1
|
Gray CW, Coster ACF. Deciphering Akt activation: Insights from a mean-field model. Math Biosci 2025; 384:109434. [PMID: 40222591 DOI: 10.1016/j.mbs.2025.109434] [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: 10/27/2024] [Revised: 03/23/2025] [Accepted: 03/25/2025] [Indexed: 04/15/2025]
Abstract
Being at the right place at the right time is vital for any signalling system component. Akt/PKB is a well-known low-threshold switch in the mammalian insulin signalling pathway. The activation of Akt is essential for the uptake of glucose, however, data concerning this vital system is very sparse, particularly with regards to cellular location and activation state. Here we present a parsimonious mathematical model that captures the current experimental understanding of Akt dynamics. The system operates on two distinct timescales (signalling and physical transport), with the transportation of Akt constituting the rate-limiting step in most circumstances. The model outputs are consistent with observations of the steady state behaviour of the system and display the transient overshoot behaviour which is a necessary characteristic of the activation of Akt.
Collapse
Affiliation(s)
- Catheryn W Gray
- School of Mathematics and Statistics, The University of New South Wales, Sydney, 2052, New South Wales, Australia.
| | - Adelle C F Coster
- School of Mathematics and Statistics, The University of New South Wales, Sydney, 2052, New South Wales, Australia.
| |
Collapse
|
2
|
Liu X, Tang Y, Luo Y, Gao Y, He L. Role and mechanism of specialized pro-resolving mediators in obesity-associated insulin resistance. Lipids Health Dis 2024; 23:234. [PMID: 39080624 PMCID: PMC11290132 DOI: 10.1186/s12944-024-02207-9] [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: 03/15/2024] [Accepted: 07/07/2024] [Indexed: 08/02/2024] Open
Abstract
With the changing times, obesity has become a characteristic epidemic in the context of the current era. Insulin resistance (IR) is most commonly caused by obesity, and IR is a common basis of the pathogenesis of many diseases such as cardiovascular disease, nonalcoholic fatty liver disease, and type 2 diabetes, which seriously threaten human life, as well as health. A major pathogenetic mechanism of obesity-associated IR has been found to be chronic low-grade inflammation in adipose tissue. Specialized pro-resolving mediators (SPMs) are novel lipid mediators that both function as "stop signals" for inflammatory reaction and promote inflammation to subside. In this article, we summarize the pathogenesis of obesity-associated IR and its treatments and outline the classification and biosynthesis of SPMs and their mechanisms and roles in the treatment of obesity-associated IR in order to explore the potential of SPMs for treating metabolic diseases linked with obesity-associated IR.
Collapse
Affiliation(s)
- Xinru Liu
- College of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yu Tang
- College of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yuanyuan Luo
- College of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yongxiang Gao
- College of International Education, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Lisha He
- College of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| |
Collapse
|
3
|
Mann G, Riddell MC, Adegoke OAJ. Effects of Acute Muscle Contraction on the Key Molecules in Insulin and Akt Signaling in Skeletal Muscle in Health and in Insulin Resistant States. DIABETOLOGY 2022; 3:423-446. [DOI: 10.3390/diabetology3030032] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
Abstract
Insulin signaling plays a key role in glucose uptake, glycogen synthesis, and protein and lipid synthesis. In insulin-resistant states like obesity and type 2 diabetes mellitus, these processes are dysregulated. Regular physical exercise is a potential therapeutic strategy against insulin resistance, as an acute bout of exercise increases glucose disposal during the activity and for hours into recovery. Chronic exercise increases the activation of proteins involved in insulin signaling and increases glucose transport, even in insulin resistant states. Here, we will focus on the effect of acute exercise on insulin signaling and protein kinase B (Akt) pathways. Activation of proximal proteins involved in insulin signaling (insulin receptor, insulin receptor substrate-1 (IRS-1), phosphoinoside-3 kinase (PI3K)) are unchanged in response to acute exercise/contraction, while activation of Akt and of its substrates, TBC1 domain family 1 (TBC1D1), and TBC domain family 4 (TBC1D4) increases in response to such exercise/contraction. A wide array of Akt substrates is also regulated by exercise. Additionally, AMP-activated protein kinase (AMPK) seems to be a main mediator of the benefits of exercise on skeletal muscle. Questions persist on how mTORC1 and AMPK, two opposing regulators, are both upregulated after an acute bout of exercise.
Collapse
Affiliation(s)
- Gagandeep Mann
- School of Kinesiology and Health Science, York University, Toronto, ON M3J 1P3, Canada
| | - Michael C. Riddell
- School of Kinesiology and Health Science, York University, Toronto, ON M3J 1P3, Canada
| | | |
Collapse
|
4
|
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
|
5
|
Zhang Y, Wei X, Sun Q, Qian W, Liu X, Li J, Long Y, Wan X. Different Types and Functional Effects of Probiotics on Human Health through Regulating Glucose Homeostasis. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2021; 69:14781-14791. [PMID: 34855398 DOI: 10.1021/acs.jafc.1c04291] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
With the increasing improvement of people's living standards, hyperglycemia has become one of the most frequent diseases in the world. The current drug therapy may have some negative effects and even cause some complications. As one of the most popular functional ingredients, probiotic bacteria have been proven to play important roles in balancing the glucose homeostasis level in animal and human clinic trials. In this perspective, we sorted three types of probiotics, discussed probiotic safety evaluation, and listed the known probiotic functional foods that assist to control glucose homeostasis. Then, the further summarization of the mechanisms on how probiotic bacteria could regulate glucose homeostasis and the developing trend of probiotic functional foods were discussed.
Collapse
Affiliation(s)
- Yong Zhang
- Zhongzhi International Institute of Agricultural Biosciences, Shunde Graduate School, Research Center of Biology and Agriculture, University of Science and Technology Beijing (USTB), Beijing 100024, People's Republic of China
- Beijing Beike Institute of Precision Medicine and Health Technology, Beijing 100192, People's Republic of China
- Beijing Engineering Laboratory of Main Crop Bio-Tech Breeding, Beijing International Science and Technology Cooperation Base of Bio-Tech Breeding, Beijing Solidwill Sci-Tech Company, Limited, Beijing 100192, People's Republic of China
| | - Xun Wei
- Zhongzhi International Institute of Agricultural Biosciences, Shunde Graduate School, Research Center of Biology and Agriculture, University of Science and Technology Beijing (USTB), Beijing 100024, People's Republic of China
- Beijing Beike Institute of Precision Medicine and Health Technology, Beijing 100192, People's Republic of China
- Beijing Engineering Laboratory of Main Crop Bio-Tech Breeding, Beijing International Science and Technology Cooperation Base of Bio-Tech Breeding, Beijing Solidwill Sci-Tech Company, Limited, Beijing 100192, People's Republic of China
| | - Qian Sun
- Zhongzhi International Institute of Agricultural Biosciences, Shunde Graduate School, Research Center of Biology and Agriculture, University of Science and Technology Beijing (USTB), Beijing 100024, People's Republic of China
| | - Weiyi Qian
- Zhongzhi International Institute of Agricultural Biosciences, Shunde Graduate School, Research Center of Biology and Agriculture, University of Science and Technology Beijing (USTB), Beijing 100024, People's Republic of China
- Beijing Beike Institute of Precision Medicine and Health Technology, Beijing 100192, People's Republic of China
| | - Xinjie Liu
- Zhongzhi International Institute of Agricultural Biosciences, Shunde Graduate School, Research Center of Biology and Agriculture, University of Science and Technology Beijing (USTB), Beijing 100024, People's Republic of China
- Beijing Beike Institute of Precision Medicine and Health Technology, Beijing 100192, People's Republic of China
- Beijing Engineering Laboratory of Main Crop Bio-Tech Breeding, Beijing International Science and Technology Cooperation Base of Bio-Tech Breeding, Beijing Solidwill Sci-Tech Company, Limited, Beijing 100192, People's Republic of China
| | - Jinping Li
- Beijing Beike Institute of Precision Medicine and Health Technology, Beijing 100192, People's Republic of China
- Beijing Engineering Laboratory of Main Crop Bio-Tech Breeding, Beijing International Science and Technology Cooperation Base of Bio-Tech Breeding, Beijing Solidwill Sci-Tech Company, Limited, Beijing 100192, People's Republic of China
| | - Yan Long
- Zhongzhi International Institute of Agricultural Biosciences, Shunde Graduate School, Research Center of Biology and Agriculture, University of Science and Technology Beijing (USTB), Beijing 100024, People's Republic of China
- Beijing Beike Institute of Precision Medicine and Health Technology, Beijing 100192, People's Republic of China
- Beijing Engineering Laboratory of Main Crop Bio-Tech Breeding, Beijing International Science and Technology Cooperation Base of Bio-Tech Breeding, Beijing Solidwill Sci-Tech Company, Limited, Beijing 100192, People's Republic of China
| | - Xiangyuan Wan
- Zhongzhi International Institute of Agricultural Biosciences, Shunde Graduate School, Research Center of Biology and Agriculture, University of Science and Technology Beijing (USTB), Beijing 100024, People's Republic of China
- Beijing Beike Institute of Precision Medicine and Health Technology, Beijing 100192, People's Republic of China
- Beijing Engineering Laboratory of Main Crop Bio-Tech Breeding, Beijing International Science and Technology Cooperation Base of Bio-Tech Breeding, Beijing Solidwill Sci-Tech Company, Limited, Beijing 100192, People's Republic of China
| |
Collapse
|
6
|
Zhang Y, He L, Chen X, Shentu P, Xu Y, Jiao J. Omega-3 polyunsaturated fatty acids promote SNAREs mediated GLUT4 vesicle docking and fusion. J Nutr Biochem 2021; 101:108912. [PMID: 34801692 DOI: 10.1016/j.jnutbio.2021.108912] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 09/24/2021] [Accepted: 10/08/2021] [Indexed: 11/27/2022]
Abstract
Glucose homeostasis imbalance and insulin resistance (IR) are major contributors to the incidence of type 2 diabetes. Omega-3 polyunsaturated fatty acids (PUFAs) are key ingredients for maintaining cellular functions and improving insulin sensitivity. However, how omega-3 PUFAs modulate the dynamic process of glucose transport at the cellular level remains unclear. Here we unraveled eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) may regulate the glucose transporter 4 (GLUT4) vesicle trafficking in both normal and IR adipocytes. Both omega-3 PUFAs significantly increase glucose consumption within a range of 10-32% in the basal state. Furthermore, both EPA (200 μM) and DHA (100 μM) may significantly promote the serine/threonine protein kinase (Akt) phosphorylation by 70% and 40% in the physiological state of adipocytes, respectively. Both omega-3 PUFAs significantly advanced the Akt phosphorylation in a dose-dependent way and showed a ∼2-fold increase at the dose of 200 μM in the IR pathological state. However, they could not up-regulate the expression of GLUT4 and insulin-regulated aminopeptidase protein. We further revealed that both omega-3 PUFAs dynamically promote insulin-stimulated GLUT4 vesicle translocation and soluble N-ethylmaleimide-sensitive factor attachment protein receptor mediated vesicle docking and fusion to the plasma membrane via specifically modulating the expression of vesicle-associated membrane protein 2. Understanding the mechanisms by which omega-3 PUFAs modulate cellular metabolism and IR in peripheral tissues may provide novel insights into the potential impact of omega-3 PUFAs on the metabolic function and the management of IR.
Collapse
Affiliation(s)
- Yu Zhang
- National Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agro-Food Processing, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, Zhejiang, China
| | - Lilin He
- National Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agro-Food Processing, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, Zhejiang, China
| | - Xiaoqian Chen
- National Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agro-Food Processing, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, Zhejiang, China
| | - Ping Shentu
- Key Laboratory for Biomedical Engineering of Ministry of Education, Zhejiang Provincial Key Laboratory of Cardio-Cerebral Vascular Detection Technology and Medicinal Effectiveness Appraisal, Department of Biomedical Engineering, Zhejiang University, Hangzhou, Zhejiang, China
| | - Yingke Xu
- Key Laboratory for Biomedical Engineering of Ministry of Education, Zhejiang Provincial Key Laboratory of Cardio-Cerebral Vascular Detection Technology and Medicinal Effectiveness Appraisal, Department of Biomedical Engineering, Zhejiang University, Hangzhou, Zhejiang, China; Department of Endocrinology, The Affiliated Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
| | - Jingjing Jiao
- Department of Nutrition, School of Public Health, Department of Clinical Nutrition, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
| |
Collapse
|
7
|
Ramos PA, Lytle KA, Delivanis D, Nielsen S, LeBrasseur NK, Jensen MD. Insulin-Stimulated Muscle Glucose Uptake and Insulin Signaling in Lean and Obese Humans. J Clin Endocrinol Metab 2021; 106:e1631-e1646. [PMID: 33382888 PMCID: PMC7993573 DOI: 10.1210/clinem/dgaa919] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Indexed: 12/29/2022]
Abstract
PURPOSE Skeletal muscle is the primary site for insulin-stimulated glucose disposal, and muscle insulin resistance is central to abnormal glucose metabolism in obesity. Whether muscle insulin signaling to the level of Akt/AS160 is intact in insulin-resistant obese humans is controversial. METHODS We defined a linear range of insulin-stimulated systemic and leg glucose uptake in 14 obese and 14 nonobese volunteers using a 2-step insulin clamp (Protocol 1) and then examined the obesity-related defects in muscle insulin action in 16 nonobese and 25 obese male and female volunteers matched for fitness using a 1-step, hyperinsulinemic, euglycemic clamp coupled with muscle biopsies (Protocol 2). RESULTS Insulin-stimulated glucose disposal (Si) was reduced by > 60% (P < 0.0001) in the obese group in Protocol 2; however, the phosphorylation of Akt and its downstream effector AS160 were not different between nonobese and obese groups. The increase in phosphorylation of Akt2 in response to insulin was positively correlated with Si for both the nonobese (r = 0.53, P = 0.03) and the obese (r = 0.55, P = 0.01) groups. Total muscle GLUT4 protein was 17% less (P < 0.05) in obese subjects. CONCLUSIONS We suggest that reduced muscle glucose uptake in obesity is not due to defects in the insulin signaling pathway at the level of Akt/AS160, which suggests there remain significant gaps in our knowledge of muscle insulin resistance in obesity. Our data imply that models of acute lipotoxicity do not replicate the pathophysiology of obesity.
