type III domain containing 4 (sFNDC4) has anti-inflammatory effects and improves insulin sensitivity. Here the authors show that liver derived sFNDC4 signals through adipose tissue GPCR GPR116 to promote insulin-mediated glucose uptake.
Collapse
|
25
|
Yoon SY, Yu JS, Hwang JY, So HM, Seo SO, Kim JK, Jang TS, Chung SJ, Kim KH. Phloridzin Acts as an Inhibitor of Protein-Tyrosine Phosphatase MEG2 Relevant to Insulin Resistance. Molecules 2021; 26:molecules26061612. [PMID: 33799458 PMCID: PMC7998658 DOI: 10.3390/molecules26061612] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 03/06/2021] [Accepted: 03/07/2021] [Indexed: 11/16/2022] Open
Abstract
Inhibition of the megakaryocyte protein tyrosine phosphatase 2 (PTP-MEG2, also named PTPN9) activity has been shown to be a potential therapeutic strategy for the treatment of type 2 diabetes. Previously, we reported that PTP-MEG2 knockdown enhances adenosine monophosphate activated protein kinase (AMPK) phosphorylation, suggesting that PTP-MEG2 may be a potential antidiabetic target. In this study, we found that phloridzin, isolated from Ulmus davidiana var. japonica, inhibits the catalytic activity of PTP-MEG2 (half-inhibitory concentration, IC50 = 32 ± 1.06 μM) in vitro, indicating that it could be a potential antidiabetic drug candidate. Importantly, phloridzin stimulated glucose uptake by differentiated 3T3-L1 adipocytes and C2C12 muscle cells compared to that by the control cells. Moreover, phloridzin led to the enhanced phosphorylation of AMPK and Akt relevant to increased insulin sensitivity. Importantly, phloridzin attenuated palmitate-induced insulin resistance in C2C12 muscle cells. We also found that phloridzin did not accelerate adipocyte differentiation, suggesting that phloridzin improves insulin sensitivity without significant lipid accumulation. Taken together, our results demonstrate that phloridzin, an inhibitor of PTP-MEG2, stimulates glucose uptake through the activation of both AMPK and Akt signaling pathways. These results strongly suggest that phloridzin could be used as a potential therapeutic candidate for the treatment of type 2 diabetes.
Collapse
Affiliation(s)
- Sun-Young Yoon
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Korea; (S.-Y.Y.); (J.S.Y.); (J.Y.H.); (H.M.S.); (S.O.S.)
- Department of Cosmetic Science, Kwangju Women’s University, Gwangju 62396, Korea
| | - Jae Sik Yu
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Korea; (S.-Y.Y.); (J.S.Y.); (J.Y.H.); (H.M.S.); (S.O.S.)
| | - Ji Young Hwang
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Korea; (S.-Y.Y.); (J.S.Y.); (J.Y.H.); (H.M.S.); (S.O.S.)
| | - Hae Min So
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Korea; (S.-Y.Y.); (J.S.Y.); (J.Y.H.); (H.M.S.); (S.O.S.)
| | - Seung Oh Seo
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Korea; (S.-Y.Y.); (J.S.Y.); (J.Y.H.); (H.M.S.); (S.O.S.)
| | - Jung Kyu Kim
- School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Korea;
| | - Tae Su Jang
- Department of Medicine, Dankook University, Cheonan, Chungnam 31116, Korea;
| | - Sang J. Chung
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Korea; (S.-Y.Y.); (J.S.Y.); (J.Y.H.); (H.M.S.); (S.O.S.)
- Correspondence: (S.J.C.); (K.H.K.); Tel.: +82-31-290-7703 (S.J.C.); +82-31-290-7700 (K.H.K.)
| | - Ki Hyun Kim
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Korea; (S.-Y.Y.); (J.S.Y.); (J.Y.H.); (H.M.S.); (S.O.S.)
- Correspondence: (S.J.C.); (K.H.K.); Tel.: +82-31-290-7703 (S.J.C.); +82-31-290-7700 (K.H.K.)
| |
Collapse
|
26
|
Giam YH, Shoemark A, Chalmers JD. Neutrophil dysfunction in bronchiectasis: an emerging role for immunometabolism. Eur Respir J 2021; 58:13993003.03157-2020. [DOI: 10.1183/13993003.03157-2020] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Accepted: 01/12/2021] [Indexed: 12/22/2022]
Abstract
Bronchiectasis is a heterogenous disease with multiple underlying causes. The pathophysiology is poorly understood but neutrophilic inflammation and dysfunctional killing of pathogens is believed to be key. There are, however, no licensed therapies for bronchiectasis that directly target neutrophilic inflammation. In this review, we discuss our current understanding of neutrophil dysfunction and therapeutic targeting in bronchiectasis. Immunometabolic reprogramming, a process through which inflammation changes inflammatory cell behaviour by altering intracellular metabolic pathways, is increasingly recognised across multiple inflammatory and autoimmune diseases. Here, we show evidence that much of the neutrophil dysfunction observed in bronchiectasis is consistent with immunometabolic reprogramming. Previous attempts at developing therapies targeting neutrophils have focused on reducing neutrophil numbers, resulting in increased frequency of infections. New approaches are needed and we propose that targeting metabolism could theoretically reverse neutrophil dysfunction and dysregulated inflammation. As an exemplar, 5' adenosine monophosphate (AMP)-activated protein kinase (AMPK) activation has already been shown to reverse phagocytic dysfunction and neutrophil extracellular trap (NET) formation in models of pulmonary disease. AMPK modulates multiple metabolic pathways, including glycolysis which is critical for energy generation in neutrophils. AMPK activators can reverse metabolic reprogramming and are already in clinical use and/or development. We propose the need for a new immunomodulatory approach, rather than an anti-inflammatory approach, to enhance bacterial clearance and reduce bronchiectasis disease severity.
Collapse
|
27
|
Komakula SB, Tiwari AK, Singh S. A novel quantitative assay for analysis of GLUT4 translocation using high content screening. Biomed Pharmacother 2021; 133:111032. [PMID: 33378945 DOI: 10.1016/j.biopha.2020.111032] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 11/11/2020] [Accepted: 11/15/2020] [Indexed: 12/23/2022] Open
Abstract
Insulin resistance is associated with obesity and can lead to several metabolic disorders including type II diabetes, nonalcoholic fatty liver disease and cardiovascular problems. Search for the small molecules which can either induce or mimic the insulin action are of great interest and can be utilized to manage insulin resistance. There are several dietary phytochemicals which can potentially have insulinomimetic action. Nevertheless, high throughput screening methods to test efficiency of small molecules to act as an insulinomimetic are not fully established. In this paper we have performed chemical screen analysis based on GLUT4 translocation using a cell line CHO-HIRC-myc-GLUT4 eGFP that expresses GLUT4-GFP in association with human Insulin receptor. We have established a high content screening-based method which can track and quantify the GLUT4 translocation from perinuclear area to the cell membrane. The assay involves measuring fluorescence intensity in a defined perinuclear area and a defined area along the cell membrane; and the results are expressed as the ratio of fluorescence intensity in the perinuclear to membrane area. The assay could collect real time data of GLUT4 translocation from thousand of cells/ sample and from many such samples in one experiment. We validated the assay using Insulin, insulin mimics/sensitizers and insulin inhibitors. The agonist or antagonists were analyzed for their ability to enhance or block the GLUT4 translocation independent of insulin. The outcome of the assay was correlated by performing glucose uptake assay using differentiated 3T3L1 cells. Using this platform we further identified several plant extracts which had the insulin mimetic action. We confirmed that these plant extracts were non-toxic to the beta cells using RIN mf5cells and 3T3L1 cells. We have identified plant extracts with the potential insulinomimetic action using novel high-content screening approach; these can be further tested for their efficiency in-vivo in pre-clinical trials.
Collapse
Affiliation(s)
- SaiSantosh Babu Komakula
- CSIR-Centre for Cellular and Molecular Biology, Hyderabad, India; Department of Experimental Biology, Wrocław University of Environmental and Life Sciences, Wroclaw, Poland
| | | | - Shashi Singh
- CSIR-Centre for Cellular and Molecular Biology, Hyderabad, India.
| |
Collapse
|
28
|
Gray CW, Coster AC. Models of Membrane-Mediated Processes: Cascades and Cycles in Insulin Action. SYSTEMS MEDICINE 2021. [DOI: 10.1016/b978-0-12-801238-3.11348-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
|
29
|
Characterization of viral insulins reveals white adipose tissue-specific effects in mice. Mol Metab 2020; 44:101121. [PMID: 33220491 PMCID: PMC7770979 DOI: 10.1016/j.molmet.2020.101121] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 11/05/2020] [Accepted: 11/16/2020] [Indexed: 12/11/2022] Open
Abstract
Objective Members of the insulin/insulin-like growth factor (IGF) superfamily are well conserved across the evolutionary tree. We recently showed that four viruses in the Iridoviridae family possess genes that encode proteins highly homologous to human insulin/IGF-1. Using chemically synthesized single-chain (sc), i.e., IGF-1-like, forms of the viral insulin/IGF-1-like peptides (VILPs), we previously showed that they can stimulate human receptors. Because these peptides possess potential cleavage sites to form double chain (dc), i.e., more insulin-like, VILPs, in this study, we have characterized dc forms of VILPs for Grouper iridovirus (GIV), Singapore grouper iridovirus (SGIV) and Lymphocystis disease virus-1 (LCDV-1) for the first time. Methods The dcVILPs were chemically synthesized. Using murine fibroblast cell lines overexpressing insulin receptor (IR-A or IR-B) or IGF1R, we first determined the binding affinity of dcVILPs to the receptors and characterized post-receptor signaling. Further, we used C57BL/6J mice to study the effect of dcVILPs on lowering blood glucose. We designed a 3-h dcVILP in vivo infusion experiment to determine the glucose uptake in different tissues. Results GIV and SGIV dcVILPs bind to both isoforms of human insulin receptor (IR-A and IR-B) and to the IGF1R, and for the latter, show higher affinity than human insulin. These dcVILPs stimulate IR and IGF1R phosphorylation and post-receptor signaling in vitro and in vivo. Both GIV and SGIV dcVILPs stimulate glucose uptake in mice. In vivo infusion experiments revealed that while insulin (0.015 nmol/kg/min) and GIV dcVILP (0.75 nmol/kg/min) stimulated a comparable glucose uptake in heart and skeletal muscle and brown adipose tissue, GIV dcVILP stimulated 2-fold higher glucose uptake in white adipose tissue (WAT) compared to insulin. This was associated with increased Akt phosphorylation and glucose transporter type 4 (GLUT4) gene expression compared to insulin in WAT. Conclusions Our results show that GIV and SGIV dcVILPs are active members of the insulin superfamily with unique characteristics. Elucidating the mechanism of tissue specificity for GIV dcVILP will help us to better understand insulin action, design new analogs that specifically target the tissues and provide new insights into their potential role in disease. Viral insulin/IGF1-like peptides (VILPs) are microbial members of the insulin superfamily. VILPs bind to human IR and IGF1R and stimulate post-receptor signaling. Grouper iridovirus (GIV) VILP has white adipose tissue (WAT)-specific characteristics. GIV VILP stimulates increased glucose uptake in WAT via increased GLUT4 expression.
Collapse
|
30
|
Mitchell R, Mikolajczak M, Kersten C, Fleetwood-Walker S. ErbB1-dependent signalling and vesicular trafficking in primary afferent nociceptors associated with hypersensitivity in neuropathic pain. Neurobiol Dis 2020; 142:104961. [DOI: 10.1016/j.nbd.2020.104961] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 05/26/2020] [Accepted: 06/08/2020] [Indexed: 02/06/2023] Open
|
31
|
Machado A, Pouzolles M, Gailhac S, Fritz V, Craveiro M, López-Sánchez U, Kondo T, Pala F, Bosticardo M, Notarangelo LD, Petit V, Taylor N, Zimmermann VS. Phosphate Transporter Profiles in Murine and Human Thymi Identify Thymocytes at Distinct Stages of Differentiation. Front Immunol 2020; 11:1562. [PMID: 32793218 PMCID: PMC7387685 DOI: 10.3389/fimmu.2020.01562] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 06/15/2020] [Indexed: 12/22/2022] Open
Abstract
Thymocyte differentiation is dependent on the availability and transport of metabolites in the thymus niche. As expression of metabolite transporters is a rate-limiting step in nutrient utilization, cell surface transporter levels generally reflect the cell's metabolic state. The GLUT1 glucose transporter is upregulated on actively dividing thymocytes, identifying thymocytes with an increased metabolism. However, it is not clear whether transporters of essential elements such as phosphate are modulated during thymocyte differentiation. While PiT1 and PiT2 are both phosphate transporters in the SLC20 family, we show here that they exhibit distinct expression profiles on both murine and human thymocytes. PiT2 expression distinguishes thymocytes with high metabolic activity, identifying immature murine double negative (CD4−CD8−) DN3b and DN4 thymocyte blasts as well as immature single positive (ISP) CD8 thymocytes. Notably, the absence of PiT2 expression on RAG2-deficient thymocytes, blocked at the DN3a stage, strongly suggests that high PiT2 expression is restricted to thymocytes having undergone a productive TCRβ rearrangement at the DN3a/DN3b transition. Similarly, in the human thymus, PiT2 was upregulated on early post-β selection CD4+ISP and TCRαβ−CD4hiDP thymocytes co-expressing the CD71 transferrin receptor, a marker of metabolic activity. In marked contrast, expression of the PiT1 phosphate importer was detected on mature CD3+ murine and human thymocytes. Notably, PiT1 expression on CD3+DN thymocytes was identified as a biomarker of an aging thymus, increasing from 8.4 ± 1.5% to 42.4 ± 9.4% by 1 year of age (p < 0.0001). We identified these cells as TCRγδ and, most significantly, NKT, representing 77 ± 9% of PiT1+DN thymocytes by 1 year of age (p < 0.001). Thus, metabolic activity and thymic aging are associated with distinct expression profiles of the PiT1 and PiT2 phosphate transporters.