Collapse
Affiliation(s)
- Paola A Ramos
- Endocrine Research Unit, Mayo Clinic, Rochester, MN, USA
| | - Kelli A Lytle
- Endocrine Research Unit, Mayo Clinic, Rochester, MN, USA
| | | | - Søren Nielsen
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus C, Denmark
| | | | - Michael D Jensen
- Endocrine Research Unit, Mayo Clinic, Rochester, MN, USA
- Correspondence: Michael D. Jensen, MD, Division of Endocrinology, Mayo Clinic, 200 First St SW, Joseph Rm 5–194, Rochester MN 55905, USA.
| |
Collapse
|
8
|
Norris D, Yang P, Shin SY, Kearney AL, Kim HJ, Geddes T, Senior AM, Fazakerley DJ, Nguyen LK, James DE, Burchfield JG. Signaling Heterogeneity is Defined by Pathway Architecture and Intercellular Variability in Protein Expression. iScience 2021; 24:102118. [PMID: 33659881 PMCID: PMC7892930 DOI: 10.1016/j.isci.2021.102118] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 01/07/2021] [Accepted: 01/22/2021] [Indexed: 12/12/2022] Open
Abstract
Insulin's activation of PI3K/Akt signaling, stimulates glucose uptake by enhancing delivery of GLUT4 to the cell surface. Here we examined the origins of intercellular heterogeneity in insulin signaling. Akt activation alone accounted for ~25% of the variance in GLUT4, indicating that additional sources of variance exist. The Akt and GLUT4 responses were highly reproducible within the same cell, suggesting the variance is between cells (extrinsic) and not within cells (intrinsic). Generalized mechanistic models (supported by experimental observations) demonstrated that the correlation between the steady-state levels of two measured signaling processes decreases with increasing distance from each other and that intercellular variation in protein expression (as an example of extrinsic variance) is sufficient to account for the variance in and between Akt and GLUT4. Thus, the response of a population to insulin signaling is underpinned by considerable single-cell heterogeneity that is largely driven by variance in gene/protein expression between cells. Insulin signaling is heterogeneous between cells in the same population The temporal response of signaling components within a cell is highly reproducible Upstream responses (Akt) can only partially predict downstream response (GLUT4) Protein expression variance is a driver of intercellular signaling heterogeneity
Collapse
Affiliation(s)
- Dougall Norris
- Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia.,School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW 2006, Australia
| | - Pengyi Yang
- Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia.,School of Mathematics and Statistics, The University of Sydney, Sydney, NSW 2006, Australia.,Computational Systems Biology Group, Children's Medical Research Institute, Faculty of Medicine and Health, The University of Sydney, Westmead, NSW 2145, Australia
| | - Sung-Young Shin
- Department of Biochemistry and Molecular Biology, School of Biomedical Sciences, Monash University, Clayton, VIC 3800, Australia.,Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia
| | - Alison L Kearney
- Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia.,School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW 2006, Australia
| | - Hani Jieun Kim
- Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia.,School of Mathematics and Statistics, The University of Sydney, Sydney, NSW 2006, Australia.,Computational Systems Biology Group, Children's Medical Research Institute, Faculty of Medicine and Health, The University of Sydney, Westmead, NSW 2145, Australia
| | - Thomas Geddes
- Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia.,School of Mathematics and Statistics, The University of Sydney, Sydney, NSW 2006, Australia.,Computational Systems Biology Group, Children's Medical Research Institute, Faculty of Medicine and Health, The University of Sydney, Westmead, NSW 2145, Australia
| | - Alistair M Senior
- Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia.,School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW 2006, Australia
| | - Daniel J Fazakerley
- Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia
| | - Lan K Nguyen
- Department of Biochemistry and Molecular Biology, School of Biomedical Sciences, Monash University, Clayton, VIC 3800, Australia.,Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia
| | - David E James
- Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia.,School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW 2006, Australia.,Sydney Medical School, The University of Sydney, Sydney, NSW 2006, Australia
| | - James G Burchfield
- Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia.,School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW 2006, Australia
| |
Collapse
|
9
|
Brumfield A, Chaudhary N, Molle D, Wen J, Graumann J, McGraw TE. Insulin-promoted mobilization of GLUT4 from a perinuclear storage site requires RAB10. Mol Biol Cell 2021; 32:57-73. [PMID: 33175605 PMCID: PMC8098823 DOI: 10.1091/mbc.e20-06-0356] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 10/30/2020] [Accepted: 11/04/2020] [Indexed: 12/05/2022] Open
Abstract
Insulin controls glucose uptake into muscle and fat cells by inducing a net redistribution of glucose transporter 4 (GLUT4) from intracellular storage to the plasma membrane (PM). The TBC1D4-RAB10 signaling module is required for insulin-stimulated GLUT4 translocation to the PM, although where it intersects GLUT4 traffic was unknown. Here we demonstrate that TBC1D4-RAB10 functions to control GLUT4 mobilization from a trans-Golgi network (TGN) storage compartment, establishing that insulin, in addition to regulating the PM proximal effects of GLUT4-containing vesicles docking to and fusion with the PM, also directly regulates the behavior of GLUT4 deeper within the cell. We also show that GLUT4 is retained in an element/domain of the TGN from which newly synthesized lysosomal proteins are targeted to the late endosomes and the ATP7A copper transporter is translocated to the PM by elevated copper. Insulin does not mobilize ATP7A nor does copper mobilize GLUT4, and RAB10 is not required for copper-elicited ATP7A mobilization. Consequently, GLUT4 intracellular sequestration and mobilization by insulin is achieved, in part, through utilizing a region of the TGN devoted to specialized cargo transport in general rather than being specific for GLUT4. Our results define the GLUT4-containing region of the TGN as a sorting and storage site from which different cargo are mobilized by distinct signals through unique molecular machinery.
Collapse
Affiliation(s)
| | - Natasha Chaudhary
- Department of Biochemistry, Weill Cornell Medical College, New York, NY 10065
| | - Dorothee Molle
- Department of Biochemistry, Weill Cornell Medical College, New York, NY 10065
| | - Jennifer Wen
- Department of Biochemistry, Weill Cornell Medical College, New York, NY 10065
| | - Johannes Graumann
- Weill Cornell Medical College in Qatar, Education City, 24144 Doha, State of Qatar
| | - Timothy E. McGraw
- Department of Biochemistry, Weill Cornell Medical College, New York, NY 10065
- Department of Cardiothoracic Surgery, Weill Cornell Medical College, New York, NY 10065
| |
Collapse
|
10
|
Shrestha MM, Lim CY, Bi X, Robinson RC, Han W. Tmod3 Phosphorylation Mediates AMPK-Dependent GLUT4 Plasma Membrane Insertion in Myoblasts. Front Endocrinol (Lausanne) 2021; 12:653557. [PMID: 33959097 PMCID: PMC8095187 DOI: 10.3389/fendo.2021.653557] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 03/22/2021] [Indexed: 12/28/2022] Open
Abstract
Insulin and muscle contractions mediate glucose transporter 4 (GLUT4) translocation and insertion into the plasma membrane (PM) for glucose uptake in skeletal muscles. Muscle contraction results in AMPK activation, which promotes GLUT4 translocation and PM insertion. However, little is known regarding AMPK effectors that directly regulate GLUT4 translocation. We aim to identify novel AMPK effectors in the regulation of GLUT4 translocation. We performed biochemical, molecular biology and fluorescent microscopy imaging experiments using gain- and loss-of-function mutants of tropomodulin 3 (Tmod3). Here we report Tmod3, an actin filament capping protein, as a novel AMPK substrate and an essential mediator of AMPK-dependent GLUT4 translocation and glucose uptake in myoblasts. Furthermore, Tmod3 plays a key role in AMPK-induced F-actin remodeling and GLUT4 insertion into the PM. Our study defines Tmod3 as a key AMPK effector in the regulation of GLUT4 insertion into the PM and glucose uptake in muscle cells, and offers new mechanistic insights into the regulation of glucose homeostasis.
Collapse
Affiliation(s)
- Man Mohan Shrestha
- Laboratory of Metabolic Medicine, Singapore Bioimaging Consortium, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Chun-Yan Lim
- Laboratory of Metabolic Medicine, Singapore Bioimaging Consortium, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Xuezhi Bi
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Robert C. Robinson
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Weiping Han
- Laboratory of Metabolic Medicine, Singapore Bioimaging Consortium, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- *Correspondence: Weiping Han,
| |
Collapse
|
11
|
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
|
12
|
Gray CW, Coster ACF. From insulin to Akt: Time delays and dominant processes. J Theor Biol 2020; 507:110454. [PMID: 32822700 DOI: 10.1016/j.jtbi.2020.110454] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 07/14/2020] [Accepted: 08/14/2020] [Indexed: 11/27/2022]
Abstract
Akt/PKB regulates numerous processes in the mammalian cell, including cell survival and proliferation, and glucose uptake in response to insulin. Abnormalities in Akt signalling are linked to the development of Type 2 diabetes, cardio-vascular disease, and cancer. In the absence of insulin, Akt is predominantly found in the inactive state in the cytosol. Following insulin stimulation, Akt translocates to the plasma membrane, docks, and is phosphorylated to take on the active conformation. In turn, the activated Akt travels to and phosphorylates its many downstream substrates. Although crucial to the activation process, the translocation of Akt from the cytosol to the plasma membrane is currently not well understood. Here we detail the parameter optimisation of a mathematical model of Akt translocation to experimental data. We have quantified the time delay between the application of insulin and the downstream Akt translocation response, indicating the constraints on the timing of the intermediate processes. A delay of approximately 0.4 min prior to the Akt response was determined for the application of 1 nM insulin to cells in the basal state, whereas it was found that a further transition from physiological insulin to higher stimuli did not incur a delay. Furthermore, our investigation indicates that the dominant processes regulating the appearance of Akt at the plasma membrane differ with the insulin level. For physiological insulin, the rate limiting step was the release of Akt to the plasma membrane in response to the insulin signal. In contrast, at high insulin levels, regulation of the recycling of Akt from the plasma membrane to the cytosol was also required.
Collapse
Affiliation(s)
- Catheryn W Gray
- School of Mathematics and Statistics, UNSW Sydney Australia.
| | | |
Collapse
|
13
|
Khandelwal M, Manglani K, Gupta S, Tiku AB. Gamma radiation improves AD pathogenesis in APP/PS1 mouse model by potentiating insulin sensitivity. Heliyon 2020; 6:e04499. [PMID: 32775714 PMCID: PMC7399127 DOI: 10.1016/j.heliyon.2020.e04499] [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: 11/15/2019] [Revised: 04/23/2020] [Accepted: 07/15/2020] [Indexed: 11/30/2022] Open
Abstract
Alzheimer's disease (AD) is the largest unmet medical complication. The devastation caused by the disease can be assumed from the disease symptoms like speech impairment, loss of self-awareness, acute memory loss etc. The individuals suffering from AD completely depend on caregivers and have to bear the high cost of treatment which increases the socio-economic burden on the society. Recent studies have shown that radiation exposure can have therapeutic effects when given in suitable amount for a specific time period. Therefore, we investigated the role of gamma irradiation in AD pathogenesis. The effect of radiation on amelioration of disease progression was studied in AD transgenic mice model (APP/PS1). Our in-vivo studies using APP/PS1 mice demonstrated that a single dose of 4.0 Gy gamma irradiation improves AD associated behavioral impairment. Radiation exposure also increased the level of anti-oxidant enzymes and reduced the astrocyte activation in the brain of APP/PS1 mice. A significant reduction was observed in AD associated proteins (APP, pTau, BACE) and neurofibrillary tangle formations (NFTs). Exposure to a single dose of 4 Gy gamma radiation also increased glucose metabolic functionality in AD transgenic mouse model. The kinases involved in insulin signaling such as GSK, ERK and JNK were also found to be modulated. However, an increased level of GSK3β (ser 9) was observed, which could be responsible for downregulating ERK and JNK phosphorylation. This resulted in a decrease in neurofibrillary tangle formations and amyloid deposition. The reduced hyperphosphorylation of Tau can be attributed to the increased level of GSK3β (ser 9) downregulating ERK and JNK phosphorylation. Thus, a single dose of 4 Gy gamma irradiation was found to have therapeutic benefits in treating AD via potentiating insulin signaling in APP/PS1 transgenic mice.
Collapse
Affiliation(s)
- Mayuri Khandelwal
- Molecular Science Laboratory, National Institute of Immunology, New Delhi, 110067, India.,Radiation and Cancer Therapeutics Lab, School of Life Sciences, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Kapil Manglani
- Molecular Science Laboratory, National Institute of Immunology, New Delhi, 110067, India
| | - Sarika Gupta
- Molecular Science Laboratory, National Institute of Immunology, New Delhi, 110067, India
| | - Ashu Bhan Tiku
- Radiation and Cancer Therapeutics Lab, School of Life Sciences, Jawaharlal Nehru University, New Delhi, 110067, India
| |
Collapse
|
14
|
Morris S, Geoghegan ND, Sadler JBA, Koester AM, Black HL, Laub M, Miller L, Heffernan L, Simpson JC, Mastick CC, Cooper J, Gadegaard N, Bryant NJ, Gould GW. Characterisation of GLUT4 trafficking in HeLa cells: comparable kinetics and orthologous trafficking mechanisms to 3T3-L1 adipocytes. PeerJ 2020; 8:e8751. [PMID: 32185116 PMCID: PMC7060922 DOI: 10.7717/peerj.8751] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 02/14/2020] [Indexed: 12/18/2022] Open
Abstract
Insulin-stimulated glucose transport is a characteristic property of adipocytes and muscle cells and involves the regulated delivery of glucose transporter (GLUT4)-containing vesicles from intracellular stores to the cell surface. Fusion of these vesicles results in increased numbers of GLUT4 molecules at the cell surface. In an attempt to overcome some of the limitations associated with both primary and cultured adipocytes, we expressed an epitope- and GFP-tagged version of GLUT4 (HA–GLUT4–GFP) in HeLa cells. Here we report the characterisation of this system compared to 3T3-L1 adipocytes. We show that insulin promotes translocation of HA–GLUT4–GFP to the surface of both cell types with similar kinetics using orthologous trafficking machinery. While the magnitude of the insulin-stimulated translocation of GLUT4 is smaller than mouse 3T3-L1 adipocytes, HeLa cells offer a useful, experimentally tractable, human model system. Here, we exemplify their utility through a small-scale siRNA screen to identify GOSR1 and YKT6 as potential novel regulators of GLUT4 trafficking in human cells.
Collapse
Affiliation(s)
- Silke Morris
- Institute of Molecular Cell and Systems Biology, University of Glasgow, Glasgow, UK
| | | | - Jessica B A Sadler
- Institute of Molecular Cell and Systems Biology, University of Glasgow, Glasgow, UK
| | - Anna M Koester
- Institute of Molecular Cell and Systems Biology, University of Glasgow, Glasgow, UK
| | | | - Marco Laub
- Institute of Molecular Cell and Systems Biology, University of Glasgow, Glasgow, UK
| | - Lucy Miller
- Institute of Molecular Cell and Systems Biology, University of Glasgow, Glasgow, UK
| | - Linda Heffernan
- School of Biology & Environmental Science, University College Dublin, Dublin, Ireland
| | - Jeremy C Simpson
- School of Biology & Environmental Science, University College Dublin, Dublin, Ireland
| | | | - Jon Cooper
- School of Engineering, University of Glasgow, Glasgow, UK
| | | | - Nia J Bryant
- Department of Biology, University of York, York, UK
| | - Gwyn W Gould
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| |
Collapse
|
15
|
Zhang Y, Yan LS, Ding Y, Cheng BCY, Luo G, Kong J, Liu TH, Zhang SF. Edgeworthia gardneri (Wall.) Meisn. Water Extract Ameliorates Palmitate Induced Insulin Resistance by Regulating IRS1/GSK3β/FoxO1 Signaling Pathway in Human HepG2 Hepatocytes. Front Pharmacol 2020; 10:1666. [PMID: 32082162 PMCID: PMC7002394 DOI: 10.3389/fphar.2019.01666] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Accepted: 12/19/2019] [Indexed: 12/12/2022] Open
Abstract
The flower of Edgeworthia gardneri (Wall.) Meisn is commonly used in beverage products in Tibet and has potential health benefits for diabetes. However, the mechanisms underlying anti-insulin resistance (IR) action of the flower of E. gardneri are not fully understood. This study aims to investigate the effects of the water extract of the flower of E. gardneri (WEE) on IR in palmitate (PA)-exposed HepG2 hepatocytes. WEE was characterized by UPLC analysis. PA-treated HepG2 cells were selected as the IR cell model. The cell viability was determined using MTT assay. Moreover, the glucose consumption and production were measured by glucose oxidase method. The glucose uptake and glycogen content were determined by the 2-NBDG (2-deoxy-2-[(7-nitro-2,1,3-benzoxadiazol-4-yl) amino]-D-glucose) glucose uptake assay and anthrone-sulfuric acid assay, respectively. The intracellular triglyceride content was detected by oxidative enzymic method. Protein levels were examined by Western blotting. Nuclear localization of FoxO1 was detected using immunofluorescence analyses and Western blotting. The expression of FoxO1 target genes was detected by quantitative real-time polymerase chain reaction (qRT-PCR). The viability of PA-treated HepG2 cells was concentration-dependently increased by incubation with WEE for 24 h. WEE treatment remarkably increased the consumption and uptake of glucose in PA-exposed HepG2 cells. Moreover, treatment with WEE significantly decreased the PA-induced over-production of glucose in HepG2 cells. After exposure of HepG2 cells with PA and WEE, the glycogen content was significantly elevated. The phosphorylation and total levels of IRβ, IRS1, and Akt were upregulated by WEE treatment in PA-exposed HepG2 cells. The phosphorylation of GSK3β was elevated after WEE treatment in PA-treated cells. WEE treatment also concentration-dependently downregulated the phosphorylated CREB, ERK, c-Jun, p38 and JNK in PA-exposed HepG2 cells. Furthermore, the nuclear protein level and nuclear translocation of FoxO1 were also suppressed by WEE. Additionally, PA-induced changes of FoxO1 targeted genes were also attenuated by WEE treatment. The GLUT2 and GLUT4 translocation were also promoted by WEE treatment in PA-treated HepG2 cells. Taken together, WEE has potential anti-IR effect in PA-exposed HepG2 cells; the underlying mechanism of this action may be associated with the regulation of IRS1/GSK3β/FoxO1 signaling pathway. This study provides a pharmacological basis for the application of WEE in the treatment of metabolic diseases such as type 2 diabetes mellitus.