Collapse
Affiliation(s)
- Alice Machado
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health (NIH), Bethesda, MD, United States.,Institut de Génétique Moléculaire de Montpellier, University of Montpellier, CNRS, Montpellier, France
| | - Marie Pouzolles
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health (NIH), Bethesda, MD, United States
| | - Sarah Gailhac
- Institut de Génétique Moléculaire de Montpellier, University of Montpellier, CNRS, Montpellier, France
| | - Vanessa Fritz
- Institut de Génétique Moléculaire de Montpellier, University of Montpellier, CNRS, Montpellier, France
| | - Marco Craveiro
- Institut de Génétique Moléculaire de Montpellier, University of Montpellier, CNRS, Montpellier, France
| | - Uriel López-Sánchez
- Institut de Génétique Moléculaire de Montpellier, University of Montpellier, CNRS, Montpellier, France
| | - Taisuke Kondo
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health (NIH), Bethesda, MD, United States
| | - Francesca Pala
- Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD, United States
| | - Marita Bosticardo
- Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD, United States
| | - Luigi D Notarangelo
- Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD, United States
| | | | - Naomi Taylor
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health (NIH), Bethesda, MD, United States.,Institut de Génétique Moléculaire de Montpellier, University of Montpellier, CNRS, Montpellier, France
| | - Valérie S Zimmermann
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health (NIH), Bethesda, MD, United States.,Institut de Génétique Moléculaire de Montpellier, University of Montpellier, CNRS, Montpellier, France
| |
Collapse
|
32
|
Luo J, Ting CY, Li Y, McQueen P, Lin TY, Hsu CP, Lee CH. Antagonistic regulation by insulin-like peptide and activin ensures the elaboration of appropriate dendritic field sizes of amacrine neurons. eLife 2020; 9:50568. [PMID: 32175842 PMCID: PMC7075694 DOI: 10.7554/elife.50568] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Accepted: 03/05/2020] [Indexed: 01/09/2023] Open
Abstract
Establishing appropriate sizes and shapes of dendritic arbors is critical for proper wiring of the central nervous system. Here we report that Insulin-like Peptide 2 (DILP2) locally activates transiently expressed insulin receptors in the central dendrites of Drosophila Dm8 amacrine neurons to positively regulate dendritic field elaboration. We found DILP2 was expressed in L5 lamina neurons, which have axonal terminals abutting Dm8 dendrites. Proper Dm8 dendrite morphogenesis and synapse formation required insulin signaling through TOR (target of rapamycin) and SREBP (sterol regulatory element-binding protein), acting in parallel with previously identified negative regulation by Activin signaling to provide robust control of Dm8 dendrite elaboration. A simulation of dendritic growth revealed trade-offs between dendritic field size and robustness when branching and terminating kinetic parameters were constant, but dynamic modulation of the parameters could mitigate these trade-offs. We suggest that antagonistic DILP2 and Activin signals from different afferents appropriately size Dm8 dendritic fields.
Collapse
Affiliation(s)
- Jiangnan Luo
- Section on Neuronal Connectivity, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, United States
| | - Chun-Yuan Ting
- Section on Neuronal Connectivity, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, United States
| | - Yan Li
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan, Republic of China
| | - Philip McQueen
- Mathematical and Statistical Computing Laboratory, Center for Information Technology, National Institutes of Health, Bethesda, United States
| | - Tzu-Yang Lin
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan, Republic of China
| | - Chao-Ping Hsu
- Institute of Chemistry, Academia Sinica, Taipei, Taiwan, Republic of China.,Genome and Systems Biology Degree Program, National Taiwan University and Academia Sinica, Taipei, Taiwan, Republic of China
| | - Chi-Hon Lee
- Section on Neuronal Connectivity, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, United States.,Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan, Republic of China
| |
Collapse
|
33
|
Zhao P, Tian D, Song G, Ming Q, Liu J, Shen J, Liu QH, Yang X. Neferine Promotes GLUT4 Expression and Fusion With the Plasma Membrane to Induce Glucose Uptake in L6 Cells. Front Pharmacol 2019; 10:999. [PMID: 31551792 PMCID: PMC6737894 DOI: 10.3389/fphar.2019.00999] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 08/06/2019] [Indexed: 11/21/2022] Open
Abstract
Glucose transporter 4 (GLUT4) is involved in regulating glucose uptake in striated muscle, liver, and adipose tissue. Neferine is a dibenzyl isoquinoline alkaloid derived from dietary lotus seeds and has multiple pharmacological effects. Therefore, this study investigated neferine’s role in glucose translocation to cell surface, glucose uptake, and GLUT4 expression. In our study, neferine upregulated GLUT4 expression, induced GLUT4 plasma membrane fusion, increased intracellular Ca2+, promoted glucose uptake, and alleviated insulin resistance in L6 cells. Furthermore, neferine significantly activated phosphorylation of AMP-activated protein kinase (AMPK) and protein kinase C (PKC). AMPK and PKC inhibitors blocked neferine-induced GLUT4 expression and increased intracellular Ca2+. While neferine-induced GLUT4 expression and intracellular Ca2+ were inhibited by G protein and PLC inhibitors, only intracellular Ca2+ was inhibited by inositol trisphosphate receptor (IP3R) inhibitors. Thus, neferine promoted GLUT4 expression via the G protein-PLC-PKC and AMPK pathways, inducing GLUT4 plasma membrane fusion and subsequent glucose uptake and increasing intracellular Ca2+ through the G protein-PLC-IP3-IP3R pathway. Treatment with 0 mM extracellular Ca2+ + Ca2+ chelator did not inhibit neferine-induced GLUT4 expression but blocked neferine-induced GLUT4 plasma membrane fusion and glucose uptake, suggesting the latter two are Ca2+-dependent. Therefore, we conclude that neferine is a potential treatment for type 2 diabetes.
Collapse
Affiliation(s)
- Ping Zhao
- Institute for Medical Biology & Hubei Provincial Key Laboratory for Protection and Application of Special Plants in the Wuling Area of China, College of Life Sciences, South-Central University for Nationalities, Wuhan, China.,National Demonstration Center for Experimental Ethnopharmacology Education, South-Central University for Nationalities, Wuhan, China.,Hubei Medical Biology International Science and Technology Cooperation Base, Wuhan, China
| | - Di Tian
- Institute for Medical Biology & Hubei Provincial Key Laboratory for Protection and Application of Special Plants in the Wuling Area of China, College of Life Sciences, South-Central University for Nationalities, Wuhan, China
| | - Guanjun Song
- Institute for Medical Biology & Hubei Provincial Key Laboratory for Protection and Application of Special Plants in the Wuling Area of China, College of Life Sciences, South-Central University for Nationalities, Wuhan, China
| | - Qian Ming
- Institute for Medical Biology & Hubei Provincial Key Laboratory for Protection and Application of Special Plants in the Wuling Area of China, College of Life Sciences, South-Central University for Nationalities, Wuhan, China
| | - Jia Liu
- Institute for Medical Biology & Hubei Provincial Key Laboratory for Protection and Application of Special Plants in the Wuling Area of China, College of Life Sciences, South-Central University for Nationalities, Wuhan, China
| | - Jinhua Shen
- Institute for Medical Biology & Hubei Provincial Key Laboratory for Protection and Application of Special Plants in the Wuling Area of China, College of Life Sciences, South-Central University for Nationalities, Wuhan, China.,Hubei Medical Biology International Science and Technology Cooperation Base, Wuhan, China
| | - Qing-Hua Liu
- Institute for Medical Biology & Hubei Provincial Key Laboratory for Protection and Application of Special Plants in the Wuling Area of China, College of Life Sciences, South-Central University for Nationalities, Wuhan, China.,Hubei Medical Biology International Science and Technology Cooperation Base, Wuhan, China
| | - Xinzhou Yang
- National Demonstration Center for Experimental Ethnopharmacology Education, South-Central University for Nationalities, Wuhan, China.,School of Pharmaceutical Sciences, South-Central University for Nationalities, Wuhan, China
| |
Collapse
|
34
|
Cytosolic N- and C-Termini of the Aspergillus nidulans FurE Transporter Contain Distinct Elements that Regulate by Long-Range Effects Function and Specificity. J Mol Biol 2019; 431:3827-3844. [DOI: 10.1016/j.jmb.2019.07.013] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Revised: 07/04/2019] [Accepted: 07/04/2019] [Indexed: 01/05/2023]
|
35
|
Yoon SY, Kang HJ, Ahn D, Hwang JY, Kwon SJ, Chung SJ. Identification of chebulinic acid as a dual targeting inhibitor of protein tyrosine phosphatases relevant to insulin resistance. Bioorg Chem 2019; 90:103087. [PMID: 31284101 DOI: 10.1016/j.bioorg.2019.103087] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 06/17/2019] [Accepted: 06/25/2019] [Indexed: 01/16/2023]
Abstract
Natural products as antidiabetic agents have been shown to stimulate insulin signaling via the inhibition of the protein tyrosine phosphatases relevant to insulin resistance. Previously, we have identified PTPN9 and DUSP9 as potential antidiabetic targets and a multi-targeting natural product thereof. In this study, knockdown of PTPN11 increased AMPK phosphorylation in differentiated C2C12 muscle cells by 3.8 fold, indicating that PTPN11 could be an antidiabetic target. Screening of a library of 658 natural products against PTPN9, DUSP9, or PTPN11 identified chebulinic acid (CA) as a strong allosteric inhibitor with a slow cooperative binding to PTPN9 (IC50 = 34 nM) and PTPN11 (IC50 = 37 nM), suggesting that it would be a potential antidiabetic candidate. Furthermore, CA stimulated glucose uptake and resulted in increased AMP-activated protein kinase (AMPK) phosphorylation. Taken together, we demonstrated that CA increased glucose uptake as a dual inhibitor of PTPN9 and PTPN11 through activation of the AMPK signaling pathway. These results strongly suggest that CA could be used as a potential therapeutic candidate for the treatment of type 2 diabetes.
Collapse
Affiliation(s)
- Sun-Young Yoon
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Hyo Jin Kang
- Department of Chemistry, Dongguk University, Seoul 100-715, Republic of Korea
| | - Dohee Ahn
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Ji Young Hwang
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Se Jeong Kwon
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Sang J Chung
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea.
| |
Collapse
|
36
|
Wang H, Zhang J, You G. The mechanistic links between insulin and human organic anion transporter 4. Int J Pharm 2019; 555:165-174. [PMID: 30453017 DOI: 10.1016/j.ijpharm.2018.11.040] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Revised: 11/10/2018] [Accepted: 11/15/2018] [Indexed: 11/18/2022]
Abstract
Human organic anion transporter 4 (hOAT4) belongs to a class of organic anion transporters that exert critical function in the secretion, absorption, and distribution of numerous drugs in the body, such as anti-viral drugs, anti-cancer therapeutics, antibiotics, antihypertensive medicine, and anti-inflammatory drugs. hOAT4 is richly existent in the kidney and placenta. We previously established that serum- and glucocorticoid-inducible kinases (sgk) stimulate hOAT4 expression and transport activity by abrogating the inhibitory effect of a ubiquitin ligase Nedd4-2. Insulin is one of the upstream signaling molecules for sgk. We therefore investigated the effect of insulin on hOAT4 function. We showed that insulin stimulated hOAT4 expression and transport activity, and the action of insulin was abolished in cells overexpressing Nedd4-2-specific siRNA to knockdown the endogenous Nedd4-2. We further showed that insulin phosphorylated serine 327 on Nedd4-2 and weakened the interaction between hOAT4 and Nedd4-2. Interestingly, in cells overexpressing sgk2, the stimulatory effect of insulin on hOAT4 was diminished. In addition, the stimulatory effect of insulin on hOAT4 was blocked by wortmannin and buparlisib, two PI3K inhibitors. In conclusion, our study demonstrated that insulin stimulates hOAT4 expression and transport activity by abrogating the inhibition effect of Nedd4-2 on the transporter. Moreover, insulin regulates hOAT4 by competing with sgk2 rather than through sgk2.
Collapse
Affiliation(s)
- Haoxun Wang
- Department of Pharmaceutics, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| | - Jinghui Zhang
- Department of Pharmaceutics, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| | - Guofeng You
- Department of Pharmaceutics, Rutgers, The State University of New Jersey, Piscataway, NJ, USA.