Collapse
Affiliation(s)
- Yi Zhang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Li Shan Yan
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Yu Ding
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Brian Chi Yan Cheng
- College of Professional and Continuing Education, Hong Kong Polytechnic University, Hong Kong, China
- Chinese Medicine Department of Quality Healthcare Medical Services , Hong Kong, China
| | - Gan Luo
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Jing Kong
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Tong Hua Liu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
- Tibetan Medicine Department of Tibetan Traditional Medical College, Lhasa, China
| | - Shuo Feng Zhang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
- Tibetan Medicine Department of Tibetan Traditional Medical College, Lhasa, China
| |
Collapse
|
16
|
Abstract
PURPOSE OF REVIEW Obesity is a major risk factor for type 2 diabetes. Although adipose tissue allows storage of excess calories in periods of overnutrition, in obesity, adipose tissue metabolism becomes dysregulated and can promote metabolic diseases. This review discusses recent advances in understandings how adipocyte metabolism impacts metabolic homeostasis. RECENT FINDINGS The ability of adipocytes to synthesize lipids from glucose is a marker of metabolic fitness, e.g., low de novo lipogenesis (DNL) in adipocytes correlates with insulin resistance in obesity. Adipocyte DNL may promote synthesis of special "insulin sensitizing" signaling lipids that act hormonally. However, each metabolic intermediate in the DNL pathway (i.e., citrate, acetyl-CoA, malonyl-CoA, and palmitate) also has second messenger functions. Mounting evidence suggests these signaling functions may also be important for maintaining healthy adipocytes. While adipocyte DNL contributes to lipid storage, lipid precursors may have additional second messenger functions critical for maintaining adipocyte health, and thus systemic metabolic homeostasis.
Collapse
Affiliation(s)
- Wen-Yu Hsiao
- Program in Molecular Medicine, University of Massachusetts Medical School, 373 Plantation Street, Worcester, MA, 01605, USA
| | - David A Guertin
- Program in Molecular Medicine, University of Massachusetts Medical School, 373 Plantation Street, Worcester, MA, 01605, USA.
| |
Collapse
|
17
|
Kawakami M, Yokota-Nakagi N, Takamata A, Morimoto K. Endurance running exercise is an effective alternative to estradiol replacement for restoring hyperglycemia through TBC1D1/GLUT4 pathway in skeletal muscle of ovariectomized rats. J Physiol Sci 2019; 69:1029-1040. [PMID: 31782092 PMCID: PMC10717071 DOI: 10.1007/s12576-019-00723-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 10/20/2019] [Indexed: 01/24/2023]
Abstract
Menopause is a risk factor for impaired glucose metabolism. Alternative treatment of estrogen for postmenopausal women is required. The present study was designed to investigate the effects of 5-week endurance running exercise (Ex) by treadmill on hyperglycemia and signal pathway components mediating glucose transport in ovariectomized (OVX) placebo-treated rats, compared with 4-week 17β-estradiol (E2) replacement or pair-feeding (PF) to the E2 group. Ex improved the hyperglycemia and insulin resistance index in OVX rats as much as E2 or PF did. However, Ex had no effect on body weight gain in the OVX rats. Moreover, Ex enhanced the levels of GLUT4 and phospho-TBC1D1 proteins in the gastrocnemius of the OVX rats, but E2 or PF did not. Instead, the E2 increased the Akt2/AS160 expression and activation in the OVX rats. This study suggests that endurance Ex training restored hyperglycemia through the TBC1D1/GLUT4 pathway in muscle by an alternative mechanism to E2 replacement.
Collapse
Affiliation(s)
- Mizuho Kawakami
- Department of Environmental Health, Faculty of Human Life and Environment, Nara Women's University, Kita-Uoya Nishi-machi, Nara, 630-8506, Japan
| | - Naoko Yokota-Nakagi
- Department of Environmental Health, Faculty of Human Life and Environment, Nara Women's University, Kita-Uoya Nishi-machi, Nara, 630-8506, Japan
| | - Akira Takamata
- Department of Environmental Health, Faculty of Human Life and Environment, Nara Women's University, Kita-Uoya Nishi-machi, Nara, 630-8506, Japan
| | - Keiko Morimoto
- Department of Environmental Health, Faculty of Human Life and Environment, Nara Women's University, Kita-Uoya Nishi-machi, Nara, 630-8506, Japan.
| |
Collapse
|
18
|
Gorres-Martens BK, Field TJ, Schmidt ER, Munger KA. Exercise prevents HFD- and OVX-induced type 2 diabetes risk factors by decreasing fat storage and improving fuel utilization. Physiol Rep 2019; 6:e13783. [PMID: 29981201 PMCID: PMC6035332 DOI: 10.14814/phy2.13783] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 06/11/2018] [Accepted: 06/14/2018] [Indexed: 11/24/2022] Open
Abstract
Previous studies suggest that the loss of estrogens increase one's risk for type 2 diabetes (T2D), and combining the loss of estrogens with a high-fat diet (HFD) poses an even greater risk for T2D. The extent to which exercise can ameliorate the deleterious effects of estrogen loss combined with a HFD and the molecular mechanisms accounting for the whole body changes is currently unknown. Therefore, we fed female Wistar rats a standard diet or a HFD for 10 weeks. The rats fed the HFD were either ovariectomized (OVX) or their ovaries remained intact. A subset of the HFD/OVX rats also underwent exercise training on a motor-driven treadmill. Exercise significantly reduced the total body weight gain, periuterine white adipose tissue (WAT) weight, hyperglycemia, and hyperinsulinemia. Additionally, the ability to store fat, as measured by lipoprotein lipase (LPL) in the WAT, was increased in the HFD/OVX group; however, exercise reduced the LPL levels. Furthermore, the combination of the HFD with OVX decreased the WAT citrate synthase protein level, which was increased with exercise. These data suggest that even during the combined HFD/OVX physiological state, exercise can decrease several risk factors associated with T2D, decrease fat storage, and increase fuel utilization.
Collapse
Affiliation(s)
| | - Tyler J Field
- Exercise and Sport Sciences Department, Augustana University, Sioux Falls, South Dakota
| | - Emma R Schmidt
- Exercise and Sport Sciences Department, Augustana University, Sioux Falls, South Dakota
| | - Karen A Munger
- Research & Development, Sioux Falls VA Health Care System, Sioux Falls, South Dakota
| |
Collapse
|
19
|
Wang H, Arias EB, Oki K, Pataky MW, Almallouhi JA, Cartee GD. Fiber type-selective exercise effects on AS160 phosphorylation. Am J Physiol Endocrinol Metab 2019; 316:E837-E851. [PMID: 30835507 PMCID: PMC6580176 DOI: 10.1152/ajpendo.00528.2018] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Earlier research using muscle tissue demonstrated that postexercise elevation in insulin-stimulated glucose uptake (ISGU) occurs concomitant with greater insulin-stimulated Akt substrate of 160 kDa (AS160) phosphorylation (pAS160) on sites that regulate ISGU. Because skeletal muscle is a heterogeneous tissue, we previously isolated myofibers from rat epitrochlearis to assess fiber type-selective ISGU. Exercise induced greater ISGU in type I, IIA, IIB, and IIBX but not IIX fibers. This study tested if exercise effects on pAS160 correspond with previously published fiber type-selective exercise effects on ISGU. Rats were studied immediately postexercise (IPEX) or 3.5 h postexercise (3.5hPEX) with time-matched sedentary controls. Myofibers dissected from the IPEX experiment were analyzed for fiber type (myosin heavy chain isoform expression) and key phosphoproteins. Isolated muscles from the 3.5hPEX experiment were incubated with or without insulin. Myofibers (3.5hPEX) were analyzed for fiber type, key phosphoproteins, and GLUT4 protein abundance. We hypothesized that insulin-stimulated pAS160 at 3.5hPEX would exceed sedentary controls only in fiber types characterized by greater ISGU postexercise. Values for phosphorylation of AMP-activated kinase substrates (acetyl CoA carboxylaseSer79 and AS160Ser704) from IPEX muscles exceeded sedentary values in each fiber type, suggesting exercise recruitment of all fiber types. Values for pAS160Thr642 and pAS160Ser704 from insulin-stimulated muscles 3.5hPEX exceeded sedentary values for type I, IIA, IIB, and IIBX but not IIX fibers. GLUT4 abundance was unaltered 3.5hPEX in any fiber type. These results advanced understanding of exercise-induced insulin sensitization by providing compelling support for the hypothesis that enhanced insulin-stimulated phosphorylation of AS160 is linked to elevated ISGU postexercise at a fiber type-specific level independent of altered GLUT4 expression.
Collapse
Affiliation(s)
- Haiyan Wang
- Muscle Biology Laboratory, School of Kinesiology, University of Michigan , Ann Arbor, Michigan
| | - Edward B Arias
- Muscle Biology Laboratory, School of Kinesiology, University of Michigan , Ann Arbor, Michigan
| | - Kentaro Oki
- Muscle Biology Laboratory, School of Kinesiology, University of Michigan , Ann Arbor, Michigan
| | - Mark W Pataky
- Muscle Biology Laboratory, School of Kinesiology, University of Michigan , Ann Arbor, Michigan
| | - Jalal A Almallouhi
- Muscle Biology Laboratory, School of Kinesiology, University of Michigan , Ann Arbor, Michigan
| | - Gregory D Cartee
- Muscle Biology Laboratory, School of Kinesiology, University of Michigan , Ann Arbor, Michigan
- Department of Molecular and Integrative Physiology, University of Michigan , Ann Arbor, Michigan
- Institute of Gerontology, University of Michigan , Ann Arbor, Michigan
| |
Collapse
|
20
|
Barzilai-Tutsch H, Dewulf M, Lamaze C, Butler Browne G, Pines M, Halevy O. A promotive effect for halofuginone on membrane repair and synaptotagmin-7 levels in muscle cells of dysferlin-null mice. Hum Mol Genet 2019; 27:2817-2829. [PMID: 29771357 DOI: 10.1093/hmg/ddy185] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Accepted: 05/09/2018] [Indexed: 11/14/2022] Open
Abstract
In the absence of dysferlin, skeletal muscle cells fail to reseal properly after injury, resulting in slow progress of the dysferlinopathy muscular dystrophy (MD). Halofuginone, a leading agent in preventing fibrosis in MDs, was tested for its effects on membrane resealing post-injury. A hypo-osmotic shock assay on myotubes derived from wild-type (Wt) and dysferlin-null (dysf-/-) mice revealed that pre-treatment with halofuginone reduces the percentage of membrane-ruptured myotubes only in dysf-/- myotubes. In laser-induced injury of isolated myofibers, halofuginone decreased the amount of FM1-43 at the injury site of dysf-/- myofibers while having no effect on Wt myofibers. These results implicate halofuginone in ameliorating muscle-cell membrane integrity in dysf-/- mice. Halofuginone increased lysosome scattering across the cytosol of dysf-/- primary myoblasts, in a protein kinase/extracellular signal-regulated protein kinase and phosphoinositide 3 kinase/Akt-dependent manner, in agreement with an elevation in lysosomal exocytotic activity in these cells. A spatial- and age-dependent synaptotagmin-7 (Syt-7) expression pattern was shown in dysf-/- versus Wt mice, suggesting that these pattern alterations are related to the disease progress and that sytnaptotagmin-7 may be compensating for the lack of dysferlin at least with regard to membrane resealing post-injury. While halofuginone did not affect patch-repair-complex key proteins, it further enhanced Syt-7 levels and its spread across the cytosol in dysf-/- myofibers and muscle tissue, and increased its co-localization with lysosomes. Together, the data imply a novel role for halofuginone in membrane-resealing events with Syt-7 possibly taking part in these events.
Collapse
Affiliation(s)
- Hila Barzilai-Tutsch
- Department of Animal Sciences, The Hebrew University of Jerusalem, Rehovot 76100, Israel
| | - Melissa Dewulf
- Membrane Dynamics and Mechanics of Intracellular Signaling Laboratory, Institut Curie-Centre de Recherche, PSL Research University, INSERM U1143, Centre national de la recherche scientifique, UMR 3666, Paris, France
| | - Christophe Lamaze
- Membrane Dynamics and Mechanics of Intracellular Signaling Laboratory, Institut Curie-Centre de Recherche, PSL Research University, INSERM U1143, Centre national de la recherche scientifique, UMR 3666, Paris, France
| | - Gillian Butler Browne
- Center for Research in Myology, CNRS FRE 3617, UPMC Univ Paris 06, UM76, INSERM U974, Sorbonne Universités, Paris, France
| | - Mark Pines
- The Volcani Center, Institute of Animal Science, Bet Dagan 52505, Israel
| | - Orna Halevy
- Department of Animal Sciences, The Hebrew University of Jerusalem, Rehovot 76100, Israel
| |
Collapse
|
21
|
Fu S, Zhu S, Tian C, Bai S, Zhang J, Zhan C, Xie D, Wang L, Li Z, Li J, Zhang H, Zhou R, Tian Z, Xu T, Bai L. Immunometabolism regulates TCR recycling and iNKT cell functions. Sci Signal 2019; 12:12/570/eaau1788. [PMID: 30808817 DOI: 10.1126/scisignal.aau1788] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Invariant natural killer T (iNKT) cells are innate-like T lymphocytes that express an invariant T cell receptor (TCR), which recognizes glycolipid antigens presented on CD1d molecules. These cells are phenotypically and functionally distinct from conventional T cells. When we characterized the metabolic activity of iNKT cells, consistent with their activated phenotype, we found that they had much less mitochondrial respiratory capacity but increased glycolytic activity in comparison to naïve conventional CD4+ T cells. After TCR engagement, iNKT cells further increased aerobic glycolysis, which was important for the expression of interferon-γ (IFN-γ). Glycolytic metabolism promoted the translocation of hexokinase-II to mitochondria and the activation of mammalian target of rapamycin complex 2 (mTORC2). Inhibiting glycolysis reduced the activity of Akt and PKCθ, which inhibited TCR recycling and accumulation within the immune synapse. Diminished TCR accumulation in the immune synapse reduced the activation of proximal and distal TCR signaling pathways and IFN-γ production in activated iNKT cells. Our studies demonstrate that glycolytic metabolism augments TCR signaling duration and IFN-γ production in iNKT cells by increasing TCR recycling.