| |
Collapse
|
37
|
Brewer PD, Romenskaia I, Mastick CC. A high-throughput chemical-genetics screen in murine adipocytes identifies insulin-regulatory pathways. J Biol Chem 2018; 294:4103-4118. [PMID: 30591588 DOI: 10.1074/jbc.ra118.006986] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Revised: 12/26/2018] [Indexed: 12/18/2022] Open
Abstract
Pathways linking activation of the insulin receptor to downstream targets of insulin have traditionally been studied using a candidate gene approach. To elucidate additional pathways regulating insulin activity, we performed a forward chemical-genetics screen based on translocation of a glucose transporter 4 (Glut4) reporter expressed in murine 3T3-L1 adipocytes. To identify compounds with known targets, we screened drug-repurposing and natural product libraries. We identified, confirmed, and validated 64 activators and 65 inhibitors that acutely increase or rapidly decrease cell-surface Glut4 in adipocytes stimulated with submaximal insulin concentrations. These agents were grouped by target, chemical class, and mechanism of action. All groups contained multiple hits from a single drug class, and several comprised multiple structurally unrelated hits for a single target. Targets include the β-adrenergic and adenosine receptors. Agonists of these receptors increased and inverse agonists/antagonists decreased cell-surface Glut4 independently of insulin. Additional activators include insulin sensitizers (thiazolidinediones), insulin mimetics, dis-inhibitors (the mTORC1 inhibitor rapamycin), cardiotonic steroids (the Na+/K+-ATPase inhibitor ouabain), and corticosteroids (dexamethasone). Inhibitors include heterocyclic amines (tricyclic antidepressants) and 21 natural product supplements and herbal extracts. Mechanisms of action include effects on Glut4 trafficking, signal transduction, inhibition of protein synthesis, and dissipation of proton gradients. Two pathways that acutely regulate Glut4 translocation were discovered: de novo protein synthesis and endocytic acidification. The mechanism of action of additional classes of activators (tanshinones, dalbergiones, and coumarins) and inhibitors (flavonoids and resveratrol) remains to be determined. These tools are among the most sensitive, responsive, and reproducible insulin-activity assays described to date.
Collapse
Affiliation(s)
- Paul Duffield Brewer
- From the Department of Pharmacology, University of Nevada, Reno School of Medicine, Reno, Nevada 89557
| | - Irina Romenskaia
- From the Department of Pharmacology, University of Nevada, Reno School of Medicine, Reno, Nevada 89557
| | - Cynthia Corley Mastick
- From the Department of Pharmacology, University of Nevada, Reno School of Medicine, Reno, Nevada 89557
| |
Collapse
|
38
|
Ayala CI, Kim J, Neufeld TP. Rab6 promotes insulin receptor and cathepsin trafficking to regulate autophagy induction and activity in Drosophila. J Cell Sci 2018; 131:jcs.216127. [PMID: 30111579 DOI: 10.1242/jcs.216127] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Accepted: 08/02/2018] [Indexed: 01/07/2023] Open
Abstract
The self-degradative process of autophagy is important for energy homeostasis and cytoplasmic renewal. This lysosome-mediated pathway is negatively regulated by the target of rapamycin kinase (TOR) under basal conditions, and requires the vesicle trafficking machinery regulated by Rab GTPases. However, the interactions between autophagy, TOR and Rab proteins remain incompletely understood in vivo Here, we identify Rab6 as a critical regulator of the balance between TOR signaling and autolysosome function. Loss of Rab6 causes an accumulation of enlarged autophagic vesicles resulting in part from a failure to deliver lysosomal hydrolases, rendering autolysosomes with a reduced degradative capacity and impaired turnover. Additionally, Rab6-deficient cells are reduced in size and display defective insulin-TOR signaling as a result of mis-sorting and internalization of the insulin receptor. Our findings suggest that Rab6 acts to maintain the reciprocal regulation between autophagy and TOR activity during distinct nutrient states, thereby balancing autophagosome production and turnover to avoid autophagic stress.
Collapse
Affiliation(s)
- Carlos I Ayala
- Department of Genetics, Cell Biology and Development, 6-160 Jackson Hall, 321 Church St. SE, University of Minnesota, Minneapolis, MN 55455, USA
| | - Jung Kim
- Department of Molecular and Cell Biology, University of California at Berkeley, Berkeley, CA 94720, USA
| | - Thomas P Neufeld
- Department of Genetics, Cell Biology and Development, 6-160 Jackson Hall, 321 Church St. SE, University of Minnesota, Minneapolis, MN 55455, USA
| |
Collapse
|
39
|
Chronic noise-exposure exacerbates insulin resistance and promotes the manifestations of the type 2 diabetes in a high-fat diet mouse model. PLoS One 2018; 13:e0195411. [PMID: 29601606 PMCID: PMC5877872 DOI: 10.1371/journal.pone.0195411] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Accepted: 03/21/2018] [Indexed: 12/30/2022] Open
Abstract
Epidemiological studies have revealed that noise exposure was associated with an increased risk of type 2 diabetes mellitus (T2DM). However, the exact nature of that association remains to be elucidated. The present study is designed to examine the effects of chronic noise exposure on the development of T2DM in combination with a high-fat-diet (HFD) in mice. Here we show that chronic noise exposure at 85 dB SPL (4 h /day, below the safety limit for occupational noise exposure) exaggerated multiple metabolic abnormalities induced by HFD in C57BL/6J male mice, including worsened glucose intolerance, insulin resistance, fasting hyperglycemia and dyslipidemia. Furthermore, noise exposure exhibited a paradoxical impact on fat accumulation and circulating levels of free fatty acid, indicating a potential stimulating effect of noise on lipolysis. These results provide first in vivo supporting evidence for the causative role of noise exposure in diabetogenesis and pinpoint a noise-associated increase in blood free fatty acid levels as a possible mediator accelerating the effect of noise on the development of insulin resistance and T2DM.
Collapse
|
40
|
Sun B, Zhong Z, Wang F, Xu J, Xu F, Kong W, Ling Z, Shu N, Li Y, Wu T, Zhang M, Zhu L, Liu X, Liu L. Atorvastatin impaired glucose metabolism in C2C12 cells partly via inhibiting cholesterol-dependent glucose transporter 4 translocation. Biochem Pharmacol 2018; 150:108-119. [PMID: 29338971 DOI: 10.1016/j.bcp.2018.01.021] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Accepted: 01/08/2018] [Indexed: 10/18/2022]
Abstract
Skeletal muscle accounts for approximately 75% of glucose disposal in body and statins impair glucose metabolism. We aimed to investigate the effect of atorvastatin on glucose metabolism in C2C12 cells. Glucose metabolism and expression of glucose transporter 4 (GLUT4) and hexokinase II (HXKII) were measured following incubation with atorvastatin or pravastatin. Roles of cholesterol in atorvastatin-induced glucose metabolism impairment were investigated via adding cholesterol or mevalonic acid and confirmed by cholesterol depletion with methyl-β-cyclodextrin. Hypercholesterolemia mice induced by high fat diet (HFD) feeding, orally received atorvastatin (6 and 12 mg/kg) or pravastatin (12 mg/kg) for 22 days. Results showed that atorvastatin not pravastatin concentration-dependently impaired glucose consumption, glucose uptake and GLUT4 membrane translocation in C2C12 cells without affecting expression of HXKII or total GLUT4 protein. The atorvastatin-induced alterations were reversed by cholesterol or mevalonic acid. Cholesterol depletion exerted similar impact to atorvastatin, which could be alleviated by cholesterol supplement. Glucose consumption or GLUT4 translocation was positively associated with cellular cholesterol levels. In HFD mice, atorvastatin not pravastatin significantly increased blood glucose levels following glucose or insulin dose and decreased expression of membrane not total GLUT4 protein in muscle. Glucose exposure following glucose or insulin dose was negatively correlated to muscular free cholesterol concentration. Expression of membrane GLUT4 protein was positively related to free cholesterol in muscle. In conclusion, atorvastatin impaired glucose utilization in muscle cells partly via inhibiting GLUT4 membrane translocation due to inhibition of cholesterol synthesis by atorvastatin, at least, partly contributing to glucose intolerance in HFD mice.
Collapse
Affiliation(s)
- Binbin Sun
- Center of Drug Metabolism and Pharmacokinetics, College of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Zeyu Zhong
- Center of Drug Metabolism and Pharmacokinetics, College of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Fan Wang
- Center of Drug Metabolism and Pharmacokinetics, College of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Jiong Xu
- Center of Drug Metabolism and Pharmacokinetics, College of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Feng Xu
- Center of Drug Metabolism and Pharmacokinetics, College of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Weimin Kong
- Center of Drug Metabolism and Pharmacokinetics, College of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Zhaoli Ling
- Center of Drug Metabolism and Pharmacokinetics, College of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Nan Shu
- Center of Drug Metabolism and Pharmacokinetics, College of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Ying Li
- Center of Drug Metabolism and Pharmacokinetics, College of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Tong Wu
- Center of Drug Metabolism and Pharmacokinetics, College of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Mian Zhang
- Center of Drug Metabolism and Pharmacokinetics, College of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Liang Zhu
- Center of Drug Metabolism and Pharmacokinetics, College of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Xiaodong Liu
- Center of Drug Metabolism and Pharmacokinetics, College of Pharmacy, China Pharmaceutical University, Nanjing 210009, China.
| | - Li Liu
- Center of Drug Metabolism and Pharmacokinetics, College of Pharmacy, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
41
|
Norris DM, Geddes TA, James DE, Fazakerley DJ, Burchfield JG. Glucose Transport: Methods for Interrogating GLUT4 Trafficking in Adipocytes. Methods Mol Biol 2018; 1713:193-215. [PMID: 29218527 DOI: 10.1007/978-1-4939-7507-5_15] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
In this chapter we detail methods for the systematic dissection of GLUT4 trafficking. The methods described have been optimized for cultured 3T3-L1 adipocytes, but can be readily adapted to other cell types.
Collapse
Affiliation(s)
- Dougall M Norris
- School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW, 2006, Australia
- The Charles Perkins Centre, The University of Sydney, Sydney, 2006, NSW, Australia
| | - Tom A Geddes
- School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW, 2006, Australia
- The Charles Perkins Centre, The University of Sydney, Sydney, 2006, NSW, Australia
| | - David E James
- School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW, 2006, Australia
- Sydney Medical School, The University of Sydney, Sydney, NSW, 2006, Australia
- The Charles Perkins Centre, The University of Sydney, Sydney, 2006, NSW, Australia
| | - Daniel J Fazakerley
- School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW, 2006, Australia
- The Charles Perkins Centre, The University of Sydney, Sydney, 2006, NSW, Australia
| | - James G Burchfield
- School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW, 2006, Australia.
- The Charles Perkins Centre, The University of Sydney, Sydney, 2006, NSW, Australia.
| |
Collapse
|
42
|
Bridges RJ, Bradbury NA. Cystic Fibrosis, Cystic Fibrosis Transmembrane Conductance Regulator and Drugs: Insights from Cellular Trafficking. Handb Exp Pharmacol 2018; 245:385-425. [PMID: 29460152 DOI: 10.1007/164_2018_103] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The eukaryotic cell is organized into membrane-delineated compartments that are characterized by specific cadres of proteins sustaining biochemically distinct cellular processes. The appropriate subcellular localization of proteins is key to proper organelle function and provides a physiological context for cellular processes. Disruption of normal trafficking pathways for proteins is seen in several genetic diseases, where a protein's absence for a specific subcellular compartment leads to organelle disruption, and in the context of an individual, a disruption of normal physiology. Importantly, several drug therapies can also alter protein trafficking, causing unwanted side effects. Thus, a deeper understanding of trafficking pathways needs to be appreciated as novel therapeutic modalities are proposed. Despite the promising efficacy of novel therapeutic agents, the intracellular bioavailability of these compounds has proved to be a potential barrier, leading to failures in treatments for various diseases and disorders. While endocytosis of drug moieties provides an efficient means of getting material into cells, the subsequent release and endosomal escape of materials into the cytosol where they need to act has been a barrier. An understanding of cellular protein/lipid trafficking pathways has opened up strategies for increasing drug bioavailability. Approaches to enhance endosomal exit have greatly increased the cytosolic bioavailability of drugs and will provide a means of investigating previous drugs that may have been shelved due to their low cytosolic concentration.
Collapse
Affiliation(s)
- Robert J Bridges
- Department of Physiology and Biophysics, Chicago Medical School, North Chicago, IL, USA
| | - Neil A Bradbury
- Department of Physiology and Biophysics, Chicago Medical School, North Chicago, IL, USA.
| |
Collapse
|
43
|
Xian HM, Che H, Qin Y, Yang F, Meng SY, Li XG, Bai YL, Wang LH. Coriolus versicolor aqueous extract ameliorates insulin resistance with PI3K/Akt and p38 MAPK signaling pathways involved in diabetic skeletal muscle. Phytother Res 2017; 32:551-560. [PMID: 29243310 DOI: 10.1002/ptr.6007] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 09/13/2017] [Accepted: 11/13/2017] [Indexed: 01/16/2023]
Affiliation(s)
- Hui-min Xian
- Department of Endocrinology; The Second affiliated Hospital of Harbin Medical University; Harbin Heilongjiang Province 150001 China
| | - Hui Che
- Department of Endocrinology; The Second affiliated Hospital of Harbin Medical University; Harbin Heilongjiang Province 150001 China
| | - Ying Qin
- Department of Endocrinology; The Second affiliated Hospital of Harbin Medical University; Harbin Heilongjiang Province 150001 China
- Department of Pharmacology; College of Pharmacy, Harbin Medical University; Harbin Heilongjiang Province 150001 China
| | - Fan Yang
- Department of Endocrinology; The Second affiliated Hospital of Harbin Medical University; Harbin Heilongjiang Province 150001 China
| | - Song-yan Meng
- Department of Endocrinology; The Second affiliated Hospital of Harbin Medical University; Harbin Heilongjiang Province 150001 China
| | - Xiao-guang Li
- Academician Workstation; Harbin Medical University; Harbin Heilongjiang Province 150001 China
| | - Yun-long Bai
- Department of Pharmacology; College of Pharmacy, Harbin Medical University; Harbin Heilongjiang Province 150001 China
- Institute of Chronic Disease; Heilongjiang Academy of Medical Science; Harbin Heilongjiang Province 150001 China
| | - Li-hong Wang
- Department of Endocrinology; The Second affiliated Hospital of Harbin Medical University; Harbin Heilongjiang Province 150001 China
- Institute of Chronic Disease; Heilongjiang Academy of Medical Science; Harbin Heilongjiang Province 150001 China
| |
Collapse
|
44
|
Nagami M, Ito Y, Nagasawa T. Phenethyl isothiocyanate protects against H 2O 2-induced insulin resistance in 3T3-L1 adipocytes. Biosci Biotechnol Biochem 2017; 81:2195-2203. [PMID: 28899227 DOI: 10.1080/09168451.2017.1372181] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Obesity is associated with systemic oxidative stress and leads to insulin resistance. Phenethyl isothiocyanate (PEITC), a natural dietary isothiocyanate, has been shown to have beneficial effects in improving cellular defense activities against oxidative stress through activation of nuclear factor erythroid-2 related factor 2 (Nrf2) pathway. However, little evidence exists if the antioxidative activity has beneficial effects on glucose metabolism. Here, we tested the preventive potential of PEITC for impaired insulin-induced glucose uptake by oxidative stress in 3T3-L1 adipocytes. Treatment with PEITC increased the expression of antioxidative enzymes regulated by Nrf2 such as γ-glutamylcysteine-synthetase, heme oxygenase 1, NAD(P)H:quinone oxidoreductase 1 and glutathione S-transferase, and reduced oxidative stress induced by H2O2. Furthermore, PEITC restored impaired insulin-stimulated glucose uptake, translocation of glucose transporter 4 and insulin signaling by H2O2. These results indicate that PEITC protected insulin-regulated glucose metabolism impaired by oxidative stress through the antioxidative activity in 3T3-L1 adipocytes.