Collapse
Affiliation(s)
- Sicheng Fu
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei 230027, China.,Innovation Center for Cell Signaling Network, Hefei 230027, China
| | - Shasha Zhu
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China.,Anhui Province Key Laboratory of Reproductive Health and Genetics, Anhui Medical University, Hefei 230022, China
| | - Chenxi Tian
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei 230027, China.,Innovation Center for Cell Signaling Network, Hefei 230027, China
| | - Shiyu Bai
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei 230027, China.,Innovation Center for Cell Signaling Network, Hefei 230027, China
| | - Jiqian Zhang
- Department of Anesthesiology, the First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei 230022, China
| | - Chonglun Zhan
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei 230027, China.,Innovation Center for Cell Signaling Network, Hefei 230027, China
| | - Di Xie
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei 230027, China.,Innovation Center for Cell Signaling Network, Hefei 230027, China
| | - Lu Wang
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei 230027, China.,Innovation Center for Cell Signaling Network, Hefei 230027, China
| | - Zonghong Li
- National Key Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Jie Li
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei 230027, China
| | - Huimin Zhang
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei 230027, China.,Innovation Center for Cell Signaling Network, Hefei 230027, China
| | - Rongbin Zhou
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei 230027, China.,Innovation Center for Cell Signaling Network, Hefei 230027, China
| | - Zhigang Tian
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei 230027, China
| | - Tao Xu
- National Key Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Li Bai
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei 230027, China. .,Innovation Center for Cell Signaling Network, Hefei 230027, China
| |
Collapse
|
22
|
Kupreeva M, Diane A, Lehner R, Watts R, Ghosh M, Proctor S, Vine D. Effect of metformin and flutamide on insulin, lipogenic and androgen-estrogen signaling, and cardiometabolic risk in a PCOS-prone metabolic syndrome rodent model. Am J Physiol Endocrinol Metab 2019; 316:E16-E33. [PMID: 30153063 PMCID: PMC6417686 DOI: 10.1152/ajpendo.00018.2018] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Revised: 08/10/2018] [Accepted: 08/22/2018] [Indexed: 02/06/2023]
Abstract
Polycystic ovary syndrome (PCOS) is highly associated with cardiometabolic risk and the metabolic syndrome (MetS), predisposing women to increased risk of developing type 2 diabetes and cardiovascular disease. Metformin is commonly used to treat insulin resistance-glucose intolerance, and flutamide, an androgen receptor (AR) antagonist, is used to target hyperandrogenemia and dyslipidemia. Currently, the physiological mechanism of action of these treatments on androgen, lipidogenic, and insulin signaling pathways remains unclear in PCOS. The aim of this study was to investigate the effects and mechanisms of action of metformin and flutamide on plasma lipid-apolipoprotein (Apo)B-lipoprotein and insulin-glucose metabolism, and endocrine-reproductive indices in a PCOS-prone MetS rodent model. PCOS-prone rodents were treated with metformin (300 mg/kg body wt), flutamide (30 mg/kg body wt), or metformin + flutamide combination treatment for 6 wk. Metformin was shown to improve fasting insulin and HOMA-IR, whereas flutamide and combination treatment were shown to reduce plasma triglycerides, ApoB48, and ApoB100, and this was associated with decreased intestinal secretion of ApoB48/triglyceride. Flutamide and metformin were shown to reduce plasma androgen indices and to improve ovarian primary and preovulatory follicle frequency. Metformin treatment increased hepatic estrogen receptor (ER)α, and metformin-flutamide decreased intestinal AR and increased ERα mRNA expression. Metformin-flutamide treatment upregulated hepatic and intestinal insulin signaling, including insulin receptor, MAPK1, and AKT2. In conclusion, cardiometabolic risk factors, in particular ApoB-hypertriglyceridemia, are independently modulated via the AR, and understanding the contribution of AR and insulin-signaling pathways further may facilitate the development of targeted interventions in high-risk women with PCOS and MetS.
Collapse
Affiliation(s)
- M. Kupreeva
- Metabolic and Cardiovascular Disease Laboratory, Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, Canada
| | - A. Diane
- Metabolic and Cardiovascular Disease Laboratory, Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, Canada
| | - R. Lehner
- Group on Molecular Cell Biology of Lipids, University of Alberta, Edmonton, Alberta, Canada
| | - R. Watts
- Group on Molecular Cell Biology of Lipids, University of Alberta, Edmonton, Alberta, Canada
| | - M. Ghosh
- Division of Endocrinology and Metabolism, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - S. Proctor
- Metabolic and Cardiovascular Disease Laboratory, Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, Canada
- Group on Molecular Cell Biology of Lipids, University of Alberta, Edmonton, Alberta, Canada
| | - D. Vine
- Metabolic and Cardiovascular Disease Laboratory, Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, Canada
- Group on Molecular Cell Biology of Lipids, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
23
|
Hatakeyama H, Morino T, Ishii T, Kanzaki M. Cooperative actions of Tbc1d1 and AS160/Tbc1d4 in GLUT4-trafficking activities. J Biol Chem 2018; 294:1161-1172. [PMID: 30482843 DOI: 10.1074/jbc.ra118.004614] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Revised: 11/13/2018] [Indexed: 12/28/2022] Open
Abstract
AS160 and Tbc1d1 are key Rab GTPase-activating proteins (RabGAPs) that mediate release of static GLUT4 in response to insulin or exercise-mimetic stimuli, respectively, but their cooperative regulation and its underlying mechanisms remain unclear. By employing GLUT4 nanometry with cell-based reconstitution models, we herein analyzed the functional cooperative activities of the RabGAPs. When both RabGAPs are present, Tbc1d1 functionally dominates AS160, and stimuli-inducible GLUT4 release relies on Tbc1d1-evoking proximal stimuli, such as AICAR and intracellular Ca2+ Detailed functional assessments with varying expression ratios revealed that AS160 modulates sensitivity to external stimuli in Tbc1d1-mediated GLUT4 release. For example, Tbc1d1-governed GLUT4 release triggered by Ca2+ plus insulin occurred more efficiently than that in cells with little or no AS160. Series of mutational analyses revealed that these synergizing actions rely on the phosphotyrosine-binding 1 (PTB1) and calmodulin-binding domains of Tbc1d1 as well as key phosphorylation sites of both AS160 (Thr642) and Tbc1d1 (Ser237 and Thr596). Thus, the emerging cooperative governance relying on the multiple regulatory nodes of both Tbc1d1 and AS160, functioning together, plays a key role in properly deciphering biochemical signals into a physical GLUT4 release process in response to insulin, exercise, and the two in combination.
Collapse
Affiliation(s)
- Hiroyasu Hatakeyama
- Frontier Research Institute for Interdisciplinary Sciences, Sendai 980-8579, Japan; Graduate School of Biomedical Engineering, Sendai 980-8579, Japan
| | - Taisuke Morino
- Department of Information and Intelligent Systems, Tohoku University, Sendai 980-8579, Japan
| | - Takuya Ishii
- Department of Information and Intelligent Systems, Tohoku University, Sendai 980-8579, Japan
| | - Makoto Kanzaki
- Graduate School of Biomedical Engineering, Sendai 980-8579, Japan; Department of Information and Intelligent Systems, Tohoku University, Sendai 980-8579, Japan.
| |
Collapse
|
24
|
Chemical biology probes of mammalian GLUT structure and function. Biochem J 2018; 475:3511-3534. [PMID: 30459202 PMCID: PMC6243331 DOI: 10.1042/bcj20170677] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Revised: 10/11/2018] [Accepted: 10/11/2018] [Indexed: 12/14/2022]
Abstract
The structure and function of glucose transporters of the mammalian GLUT family of proteins has been studied over many decades, and the proteins have fascinated numerous research groups over this time. This interest is related to the importance of the GLUTs as archetypical membrane transport facilitators, as key limiters of the supply of glucose to cell metabolism, as targets of cell insulin and exercise signalling and of regulated membrane traffic, and as potential drug targets to combat cancer and metabolic diseases such as type 2 diabetes and obesity. This review focusses on the use of chemical biology approaches and sugar analogue probes to study these important proteins.
Collapse
|
25
|
Crosstalk in transition: the translocation of Akt. J Math Biol 2018; 78:919-942. [PMID: 30306249 DOI: 10.1007/s00285-018-1297-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 09/17/2018] [Indexed: 12/30/2022]
Abstract
Akt/PKB is an important crosstalk node at the junction between a number of major signalling pathways in the mammalian cell. As a significant nutrient sensor, Akt plays a central role in many cellular processes, including cell growth, cell survival and glucose metabolism. The dysregulation of Akt signalling is implicated in the development of many diseases, from diabetes to cancer. The translocation of Akt from cytosol to plasma membrane is a crucial step in Akt activation. Akt is initially synthesized on the endoplasmic reticulum, but translocates to the plasma membrane (PM) in response to insulin stimulation, where it may be activated. The Akt is then recycled to the cytoplasm. The activated Akt may propagate signals to downstream substrates both at the PM and in the cytosol, hence understanding the translocation dynamics is an important step in dissecting the signalling system. At the present time, however, knowledge concerning the translocation of either activated and unactivated Akt is scant. Here we present a simple, deterministic, three-compartment ordinary differential equation model of Akt translocation in vitro. This model can reproduce the salient features of Akt translocation in a manner consistent with the experimental data. Furthermore, we demonstrate that this system is equivalent to a damped harmonic oscillator, and analyse the steady state and transient behaviour of the model over the entire parameter space.
Collapse
|
26
|
Petersen MC, Shulman GI. Mechanisms of Insulin Action and Insulin Resistance. Physiol Rev 2018; 98:2133-2223. [PMID: 30067154 PMCID: PMC6170977 DOI: 10.1152/physrev.00063.2017] [Citation(s) in RCA: 1682] [Impact Index Per Article: 240.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Revised: 03/22/2018] [Accepted: 03/24/2018] [Indexed: 12/15/2022] Open
Abstract
The 1921 discovery of insulin was a Big Bang from which a vast and expanding universe of research into insulin action and resistance has issued. In the intervening century, some discoveries have matured, coalescing into solid and fertile ground for clinical application; others remain incompletely investigated and scientifically controversial. Here, we attempt to synthesize this work to guide further mechanistic investigation and to inform the development of novel therapies for type 2 diabetes (T2D). The rational development of such therapies necessitates detailed knowledge of one of the key pathophysiological processes involved in T2D: insulin resistance. Understanding insulin resistance, in turn, requires knowledge of normal insulin action. In this review, both the physiology of insulin action and the pathophysiology of insulin resistance are described, focusing on three key insulin target tissues: skeletal muscle, liver, and white adipose tissue. We aim to develop an integrated physiological perspective, placing the intricate signaling effectors that carry out the cell-autonomous response to insulin in the context of the tissue-specific functions that generate the coordinated organismal response. First, in section II, the effectors and effects of direct, cell-autonomous insulin action in muscle, liver, and white adipose tissue are reviewed, beginning at the insulin receptor and working downstream. Section III considers the critical and underappreciated role of tissue crosstalk in whole body insulin action, especially the essential interaction between adipose lipolysis and hepatic gluconeogenesis. The pathophysiology of insulin resistance is then described in section IV. Special attention is given to which signaling pathways and functions become insulin resistant in the setting of chronic overnutrition, and an alternative explanation for the phenomenon of ‟selective hepatic insulin resistanceˮ is presented. Sections V, VI, and VII critically examine the evidence for and against several putative mediators of insulin resistance. Section V reviews work linking the bioactive lipids diacylglycerol, ceramide, and acylcarnitine to insulin resistance; section VI considers the impact of nutrient stresses in the endoplasmic reticulum and mitochondria on insulin resistance; and section VII discusses non-cell autonomous factors proposed to induce insulin resistance, including inflammatory mediators, branched-chain amino acids, adipokines, and hepatokines. Finally, in section VIII, we propose an integrated model of insulin resistance that links these mediators to final common pathways of metabolite-driven gluconeogenesis and ectopic lipid accumulation.
Collapse
Affiliation(s)
- Max C Petersen
- Departments of Internal Medicine and Cellular & Molecular Physiology, Howard Hughes Medical Institute, Yale University School of Medicine , New Haven, Connecticut
| | - Gerald I Shulman
- Departments of Internal Medicine and Cellular & Molecular Physiology, Howard Hughes Medical Institute, Yale University School of Medicine , New Haven, Connecticut
| |
Collapse
|
27
|
Insulin signaling acts in adult adipocytes via GSK-3β and independently of FOXO to control Drosophila female germline stem cell numbers. Dev Biol 2018; 440:31-39. [PMID: 29729259 DOI: 10.1016/j.ydbio.2018.04.028] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Revised: 04/30/2018] [Accepted: 04/30/2018] [Indexed: 12/14/2022]
Abstract
Tissue-specific stem cells are tied to the nutritional and physiological environment of adult organisms. Adipocytes have key endocrine and nutrient-sensing roles and have emerged as major players in relaying dietary information to regulate other organs. For example, previous studies in Drosophila melanogaster revealed that amino acid sensing as well as diet-dependent metabolic pathways function in adipocytes to influence the maintenance of female germline stem cells (GSCs). How nutrient-sensing pathways acting within adipocytes influence adult stem cell lineages, however, is just beginning to be elucidated. Here, we report that insulin/insulin-like growth factor signaling in adipocytes promotes GSC maintenance, early germline cyst survival, and vitellogenesis. Further, adipocytes use distinct mechanisms downstream of insulin receptor activation to control these aspects of oogenesis, all of which are independent of FOXO. We find that GSC maintenance is modulated by Akt1 through GSK-3β, early germline cyst survival is downstream of adipocyte Akt1 but independent of GSK-3β, and vitellogenesis is regulated through an Akt1-independent pathway in adipocytes. These results indicate that, in addition to employing different types of nutrient sensing, adipocytes can use distinct axes of a single nutrient-sensing pathway to regulate multiple stages of the GSC lineage in the ovary.
Collapse
|
28
|
Action of Phytochemicals on Insulin Signaling Pathways Accelerating Glucose Transporter (GLUT4) Protein Translocation. Molecules 2018; 23:molecules23020258. [PMID: 29382104 PMCID: PMC6017132 DOI: 10.3390/molecules23020258] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 01/15/2018] [Accepted: 01/23/2018] [Indexed: 11/17/2022] Open
Abstract
Diabetes is associated with obesity, generally accompanied by a chronic state of oxidative stress and redox imbalances which are implicated in the progression of micro- and macro-complications like heart disease, stroke, dementia, cancer, kidney failure and blindness. All these complications rise primarily due to consistent high blood glucose levels. Insulin and glucagon help to maintain the homeostasis of glucose and lipids through signaling cascades. Pancreatic hormones stimulate translocation of the glucose transporter isoform 4 (GLUT4) from an intracellular location to the cell surface and facilitate the rapid insulin-dependent storage of glucose in muscle and fat cells. Malfunction in glucose uptake mechanisms, primarily contribute to insulin resistance in type 2 diabetes. Plant secondary metabolites, commonly known as phytochemicals, are reported to have great benefits in the management of type 2 diabetes. The role of phytochemicals and their action on insulin signaling pathways through stimulation of GLUT4 translocation is crucial to understand the pathogenesis of this disease in the management process. This review will summarize the effects of phytochemicals and their action on insulin signaling pathways accelerating GLUT4 translocation based on the current literature.