Collapse
Affiliation(s)
- Moe Nagami
- a Department of Biological Chemistry and Food Science, Graduate School of Agriculture , Iwate University , Morioka , Japan
| | - Yoshiaki Ito
- a Department of Biological Chemistry and Food Science, Graduate School of Agriculture , Iwate University , Morioka , Japan
| | - Takashi Nagasawa
- a Department of Biological Chemistry and Food Science, Graduate School of Agriculture , Iwate University , Morioka , Japan
| |
Collapse
|
45
|
Bergqvist N, Nyman E, Cedersund G, Stenkula KG. A systems biology analysis connects insulin receptor signaling with glucose transporter translocation in rat adipocytes. J Biol Chem 2017; 292:11206-11217. [PMID: 28495883 DOI: 10.1074/jbc.m117.787515] [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: 03/21/2017] [Revised: 05/03/2017] [Indexed: 12/26/2022] Open
Abstract
Type 2 diabetes is characterized by insulin resistance, which arises from malfunctions in the intracellular insulin signaling network. Knowledge of the insulin signaling network is fragmented, and because of the complexity of this network, little consensus has emerged for the structure and importance of the different branches of the network. To help overcome this complexity, systems biology mathematical models have been generated for predicting both the activation of the insulin receptor (IR) and the redistribution of glucose transporter 4 (GLUT4) to the plasma membrane. Although the insulin signal transduction between IR and GLUT4 has been thoroughly studied with modeling and time-resolved data in human cells, comparable analyses in cells from commonly used model organisms such as rats and mice are lacking. Here, we combined existing data and models for rat adipocytes with new data collected for the signaling network between IR and GLUT4 to create a model also for their interconnections. To describe all data (>140 data points), the model needed three distinct pathways from IR to GLUT4: (i) via protein kinase B (PKB) and Akt substrate of 160 kDa (AS160), (ii) via an AS160-independent pathway from PKB, and (iii) via an additional pathway from IR, e.g. affecting the membrane constitution. The developed combined model could describe data not used for training the model and was used to generate predictions of the relative contributions of the pathways from IR to translocation of GLUT4. The combined model provides a systems-level understanding of insulin signaling in rat adipocytes, which, when combined with corresponding models for human adipocytes, may contribute to model-based drug development for diabetes.
Collapse
Affiliation(s)
| | - Elin Nyman
- From the Departments of Biomedical Engineering and.,Cardiovascular and Metabolic Diseases Innovative Medicines and Early Development Biotech Unit, AstraZeneca R&D, SE431 83 Gothenburg, Sweden, and
| | - Gunnar Cedersund
- From the Departments of Biomedical Engineering and .,Clinical and Experimental Medicine and Biomedical Engineering, Linköping University, SE581 85 Linköping, Sweden
| | - Karin G Stenkula
- Glucose Transport and Protein Trafficking, Department of Experimental Medical Sciences, Biomedical Centre, Lund University, SE221 84 Lund, Sweden
| |
Collapse
|
46
|
Faecalibacterium prausnitzii treatment improves hepatic health and reduces adipose tissue inflammation in high-fat fed mice. ISME JOURNAL 2017; 11:1667-1679. [PMID: 28375212 DOI: 10.1038/ismej.2017.24] [Citation(s) in RCA: 168] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Revised: 11/10/2016] [Accepted: 01/27/2017] [Indexed: 02/06/2023]
Abstract
Faecalibacterium prausnitzii is considered as one of the most important bacterial indicators of a healthy gut. We studied the effects of oral F. prausnitzii treatment on high-fat fed mice. Compared to the high-fat control mice, F. prausnitzii-treated mice had lower hepatic fat content, aspartate aminotransferase and alanine aminotransferase, and increased fatty acid oxidation and adiponectin signaling in liver. Hepatic lipidomic analyses revealed decreases in several species of triacylglycerols, phospholipids and cholesteryl esters. Adiponectin expression was increased in the visceral adipose tissue, and the subcutaneous and visceral adipose tissues were more insulin sensitive and less inflamed in F. prausnitzii-treated mice. Further, F. prausnitzii treatment increased muscle mass that may be linked to enhanced mitochondrial respiration, modified gut microbiota composition and improved intestinal integrity. Our findings show that F. prausnitzii treatment improves hepatic health, and decreases adipose tissue inflammation in mice and warrant the need for further studies to discover its therapeutic potential.
Collapse
|
47
|
Mishraki-Berkowitz T, Aserin A, Garti N. Structural properties and release of insulin-loaded reverse hexagonal (HII) liquid crystalline mesophase. J Colloid Interface Sci 2017; 486:184-193. [DOI: 10.1016/j.jcis.2016.09.074] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Revised: 09/28/2016] [Accepted: 09/28/2016] [Indexed: 12/15/2022]
|
48
|
Au DT, Strickland DK, Muratoglu SC. The LDL Receptor-Related Protein 1: At the Crossroads of Lipoprotein Metabolism and Insulin Signaling. J Diabetes Res 2017; 2017:8356537. [PMID: 28584820 PMCID: PMC5444004 DOI: 10.1155/2017/8356537] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Accepted: 04/11/2017] [Indexed: 12/30/2022] Open
Abstract
The metabolic syndrome is an escalating worldwide public health concern. Defined by a combination of physiological, metabolic, and biochemical factors, the metabolic syndrome is used as a clinical guideline to identify individuals with a higher risk for type 2 diabetes and cardiovascular disease. Although risk factors for type 2 diabetes and cardiovascular disease have been known for decades, the molecular mechanisms involved in the pathophysiology of these diseases and their interrelationship remain unclear. The LDL receptor-related protein 1 (LRP1) is a large endocytic and signaling receptor that is widely expressed in several tissues. As a member of the LDL receptor family, LRP1 is involved in the clearance of chylomicron remnants from the circulation and has been demonstrated to be atheroprotective. Recently, studies have shown that LRP1 is involved in insulin receptor trafficking and regulation and glucose metabolism. This review summarizes the role of tissue-specific LRP1 in insulin signaling and its potential role as a link between lipoprotein and glucose metabolism in diabetes.
Collapse
Affiliation(s)
- Dianaly T. Au
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Dudley K. Strickland
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, USA
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD, USA
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Selen C. Muratoglu
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, USA
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, USA
- *Selen C. Muratoglu:
| |
Collapse
|
49
|
Xie B, Chen Q, Chen L, Sheng Y, Wang HY, Chen S. The Inactivation of RabGAP Function of AS160 Promotes Lysosomal Degradation of GLUT4 and Causes Postprandial Hyperglycemia and Hyperinsulinemia. Diabetes 2016; 65:3327-3340. [PMID: 27554475 DOI: 10.2337/db16-0416] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Accepted: 08/16/2016] [Indexed: 11/13/2022]
Abstract
The AS160 (Akt substrate of 160 kDa) is a Rab-GTPase activating protein (RabGAP) with several other functional domains, and its deficiency in mice or human patients lowers GLUT4 protein levels and causes severe insulin resistance. How its deficiency causes diminished GLUT4 proteins remains unknown. We found that the deletion of AS160 decreased GLUT4 levels in a cell/tissue-autonomous manner. Consequently, skeletal muscle-specific deletion of AS160 caused postprandial hyperglycemia and hyperinsulinemia. The pathogenic effects of AS160 deletion are mainly, if not exclusively, due to the loss of its RabGAP function since the RabGAP-inactive AS160R917K mutant mice phenocopied the AS160 knockout mice. The inactivation of RabGAP of AS160 promotes lysosomal degradation of GLUT4, and the inhibition of lysosome function could restore GLUT4 protein levels. Collectively, these findings demonstrate that the RabGAP activity of AS160 maintains GLUT4 protein levels in a cell/tissue-autonomous manner and its inactivation causes lysosomal degradation of GLUT4 and postprandial hyperglycemia and hyperinsulinemia.
Collapse
Affiliation(s)
- Bingxian Xie
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing University, Pukou District, Nanjing, China
| | - Qiaoli Chen
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing University, Pukou District, Nanjing, China
| | - Liang Chen
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing University, Pukou District, Nanjing, China
| | - Yang Sheng
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing University, Pukou District, Nanjing, China
| | - Hong Yu Wang
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing University, Pukou District, Nanjing, China
- Collaborative Innovation Center of Genetics and Development, Shanghai, China
| | - Shuai Chen
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing University, Pukou District, Nanjing, China
- Collaborative Innovation Center of Genetics and Development, Shanghai, China
| |
Collapse
|
50
|
Liu L, Wang F, Lu H, Cao S, Du Z, Wang Y, Feng X, Gao Y, Zha M, Guo M, Sun Z, Wang J. Effects of Noise Exposure on Systemic and Tissue-Level Markers of Glucose Homeostasis and Insulin Resistance in Male Mice. ENVIRONMENTAL HEALTH PERSPECTIVES 2016; 124:1390-1398. [PMID: 27128844 PMCID: PMC5010391 DOI: 10.1289/ehp162] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Revised: 01/24/2016] [Accepted: 04/12/2016] [Indexed: 06/05/2023]
Abstract
BACKGROUND Epidemiological studies have indicated that noise exposure is associated with an increased risk of type 2 diabetes mellitus (T2DM). However, the nature of the connection between noise exposure and T2DM remains to be explored. OBJECTIVES We explored whether and how noise exposure affects glucose homeostasis in mice as the initial step toward T2DM development. METHODS Male ICR mice were randomly assigned to one of four groups: the control group and three noise groups (N20D, N10D, and N1D), in which the animals were exposed to white noise at 95 decibel sound pressure level (dB SPL) for 4 hr per day for 20 successive days, 10 successive days, or 1 day, respectively. Glucose tolerance and insulin sensitivity were evaluated 1 day, 1 week, and 1 month after the final noise exposure (1DPN, 1WPN, and 1MPN). Standard immunoblots, immunohistochemical methods, and enzyme-linked immunosorbent assays (ELISA) were performed to assess insulin signaling in skeletal muscle, the morphology of β cells, and plasma corticosterone levels. RESULTS Noise exposure for 1 day caused transient glucose intolerance and insulin resistance, whereas noise exposure for 10 and 20 days had no effect on glucose tolerance but did cause prolonged insulin resistance and an increased insulin response to glucose challenge. Akt phosphorylation and GLUT4 translocation in response to exogenous insulin were decreased in the skeletal muscle of noise-exposed animals. CONCLUSIONS Noise exposure at 95 dB SPL caused insulin resistance in male ICR mice, which was prolonged with longer noise exposure and was likely related to the observed blunted insulin signaling in skeletal muscle. CITATION Liu L, Wang F, Lu H, Cao S, Du Z, Wang Y, Feng X, Gao Y, Zha M, Guo M, Sun Z, Wang J. 2016. Effects of noise exposure on systemic and tissue-level markers of glucose homeostasis and insulin resistance in male mice. Environ Health Perspect 124:1390-1398; http://dx.doi.org/10.1289/EHP162.
Collapse
Affiliation(s)
- Lijie Liu
- Department of Physiology, Medical College, Southeast University, Nanjing, China
| | - Fanfan Wang
- Institute of Life Sciences, Southeast University, Nanjing, China
| | - Haiying Lu
- Institute of Life Sciences, Southeast University, Nanjing, China
| | - Shuangfeng Cao
- Institute of Life Sciences, Southeast University, Nanjing, China
| | - Ziwei Du
- Medical College, Southeast University, Nanjing, China
| | - Yongfang Wang
- Medical College, Southeast University, Nanjing, China
| | - Xian Feng
- Medical College, Southeast University, Nanjing, China
| | - Ye Gao
- Medical College, Southeast University, Nanjing, China
| | - Mingming Zha
- Medical College, Southeast University, Nanjing, China
| | - Min Guo
- Medical College, Southeast University, Nanjing, China
| | - Zilin Sun
- Department of Endocrinology, Medical College, Affiliated ZhongDa Hospital of Southeast University, Nanjing, China
| | - Jian Wang
- Department of Physiology, Medical College, Southeast University, Nanjing, China
- School of Human Communication Disorders, Dalhousie University, Halifax, Nova Scotia, Canada
| |
Collapse
|