Collapse
|
29
|
Whitfield J, Paglialunga S, Smith BK, Miotto PM, Simnett G, Robson HL, Jain SS, Herbst EAF, Desjardins EM, Dyck DJ, Spriet LL, Steinberg GR, Holloway GP. Ablating the protein TBC1D1 impairs contraction-induced sarcolemmal glucose transporter 4 redistribution but not insulin-mediated responses in rats. J Biol Chem 2017; 292:16653-16664. [PMID: 28808062 DOI: 10.1074/jbc.m117.806786] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Revised: 08/10/2017] [Indexed: 12/28/2022] Open
Abstract
TBC1 domain family member 1 (TBC1D1), a Rab GTPase-activating protein and paralogue of Akt substrate of 160 kDa (AS160), has been implicated in both insulin- and 5-aminoimidazole-4-carboxamide ribonucleotide formyltransferase/IMP cyclohydrolase-mediated glucose transporter type 4 (GLUT4) translocation. However, the role of TBC1D1 in contracting muscle remains ambiguous. We therefore explored the metabolic consequence of ablating TBC1D1 in both resting and contracting skeletal muscles, utilizing a rat TBC1D1 KO model. Although insulin administration rapidly increased (p < 0.05) plasma membrane GLUT4 content in both red and white gastrocnemius muscles, the TBC1D1 ablation did not alter this response nor did it affect whole-body insulin tolerance, suggesting that TBC1D1 is not required for insulin-induced GLUT4 trafficking events. Consistent with findings in other models of altered TBC1D1 protein levels, whole-animal and ex vivo skeletal muscle fat oxidation was increased in the TBC1D1 KO rats. Although there was no change in mitochondrial content in the KO rats, maximal ADP-stimulated respiration was higher in permeabilized muscle fibers, which may contribute to the increased reliance on fatty acids in resting KO animals. Despite this increase in mitochondrial oxidative capacity, run time to exhaustion at various intensities was impaired in the KO rats. Moreover, contraction-induced increases in sarcolemmal GLUT4 content and glucose uptake were lower in the white gastrocnemius of the KO animals. Altogether, our results highlight a critical role for TBC1D1 in exercise tolerance and contraction-mediated translocation of GLUT4 to the plasma membrane in skeletal muscle.
Collapse
Affiliation(s)
- Jamie Whitfield
- From the Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario N1G 2W1, Canada and
| | - Sabina Paglialunga
- From the Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario N1G 2W1, Canada and
| | - Brennan K Smith
- Division of Endocrinology and Metabolism, Department of Medicine, and
| | - Paula M Miotto
- From the Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario N1G 2W1, Canada and
| | - Genevieve Simnett
- From the Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario N1G 2W1, Canada and
| | - Holly L Robson
- From the Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario N1G 2W1, Canada and
| | - Swati S Jain
- From the Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario N1G 2W1, Canada and
| | - Eric A F Herbst
- From the Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario N1G 2W1, Canada and
| | - Eric M Desjardins
- Division of Endocrinology and Metabolism, Department of Medicine, and
| | - David J Dyck
- From the Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario N1G 2W1, Canada and
| | - Lawrence L Spriet
- From the Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario N1G 2W1, Canada and
| | - Gregory R Steinberg
- Division of Endocrinology and Metabolism, Department of Medicine, and.,Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario L8N 3Z5, Canada
| | - Graham P Holloway
- From the Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario N1G 2W1, Canada and
| |
Collapse
|
30
|
Diehl T, Mullins R, Kapogiannis D. Insulin resistance in Alzheimer's disease. Transl Res 2017; 183:26-40. [PMID: 28034760 PMCID: PMC5393926 DOI: 10.1016/j.trsl.2016.12.005] [Citation(s) in RCA: 90] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Revised: 12/05/2016] [Accepted: 12/06/2016] [Indexed: 12/14/2022]
Abstract
The links between systemic insulin resistance (IR), brain-specific IR, and Alzheimer's disease (AD) have been an extremely productive area of current research. This review will cover the fundamentals and pathways leading to IR, its connection to AD via cellular mechanisms, the most prominent methods and models used to examine it, an introduction to the role of extracellular vesicles (EVs) as a source of biomarkers for IR and AD, and an overview of modern clinical studies on the subject. To provide additional context, we also present a novel analysis of the spatial correlation of gene expression in the brain with the aid of Allen Human Brain Atlas data. Ultimately, examining the relation between IR and AD can be seen as a means of advancing the understanding of both disease states, with IR being a promising target for therapeutic strategies in AD treatment. In conclusion, we highlight the therapeutic potential of targeting brain IR in AD and the main strategies to pursue this goal.
Collapse
Affiliation(s)
- Thomas Diehl
- Laboratory of Neurosciences, Intramural Research Program, National Institute on Aging/National Institutes of Health (NIA/NIH), Baltimore, MD
| | - Roger Mullins
- Laboratory of Neurosciences, Intramural Research Program, National Institute on Aging/National Institutes of Health (NIA/NIH), Baltimore, MD
| | - Dimitrios Kapogiannis
- Laboratory of Neurosciences, Intramural Research Program, National Institute on Aging/National Institutes of Health (NIA/NIH), Baltimore, MD.
| |
Collapse
|
31
|
Zhou X, Shentu P, Xu Y. Spatiotemporal Regulators for Insulin-Stimulated GLUT4 Vesicle Exocytosis. J Diabetes Res 2017; 2017:1683678. [PMID: 28529958 PMCID: PMC5424486 DOI: 10.1155/2017/1683678] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Revised: 03/21/2017] [Accepted: 04/03/2017] [Indexed: 11/30/2022] Open
Abstract
Insulin increases glucose uptake and storage in muscle and adipose cells, which is accomplished through the mobilization of intracellular GLUT4 storage vesicles (GSVs) to the cell surface upon stimulation. Importantly, the dysfunction of insulin-regulated GLUT4 trafficking is strongly linked with peripheral insulin resistance and type 2 diabetes in human. The insulin signaling pathway, key signaling molecules involved, and precise trafficking itinerary of GSVs are largely identified. Understanding the interaction between insulin signaling molecules and key regulatory proteins that are involved in spatiotemporal regulation of GLUT4 vesicle exocytosis is of great importance to explain the pathogenesis of diabetes and may provide new potential therapeutic targets.
Collapse
Affiliation(s)
- Xiaoxu Zhou
- Department of Biomedical Engineering, Key Laboratory for Biomedical Engineering of Ministry of Education, Zhejiang Provincial Key Laboratory of Cardio-Cerebral Vascular Detection Technology and Medicinal Effectiveness Appraisal, Zhejiang University, Hangzhou 310027, China
| | - Ping Shentu
- Department of Biomedical Engineering, Key Laboratory for Biomedical Engineering of Ministry of Education, Zhejiang Provincial Key Laboratory of Cardio-Cerebral Vascular Detection Technology and Medicinal Effectiveness Appraisal, Zhejiang University, Hangzhou 310027, China
| | - Yingke Xu
- Department of Biomedical Engineering, Key Laboratory for Biomedical Engineering of Ministry of Education, Zhejiang Provincial Key Laboratory of Cardio-Cerebral Vascular Detection Technology and Medicinal Effectiveness Appraisal, Zhejiang University, Hangzhou 310027, China
- *Yingke Xu:
| |
Collapse
|
32
|
Beg M, Srivastava A, Shankar K, Varshney S, Rajan S, Gupta A, Kumar D, Gaikwad AN. PPP2R5B, a regulatory subunit of PP2A, contributes to adipocyte insulin resistance. Mol Cell Endocrinol 2016; 437:97-107. [PMID: 27521959 DOI: 10.1016/j.mce.2016.08.016] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Revised: 07/04/2016] [Accepted: 08/09/2016] [Indexed: 12/29/2022]
Abstract
Insulin resistance is associated with deregulation of insulin signaling owing to the chronic exposure of insulin (hyperinsulinemia) to the tissues. Phosphorylation and dephosphorylation events in insulin signaling pathway play an essential role in signal transduction and glucose uptake. Amongst all, Akt protein is considered to be central to the overall insulin signaling proteins. In glucose responsive tissues like adipose and muscles, activation of Akt is responsible for triggering GLUT4 translocation and glucose transport. Several phosphatases such as PTEN, PP2A have been reported to be involved in dephosphorylation and inactivation of Akt protein. We have identified increased PP2A activity during state of chronic hyperinsulinemia exposure along-with development of adipocyte insulin resistance. This increased phosphatase activity leads activation of cAMP/PKA axis, which in turn increased cAMP levels in insulin resistant (IR) adipocytes. Okadaic acid, an inhibitor of PP2A restored and increased insulin stimulated glucose uptake in insulin resistant (IR) and insulin sensitive (IS) adipocytes respectively. In IS adipocyte, chemical activation of PP2A through MG132 and FTY720 showed decreased insulin sensitivity corroborated with decreased Akt phosphorylation and glucose uptake. We also observed an increased expression of PP2A-B (regulatory) subunit in IR adipocytes. We found PPP2R5B, a regulatory subunit of PP2A is responsible for the dephosphorylation and inactivation of Akt protein. Increased expression of PPP2R5B was also confirmed in white adipose tissue of high fat diet induced IR mice model. Overexpression and suppression strategies confirmed the role of PPP2R5B in regulating insulin signaling. Thus, we conclude that PPP2R5B, a B subunit of PP2A is a negative regulator of Akt phosphorylation contributing partly to the chronic hyperinsulinemia induced insulin resistance in adipocytes.
Collapse
Affiliation(s)
- Muheeb Beg
- Division of Pharmacology, CSIR-Central Drug Research Institute, Lucknow, 226031, India
| | - Ankita Srivastava
- Division of Pharmacology, CSIR-Central Drug Research Institute, Lucknow, 226031, India; Academy of Scientific and Innovative Research, CSIR-CDRI, India
| | - Kripa Shankar
- Division of Pharmacology, CSIR-Central Drug Research Institute, Lucknow, 226031, India
| | - Salil Varshney
- Division of Pharmacology, CSIR-Central Drug Research Institute, Lucknow, 226031, India
| | - Sujith Rajan
- Division of Pharmacology, CSIR-Central Drug Research Institute, Lucknow, 226031, India; Academy of Scientific and Innovative Research, CSIR-CDRI, India
| | - Abhishek Gupta
- Division of Pharmacology, CSIR-Central Drug Research Institute, Lucknow, 226031, India
| | - Durgesh Kumar
- Division of Pharmacology, CSIR-Central Drug Research Institute, Lucknow, 226031, India; Academy of Scientific and Innovative Research, CSIR-CDRI, India
| | - Anil N Gaikwad
- Division of Pharmacology, CSIR-Central Drug Research Institute, Lucknow, 226031, India; Academy of Scientific and Innovative Research, CSIR-CDRI, India.
| |
Collapse
|
33
|
Gao L, Chen J, Gao J, Wang H, Xiong W. Super-resolution microscopy reveals the insulin-resistance-regulated reorganization of GLUT4 on plasma membranes. J Cell Sci 2016; 130:396-405. [PMID: 27888215 DOI: 10.1242/jcs.192450] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Accepted: 11/11/2016] [Indexed: 02/01/2023] Open
Abstract
GLUT4 (also known as SLC2A4) is essential for glucose uptake in skeletal muscles and adipocytes, which play central roles in whole-body glucose metabolism. Here, using direct stochastic optical reconstruction microscopy (dSTORM) to investigate the characteristics of plasma-membrane-fused GLUT4 at the single-molecule level, we have demonstrated that insulin and insulin resistance regulate the spatial organization of GLUT4 in adipocytes. Stimulation with insulin shifted the balance of GLUT4 on the plasma membrane toward a more dispersed configuration. In contrast, insulin resistance induced a more clustered distribution of GLUT4 and increased the mean number of molecules per cluster. Furthermore, our data demonstrate that the F5QQI motif and lipid rafts mediate the maintenance of GLUT4 clusters on the plasma membrane. Mutation of F5QQI (F5QQA-GLUT4) induced a more clustered distribution of GLUT4; moreover, destruction of lipid rafts in adipocytes expressing F5QQA-GLUT4 dramatically decreased the percentage of large clusters and the mean number of molecules per cluster. In conclusion, our data clarify the effects of insulin stimulation or insulin resistance on GLUT4 reorganization on the plasma membrane and reveal new pathogenic mechanisms of insulin resistance.
Collapse
Affiliation(s)
- Lan Gao
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, Yunnan 650201, P.R. China.,Graduate University of Chinese Academy of Sciences, Beijing 100049, P.R. China
| | - Junling Chen
- Graduate University of Chinese Academy of Sciences, Beijing 100049, P.R. China.,State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilil 130022, P.R. China
| | - Jing Gao
- Graduate University of Chinese Academy of Sciences, Beijing 100049, P.R. China.,State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilil 130022, P.R. China
| | - Hongda Wang
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilil 130022, P.R. China
| | - Wenyong Xiong
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, Yunnan 650201, P.R. China
| |
Collapse
|
34
|
Chanda D, Luiken JJFP, Glatz JFC. Signaling pathways involved in cardiac energy metabolism. FEBS Lett 2016; 590:2364-74. [PMID: 27403883 DOI: 10.1002/1873-3468.12297] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Revised: 07/09/2016] [Accepted: 07/11/2016] [Indexed: 11/09/2022]
Abstract
Various signaling pathways coordinate energy metabolism and contractile function in the heart. Myocardial uptake of long-chain fatty acids largely occurs by facilitated diffusion, involving the membrane-associated protein, CD36. Glucose uptake, the rate-limiting step in glucose utilization, is mediated predominantly by the glucose transporter protein, GLUT4. Insulin and contraction-mediated AMPK signaling each are implicated in tightly regulating these myocardial 'gate-keepers' of energy balance, that is, CD36 and GLUT4. The insulin and AMPK signaling cascades are complex and their cross-talk is only beginning to be understood. Moreover, transcriptional regulation of the CD36 and GLUT4 is significantly understudied. This review focuses on recent advances on the role of these signaling pathways and transcription factors involved in the regulation of CD36 and GLUT4.
Collapse
Affiliation(s)
- Dipanjan Chanda
- Department of Genetics and Cell Biology, CARIM School of Cardiovascular Diseases, Maastricht University, The Netherlands
| | - Joost J F P Luiken
- Department of Genetics and Cell Biology, CARIM School of Cardiovascular Diseases, Maastricht University, The Netherlands
| | - Jan F C Glatz
- Department of Genetics and Cell Biology, CARIM School of Cardiovascular Diseases, Maastricht University, The Netherlands
| |
Collapse
|
35
|
Bruno J, Brumfield A, Chaudhary N, Iaea D, McGraw TE. SEC16A is a RAB10 effector required for insulin-stimulated GLUT4 trafficking in adipocytes. J Cell Biol 2016; 214:61-76. [PMID: 27354378 PMCID: PMC4932369 DOI: 10.1083/jcb.201509052] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Accepted: 06/08/2016] [Indexed: 12/19/2022] Open
Abstract
Sec16A is known to be required for COPII vesicle formation from the ER. Here, Bruno et al. show that, independent of its role at the ER, Sec16A is a RAB10 effector involved in the insulin-stimulated formation of specialized transport vesicles that ferry the GLUT4 glucose transporter to the plasma membrane of adipocytes. RAB10 is a regulator of insulin-stimulated translocation of the GLUT4 glucose transporter to the plasma membrane (PM) of adipocytes, which is essential for whole-body glucose homeostasis. We establish SEC16A as a novel RAB10 effector in this process. Colocalization of SEC16A with RAB10 is augmented by insulin stimulation, and SEC16A knockdown attenuates insulin-induced GLUT4 translocation, phenocopying RAB10 knockdown. We show that SEC16A and RAB10 promote insulin-stimulated mobilization of GLUT4 from a perinuclear recycling endosome/TGN compartment. We propose RAB10–SEC16A functions to accelerate formation of the vesicles that ferry GLUT4 to the PM during insulin stimulation. Because GLUT4 continually cycles between the PM and intracellular compartments, the maintenance of elevated cell-surface GLUT4 in the presence of insulin requires accelerated biogenesis of the specialized GLUT4 transport vesicles. The function of SEC16A in GLUT4 trafficking is independent of its previously characterized activity in ER exit site formation and therefore independent of canonical COPII-coated vesicle function. However, our data support a role for SEC23A, but not the other COPII components SEC13, SEC23B, and SEC31, in the insulin stimulation of GLUT4 trafficking, suggesting that vesicles derived from subcomplexes of COPII coat proteins have a role in the specialized trafficking of GLUT4.
Collapse
Affiliation(s)
- Joanne Bruno
- Department of Biochemistry, Weill Cornell Medical College, New York, NY 10065 Weill Cornell/Rockefeller/Sloan Kettering Tri-Institutional MD-PhD Program, New York, NY 10065
| | | | - Natasha Chaudhary
- Department of Biochemistry, Weill Cornell Medical College, New York, NY 10065
| | - David Iaea
- Department of Biochemistry, Weill Cornell Medical College, New York, NY 10065
| | - Timothy E McGraw
- Department of Biochemistry, Weill Cornell Medical College, New York, NY 10065 Department of Cardiothoracic Surgery, Weill Cornell Medical College, New York, NY 10065
| |
Collapse
|
36
|
Xu Y, Nan D, Fan J, Bogan JS, Toomre D. Optogenetic activation reveals distinct roles of PIP3 and Akt in adipocyte insulin action. J Cell Sci 2016; 129:2085-95. [PMID: 27076519 DOI: 10.1242/jcs.174805] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2015] [Accepted: 03/31/2016] [Indexed: 12/26/2022] Open
Abstract
Glucose transporter 4 (GLUT4; also known as SLC2A4) resides on intracellular vesicles in muscle and adipose cells, and translocates to the plasma membrane in response to insulin. The phosphoinositide 3-kinase (PI3K)-Akt signaling pathway plays a major role in GLUT4 translocation; however, a challenge has been to unravel the potentially distinct contributions of PI3K and Akt (of which there are three isoforms, Akt1-Akt3) to overall insulin action. Here, we describe new optogenetic tools based on CRY2 and the N-terminus of CIB1 (CIBN). We used these 'Opto' modules to activate PI3K and Akt selectively in time and space in 3T3-L1 adipocytes. We validated these tools using biochemical assays and performed live-cell kinetic analyses of IRAP-pHluorin translocation (IRAP is also known as LNPEP and acts as a surrogate marker for GLUT4 here). Strikingly, Opto-PIP3 largely mimicked the maximal effects of insulin stimulation, whereas Opto-Akt only partially triggered translocation. Conversely, drug-mediated inhibition of Akt only partially dampened the translocation response of Opto-PIP3 In spatial optogenetic studies, focal targeting of Akt to a region of the cell marked the sites where IRAP-pHluorin vesicles fused, supporting the idea that local Akt-mediated signaling regulates exocytosis. Taken together, these results indicate that PI3K and Akt play distinct roles, and that PI3K stimulates Akt-independent pathways that are important for GLUT4 translocation.
Collapse
Affiliation(s)
- Yingke Xu
- Department of Biomedical Engineering, MOE Key Laboratory of Biomedical Engineering, Zhejiang Provincial Key Laboratory of Cardio-Cerebral Vascular Detection Technology and Medicinal Effectiveness Appraisal, Zhejiang University, Hangzhou 310027, China Department of Cell Biology, Yale University School of Medicine, New Haven, 06510, USA
| | - Di Nan
- Department of Biomedical Engineering, MOE Key Laboratory of Biomedical Engineering, Zhejiang Provincial Key Laboratory of Cardio-Cerebral Vascular Detection Technology and Medicinal Effectiveness Appraisal, Zhejiang University, Hangzhou 310027, China
| | - Jiannan Fan
- Department of Biomedical Engineering, MOE Key Laboratory of Biomedical Engineering, Zhejiang Provincial Key Laboratory of Cardio-Cerebral Vascular Detection Technology and Medicinal Effectiveness Appraisal, Zhejiang University, Hangzhou 310027, China
| | - Jonathan S Bogan
- Department of Cell Biology, Yale University School of Medicine, New Haven, 06510, USA Section of Endocrinology and Metabolism, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520-8020, USA
| | - Derek Toomre
- Department of Cell Biology, Yale University School of Medicine, New Haven, 06510, USA
| |
Collapse
|
37
|
The Akt switch model: Is location sufficient? J Theor Biol 2016; 398:103-11. [PMID: 26992575 DOI: 10.1016/j.jtbi.2016.03.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Revised: 03/07/2016] [Accepted: 03/07/2016] [Indexed: 12/18/2022]
Abstract
Akt/PKB is a biochemical regulator that functions as an important cross-talk node between several signalling pathways in the mammalian cell. In particular, Akt is a key mediator of glucose transport in response to insulin. The phosphorylation (activation) of only a small percentage of the Akt pool of insulin-sensitive cells results in maximal translocation of glucose transporter 4 (GLUT4) to the plasma membrane (PM). This enables the diffusion of glucose into the cell. The dysregulation of Akt signalling is associated with the development of diabetes, cancer and cardiovascular disease. Akt is synthesised in the cytoplasm in the inactive state. Under the influence of insulin, it moves to the PM, where it is phosphorylated to form pAkt. Although phosphorylation occurs only at the PM, pAkt is found in many cellular locations, including the PM, the cytoplasm, and the nucleus. Indeed, the spatial distribution of pAkt within the cell appears to be an important determinant of downstream regulation. Here we present a simple, linear, four-compartment ordinary differential equation (ODE) model of Akt activation that tracks both the biochemical state and the physical location of Akt. This model embodies the main features of the activation of this important cross-talk node and is consistent with the experimental data. In particular, it allows different downstream signalling motifs without invoking separate feedback pathways. Moreover, the model is computationally tractable, readily analysed, and elucidates some of the apparent anomalies in insulin signalling via Akt.
Collapse
|
38
|
Abstract
The serine/threonine kinase Akt/PKB (protein kinase B) is key for mammalian cell growth, survival, metabolism and oncogenic transformation. The diverse level and tissue expression of its three isoforms, Akt1/PKBα, Akt2/PKBβ and Akt3/PKBγ, make it daunting to identify isoform-specific actions in vivo and even in isolated tissues/cells. To date, isoform-specific knockout and knockdown have been the best strategies to dissect their individual overall functions. In a recent article in the Biochemical Journal, Kajno et al. reported a new strategy to study isoform selectivity in cell lines. Individual Akt/PKB isoforms in 3T3-L1 pre-adipocytes are first silenced via shRNA and stable cellular clones lacking one or the other isoform are selected. The stably silenced isoform is then replaced by a mutant engineered to be refractory to inhibition by MK-2206 (Akt1(W80A) or Akt2(W80A)). Akt1(W80A) or Akt2(W80A) are functional and effectively recruited to the plasma membrane in response to insulin. The system affords the opportunity to acutely control the activity of the endogenous non-silenced isoform through timely addition of MK-2206. Using this approach, it is confirmed that Akt1/PKBα is the preferred isoform sustaining adipocyte differentiation, but both Akt1/PKBα and Akt2/PKBβ can indistinctly support insulin-dependent FoxO1 (forkhead box O1) nuclear exclusion. Surprisingly, either isoform can also support insulin-dependent glucose transporter (GLUT) 4 translocation to the membrane, in contrast with the preferential role of Akt2/PKBβ assessed by knockdown studies. The new strategy should allow analysis of the plurality of Akt/PKB functions in other cells and in response to other stimuli. It should also be amenable to high-throughput studies to speed up advances in signal transmission by this pivotal kinase.
Collapse
|
39
|
Lee YJ, Liu C, Liao M, Sukhova GK, Shirakawa J, Abdennour M, Iamarene K, Andre S, Inouye K, Clement K, Kulkarni RN, Banks AS, Libby P, Shi GP. Deficiency of FcϵR1 Increases Body Weight Gain but Improves Glucose Tolerance in Diet-Induced Obese Mice. Endocrinology 2015; 156:4047-58. [PMID: 26295369 PMCID: PMC4606759 DOI: 10.1210/en.2015-1184] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Prior studies demonstrated increased plasma IgE in diabetic patients, but the direct participation of IgE in diabetes or obesity remains unknown. This study found that plasma IgE levels correlated inversely with body weight, body mass index, and body fat mass among a population of randomly selected obese women. IgE receptor FcϵR1-deficient (Fcer1a(-/-)) mice and diet-induced obesity (DIO) mice demonstrated that FcϵR1 deficiency in DIO mice increased food intake, reduced energy expenditure, and increased body weight gain but improved glucose tolerance and glucose-induced insulin secretion. White adipose tissue from Fcer1a(-/-) mice showed an increased expression of phospho-AKT, CCAAT/enhancer binding protein-α, peroxisome proliferator-activated receptor-γ, glucose transporter-4 (Glut4), and B-cell lymphoma 2 (Bcl2) but reduced uncoupling protein 1 (UCP1) and phosphorylated c-Jun N-terminal kinase (JNK) expression, tissue macrophage accumulation, and apoptosis, suggesting that IgE reduces adipogenesis and glucose uptake but induces energy expenditure, adipocyte apoptosis, and white adipose tissue inflammation. In 3T3-L1 cells, IgE inhibited the expression of CCAAT/enhancer binding protein-α and peroxisome proliferator-activated receptor-γ, and preadipocyte adipogenesis and induced adipocyte apoptosis. IgE reduced the 3T3-L1 cell expression of Glut4, phospho-AKT, and glucose uptake, which concurred with improved glucose tolerance in Fcer1a(-/-) mice. This study established two novel pathways of IgE in reducing body weight gain in DIO mice by suppressing adipogenesis and inducing adipocyte apoptosis while worsening glucose tolerance by reducing Glut4 expression, glucose uptake, and insulin secretion.
Collapse
Affiliation(s)
- Yun-Jung Lee
- Department of Medicine (Y.-J.L., C.L., M.L., G.K.S., K.I., A.S.B., P.L., G.-P.S.), Brigham and Women's Hospital and Harvard Medical School, Department of Genetics and Complex Diseases (K.I.), School of Public Health, Harvard University, and Department of Cell Biology (J.S., R.N.K.), Joslin Diabetes Center and Harvard Medical School, Boston, Massachusetts 02115; Department of Cardiology (C.L.), Institute of Clinical Medicine, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450003, People's Republic of China; Institute of Cardiology (M.L.), Union Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan 430072, People's Republic of China; and NutriOmique team (M.A., S.A., K.C.), Institute of Cardiometabolism and Nutrition, INSERM, Unité Mixte de Recherche en Santé Unité 1166, and NutriOmique team (M.A., S.A., K.C.), Université Pierre et Marie Curie-Paris 6, Paris F-75013 France
| | - Conglin Liu
- Department of Medicine (Y.-J.L., C.L., M.L., G.K.S., K.I., A.S.B., P.L., G.-P.S.), Brigham and Women's Hospital and Harvard Medical School, Department of Genetics and Complex Diseases (K.I.), School of Public Health, Harvard University, and Department of Cell Biology (J.S., R.N.K.), Joslin Diabetes Center and Harvard Medical School, Boston, Massachusetts 02115; Department of Cardiology (C.L.), Institute of Clinical Medicine, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450003, People's Republic of China; Institute of Cardiology (M.L.), Union Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan 430072, People's Republic of China; and NutriOmique team (M.A., S.A., K.C.), Institute of Cardiometabolism and Nutrition, INSERM, Unité Mixte de Recherche en Santé Unité 1166, and NutriOmique team (M.A., S.A., K.C.), Université Pierre et Marie Curie-Paris 6, Paris F-75013 France
| | - Mengyang Liao
- Department of Medicine (Y.-J.L., C.L., M.L., G.K.S., K.I., A.S.B., P.L., G.-P.S.), Brigham and Women's Hospital and Harvard Medical School, Department of Genetics and Complex Diseases (K.I.), School of Public Health, Harvard University, and Department of Cell Biology (J.S., R.N.K.), Joslin Diabetes Center and Harvard Medical School, Boston, Massachusetts 02115; Department of Cardiology (C.L.), Institute of Clinical Medicine, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450003, People's Republic of China; Institute of Cardiology (M.L.), Union Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan 430072, People's Republic of China; and NutriOmique team (M.A., S.A., K.C.), Institute of Cardiometabolism and Nutrition, INSERM, Unité Mixte de Recherche en Santé Unité 1166, and NutriOmique team (M.A., S.A., K.C.), Université Pierre et Marie Curie-Paris 6, Paris F-75013 France
| | - Galina K Sukhova
- Department of Medicine (Y.-J.L., C.L., M.L., G.K.S., K.I., A.S.B., P.L., G.-P.S.), Brigham and Women's Hospital and Harvard Medical School, Department of Genetics and Complex Diseases (K.I.), School of Public Health, Harvard University, and Department of Cell Biology (J.S., R.N.K.), Joslin Diabetes Center and Harvard Medical School, Boston, Massachusetts 02115; Department of Cardiology (C.L.), Institute of Clinical Medicine, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450003, People's Republic of China; Institute of Cardiology (M.L.), Union Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan 430072, People's Republic of China; and NutriOmique team (M.A., S.A., K.C.), Institute of Cardiometabolism and Nutrition, INSERM, Unité Mixte de Recherche en Santé Unité 1166, and NutriOmique team (M.A., S.A., K.C.), Université Pierre et Marie Curie-Paris 6, Paris F-75013 France
| | - Jun Shirakawa
- Department of Medicine (Y.-J.L., C.L., M.L., G.K.S., K.I., A.S.B., P.L., G.-P.S.), Brigham and Women's Hospital and Harvard Medical School, Department of Genetics and Complex Diseases (K.I.), School of Public Health, Harvard University, and Department of Cell Biology (J.S., R.N.K.), Joslin Diabetes Center and Harvard Medical School, Boston, Massachusetts 02115; Department of Cardiology (C.L.), Institute of Clinical Medicine, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450003, People's Republic of China; Institute of Cardiology (M.L.), Union Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan 430072, People's Republic of China; and NutriOmique team (M.A., S.A., K.C.), Institute of Cardiometabolism and Nutrition, INSERM, Unité Mixte de Recherche en Santé Unité 1166, and NutriOmique team (M.A., S.A., K.C.), Université Pierre et Marie Curie-Paris 6, Paris F-75013 France
| | - Meriem Abdennour
- Department of Medicine (Y.-J.L., C.L., M.L., G.K.S., K.I., A.S.B., P.L., G.-P.S.), Brigham and Women's Hospital and Harvard Medical School, Department of Genetics and Complex Diseases (K.I.), School of Public Health, Harvard University, and Department of Cell Biology (J.S., R.N.K.), Joslin Diabetes Center and Harvard Medical School, Boston, Massachusetts 02115; Department of Cardiology (C.L.), Institute of Clinical Medicine, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450003, People's Republic of China; Institute of Cardiology (M.L.), Union Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan 430072, People's Republic of China; and NutriOmique team (M.A., S.A., K.C.), Institute of Cardiometabolism and Nutrition, INSERM, Unité Mixte de Recherche en Santé Unité 1166, and NutriOmique team (M.A., S.A., K.C.), Université Pierre et Marie Curie-Paris 6, Paris F-75013 France
| | - Karine Iamarene
- Department of Medicine (Y.-J.L., C.L., M.L., G.K.S., K.I., A.S.B., P.L., G.-P.S.), Brigham and Women's Hospital and Harvard Medical School, Department of Genetics and Complex Diseases (K.I.), School of Public Health, Harvard University, and Department of Cell Biology (J.S., R.N.K.), Joslin Diabetes Center and Harvard Medical School, Boston, Massachusetts 02115; Department of Cardiology (C.L.), Institute of Clinical Medicine, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450003, People's Republic of China; Institute of Cardiology (M.L.), Union Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan 430072, People's Republic of China; and NutriOmique team (M.A., S.A., K.C.), Institute of Cardiometabolism and Nutrition, INSERM, Unité Mixte de Recherche en Santé Unité 1166, and NutriOmique team (M.A., S.A., K.C.), Université Pierre et Marie Curie-Paris 6, Paris F-75013 France
| | - Sebastien Andre
- Department of Medicine (Y.-J.L., C.L., M.L., G.K.S., K.I., A.S.B., P.L., G.-P.S.), Brigham and Women's Hospital and Harvard Medical School, Department of Genetics and Complex Diseases (K.I.), School of Public Health, Harvard University, and Department of Cell Biology (J.S., R.N.K.), Joslin Diabetes Center and Harvard Medical School, Boston, Massachusetts 02115; Department of Cardiology (C.L.), Institute of Clinical Medicine, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450003, People's Republic of China; Institute of Cardiology (M.L.), Union Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan 430072, People's Republic of China; and NutriOmique team (M.A., S.A., K.C.), Institute of Cardiometabolism and Nutrition, INSERM, Unité Mixte de Recherche en Santé Unité 1166, and NutriOmique team (M.A., S.A., K.C.), Université Pierre et Marie Curie-Paris 6, Paris F-75013 France
| | - Karen Inouye
- Department of Medicine (Y.-J.L., C.L., M.L., G.K.S., K.I., A.S.B., P.L., G.-P.S.), Brigham and Women's Hospital and Harvard Medical School, Department of Genetics and Complex Diseases (K.I.), School of Public Health, Harvard University, and Department of Cell Biology (J.S., R.N.K.), Joslin Diabetes Center and Harvard Medical School, Boston, Massachusetts 02115; Department of Cardiology (C.L.), Institute of Clinical Medicine, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450003, People's Republic of China; Institute of Cardiology (M.L.), Union Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan 430072, People's Republic of China; and NutriOmique team (M.A., S.A., K.C.), Institute of Cardiometabolism and Nutrition, INSERM, Unité Mixte de Recherche en Santé Unité 1166, and NutriOmique team (M.A., S.A., K.C.), Université Pierre et Marie Curie-Paris 6, Paris F-75013 France
| | - Karine Clement
- Department of Medicine (Y.-J.L., C.L., M.L., G.K.S., K.I., A.S.B., P.L., G.-P.S.), Brigham and Women's Hospital and Harvard Medical School, Department of Genetics and Complex Diseases (K.I.), School of Public Health, Harvard University, and Department of Cell Biology (J.S., R.N.K.), Joslin Diabetes Center and Harvard Medical School, Boston, Massachusetts 02115; Department of Cardiology (C.L.), Institute of Clinical Medicine, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450003, People's Republic of China; Institute of Cardiology (M.L.), Union Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan 430072, People's Republic of China; and NutriOmique team (M.A., S.A., K.C.), Institute of Cardiometabolism and Nutrition, INSERM, Unité Mixte de Recherche en Santé Unité 1166, and NutriOmique team (M.A., S.A., K.C.), Université Pierre et Marie Curie-Paris 6, Paris F-75013 France
| | - Rohit N Kulkarni
- Department of Medicine (Y.-J.L., C.L., M.L., G.K.S., K.I., A.S.B., P.L., G.-P.S.), Brigham and Women's Hospital and Harvard Medical School, Department of Genetics and Complex Diseases (K.I.), School of Public Health, Harvard University, and Department of Cell Biology (J.S., R.N.K.), Joslin Diabetes Center and Harvard Medical School, Boston, Massachusetts 02115; Department of Cardiology (C.L.), Institute of Clinical Medicine, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450003, People's Republic of China; Institute of Cardiology (M.L.), Union Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan 430072, People's Republic of China; and NutriOmique team (M.A., S.A., K.C.), Institute of Cardiometabolism and Nutrition, INSERM, Unité Mixte de Recherche en Santé Unité 1166, and NutriOmique team (M.A., S.A., K.C.), Université Pierre et Marie Curie-Paris 6, Paris F-75013 France
| | - Alexander S Banks
- Department of Medicine (Y.-J.L., C.L., M.L., G.K.S., K.I., A.S.B., P.L., G.-P.S.), Brigham and Women's Hospital and Harvard Medical School, Department of Genetics and Complex Diseases (K.I.), School of Public Health, Harvard University, and Department of Cell Biology (J.S., R.N.K.), Joslin Diabetes Center and Harvard Medical School, Boston, Massachusetts 02115; Department of Cardiology (C.L.), Institute of Clinical Medicine, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450003, People's Republic of China; Institute of Cardiology (M.L.), Union Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan 430072, People's Republic of China; and NutriOmique team (M.A., S.A., K.C.), Institute of Cardiometabolism and Nutrition, INSERM, Unité Mixte de Recherche en Santé Unité 1166, and NutriOmique team (M.A., S.A., K.C.), Université Pierre et Marie Curie-Paris 6, Paris F-75013 France
| | - Peter Libby
- Department of Medicine (Y.-J.L., C.L., M.L., G.K.S., K.I., A.S.B., P.L., G.-P.S.), Brigham and Women's Hospital and Harvard Medical School, Department of Genetics and Complex Diseases (K.I.), School of Public Health, Harvard University, and Department of Cell Biology (J.S., R.N.K.), Joslin Diabetes Center and Harvard Medical School, Boston, Massachusetts 02115; Department of Cardiology (C.L.), Institute of Clinical Medicine, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450003, People's Republic of China; Institute of Cardiology (M.L.), Union Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan 430072, People's Republic of China; and NutriOmique team (M.A., S.A., K.C.), Institute of Cardiometabolism and Nutrition, INSERM, Unité Mixte de Recherche en Santé Unité 1166, and NutriOmique team (M.A., S.A., K.C.), Université Pierre et Marie Curie-Paris 6, Paris F-75013 France
| | - Guo-Ping Shi
- Department of Medicine (Y.-J.L., C.L., M.L., G.K.S., K.I., A.S.B., P.L., G.-P.S.), Brigham and Women's Hospital and Harvard Medical School, Department of Genetics and Complex Diseases (K.I.), School of Public Health, Harvard University, and Department of Cell Biology (J.S., R.N.K.), Joslin Diabetes Center and Harvard Medical School, Boston, Massachusetts 02115; Department of Cardiology (C.L.), Institute of Clinical Medicine, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450003, People's Republic of China; Institute of Cardiology (M.L.), Union Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan 430072, People's Republic of China; and NutriOmique team (M.A., S.A., K.C.), Institute of Cardiometabolism and Nutrition, INSERM, Unité Mixte de Recherche en Santé Unité 1166, and NutriOmique team (M.A., S.A., K.C.), Université Pierre et Marie Curie-Paris 6, Paris F-75013 France
| |
Collapse
|
40
|
Boal F, Hodgson LR, Reed SE, Yarwood SE, Just VJ, Stephens DJ, McCaffrey MW, Tavaré JM. Insulin promotes Rip11 accumulation at the plasma membrane by inhibiting a dynamin- and PI3-kinase-dependent, but Akt-independent, internalisation event. Cell Signal 2015; 28:74-82. [PMID: 26515129 PMCID: PMC4678287 DOI: 10.1016/j.cellsig.2015.10.014] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Accepted: 10/23/2015] [Indexed: 12/11/2022]
Abstract
Rip11 is a Rab11 effector protein that has been shown to be important in controlling the trafficking of several intracellular cargoes, including the fatty acid transporter FAT/CD36, V-ATPase and the glucose transporter GLUT4. We have previously demonstrated that Rip11 translocates to the plasma membrane in response to insulin and here we examine the basis of this regulated phenomenon in more detail. We show that Rip11 rapidly recycles between the cell interior and surface, and that the ability of insulin to increase the appearance of Rip11 at the cell surface involves an inhibition of Rip11 internalisation from the plasma membrane. By contrast the hormone has no effect on the rate of Rip11 translocation towards the plasma membrane. The ability of insulin to inhibit Rip11 internalisation requires dynamin and class I PI3-kinases, but is independent of the activation of the protein kinase Akt; characteristics which are very similar to the mechanism by which insulin inhibits GLUT4 endocytosis.
Collapse
Affiliation(s)
- Frédéric Boal
- School of Biochemistry, University of Bristol, Bristol, BS8 1TD, UK
| | - Lorna R Hodgson
- School of Biochemistry, University of Bristol, Bristol, BS8 1TD, UK
| | - Sam E Reed
- School of Biochemistry, University of Bristol, Bristol, BS8 1TD, UK
| | - Sophie E Yarwood
- School of Biochemistry, University of Bristol, Bristol, BS8 1TD, UK
| | - Victoria J Just
- School of Biochemistry, University of Bristol, Bristol, BS8 1TD, UK
| | - David J Stephens
- School of Biochemistry, University of Bristol, Bristol, BS8 1TD, UK
| | - Mary W McCaffrey
- Molecular Cell Biology Laboratory, School of Biochemistry and Cell Biology, Biosciences Institute, University College Cork, Cork, Ireland
| | - Jeremy M Tavaré
- School of Biochemistry, University of Bristol, Bristol, BS8 1TD, UK.
| |
Collapse
|
41
|
Roles of Akt and SGK1 in the Regulation of Renal Tubular Transport. BIOMED RESEARCH INTERNATIONAL 2015; 2015:971697. [PMID: 26491696 PMCID: PMC4600925 DOI: 10.1155/2015/971697] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/03/2015] [Accepted: 09/06/2015] [Indexed: 01/31/2023]
Abstract
A serine/threonine kinase Akt is a key mediator in various signaling pathways including regulation of renal tubular transport. In proximal tubules, Akt mediates insulin signaling via insulin receptor substrate 2 (IRS2) and stimulates sodium-bicarbonate cotransporter (NBCe1), resulting in increased sodium reabsorption. In insulin resistance, the IRS2 in kidney cortex is exceptionally preserved and may mediate the stimulatory effect of insulin on NBCe1 to cause hypertension in diabetes via sodium retention. Likewise, in distal convoluted tubules and cortical collecting ducts, insulin-induced Akt phosphorylation mediates several hormonal signals to enhance sodium-chloride cotransporter (NCC) and epithelial sodium channel (ENaC) activities, resulting in increased sodium reabsorption. Serum- and glucocorticoid-inducible kinase 1 (SGK1) mediates aldosterone signaling. Insulin can stimulate SGK1 to exert various effects on renal transporters. In renal cortical collecting ducts, SGK1 regulates the expression level of ENaC through inhibition of its degradation. In addition, SGK1 and Akt cooperatively regulate potassium secretion by renal outer medullary potassium channel (ROMK). Moreover, sodium-proton exchanger 3 (NHE3) in proximal tubules is possibly activated by SGK1. This review focuses on recent advances in understanding of the roles of Akt and SGK1 in the regulation of renal tubular transport.
Collapse
|
42
|
Snook LA, Nelson EM, Dyck DJ, Wright DC, Holloway GP. Glucose-dependent insulinotropic polypeptide directly induces glucose transport in rat skeletal muscle. Am J Physiol Regul Integr Comp Physiol 2015; 309:R295-303. [PMID: 26041107 DOI: 10.1152/ajpregu.00003.2015] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2015] [Accepted: 06/01/2015] [Indexed: 12/25/2022]
Abstract
Several gastrointestinal proteins have been identified to have insulinotropic effects, including glucose-dependent insulinotropic polypeptide (GIP); however, the direct effects of incretins on skeletal muscle glucose transport remain largely unknown. Therefore, the purpose of the current study was to examine the role of GIP on skeletal muscle glucose transport and insulin signaling in rats. Relative to a glucose challenge, a mixed glucose+lipid oral challenge increased circulating GIP concentrations, skeletal muscle Akt phosphorylation, and improved glucose clearance by ∼35% (P < 0.05). These responses occurred without alterations in serum insulin concentrations. In an incubated soleus muscle preparation, GIP directly stimulated glucose transport and increased GLUT4 accumulation on the plasma membrane in the absence of insulin. Moreover, the ability of GIP to stimulate glucose transport was mitigated by the addition of the PI 3-kinase (PI3K) inhibitor wortmannin, suggesting that signaling through PI3K is required for these responses. We also provide evidence that the combined stimulatory effects of GIP and insulin on soleus muscle glucose transport are additive. However, the specific GIP receptor antagonist (Pro(3))GIP did not attenuate GIP-stimulated glucose transport, suggesting that GIP is not signaling through its classical receptor. Together, the current data provide evidence that GIP regulates skeletal muscle glucose transport; however, the exact signaling mechanism(s) remain unknown.
Collapse
Affiliation(s)
- Laelie A Snook
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario, Canada
| | - Emery M Nelson
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario, Canada
| | - David J Dyck
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario, Canada
| | - David C Wright
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario, Canada
| | - Graham P Holloway
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario, Canada
| |
Collapse
|
43
|
Rugby-specific small-sided games training is an effective alternative to stationary cycling at reducing clinical risk factors associated with the development of type 2 diabetes: a randomized, controlled trial. PLoS One 2015; 10:e0127548. [PMID: 26030423 PMCID: PMC4452519 DOI: 10.1371/journal.pone.0127548] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2014] [Accepted: 04/10/2015] [Indexed: 12/16/2022] Open
Abstract
INTRODUCTION The present study investigated whether rugby small-sided games (SSG) could be an effective alternative to continuous stationary cycling (CYC) training at reducing clinical risk factors associated with the development of type 2 diabetes mellitus (T2DM). METHODS Thirty-three middle-aged (48.6±6.6y), inactive men were randomized into a CYC (n=11), SSG (n=11), or control (CON, n=11) group. Participants trained 3d·wk(-1) for 8 weeks, while control participants maintained normal activity and dietary patterns. Exercise duration was matched between groups, which involved CYC or SSG (four quarters, interspersed with 2-min passive recovery). Both training programs were designed to induce similar internal loads of maximal heart rate (~80-85%HRmax) and rating of perceived exertion. Pre- and post-intervention testing included dual-energy x-ray absorptiometry scan, graded exercise test, fasting 2 h oral glucose tolerance test and resting muscle biopsy. Western blotting was used to assess the content of skeletal muscle proteins associated with mitochondrial biogenesis and glucose regulation. RESULTS Both CYC and SSG increased VO2 at 80%HRmax, and reduced glycated haemoglobin, glucose area under the curve (AUC; SSG, -2.3±2.4; CYC -2.2±1.6 mmol·L(1)(120 min)(1); p<0.05), and total body fat-mass (SSG -2.6±0.9%; CYC -2.9±1.1%), compared to no change in CON (p<0.05). SSG reduced insulin AUC (-30.4±40.7 µlU·mL(1)(120 min)(1); p<0.05) and increased total body fat-free mass (1.1±1.2 kg; p<0.05), with no change in CYC or CON (P>0.05). There were no differences within or between conditions for protein content of peroxisome proliferator-activated receptor gamma coactivator-1α, sirtuin-1, p53, glucose transporter-4, protein kinase AKT/PKB, myocyte enhancer factor 2A, mitochondrial transcription factor, nuclear respiratory factor (NRF)-1, NRF-2 or mitochondrial complexes I-V (p>0.05). CONCLUSION Rugby small-sided games is an effective alternative to continuous cycling for improving metabolic risk-factors associated with the prevention of T2DM. Despite such positive adaptations in clinical risk factors, there were no changes in the content of skeletal muscle proteins associated with glucose regulation and mitochondrial biogenesis. TRIAL REGISTRATION Australian New Zealand Clinical Trial Registry ACTRN12613000874718.
Collapse
|
44
|
Glade MJ, Smith K. A glance at … exercise and glucose uptake. Nutrition 2015; 31:893-7. [PMID: 25933500 DOI: 10.1016/j.nut.2014.12.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Accepted: 12/03/2014] [Indexed: 10/24/2022]
Affiliation(s)
| | - Kyl Smith
- Progressive Laboratories Inc., Irving, Texas
| |
Collapse
|
45
|
Chen MZ, Hudson CA, Vincent EE, de Berker DAR, May MT, Hers I, Dayan CM, Andrews RC, Tavaré JM. Bariatric surgery in morbidly obese insulin resistant humans normalises insulin signalling but not insulin-stimulated glucose disposal. PLoS One 2015; 10:e0120084. [PMID: 25876175 PMCID: PMC4395354 DOI: 10.1371/journal.pone.0120084] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2014] [Accepted: 01/19/2015] [Indexed: 01/14/2023] Open
Abstract
Aims Weight-loss after bariatric surgery improves insulin sensitivity, but the underlying molecular mechanism is not clear. To ascertain the effect of bariatric surgery on insulin signalling, we examined glucose disposal and Akt activation in morbidly obese volunteers before and after Roux-en-Y gastric bypass surgery (RYGB), and compared this to lean volunteers. Materials and Methods The hyperinsulinaemic euglycaemic clamp, at five infusion rates, was used to determine glucose disposal rates (GDR) in eight morbidly obese (body mass index, BMI=47.3±2.2 kg/m2) patients, before and after RYGB, and in eight lean volunteers (BMI=20.7±0.7 kg/m2). Biopsies of brachioradialis muscle, taken at fasting and insulin concentrations that induced half-maximal (GDR50) and maximal (GDR100) GDR in each subject, were used to examine the phosphorylation of Akt-Thr308, Akt-473, and pras40, in vivo biomarkers for Akt activity. Results Pre-operatively, insulin-stimulated GDR was lower in the obese compared to the lean individuals (P<0.001). Weight-loss of 29.9±4 kg after surgery significantly improved GDR50 (P=0.004) but not GDR100 (P=0.3). These subjects still remained significantly more insulin resistant than the lean individuals (p<0.001). Weight loss increased insulin-stimulated skeletal muscle Akt-Thr308 and Akt-Ser473 phosphorylation, P=0.02 and P=0.03 respectively (MANCOVA), and Akt activity towards the substrate PRAS40 (P=0.003, MANCOVA), and in contrast to GDR, were fully normalised after the surgery (obese vs lean, P=0.6, P=0.35, P=0.46, respectively). Conclusions Our data show that although Akt activity substantially improved after surgery, it did not lead to a full restoration of insulin-stimulated glucose disposal. This suggests that a major defect downstream of, or parallel to, Akt signalling remains after significant weight-loss.
Collapse
Affiliation(s)
- Mimi Z. Chen
- School of Clinical Sciences, University of Bristol, Bristol, United Kingdom
| | - Claire A. Hudson
- School of Clinical Sciences, University of Bristol, Bristol, United Kingdom
| | - Emma E. Vincent
- School of Biochemistry, University of Bristol, Bristol, United Kingdom
| | | | - Margaret T. May
- School of Social and Community Medicine, University of Bristol, Bristol, United Kingdom
| | - Ingeborg Hers
- School of Physiology and Pharmacology, University of Bristol, Bristol, United Kingdom
| | - Colin M. Dayan
- Institute of Molecular and Experimental Medicine, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Robert C. Andrews
- School of Clinical Sciences, University of Bristol, Bristol, United Kingdom
- * E-mail:
| | - Jeremy M. Tavaré
- School of Biochemistry, University of Bristol, Bristol, United Kingdom
| |
Collapse
|
46
|
Development of a new model system to dissect isoform specific Akt signalling in adipocytes. Biochem J 2015; 468:425-34. [PMID: 25856301 PMCID: PMC4604748 DOI: 10.1042/bj20150191] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2015] [Accepted: 04/09/2015] [Indexed: 12/17/2022]
Abstract
Our study describes the development and validation of a new model system that allows for acute control of signalling by specific Akt isoforms. This model system revealed new insights into the role of Akt kinases in glucose transport and adipogenesis. Protein kinase B (Akt) kinases are critical signal transducers mediating insulin action. Genetic studies revealed that Akt1 and Akt2 signalling differentially contribute to sustain lipid and glucose homoeostasis; however Akt isoform-specific effectors remain elusive due to the lack of a suitable model system to mechanistically interrogate Akt isoform-specific signalling. To overcome those technical limitations we developed a novel model system that provides acute and specific control of signalling by Akt isoforms. We generated mutants of Akt1 and Akt2 resistant to the allosteric Akt inhibitor MK-2206. We then developed adipocyte cell lines, in which endogenous Akt1 or Akt2 has been replaced by their corresponding drug-resistant Akt mutant. Treatment of those cells with MK-2206 allowed for acute and specific control of either Akt1 or Akt2 function. Our data showed that Akt1W80A and Akt2W80A mutants are resistant to MK-2206, dynamically regulated by insulin and able to signal to Akt downstream effectors. Analyses of insulin action in this cellular system showed that Akt1 and Akt2 are both able to mediate insulin regulation of the transcription factor forkhead box O1 (FoxO1) and the glucose transporter 4 (GLUT4), revealing a redundant role for these Akt kinases in the control of glucose transport into fat cells. In contrast, Akt1 signalling is uniquely required for adipogenesis, by controlling the mitotic clonal expansion (MCE) of pre-adipocytes that precedes white adipose cell differentiation. Our data provide new insights into the role of Akt kinases in glucose transport and adipogenesis and support our model system as a valuable tool for the biochemical characterization of signalling by specific Akt isoforms.
Collapse
|
47
|
Manna P, Jain SK. Phosphatidylinositol-3,4,5-triphosphate and cellular signaling: implications for obesity and diabetes. Cell Physiol Biochem 2015; 35:1253-75. [PMID: 25721445 DOI: 10.1159/000373949] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/07/2015] [Indexed: 12/26/2022] Open
Abstract
Phosphatidylinositol-3,4,5-triphosphate (PtdIns(3,4,5)P₃) is one of the most important phosphoinositides and is capable of activating a wide range of proteins through its interaction with their specific binding domains. Localization and activation of these effector proteins regulate a number of cellular functions, including cell survival, proliferation, cytoskeletal rearrangement, intracellular vesicle trafficking, and cell metabolism. Phosphoinositides have been investigated as an important agonist-dependent second messenger in the regulation of diverse physiological events depending upon the phosphorylation status of their inositol group. Dysregulation in formation as well as metabolism of phosphoinositides is associated with various pathophysiological disorders such as inflammation, allergy, cardiovascular diseases, cancer, and metabolic diseases. Recent studies have demonstrated that the impaired metabolism of PtdIns(3,4,5)P₃ is a prime mediator of insulin resistance associated with various metabolic diseases including obesity and diabetes. This review examines the current status of the role of PtdIns(3,4,5)P₃ signaling in the regulation of various cellular functions and the implications of dysregulated PtdIns(3,4,5)P₃ signaling in obesity, diabetes, and their associated complications.
Collapse
Affiliation(s)
- Prasenjit Manna
- Department of Pediatrics, Louisiana State University Health Sciences Center, Shreveport, LA, USA
| | | |
Collapse
|
48
|
Tropomodulin3 is a novel Akt2 effector regulating insulin-stimulated GLUT4 exocytosis through cortical actin remodeling. Nat Commun 2015; 6:5951. [PMID: 25575350 PMCID: PMC4354152 DOI: 10.1038/ncomms6951] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Accepted: 11/25/2014] [Indexed: 12/19/2022] Open
Abstract
Akt2 and its downstream effectors mediate insulin-stimulated GLUT4-storage vesicle (GSV) translocation and fusion with the plasma membrane (PM). Using mass spectrometry, we identify actin-capping protein Tropomodulin 3 (Tmod3) as an Akt2-interacting partner in 3T3-L1 adipocytes. We demonstrate that Tmod3 is phosphorylated at Ser71 on insulin-stimulated Akt2 activation, and Ser71 phosphorylation is required for insulin-stimulated GLUT4 PM insertion and glucose uptake. Phosphorylated Tmod3 regulates insulin-induced actin remodelling, an essential step for GSV fusion with the PM. Furthermore, the interaction of Tmod3 with its cognate tropomyosin partner, Tm5NM1 is necessary for GSV exocytosis and glucose uptake. Together these results establish Tmod3 as a novel Akt2 effector that mediates insulin-induced cortical actin remodelling and subsequent GLUT4 membrane insertion. Our findings suggest that defects in cytoskeletal remodelling may contribute to impaired GLUT4 exocytosis and glucose uptake.
Collapse
|
49
|
Hassanpour Fard M, Naseh G, Lotfi N, Hosseini SM, Hosseini M. Effects of aqueous extract of turnip leaf (Brassica rapa) in alloxan-induced diabetic rats. AVICENNA JOURNAL OF PHYTOMEDICINE 2015; 5:148-56. [PMID: 25949956 PMCID: PMC4418064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/27/2014] [Revised: 11/05/2014] [Accepted: 11/11/2014] [Indexed: 11/01/2022]
Abstract
OBJECTIVES Turnip leaf has been used in folk medicine of Iran for the treatment of diabetes. However,so far no scientific study has been done to support its use in traditional medicine. The present study was carried out to evaluate the possible hypoglycemic efficacy of aqueous extract of turnip leaf (AETL) in diabetic rats. MATERIALS AND METHODS Alloxan-induced diabetic rats were orally treated with AETL at doses of 200 and 400 mg/kg body weight (bw) per day for 28 days. In order to evaluate the anti-diabetic activity, fasting blood glucose concentrations were determined on the 1(st), 14(th) and 29(th) days. Moreover,at the end of the study, plasma concentrations of total cholesterol, triglyceride (TG), high density lipoprotein cholesterol (HDL-c), low density lipoprotein cholesterol (LDL-c), aspartate amino transfarase (AST), and alanine amino transferase (ALT) were measured by the use of standard kits and auto-analyzer. RESULTS Both doses of AETL significantly decreased (p<0.001) blood glucose and ALT levels in diabetic rats after 28 days of administration. AETL at both doses decreased (p<0.05) plasma total cholesterol and LDL-c in diabetic rats, but they significantly decreased (p<0.05) HDL-c and increased triglycerideand AST levels in a-dose dependent manner. CONCLUSION The results showed that AETL has a dose- dependent decrease in the blood glucose in diabetic rats. However,we should not be unaware of adverse effects of AETL on lipid profiles and liver enzymes activity, especially decrease of HDL and increase of TG and AST.
Collapse
Affiliation(s)
- Mohammad Hassanpour Fard
- Department of Physiology and Pharmacology, Birjand University of Medical Sciences (BUMS), Birjand, Iran
| | | | | | | | - Mehran Hosseini
- Department of Public Health, Research Centre of Experimental Medicine, BUMS, Birjand, Iran,Corresponding Author: Tel: +989151606927, Fax:+985632433001,
| |
Collapse
|
50
|
Collino M, Benetti E, Rogazzo M, Chiazza F, Mastrocola R, Nigro D, Cutrin JC, Aragno M, Fantozzi R, Minetto MA, Thiemermann C. A non-erythropoietic peptide derivative of erythropoietin decreases susceptibility to diet-induced insulin resistance in mice. Br J Pharmacol 2014; 171:5802-15. [PMID: 25164531 PMCID: PMC4290718 DOI: 10.1111/bph.12888] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2014] [Revised: 08/01/2014] [Accepted: 08/11/2014] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND AND PURPOSE The haematopoietic activity of erythropoietin (EPO) is mediated by the classic EPO receptor (EpoR) homodimer, whereas tissue-protective effects are mediated by a heterocomplex between EpoR and the β-common receptor (βcR). Here, we investigated the effects of a novel, selective ligand of this heterocomplex - pyroglutamate helix B surface peptide (pHBSP) - in mice fed a diet enriched in sugars and saturated fats. EXPERIMENTAL APPROACH Male C57BL/6J mice were fed a high-fat high-sucrose diet (HFHS) for 22 weeks. pHBSP (30 μg·kg(-1) s.c.) was administered for the last 11 weeks. Biochemical assays, histopathological and immunohistochemical examinations and Western blotting were performed on serum and target organs (liver, kidney and skeletal muscle). KEY RESULTS Mice fed with HFHS diet exhibited insulin resistance, hyperlipidaemia, hepatic lipid accumulation and kidney dysfunction. In gastrocnemius muscle, HFHS impaired the insulin signalling pathway and reduced membrane translocation of glucose transporter type 4 and glycogen content. Treatment with pHBSP ameliorated renal function, reduced hepatic lipid deposition, and normalized serum glucose and lipid profiles. These effects were associated with an improvement in insulin sensitivity and glucose uptake in skeletal muscle. Diet-induced overproduction of the myokines IL-6 and fibroblast growth factor-21 were attenuated by pHBSP and, most importantly, pHBSP markedly enhanced mitochondrial biogenesis in skeletal muscle. CONCLUSIONS AND IMPLICATIONS Chronic treatment of mice with an EPO derivative, devoid of haematopoietic effects, improved metabolic abnormalities induced by a high-fat high-sucrose diet, by affecting several levels of the insulin signalling and inflammatory cascades within skeletal muscle, while enhancing mitochondrial biogenesis.
Collapse
Affiliation(s)
- M Collino
- Department of Drug Science and Technology, University of TurinTurin, Italy
| | - E Benetti
- Department of Drug Science and Technology, University of TurinTurin, Italy
| | - M Rogazzo
- Department of Drug Science and Technology, University of TurinTurin, Italy
| | - F Chiazza
- Department of Drug Science and Technology, University of TurinTurin, Italy
| | - R Mastrocola
- Department of Clinical and Biological Sciences, University of TurinTurin, Italy
| | - D Nigro
- Department of Clinical and Biological Sciences, University of TurinTurin, Italy
| | - J C Cutrin
- Department of Biotechnology and Sciences for the Health, University of TurinItaly
- Instituto de Investigaciones Cardiológicas, ININCA-CONICETBuenos Aires, Argentina
| | - M Aragno
- Department of Clinical and Biological Sciences, University of TurinTurin, Italy
| | - R Fantozzi
- Department of Drug Science and Technology, University of TurinTurin, Italy
| | - M A Minetto
- Division of Endocrinology, Diabetology and Metabolism, Department of Medical Sciences, University of TurinTurin, Italy
| | - C Thiemermann
- Centre for Translational Medicine and Therapeutics, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of LondonLondon, UK
| |
Collapse
|